1
|
Gao D, Bing C, Griffiths HR. Disrupted adipokine secretion and inflammatory responses in human adipocyte hypertrophy. Adipocyte 2025; 14:2485927. [PMID: 40176539 PMCID: PMC11980453 DOI: 10.1080/21623945.2025.2485927] [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: 07/02/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 04/04/2025] Open
Abstract
Adipocyte hypertrophy is a critical contributor to obesity-induced inflammation and insulin resistance. This study employed a human adipocyte hypertrophy model to investigate the adipokine release, inflammatory responses, and the intracellular singling pathways. Hypertrophic adipocytes exhibited increased lipid content and lipolysis, a decline of anti-inflammatory adipokine adiponectin release and an increase of pro-inflammatory adipokine leptin release compared to mature adipocytes. Moreover, TNFα and LPS exacerbated the decrease in adiponectin secretion by hypertrophic adipocytes while promoting the secretion of leptin, MCP-1 and IL-6, which is associated with impaired activation of p38 and JNK MAPK and persistent activation of ERK and IκBα in hypertrophic adipocytes. These altered adipokine secretions and inflammatory responses within hypertrophic adipocytes may contribute to adipocyte dysfunction in human obesity.
Collapse
Affiliation(s)
- Dan Gao
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Ministry of Education, Key Laboratory of Environment and Genes Related to Diseases Xi’an Jiaotong University, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| | - Chen Bing
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
2
|
Berbudi A, Khairani S, Tjahjadi AI. Interplay Between Insulin Resistance and Immune Dysregulation in Type 2 Diabetes Mellitus: Implications for Therapeutic Interventions. Immunotargets Ther 2025; 14:359-382. [PMID: 40196377 PMCID: PMC11974557 DOI: 10.2147/itt.s499605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a rapidly growing global health issue characterized by insulin resistance and chronic inflammation. Beyond regulating glucose homeostasis, insulin plays a pivotal role in modulating immune cell function, linking metabolic dysregulation with immune responses. This review examines the intricate relationship between insulin resistance and immune dysfunction in T2DM, focusing on how impaired insulin signaling pathways, particularly PI3K/Akt and MAPK, contribute to immune cell activation, proliferation, and chronic inflammation. Insulin resistance impacts immune cells such as T cells, B cells, macrophages, and neutrophils, leading to an imbalance between pro-inflammatory and anti-inflammatory responses. Elevated pro-inflammatory cytokines (eg, TNF-α, IL-6) and adipokines (eg, leptin, resistin) exacerbate insulin resistance, promoting a vicious cycle of metabolic and immune dysregulation. This interplay contributes to the chronic low-grade inflammation that underlies T2DM pathogenesis, further impairing insulin signaling and glucose metabolism. Restoration of insulin sensitivity is, therefore, a critical step toward correcting immune imbalance in insulin-resistant states like T2DM. Therapeutic approaches that reduce inflammation could also support improvements in insulin sensitivity, addressing both metabolic and immune disturbances simultaneously. The review also explores therapeutic strategies, including insulin therapy, targeting insulin signaling pathways, and lifestyle interventions. Insulin therapy can reduce pro-inflammatory cytokine production and enhance anti-inflammatory responses, although challenges such as potential immune suppression and hyperinsulinemia remain. Targeting key signaling pathways and transcription factors offers promising avenues for modulating immune responses, while lifestyle interventions, such as dietary modifications, physical activity, and weight management, can improve insulin sensitivity and reduce inflammation. By understanding the dual role of insulin in regulating both metabolic and immune functions, this review underscores the importance of addressing immune dysfunction as part of comprehensive T2DM management. Targeting the interconnected pathways of insulin signaling and immune regulation could lead to more effective therapeutic approaches, ultimately improving patient outcomes and reducing disease complications.
Collapse
Affiliation(s)
- Afiat Berbudi
- Department of Biomedical Sciences, Parasitology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Research Center for Care and Control of Infectious Diseases (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Universitas Padjadjaran Hospital, Sumedang, Indonesia
| | - Shafia Khairani
- Department of Biomedical Sciences, Cell Biology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Adi Imam Tjahjadi
- Research Center for Care and Control of Infectious Diseases (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Universitas Padjadjaran Hospital, Sumedang, Indonesia
- Department of Biomedical Sciences, Microbiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
3
|
Wu X, Chen H, Tian Y, Wang H, Hou H, Hu Q, Wang C. Amelioration of obesity-associated disorders using solanesol with the mitigation of NLRP3 inflammasome activation and macrophage inflammation in adipose tissue. Food Funct 2025; 16:1903-1918. [PMID: 39935386 DOI: 10.1039/d4fo05586a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Obesity and obesity-related metabolic diseases are causally linked to inflammatory activation. Proinflammatory macrophage infiltration and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome activation contribute to chronic inflammation and insulin resistance. Alleviating inflammatory responses is a reliable method to restore insulin sensitivity and reduce the severity of metabolic syndrome. Solanesol, rich in anti-inflammatory foods (potato, tomato, eggplant, chili peppers), has demonstrated anti-inflammatory properties, but whether it plays a beneficial role in obesity-induced chronic inflammation remains poorly understood. In this study, we investigated the effects of solanesol on the NLRP3 inflammasome and inflammatory responses both in vitro and in high-fat diet (HFD)-fed mice. We found that oral administration of solanesol reduced weight gain, insulin resistance, and inflammation in epididymal white adipose tissue (eWAT) in both HFD-fed obese mice and mice concurrently treated with a HFD. This effect was involved with reducing macrophage inflammation and inactivating the NLRP3 inflammasome by reducing the K+ efflux and reactive oxygen species (ROS) production in macrophages. Solanesol also reprogrammed the phenotype of inflammatory macrophages. Taken together, our study suggests that solanesol may be a promising candidate for treating obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Xiaqing Wu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, China.
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Yushan Tian
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Hongjuan Wang
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Qingyuan Hu
- China National Tobacco Quality Supervision & Test Center, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China.
- Beijing Life Science Academy, Beijing, China
| | - Congyi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, China.
| |
Collapse
|
4
|
Kim JS, Jun JH, Lee J, Park S, Kim E, Hwang SJ, Moon H, Baek SH, Kim HK, Park J, Cho Y, Han J, Kim C, Kim J, Yang HM, Lee C, Chung Y, Lee HJ, Jo DG. HDAC6 mediates NLRP3 inflammasome activation in the pathogenesis of diabetic retinopathy. Metabolism 2025; 164:156108. [PMID: 39689826 DOI: 10.1016/j.metabol.2024.156108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Diabetic retinopathy (DR), a major blindness cause in developed countries, is intricately linked to diabetes management and its duration. Here, we demonstrate that HDAC6 mediates NLRP3 inflammasome activation under diabetic conditions, leading to retinal inflammation and degeneration. METHODS This study demonstrated the therapeutic effects of HDAC6 genetic ablation, pharmacological inhibition, and HDAC6-deficient bone marrow transplantation in a diabetes model induced by streptozotocin and a high-fat diet. The therapeutic potential was evaluated from a metabolic perspective, including ocular pathologies such as retinal lesions, neovascularization, and vascular leakage. RESULTS We discovered that inhibition or genetic ablation of HDAC6 markedly alleviates DR symptoms by dampening NLRP3 inflammasome activation and mitigating retinal damage. Moreover, bone marrow transplantation from HDAC6-deficient mice into wild-type counterparts reversed DR symptoms, underscoring the significance of HDAC6 in systemic immune regulation. The study introduces a novel HDAC6 inhibitor, noted for superior bioavailability and blood-retinal barrier permeability, further highlights the therapeutic promise of targeting HDAC6 in DR. CONCLUSIONS Our findings not only underscore the crucial role of HDAC6 in the immune regulatory mechanisms underlying DR pathogenesis through NLRP3 inflammasome activation but also position HDAC6 inhibition as a promising strategy for addressing diabetic complications beyond DR.
Collapse
Affiliation(s)
- Jun-Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyun Jun
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Pharmacology, CKD Research Institute, Chong Kun Dang Pharmaceutical Co., Yongin 16995, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunyoung Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eunae Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Su Jung Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Heesu Moon
- School of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Hyun Baek
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hark Kyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jinsu Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoonsuk Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jihoon Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chanhee Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jongho Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyun-Mo Yang
- Department of Medical Chemistry, CKD Research Institute, Chong Kun Dang Pharmaceutical Co., Yongin 16995, Republic of Korea
| | - Changsik Lee
- Department of Medical Chemistry, CKD Research Institute, Chong Kun Dang Pharmaceutical Co., Yongin 16995, Republic of Korea
| | - Yeonseok Chung
- School of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea; Biomedical Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
5
|
Bahrami P, Aromolaran KA, Aromolaran AS. Proarrhythmic Lipid Inflammatory Mediators: Mechanisms in Obesity Arrhythmias. J Cell Physiol 2025; 240:e70012. [PMID: 39943721 PMCID: PMC11822244 DOI: 10.1002/jcp.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
The prevalence of obesity and associated metabolic disorders such as diabetes is rapidly increasing; therefore, concerns regarding their cardiovascular consequences, including cardiac arrhythmias, are rising. As obesity progresses, the excessively produced lipids accumulate in unconventional areas such as the epicardial adipose tissue (EAT) around the myocardium. Metabolic alterations in obesity contribute to the transformation of these ectopic fat deposits into arrhythmogenic substrates. However, despite advances in therapeutic approaches, particularly in lowering EAT volume and thickness through sodium-glucose co-transporter-2 (SGLT2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists, obese and diabetic patients still suffer from fatal arrhythmias that may lead to sudden cardiac death. Therefore, an investigation into how unappreciated underlying pathways such as lipid mediators contribute to the transformation of adipose tissues into proinflammatory and arrhythmogenic substrates is of significance. Leukotriene B4 (LTB4) is an eicosanoid derived from arachidonic acid and acts as a lipid mediator. LTB4 has recently been identified to be associated with cardiac ion channel modulations and arrhythmogenic conditions in diabetes. LTB4 increases circulatory free fatty acids (FFAs) and has been associated with adipocyte hypertrophy. LTB4 also interferes with insulin signaling pathways, instigating insulin resistance (IR). In addition, LTB4, as a potent chemoattractant, contributes to the mobilization of circulatory immune cells such as monocytes and promotes inflammatory macrophage polarization and macrophage dysfunction. Thus, this review provides a comprehensive overview of LTB4's underlying pathways in obesity; illustrates how these pathways might lead to alterations in cardiac ion channels, currents, and cardiac arrhythmias; and shows how they might pose a therapeutic target for metabolic-associated arrhythmias.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI)University of Utah School of MedicineSalt Lake CityUtahUSA
- Division of Cardiothoracic Surgery, Department of Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine ProgramUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| |
Collapse
|
6
|
Kaylan KB, Nargis T, Figatner K, Wang JE, Pratuangtham S, Chakraborty A, Casimiro I, Nadler JL, Boxer MB, Maloney DJ, Anderson RM, Mirmira RG, Tersey SA. 12-Lipoxygenase inhibition improves glucose homeostasis and obesity-associated inflammation in human gene replacement mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632274. [PMID: 39868153 PMCID: PMC11761697 DOI: 10.1101/2025.01.10.632274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Obesity-associated inflammation is characterized by macrophage infiltration into peripheral tissues, contributing to the progression of prediabetes and type 2 diabetes (T2D). The enzyme 12-lipoxygenase (12-LOX) catalyzes the formation of pro-inflammatory eicosanoids and is known to promote the migration of macrophages, yet its role in obesity-associated inflammation remains incompletely understood. Furthermore, differences between mouse and human orthologs of 12-LOX have limited efforts to study existing pharmacologic inhibitors of 12-LOX. In this study, we utilized a human gene replacement mouse model in which the gene encoding mouse 12-LOX (Alox15) is replaced by the human ALOX12 gene. As a model of obesity and dysglycemia, we administered these mice a high-fat diet. We subsequently investigated the effects of VLX-1005, a potent and selective small molecule inhibitor of human 12-LOX. Oral administration of VLX-1005 resulted in improved glucose homeostasis, decreased β cell dedifferentiation, and reduced macrophage infiltration in islets and adipose tissue. Analysis of the stromal vascular fraction from adipose tissue showed a reduction in myeloid cells and cytokine expression with VLX-1005 treatment, indicating decreased adipose tissue inflammation. In a distinct mouse model in which Alox15 was selectively deleted in myeloid cells, we observed decreased β cell dedifferentiation and reduced macrophage infiltration in both islets and adipose tissue, suggesting that the effects of VLX-1005 may relate to the inhibition of 12-LOX in macrophages. These findings highlight 12-LOX as a key factor in obesity-associated inflammation and suggest that 12-LOX inhibition could serve as a therapeutic strategy to improve glucose homeostasis and peripheral inflammation in the setting of obesity and T2D.
Collapse
Affiliation(s)
- Kerim B. Kaylan
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Titli Nargis
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Kayla Figatner
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jiayi E. Wang
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarida Pratuangtham
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Advaita Chakraborty
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Isabel Casimiro
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jerry L. Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | - Ryan M. Anderson
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Raghavendra G. Mirmira
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A. Tersey
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
7
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
8
|
Xue M, Liao Y, Jiang W. Insights into the molecular changes of adipocyte dedifferentiation and its future research opportunities. J Lipid Res 2024; 65:100644. [PMID: 39303983 PMCID: PMC11550672 DOI: 10.1016/j.jlr.2024.100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/23/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024] Open
Abstract
Recent studies have challenged the traditional belief that mature fat cells are irreversibly differentiated and revealed they can dedifferentiate into fibroblast-like cells known as dedifferentiated fat (DFAT) cells. Resembling pluripotent stem cells, DFAT cells hold great potential as a cell source for stem cell therapy. However, there is limited understanding of the specific changes that occur following adipocyte dedifferentiation and the detailed regulation of this process. This review explores the epigenetic, genetic, and phenotypic alterations associated with DFAT cell dedifferentiation, identifies potential targets for clinical regulation and discusses the current applications and challenges in the field of DFAT cell research.
Collapse
Affiliation(s)
- Mingheng Xue
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Wenqing Jiang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Abu Rached N, Dietrich JW, Ocker L, Quast DR, Stockfleth E, Bechara FG. Diabetes remission associated with optimized treatment of hidradenitis suppurativa. J Dtsch Dermatol Ges 2024; 22:1427-1429. [PMID: 39106221 DOI: 10.1111/ddg.15490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/16/2024] [Indexed: 08/09/2024]
Affiliation(s)
- Nessr Abu Rached
- International Centre for Hidradenitis suppurativa/Acne inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| | - Johannes W Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
- Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Hattingen, Germany
- Centre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Bochum, Germany
- Centre for Diabetes Technology, Catholic Hospitals Bochum, Bochum, Germany
| | - Lennart Ocker
- International Centre for Hidradenitis suppurativa/Acne inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| | - Daniel R Quast
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
- Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Hattingen, Germany
- Centre for Diabetes Technology, Catholic Hospitals Bochum, Bochum, Germany
| | - Eggert Stockfleth
- International Centre for Hidradenitis suppurativa/Acne inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| | - Falk G Bechara
- International Centre for Hidradenitis suppurativa/Acne inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
10
|
Yuan A, Liu J, Guo J, Chen F, Xu J, Chen H, Wang C, Le Y, Lu D. Calenduloside E Ameliorates Inflammatory Responses in Adipose Tissue via Sirtuin 2-NLRP3 Inflammasome Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20959-20973. [PMID: 39282743 DOI: 10.1021/acs.jafc.4c03917] [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: 09/26/2024]
Abstract
Obesity-related metabolic diseases are associated with a chronic inflammatory state. Calenduloside E (CE) is a triterpene saponin from sugar beet. In mouse models, CE reduced pro-inflammatory cytokines in white adipose tissue (WAT) and decreased macrophage infiltration of WAT. And CE inhibited pyroptosis in J774A.1 cells and WAT by inhibiting the activation of the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome. Moreover, CE could trigger the activation of Sirtuin 2 (SIRT2), leading to a decrease in the acetylation of NLRP3, particularly at the K24 site. In addition, it has been shown that CE can reduce inflammation in adipocytes that have been induced by macrophage-conditioned medium. However, the selective SIRT2 inhibitor AGK2 hindered the beneficial effects of CE. In summary, CE has the capacity to impede NLRP3-mediated pyroptosis by triggering SIRT2 activity, thus positioning CE as a promising therapeutic avenue for combating obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Aini Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jing Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fangming Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jingyi Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Medical Research Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, China
| | - Cui Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
11
|
Mahdavifard S, Shahi Z. Hepatoprotective Effect of Nicotinamide Versus Lead-Motivated Hepatotoxicity in Rats via Correcting Effect on Nuclear Factor-kβ Pathway and Glutathione Metabolism. Biol Trace Elem Res 2024; 202:4047-4053. [PMID: 38087034 DOI: 10.1007/s12011-023-03980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/27/2023] [Indexed: 07/18/2024]
Abstract
Lead (Pb) poisoning is one of the pivotal environmental issues and prompts liver dysfunction by elevating oxidative stress and inflammation. Nicotinamide (NA) deficiency enhances sensitivity to Pb toxicity. So, we investigated the effect of nicotinamide (NA) on the rat's liver histopathological and biochemical profiles in a rat model of Pb toxicity. Thirty-six rats were divided into four groups (nine rats at each): normal (N), lead toxicity (Pbt), and NA-treated N and Pbt groups. Treated groups took NA (180 mg/L in drinking water for one month). Pb intoxication was motivated in rats by acquiring 50 mg/L lead acetate in drinking water. Oxidative stress markers (advanced oxidation protein products and malondialdehyde), antioxidant markers (total glutathione, reduced glutathione to oxidized glutathione ratio, ferric ion reducing power, catalase, and paraoxonase-1), and inflammatory markers (hepatic nuclear factor-kβ expression, interleukin 1β level, and myeloperoxidase activity) in sera and liver homogenates were determined. In addition, the biochemical parameters of the liver function were measured. Finally, the liver of rats was evaluated by histopathological observation. NA corrected lead-persuaded biochemical and histopathological changes in the rat's liver. In addition, treatment decreased Pb, oxidative stress, and inflammatory markers in the sera and liver homogenates of N and Pbt groups. In addition, it elevated antioxidant markers (p < 0.001). NA prevented Pb-induced liver histopathological alternations and reduced liver dysfunction by reducing Pb, oxidative stress, and inflammation. Moreover, raising GSH/GSSG and diminishing the hepatic NF-kβ pathway are cardinal mechanisms of the treatment against Pb-motivated hepatotoxicity in rats.
Collapse
Affiliation(s)
- Sina Mahdavifard
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Zahra Shahi
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
12
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [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: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
13
|
Mahdavifard S, Nowruz N. Glutamine Defended the Kidneys Versus Lead Intoxication Via Elevating Endogenous Antioxidants, Reducing Inflammation and Carbonyl Stress, as well as Improving Insulin Resistance and Dyslipidemia. Biol Trace Elem Res 2024; 202:3141-3148. [PMID: 37776396 DOI: 10.1007/s12011-023-03887-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Kidneys are primarily sensitive to lead (Pb) poisoning due to their cardinal role in lead excretion. Then, we studied the effect of glutamine (Gln) on lead nephrotoxicity in rats by assessing the histopathological and biochemical parameters (the renal NF-kβ expression, metabolic profile, oxidative stress, inflammatory markers, methylglyoxal (MGO), and glyoxalase-I activity). Forty rats were allotted into four groups (ten rats in each): normal (N), Gln-treated N, Pb intoxication (Pbi), and Gln-treated Pbi. The treated groups took 0.1% Gln in drinking water for 1 month. To motivate lead poisoning, rats gained 50 mg/l lead acetate in drinking water for 1 month. Oxidative stress indices (total glutathione, its reduced and oxidized forms, their ratios, advanced protein oxidation products, malondialdehyde, and ferric ion reducing power) and inflammatory markers (renal nuclear factor-kβ expression, interleukin 1β level, and myeloperoxidase activity) were measured. Furthermore, metabolic profile (fasting blood sugar, insulin, insulin resistance, lipid profile, and atherogenic index) and renal dysfunction parameters were determined. Pb-induced renal histopathological alterations were investigated by a pathologist. In the kidney of Pbi rats, the glomerulus was damaged. Gln prevented kidney damage and reduced kidney dysfunction parameters. In addition, Gln decreased oxidative stress and inflammation in sera and kidney homogenates. In addition, it improved insulin resistance, dyslipidemia, and carbonyl stress (p < 0.001). Gln guarded the kidneys versus lead intoxication by improving insulin resistance and dyslipidemia, elevating antioxidant markers, and diminishing inflammation and carbonyl stress.
Collapse
Affiliation(s)
- Sina Mahdavifard
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Najafzadeh Nowruz
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
14
|
Olejnik AE, Kuźnar-Kamińska B. Association of Obesity and Severe Asthma in Adults. J Clin Med 2024; 13:3474. [PMID: 38930006 PMCID: PMC11204497 DOI: 10.3390/jcm13123474] [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: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The incidence of obesity and asthma continues to enhance, significantly impacting global public health. Adipose tissue is an organ that secretes hormones and cytokines, causes meta-inflammation, and contributes to the intensification of bronchial hyperreactivity, oxidative stress, and consequently affects the different phenotypes of asthma in obese people. As body weight increases, the risk of severe asthma increases, as well as more frequent exacerbations requiring the use of glucocorticoids and hospitalization, which consequently leads to a deterioration of the quality of life. This review discusses the relationship between obesity and severe asthma, the underlying molecular mechanisms, changes in respiratory function tests in obese people, its impact on the occurrence of comorbidities, and consequently, a different response to conventional asthma treatment. The article also reviews research on possible future therapies for severe asthma. The manuscript is a narrative review of clinical trials in severe asthma and comorbid obesity. The articles were found in the PubMed database using the keywords asthma and obesity. Studies on severe asthma were then selected for inclusion in the article. The sections: 'The classification connected with asthma and obesity', 'Obesity-related changes in pulmonary functional tests', and 'Obesity and inflammation', include studies on subjects without asthma or non-severe asthma, which, according to the authors, familiarize the reader with the pathophysiology of obesity-related asthma.
Collapse
Affiliation(s)
- Aneta Elżbieta Olejnik
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland;
| | | |
Collapse
|
15
|
Varra FN, Varras M, Varra VK, Theodosis-Nobelos P. Molecular and pathophysiological relationship between obesity and chronic inflammation in the manifestation of metabolic dysfunctions and their inflammation‑mediating treatment options (Review). Mol Med Rep 2024; 29:95. [PMID: 38606791 PMCID: PMC11025031 DOI: 10.3892/mmr.2024.13219] [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: 10/25/2023] [Accepted: 01/17/2024] [Indexed: 04/13/2024] Open
Abstract
Obesity reaches up to epidemic proportions globally and increases the risk for a wide spectrum of co‑morbidities, including type‑2 diabetes mellitus (T2DM), hypertension, dyslipidemia, cardiovascular diseases, non‑alcoholic fatty liver disease, kidney diseases, respiratory disorders, sleep apnea, musculoskeletal disorders and osteoarthritis, subfertility, psychosocial problems and certain types of cancers. The underlying inflammatory mechanisms interconnecting obesity with metabolic dysfunction are not completely understood. Increased adiposity promotes pro‑inflammatory polarization of macrophages toward the M1 phenotype, in adipose tissue (AT), with subsequent increased production of pro‑inflammatory cytokines and adipokines, inducing therefore an overall, systemic, low‑grade inflammation, which contributes to metabolic syndrome (MetS), insulin resistance (IR) and T2DM. Targeting inflammatory mediators could be alternative therapies to treat obesity, but their safety and efficacy remains to be studied further and confirmed in future clinical trials. The present review highlights the molecular and pathophysiological mechanisms by which the chronic low‑grade inflammation in AT and the production of reactive oxygen species lead to MetS, IR and T2DM. In addition, focus is given on the role of anti‑inflammatory agents, in the resolution of chronic inflammation, through the blockade of chemotactic factors, such as monocytes chemotractant protein‑1, and/or the blockade of pro‑inflammatory mediators, such as IL‑1β, TNF‑α, visfatin, and plasminogen activator inhibitor‑1, and/or the increased synthesis of adipokines, such as adiponectin and apelin, in obesity‑associated metabolic dysfunction.
Collapse
Affiliation(s)
- Fani-Niki Varra
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia 1036, Cyprus
- Medical School, Dimocritus University of Thrace, Alexandroupolis 68100, Greece
| | - Michail Varras
- Fourth Department of Obstetrics and Gynecology, ‘Elena Venizelou’ General Hospital, Athens 11521, Greece
| | | | | |
Collapse
|
16
|
Ding Y, Cao Q, Yang W, Xu J, Xiao P. Macrophage: Hidden Criminal in Therapy Resistance. J Innate Immun 2024; 16:188-202. [PMID: 38442696 PMCID: PMC10990480 DOI: 10.1159/000538212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/29/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Although substantial efforts have been made by researchers to develop drugs, a disappointing reality is that the emergence of drug resistance is an unavoidable reality for the majority of patients. In recent years, emerging evidence suggests a connection between drug resistance and immune dysregulation. SUMMARY As a ubiquitously distributed, versatile innate immune cell, macrophages play essential roles in maintaining tissue homeostasis in a steady state. Nevertheless, it is becoming aware that macrophages undermine the action of therapeutic drugs across various disease types. Reprogramming macrophage function has been proven to be effective in restoring patient responsiveness to treatment. Herein, we comprehensively reviewed how macrophages respond to drugs and the mechanisms by which they contribute to treatment unresponsiveness in cancer, inflammatory diseases, and metabolic diseases. In addition, future prospects in macrophage-based combination therapy were discussed. KEY MESSAGES Targeting macrophages is a promising strategy for overcoming drug resistance in immune disorders.
Collapse
Affiliation(s)
- Yimin Ding
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjuan Yang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Xu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
17
|
Chamanara S, Hozouri V, Irandoost E. Inhibition of NLRP3 inflammasome-A potential mechanistic therapeutic for treatment of polycystic ovary syndrome? J Biochem Mol Toxicol 2024; 38:e23592. [PMID: 38054794 DOI: 10.1002/jbt.23592] [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: 05/02/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
This review article explores the relationship between the NOD-like receptor protein 3 (NLRP3) inflammasome and the risk of developing polycystic ovary syndrome (PCOS). The NLRP3 inflammasome, a fundamental element of the innate immune system, plays a crucial role in the production of proinflammatory mediators and pyroptosis, a type inflammatory cell death. We conducted a thorough search on scientific databases to gather relevant information on this topic, utilizing relevant keywords. The reviewed studies indicated a correlation between PCOS and a higher incidence of granulosa cell (GC) death and the presence of ovarian tissue fibrosis. NLRP3 inflammasome stimulation and subsequent pyroptosis in GCs play a significant role in the pathophysiology of PCOS. Active NLRP3 inflammasome is involved in the production of inflammatory mediators like interleukin-1β (IL-1β) and IL-18, contributing to the development of PCOS, particularly in overweight patients. Therefore, inhibiting NLRP3 activation and pyroptosis could potentially offer novel therapeutic strategies for PCOS. Some limited studies have explored the use of agents with antioxidant and anti-inflammatory properties, as well as gene therapy approaches, to target the NLRP3 and pyroptosis signaling pathways. This study overview the understanding of the relationship between NLRP3 inflammasome activation, pyroptosis, and PCOS. It highlights the potential of targeting the NLRP3 inflammasome as an approach for treating PCOS. Nonetheless, further research and clinical trials are imperative to validate these results and explore the effectiveness of NLRP3 inflammasome inhibition in the management of PCOS.
Collapse
Affiliation(s)
- Solmaz Chamanara
- Department of Gynecology and Obstetrics, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Vahid Hozouri
- Internal Medicine Department, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Elnaz Irandoost
- Department of Gynecology and Obstetrics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Bailin SS, Koethe JR, Rebeiro PF. The pathogenesis of obesity in people living with HIV. Curr Opin HIV AIDS 2024; 19:6-13. [PMID: 37934696 PMCID: PMC10842175 DOI: 10.1097/coh.0000000000000834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
PURPOSE OF REVIEW The public health challenge of overweight and obesity increasingly affects people living with HIV (PWH). These effects have also accelerated as the prevalence of antiretroviral therapy (ART) use has increased among PWH. It is therefore also critical that we examine and understand the pathogenesis of obesity among PWH.This review will aim to summarize relevant and recent literature related to the risks of weight gain and obesity associated with HIV disease progression, cardiometabolic disease, and multimorbidity among PWH. Further, we will discuss adipose tissue changes associated with weight gain and obesity and how these changes relate to metabolic complications. RECENT FINDINGS Several observational and experimental studies in recent years have evaluated the role of contemporary ART regimens, particularly integrase strand transfer inhibitors (INSTIs) and tenofovir alafenamide (TAF), as contributors to weight gain, obesity, and cardiometabolic disease, though the mechanisms remain unclear. Metabolic dysregulation has also been linked to ectopic fat deposition and alterations in innate and adaptive immune cell populations in adipose tissue that accompany HIV and obesity. These factors continue to contribute to an increasing burden of metabolic diseases in an aging HIV population. SUMMARY Obesity accompanies an increasing burden of metabolic disease among PWH, and understanding the role of fat partitioning and HIV and ART-related adipose tissue dysfunction may guide prevention and treatment strategies.
Collapse
Affiliation(s)
| | - John R Koethe
- Department of Medicine, Division of Infectious Diseases
| | - Peter F Rebeiro
- Department of Medicine, Division of Infectious Diseases
- Department of Medicine, Division of Epidemiology
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Engin AB. Message Transmission Between Adipocyte and Macrophage in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:273-295. [PMID: 39287855 DOI: 10.1007/978-3-031-63657-8_9] [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
Obesity is characterized by the chronic low-grade activation of the innate immune system. In this respect, macrophage-elicited metabolic inflammation and adipocyte-macrophage interaction have primary importance in obesity. Large quantity of macrophages is accumulated by different mechanisms in obese adipose tissue. Hypertrophic adipocyte-derived chemotactic monocyte chemoattractant protein-1 (MCP-1)/C-C chemokine receptor 2 (CCR2) pathway promotes more macrophage accumulation into the obese adipose tissue. However, obesity-induced changes in adipose tissue macrophage density are mainly dependent on increases in the triple-positive cluster of differentiation (CD)11b+ F4/80+ CD11c+ adipose tissue macrophage subpopulation. As epigenetic regulators, microRNAs (miRNAs) are one of the most important mediators of obesity. miRNAs are expressed by adipocytes as well as macrophages and regulate inflammation with the expression of target genes. A paracrine loop involving free fatty acids and tumor necrosis factor-alpha (TNF-α) between adipocytes and macrophages establishes a vicious cycle that aggravates inflammatory changes in the adipose tissue. Adipocyte-specific caspase-1 and production of interleukin-1beta (IL-1β) by macrophages; both adipocyte and macrophage induction by toll-like receptor-4 (TLR4) through nuclear factor-kappaB (NF-κB) activation; free fatty acid-induced and TLR-mediated activation of c-Jun N-terminal kinase (JNK)-related pro-inflammatory pathways in CD11c+ immune cells; are effective in mutual message transmission between adipocyte and macrophage and in the development of adipose tissue inflammation. Thus, the metabolic status of adipocytes and their released exosomes are important determinants of macrophage inflammatory output. However, old adipocytes are removed by macrophages through trogocytosis or sending an "eat me" signal. As a single miRNA can be able to regulate a variety of target genes and signaling pathways, reciprocal transfer of miRNAs between adipocytes and macrophages via miRNA-loaded exosomes reorganizes the different stages of obesity. Changes in the expression of circulating miRNAs because of obesity progression or anti-obesity treatment indicate that miRNAs could be used as potential biomarkers. Therefore, it is believed that targeting macrophage-associated miRNAs with anti-obesity miRNA-loaded nano-carriers may be successful in the attenuation of both obesity and adipose tissue inflammation in clinical practice. Moreover, miRNA-containing exosomes and transferable mitochondria between the adipocyte and macrophage are investigated as new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
20
|
Caslin HL, Bolus WR, Thomas C, Toki S, Norlander AE, Peebles RS, Hasty AH. Bovine Serum Albumin Elicits IL-33-Dependent Adipose Tissue Eosinophilia: Potential Relevance to Ovalbumin-induced Models of Allergic Disease. Immunohorizons 2023; 7:842-852. [PMID: 38095595 PMCID: PMC10759155 DOI: 10.4049/immunohorizons.2300061] [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: 08/23/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
All cells of the immune system reside in adipose tissue (AT), and increasing type 2 immune cells may be a therapeutic strategy to improve metabolic health. In our previous study using i.p. IL-5 injections to increase eosinophils, we observed that a standard vehicle control of 0.1% BSA also elicited profound AT eosinophilia. In this study, we aimed to determine whether BSA-induced AT eosinophilia results in metabolic benefits in murine models of diet-induced obesity. I.p. 0.1% BSA injections increased AT eosinophils after 4 wk. Despite elevating eosinophils to >50% of immune cells in the AT, body weight and glucose tolerance were not different between groups. Interestingly, BSA elicited epithelial IL-33 production, as well as gene expression for type 2 cytokines and IgE production that were dependent on IL-33. Moreover, multiple models of OVA sensitization also drove AT eosinophilia. Following transplantation of a donor fat pad with BSA-induced eosinophilia, OVA-sensitized recipient mice had higher numbers of bronchoalveolar lavage eosinophils that were recipient derived. Interestingly, lungs of recipient mice contained eosinophils, macrophages, and CD8 T cells from the donor AT. These trafficked similarly from BSA- and non-BSA-treated AT, suggesting even otherwise healthy AT serves as a reservoir of immune cells capable of migrating to the lungs. In conclusion, our studies suggest that i.p. injections of BSA and OVA induce an allergic response in the AT that elicits eosinophil recruitment, which may be an important consideration for those using OVA in animal models of allergic disease.
Collapse
Affiliation(s)
- Heather L. Caslin
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Department of Health and Human Performance, University of Houston, Houston, TX
| | - W. Reid Bolus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA
| | - Christopher Thomas
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Shinji Toki
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Allison E. Norlander
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| |
Collapse
|
21
|
Stenberg K, Novotny GW, Lutz TA, Mandrup-Poulsen T, Bjørnvad CR. Obesity-induced changes in gene expression in feline adipose and skeletal muscle tissue. J Anim Physiol Anim Nutr (Berl) 2023; 107:1262-1278. [PMID: 36591865 DOI: 10.1111/jpn.13802] [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: 03/21/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023]
Abstract
Indoor-confined cats are prone to developing obesity due to a sedentary life and an energy intake exceeding energy requirements. As in humans, feline obesity decreases insulin sensitivity and increases the risk of developing feline diabetes mellitus, but the pathophysiological mechanisms are currently poorly understood. Human obesity-related metabolic alterations seem to relate to changes in the expression of genes involved in glucose metabolism, insulin action and inflammation. The objective of the current study was to investigate changes in the expression of genes relating to obesity, glucose metabolism and inflammation in cats with non-experimentally induced obesity. Biopsies from the sartorius muscle and subcutaneous adipose tissue were obtained from 73 healthy, neutered, indoor-confined domestic shorthaired cats ranging from lean to obese. Quantification of obesity-related gene expression levels relative to glyceraldehyde-3-phosphate dehydrogenase was performed by quantitative real-time polymerase chain reaction. A negative association between obesity and adiponectin expression was observed in the adipose tissue (mean ± SD; normal weight, 27.30 × 10-3 ± 77.14 × 10-3 ; overweight, 2.89 × 10-3 ± 0.38 × 10-3 and obese, 2.93 × 10-3 ± 4.20 × 10-3 , p < 0.05). In muscle, the expression of peroxisome proliferative activated receptor-γ2 and plasminogen activator inhibitor-1 was increased in the obese compared to the normal-weight cats, and resistin was increased in the normal-weight compared to the overweight cats. There were no detectable obesity-related changes in the messenger RNA levels of inflammatory cytokines. In conclusion, a possible obesity-related low-grade inflammation caused by increased expression of key proinflammatory regulators was not observed. This could imply that the development of feline obesity and ensuing insulin resistance may not be based on tissue-derived inflammation, but caused by several determining factors, many of which still need further investigation.
Collapse
Affiliation(s)
- Kathrine Stenberg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Guy W Novotny
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Departments of Hematology/Pathology, Herlev Hospital, Herlev, Denmark
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse-Faculty University of Zurich, Zurich, Switzerland
| | | | - Charlotte Reinhard Bjørnvad
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
22
|
Lin YC, Hou YC, Wang HC, Shan YS. New insights into the role of adipocytes in pancreatic cancer progression: paving the way towards novel therapeutic targets. Theranostics 2023; 13:3925-3942. [PMID: 37554282 PMCID: PMC10405844 DOI: 10.7150/thno.82911] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/21/2023] [Indexed: 08/10/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies across the world, which is due to delayed diagnosis and resistance to current therapies. The interactions between pancreatic tumor cells and their tumor microenvironment (TME) allow cancer cells to escape from anti-cancer therapies, leading to difficulties in treating PC. With endocrine function and lipid storage capacity, adipose tissue can maintain energy homeostasis. Direct or indirect interaction between adipocytes and PC cells leads to adipocyte dysfunction characterized by morphological change, fat loss, abnormal adipokine secretion, and fibroblast-like transformation. Various adipokines released from dysfunctional adipocytes have been reported to promote proliferation, invasion, metastasis, stemness, and chemoresistance of PC cells via different mechanisms. Additional lipid outflow from adipocytes can be taken into the TME and thus alter the metabolism in PC cells and surrounding stromal cells. Besides, the trans-differentiation potential enables adipocytes to turn into various cell types, which may give rise to an inflammatory response as well as extracellular matrix reorganization to modulate tumor burden. Understanding the molecular basis behind the protumor functions of adipocytes in PC may offer new therapeutic targets.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Hao-Chen Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Medical Imaging Center, Innovation Headquarter, National Cheng Kung University; Tainan 704, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| |
Collapse
|
23
|
Zhong J, Zhao G, Edwards S, Tran J, Rajagopalan S, Rao X. Particulate air pollution exaggerates diet-induced insulin resistance through NLRP3 inflammasome in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 328:121603. [PMID: 37062408 PMCID: PMC10164710 DOI: 10.1016/j.envpol.2023.121603] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023]
Abstract
Air particulate matter 2.5 (PM2.5) has been demonstrated to exaggerate insulin resistance in both human and animal studies. However, the exact molecular mechanisms remain elusive. This study sought to assess the role of NLRP3 inflammasome in PM2.5 exposure-induced insulin resistance and explore the underlying mechanisms. Wild-type (WT), Nlrp3-/-, Tlr4Lps-d, or Nrf2-/- mice, on a normal diet or high-fat diet (HFD), were exposed to PM2.5 or filtered air (FA) in a whole-body exposure facility. Priming (first signal) and assembly (second signal) of NLRP3 inflammasome activation were assessed by measuring the transcription of Nlrp3/Il-1β and detecting the activity of caspase-1 and secretion of IL-1β. We found PM2.5 exposure exaggerated insulin resistance and increased IL-1β production in the HFD-fed WT mice, but not Nlrp3-/- mice. Gene expressions of Nlrp3 and Il-1β in the lungs and peritoneal macrophages were upregulated in WT mice exposed to PM2.5. When stimulated with LPS (first signal) or monosodium urate (second signal), PM2.5 exposure was able to enhance the activity of caspase-1 and IL-1β secretion, suggesting that PM2.5 may serve as a stimulus of either the first or second signal for NLRP3 inflammasome activation. Effects of PM2.5 on caspase-1 activation and IL-1β secretion were partially blocked in Tlr4Lps-d mice. Reactive oxygen species (ROS), co-localization of NLRP3 and mitochondria, and secondary lysosomes in macrophages were increased after PM2.5 exposure, while deficiency of antioxidant gene Nrf2 in mice significantly enhanced PM2.5-induced secretion of IL-1β. Imaging flow cytometry and transmission electron microscopy demonstrated an engulfment of PM2.5 particles by macrophages, while suppression of phagocytosis by cytochalasin D abolished PM2.5-induced transcription of Nlrp3/Il-1β. Our results demonstrated a critical role of NLRP3 inflammasome in PM2.5 exaggerated insulin resistance, and multiple pathways in the first and second signals of NLRP3 inflammasome activation may be involved.
Collapse
Affiliation(s)
- Jixin Zhong
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China; Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, United States
| | - Gang Zhao
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97239, United States; Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Sabrina Edwards
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97239, United States
| | - Joanne Tran
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97239, United States; Pacific Northwest University of Health Science, Yakima, WA, 98901, United States
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, United States
| | - Xiaoquan Rao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China; Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, 44106, United States; Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, 97239, United States.
| |
Collapse
|
24
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
25
|
Hosseini E, Kohan-Ghadr HR, Bazrafkan M, Amorim CA, Askari M, Zakeri A, Mousavi SN, Kafaeinezhad R, Afradiasbagharani P, Esfandyari S, Nazari M. Rescuing fertility during COVID-19 infection: exploring potential pharmacological and natural therapeutic approaches for comorbidity, by focusing on NLRP3 inflammasome mechanism. J Assist Reprod Genet 2023; 40:1173-1185. [PMID: 36892705 PMCID: PMC9995769 DOI: 10.1007/s10815-023-02768-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/28/2023] [Indexed: 03/10/2023] Open
Abstract
The respiratory system was primarily considered the only organ affected by Coronavirus disease 2019 (COVID-19). As the pandemic continues, there is an increasing concern from the scientific community about the future effects of the virus on male and female reproductive organs, infertility, and, most significantly, its impact on the future generation. The general presumption is that if the primary clinical symptoms of COVID-19 are not controlled, we will face several challenges, including compromised infertility, infection-exposed cryopreserved germ cells or embryos, and health complications in future generations, likely connected to the COVID-19 infections of parents and ancestors. In this review article, we dedicatedly studied severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) virology, its receptors, and the effect of the virus to induce the activation of inflammasome as the main arm of the innate immune response. Among inflammasomes, nucleotide oligomerization domain-like receptor protein, pyrin domain containing 3 (NLRP3) inflammasome pathway activation is partly responsible for the inflicted damages in both COVID-19 infection and some reproductive disorders, so the main focus of the discussion is on NLRP3 inflammasome in the pathogenesis of COVID-19 infection alongside in the reproductive biology. In addition, the potential effects of the virus on male and female gonad functions were discussed, and we further explored the potential natural and pharmacological therapeutic approaches for comorbidity via NLRP3 inflammasome neutralization to develop a hypothesis for averting the long-term repercussions of COVID-19. Since activation of the NLRP3 inflammasome pathway contributes to the damage caused by COVID-19 infection and some reproductive disorders, NLRP3 inflammasome inhibitors have a great potential to be considered candidates for alleviating the pathological effects of the COVID-19 infection on the germ cells and reproductive tissues. This would impede the subsequent massive wave of infertility that may threaten the patients.
Collapse
Affiliation(s)
- Elham Hosseini
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Obstetrics and Gynecology, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI USA
| | - Mahshid Bazrafkan
- Reproductive Biotechnology Research Center, Avicenna Research Institute (ARI), ACECR, Tehran, Iran
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maryam Askari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Armin Zakeri
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyedeh Neda Mousavi
- Department of Nutrition, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Raheleh Kafaeinezhad
- Department of Biology, Faculty of Basic Sciences, University of Maragheh, Maragheh, Iran
| | | | - Sahar Esfandyari
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Mahboobeh Nazari
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
26
|
Li X, Ren Y, Chang K, Wu W, Griffiths HR, Lu S, Gao D. Adipose tissue macrophages as potential targets for obesity and metabolic diseases. Front Immunol 2023; 14:1153915. [PMID: 37153549 PMCID: PMC10154623 DOI: 10.3389/fimmu.2023.1153915] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Macrophage infiltration into adipose tissue is a key pathological factor inducing adipose tissue dysfunction and contributing to obesity-induced inflammation and metabolic disorders. In this review, we aim to present the most recent research on macrophage heterogeneity in adipose tissue, with a focus on the molecular targets applied to macrophages as potential therapeutics for metabolic diseases. We begin by discussing the recruitment of macrophages and their roles in adipose tissue. While resident adipose tissue macrophages display an anti-inflammatory phenotype and promote the development of metabolically favorable beige adipose tissue, an increase in pro-inflammatory macrophages in adipose tissue has negative effects on adipose tissue function, including inhibition of adipogenesis, promotion of inflammation, insulin resistance, and fibrosis. Then, we presented the identities of the newly discovered adipose tissue macrophage subtypes (e.g. metabolically activated macrophages, CD9+ macrophages, lipid-associated macrophages, DARC+ macrophages, and MFehi macrophages), the majority of which are located in crown-like structures within adipose tissue during obesity. Finally, we discussed macrophage-targeting strategies to ameliorate obesity-related inflammation and metabolic abnormalities, with a focus on transcriptional factors such as PPARγ, KLF4, NFATc3, and HoxA5, which promote macrophage anti-inflammatory M2 polarization, as well as TLR4/NF-κB-mediated inflammatory pathways that activate pro-inflammatory M1 macrophages. In addition, a number of intracellular metabolic pathways closely associated with glucose metabolism, oxidative stress, nutrient sensing, and circadian clock regulation were examined. Understanding the complexities of macrophage plasticity and functionality may open up new avenues for the development of macrophage-based treatments for obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Xirong Li
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yakun Ren
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Kewei Chang
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| | - Wenlong Wu
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Shemin Lu
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Dan Gao
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| |
Collapse
|
27
|
Liu DF, Zhang XY, Zhou RF, Cai L, Yan DM, Lan LJ, He SH, Tang H. Glucose metabolism continuous deteriorating in male patients with human immunodeficiency virus accepted antiretroviral therapy for 156 weeks. World J Diabetes 2023; 14:299-312. [PMID: 37035225 PMCID: PMC10075030 DOI: 10.4239/wjd.v14.i3.299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND The dynamic characteristics of glucose metabolism and its risk factors in patients living with human immunodeficiency virus (PLWH) who accepted primary treatment with the efavirenz (EFV) plus lamivudine (3TC) plus tenofovir (TDF) (EFV + 3TC + TDF) regimen are unclear and warrant investigation.
AIM To study the long-term dynamic characteristics of glucose metabolism and its contributing factors in male PLWH who accepted primary treatment with the EFV + 3TC + TDF regimen for 156 wk.
METHODS This study was designed using a follow-up design. Sixty-one male treatment-naive PLWH, including 50 cases with normal glucose tolerance and 11 cases with prediabetes, were treated with the EFV + 3TC + TDF regimen for 156 wk. The glucose metabolism dynamic characteristics, the main risk factors and the differences among the three CD4+ count groups were analyzed.
RESULTS In treatment-naive male PLWH, regardless of whether glucose metabolism disorder was present at baseline, who accepted treatment with the EFV + 3TC + TDF regimen for 156 wk, a continuous increase in the fasting plasma glucose (FPG) level, the rate of impaired fasting glucose (IFG) and the glycosylated hemoglobin (HbA1c) level were found. These changes were not due to insulin resistance but rather to significantly reduced islet β cell function, according to the homeostasis model assessment of β cell function (HOMA-β). Moreover, the lower the baseline CD4+ T-cell count was, the higher the FPG level and the lower the HOMA-β value. Furthermore, the main risk factors for the FPG levels were the CD3+CD8+ cell count and viral load (VL), and the factors contributing to the HOMA-β values were the alanine aminotransferase level, VL and CD3+CD8+ cell count.
CONCLUSION These findings provide guidance to clinicians who are monitoring FPG levels closely and are concerned about IFG and decreased islet β cell function during antiretroviral therapy with the EFV + 3TC + TDF regimen for long-term application.
Collapse
Affiliation(s)
- Da-Feng Liu
- Department of Internal Medicine, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Xin-Yi Zhang
- Department of Endocrinology and Metabolism, Sichuan University West China Hoapital, Chengdu 610041, Sichuan Province, China
| | - Rui-Feng Zhou
- Department of Infectious Disease, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Lin Cai
- Department of Infectious Disease, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Dong-Mei Yan
- Department of Infectious Disease, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Li-Juan Lan
- Department of Internal Medicine, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Sheng-Hua He
- Department of Infectious Disease, Public Health and Clinical Center of Chengdu, Chengdu 610061, Sichuan Province, China
| | - Hong Tang
- Center of Infectious Disease, Sichuan University West China Hoapital, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
28
|
Guha Ray A, Odum OP, Wiseman D, Weinstock A. The diverse roles of macrophages in metabolic inflammation and its resolution. Front Cell Dev Biol 2023; 11:1147434. [PMID: 36994095 PMCID: PMC10041730 DOI: 10.3389/fcell.2023.1147434] [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: 01/18/2023] [Accepted: 02/14/2023] [Indexed: 03/14/2023] Open
Abstract
Macrophages are one of the most functionally diverse immune cells, indispensable to maintain tissue integrity and metabolic health. Macrophages perform a myriad of functions ranging from promoting inflammation, through inflammation resolution to restoring and maintaining tissue homeostasis. Metabolic diseases encompass a growing list of diseases which develop from a mix of genetics and environmental cues leading to metabolic dysregulation and subsequent inflammation. In this review, we summarize the contributions of macrophages to four metabolic conditions-insulin resistance and adipose tissue inflammation, atherosclerosis, non-alcoholic fatty liver disease and neurodegeneration. The role of macrophages is complex, yet they hold great promise as potential therapies to address these growing health concerns.
Collapse
Affiliation(s)
| | | | | | - Ada Weinstock
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
29
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
30
|
Chen JL, Feng ZL, Zhou F, Lou RH, Peng C, Ye Y, Lin LG. 14-Deoxygarcinol improves insulin sensitivity in high-fat diet-induced obese mice via mitigating NF-κB/Sirtuin 2-NLRP3-mediated adipose tissue remodeling. Acta Pharmacol Sin 2023; 44:434-445. [PMID: 35945312 PMCID: PMC9889782 DOI: 10.1038/s41401-022-00958-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023]
Abstract
Interleukin (IL)-1β is a culprit of adipose tissue inflammation, which in turn causes systematic inflammation and insulin resistance in obese individuals. IL-1β is mainly produced in monocytes and macrophages and marginally in adipocytes, through cleavage of the inactive pro-IL-1β precursor by caspase-1, which is activated via the NLRP3 inflammasome complex. The nuclear factor-κB (NF-κB) transcription factor is the master regulator of inflammatory responses. Brindle berry (Garcinia cambogia) has been widely used as health products for treating obesity and related metabolic disorders, but its active principles remain unclear. We previously found a series of polyisoprenylated benzophenones from brindle berry with anti-inflammatory activities. In this study we investigated whether 14-deoxygarcinol (DOG), a major polyisoprenylated benzophenone from brindle berry, alleviated adipose tissue inflammation and insulin sensitivity in high-fat diet fed mice. The mice were administered DOG (2.5, 5 mg · kg-1 · d-1, i.p.) for 4 weeks. We showed that DOG injection dose-dependently improved insulin resistance and hyperlipidemia, but not adiposity in high-fat diet-fed mice. We found that DOG injection significantly alleviated adipose tissue inflammation via preventing macrophage infiltration and pro-inflammatory polarization of macrophages, and adipose tissue fibrosis via reducing the abnormal deposition of extracellular matrix. In LPS plus nigericin-stimulated THP-1 macrophages, DOG (1.25, 2.5, 5 μM) dose-dependently suppressed the activation of NLRP3 inflammasome and NF-κB signaling pathway. We demonstrated that DOG bound to and activated the deacetylase Sirtuin 2, which in turn deacetylated and inactivated NLRP3 inflammasome to reduce IL-1β secretion. Moreover, DOG (1.25, 2.5, 5 μM) dose-dependently mitigated inflammatory responses in macrophage conditioned media-treated adipocytes and suppressed macrophage migration toward adipocytes. Taken together, DOG might be a drug candidate to treat metabolic disorders through modulation of adipose tissue remodeling.
Collapse
Affiliation(s)
- Jia-Li Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhe-Ling Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ruo-Han Lou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
| |
Collapse
|
31
|
Pavlovic D, Ristic S, Djukanovic L, Matic M, Kovacevic M, Pljesa-Ercegovac M, Hadzi-Djokic J, Savic-Radojevic A, Djukic T. The GSTO2 (rs156697) Polymorphism Modifies Diabetic Nephropathy Risk. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59010164. [PMID: 36676788 PMCID: PMC9865841 DOI: 10.3390/medicina59010164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Background and Objectives: In the development of type 2 diabetes mellitus (T2DM) and its complications, genetic and environmental factors play important roles. Diabetic nephropathy (DN), one of the major microangiopathic chronic diabetic complications, is associated with an increased risk of major cardiovascular events and all-cause mortality. The present study was designed to investigate the possible modifying effect of glutathione transferase polymorphisms (GSTM1, GSTT1, GSTP1 rs1138272/rs1695, GSTO1 rs4925 and GSTO2 rs156697) in the susceptibility to T2DM and diabetic nephropathy. Materials and Methods: GSTM1 and GSTT1 deletion polymorphisms were determined by multiplex PCR, whereas GSTO1, GSTO2, and GSTP1 polymorphisms were determined by the real-time PCR in 160 T2DM patients and 248 age- and gender-matched controls. Advanced glycation end products (AGEs) were measured by ELISA. Results: Among six investigated GST polymorphisms, a significant association between the GST genotypes and susceptibility for development of diabetes mellitus was found for the GSTM1, GSTT1, GSTP1 (rs1138272) and GSTO1 polymorphisms. When the GST genotypes’ distribution in diabetes patients was assessed in the subgroups with and without diabetic nephropathy, a significant association was found only for the GSTO2 rs156697 polymorphism. Diabetic patients, carriers of the GSTM1 null, GSTT1 null and variant GSTO1*AA genotypes, had significantly increased levels of AGEs in comparison with carriers of the GSTM1 active, GSTT1 active and referent GSTO1*CC genotypes (p < 0.001, p < 0.001, p = 0.004, respectively). Conclusions: The present study supports the hypothesis that GST polymorphisms modulate the risk of diabetes and diabetic nephropathy and influence the AGEs concentration, suggesting the potential regulatory role of these enzymes in redox homeostasis disturbances.
Collapse
Affiliation(s)
- Dragana Pavlovic
- Faculty of Medicine, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Sinisa Ristic
- Faculty of Medicine, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Ljubica Djukanovic
- Academy of Medical Sciences of Serbian Medical Society, 11000 Belgrade, Serbia
| | - Marija Matic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, 11000 Belgrade, Serbia
| | - Marijana Kovacevic
- Faculty of Medicine, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Marija Pljesa-Ercegovac
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, 11000 Belgrade, Serbia
| | | | - Ana Savic-Radojevic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, 11000 Belgrade, Serbia
| | - Tatjana Djukic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Institute of Medical and Clinical Biochemistry, 11000 Belgrade, Serbia
- Correspondence: ; Tel.: +381-11-3643273
| |
Collapse
|
32
|
Vieira-Potter VJ. Effects of Sex Hormones and Exercise on Adipose Tissue. SEX HORMONES, EXERCISE AND WOMEN 2023:55-85. [DOI: 10.1007/978-3-031-21881-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
33
|
Mahdavifard S, Sekhavatmand N. Glutamine Is a Superior Protector Against Lead-Induced Hepatotoxicity in Rats via Antioxidant, Anti-inflammatory, and Chelating Properties. Biol Trace Elem Res 2022; 200:4726-4732. [PMID: 35478087 DOI: 10.1007/s12011-021-03046-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022]
Abstract
Lead acetate-motivated oxidative stress can affect all organ systems, particularly the liver. Glutamine (Gln) has both antioxidant and chelating properties. Therefore, we investigated for the first time the effect of Gln on the biochemical and histopathological alternations in a rat model of lead toxicity. Thirty-two rats were divided into four groups (eight rats in each): untreated normal, lead poisoning, and two similar groups receiving Gln (0.1% in drinking water for 4 weeks). To induce lead poisoning, rats received 50 mg/L lead acetate in drinking water for 4 weeks. Oxidative stress indices (total glutathione, the ratio of reduced glutathione to oxidized glutathione, advanced protein oxidation products, malondialdehyde, and ferric ion reducing power) and inflammatory markers (hepatic nuclear factor-kβ expression, interleukin 1β level, and myeloperoxidase activity) were measured. Furthermore, biochemical markers of hepatotoxicity (alanine transaminase, aspartate transaminase, alkaline phosphatase, gamma-glutamyl transpeptidase, total bilirubin, total protein, albumin, and globulins) were measured. Histopathological examination evaluated lead-induced liver damage. The treatment compensated lead-induced biochemical and histopathological alternations in rat liver. Furthermore, it decreased lead acetate level, the NF-kβ gene expression, oxidative stress, and inflammatory markers. Moreover, the treatment elevated total glutathione and reduced glutathione in the sera and liver homogenates of treated groups (p < 0.001). Glutamine could protect the liver against lead intoxication via antioxidant, anti-inflammatory, and chelating properties. In addition, its downregulating effect on the hepatic NF-kβ signaling pathway confirms its hepatoprotective activity.
Collapse
Affiliation(s)
- Sina Mahdavifard
- Department of Clinical Biochemistry, Ardabil University of Medical Sciences, P.O. Box, 56189-85991, Ardabil, Iran.
| | - Negar Sekhavatmand
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
34
|
Li H, Meng Y, He S, Tan X, Zhang Y, Zhang X, Wang L, Zheng W. Macrophages, Chronic Inflammation, and Insulin Resistance. Cells 2022; 11:cells11193001. [PMID: 36230963 PMCID: PMC9562180 DOI: 10.3390/cells11193001] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
The prevalence of obesity has reached alarming levels, which is considered a major risk factor for several metabolic diseases, including type 2 diabetes (T2D), non-alcoholic fatty liver, atherosclerosis, and ischemic cardiovascular disease. Obesity-induced chronic, low-grade inflammation may lead to insulin resistance, and it is well-recognized that macrophages play a major role in such inflammation. In the current review, the molecular mechanisms underlying macrophages, low-grade tissue inflammation, insulin resistance, and T2D are described. Also, the role of macrophages in obesity-induced insulin resistance is presented, and therapeutic drugs and recent advances targeting macrophages for the treatment of T2D are introduced.
Collapse
Affiliation(s)
- He Li
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ya Meng
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuwang He
- Shandong DYNE Marine Biopharmaceutical Co., Ltd., Rongcheng 264300, China
| | - Xiaochuan Tan
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yujia Zhang
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiuli Zhang
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lulu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Correspondence: (L.W.); (W.Z.); Tel.: +86-010-63165233 (W.Z.)
| | - Wensheng Zheng
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Correspondence: (L.W.); (W.Z.); Tel.: +86-010-63165233 (W.Z.)
| |
Collapse
|
35
|
Stenberg K, Gensby L, Cremer SE, Nielsen MM, Bjørnvad CR. Analytical performance of a canine ELISA monocyte chemoattractant protein-1 assay for use in cats and evaluation of circulating levels in normal weight and obese cats. Acta Vet Scand 2022; 64:22. [PMID: 36064726 PMCID: PMC9446815 DOI: 10.1186/s13028-022-00640-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In human and murine obesity, adipose tissue dwelling macrophages and adipocytes produce monocyte chemoattractant protein-1 (MCP-1) leading to systemic low-grade inflammation. The aim of the study was to validate a canine MCP-1 ELISA assay for use in cats and to investigate whether a difference in MCP-1 concentrations could be detected between: a) cats having normal or elevated circulating serum amyloid A (SAA) levels and b) normal weight and obese cats. Serum obtained from 36 client-owned cats of various breed, age and sex with normal (n = 20) to elevated SAA (n = 16) was used for the validation of the canine MCP-1 ELISA assay. As no golden standard exists for measurement of inflammation, circulating MCP-1 concentrations were compared to SAA measurements, as an indicator of systemic inflammation. Analytical precision, dilution recovery and detection limit were calculated. A possible correlation between MCP-1 concentrations and obesity related measures (body fat percentage (BF%), insulin sensitivity and cytokine expression) were investigated in another population of 73 healthy, lean to obese, neutered domestic short-haired cats. RESULTS Intra- (2.7-4.1%) and inter-assay (2.2-3.6%) coefficient of variation and dilution recovery were acceptable, and the detection limit was 27.1 pg/mL. MCP-1 did not correlate with SAA, and there was no difference between the inflammatory (SAA > 20 mg/L) and non-inflammatory group, due to a marked overlap in MCP-1 concentrations. Circulating MCP-1 concentrations were unaffected by BF% (r2 = 2.7 × 10-6, P = 0.21) and other obesity-related markers. CONCLUSIONS The present canine ELISA assay seems to be able to measure circulating feline MCP-1. However, further studies are needed to determine its possible use for detecting inflammation in relation to disease processes or obesity-related low-grade inflammation in cats.
Collapse
Affiliation(s)
- Kathrine Stenberg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg, Denmark
| | - Line Gensby
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg, Denmark
- Present Address: AniCura Vangede Animal Hospital, Plantevej 2, 2870 Dyssegård, Denmark
| | - Signe Emilie Cremer
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg, Denmark
- Present Address: Coloplast, Holtedam 1-3, 3050 Humlebæk, Denmark
| | - Michelle Møller Nielsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg, Denmark
| | - Charlotte Reinhard Bjørnvad
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg, Denmark
| |
Collapse
|
36
|
Speelman T, Dale L, Louw A, Verhoog NJD. The Association of Acute Phase Proteins in Stress and Inflammation-Induced T2D. Cells 2022; 11:2163. [PMID: 35883605 PMCID: PMC9321356 DOI: 10.3390/cells11142163] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/06/2023] Open
Abstract
Acute phase proteins (APPs), such as plasminogen activator inhibitor-1 (PAI-1), serum amyloid A (SAA), and C-reactive protein (CRP), are elevated in type-2 diabetes (T2D) and are routinely used as biomarkers for this disease. These APPs are regulated by the peripheral mediators of stress (i.e., endogenous glucocorticoids (GCs)) and inflammation (i.e., pro-inflammatory cytokines), with both implicated in the development of insulin resistance, the main risk factor for the development of T2D. In this review we propose that APPs, PAI-1, SAA, and CRP, could be the causative rather than only a correlative link between the physiological elements of risk (stress and inflammation) and the development of insulin resistance.
Collapse
Affiliation(s)
| | | | | | - Nicolette J. D. Verhoog
- Biochemistry Department, Stellenbosch University, Van der Byl Street, Stellenbosch 7200, South Africa; (T.S.); (L.D.); (A.L.)
| |
Collapse
|
37
|
Chen C, Ren YM, Zhu JZ, Chen JL, Feng ZL, Zhang T, Ye Y, Lin LG. Ainsliadimer C, a disesquiterpenoid isolated from Ainsliaea macrocephala, ameliorates inflammatory responses in adipose tissue via Sirtuin 1-NLRP3 inflammasome axis. Acta Pharmacol Sin 2022; 43:1780-1792. [PMID: 34789920 PMCID: PMC9253034 DOI: 10.1038/s41401-021-00797-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022]
Abstract
Interleukin-1β (IL-1β), a key pro-inflammatory cytokine, is majorly produced by macrophages through NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, which has been identified as the culprit to deteriorate the inflammatory crosstalk between macrophages and adipocytes. Ainsliadimer C (AC) is a disesquiterpenoid isolated from Ainsliaea macrocephala. In the current study, we investigated the effects of AC on adipose tissue inflammation in co-culture of macrophages and adipocytes in vitro as well as in LPS-treated mice in vivo. We showed that AC (20-80 µM) dose-dependently inhibited the secretion of IL-1β from LPS plus ATP-stimulated THP-1 macrophages by inhibiting the activation of NLRP3 inflammasome. Furthermore, we found that AC treatment activated NAD+-dependent deacetylase Sirtuin 1 (SIRT1), resulting in reduced acetylation level of NLRP3. Molecular modeling analysis revealed that binding of AC to sirtuin-activating compound-binding domain increased the affinity of the substrate to the catalytic domain of SIRT1. Moreover, AC (80 µM) significantly attenuated macrophage-conditioned medium-induced inflammatory responses in 3T3-L1 adipocytes. In LPS-induced acute inflammatory mice, administration of AC (20, 60 mg·kg-1·d-1, ip) for 5 days significantly suppressed the pro-inflammatory cytokine levels in serum and epididymal white adipose tissue (eWAT), attenuated macrophage infiltration into eWAT, and mitigated adipose tissue inflammation. The beneficial effects of AC were blocked by co-administration of a selective SIRT1 inhibitor EX-527 (10 mg·kg-1·d-1). Taken together, AC suppresses NLRP3-mediated IL-1β secretion through activating SIRT1, leading to attenuated inflammation in macrophages and adipose tissue, which might be a candidate to treat obesity-associated metabolic diseases.
Collapse
Affiliation(s)
- Cheng Chen
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Yong-mei Ren
- grid.9227.e0000000119573309State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jian-zhong Zhu
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Jia-li Chen
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Zhe-ling Feng
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Tian Zhang
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Li-gen Lin
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078 China
| |
Collapse
|
38
|
Im S, Kim H, Jeong M, Yang H, Hong JY. Integrative understanding of immune-metabolic interaction. BMB Rep 2022. [PMID: 35651325 PMCID: PMC9252895 DOI: 10.5483/bmbrep.2022.55.6.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies have revealed that the immune system plays a critical role in various physiological processes beyond its classical pathogen control activity. Even under a sterile condition, various cells and tissues can utilize the immune system to meet a specific demand for proper physiological functions. Particularly, a strong link between immunity and metabolism has been identified. Studies have identified the reciprocal regulation between these two systems. For example, immune signals can regulate metabolism, and metabolism (cellular or systemic) can regulate immunity. In this review, we will summarize recent findings on this reciprocal regulation between immunity and metabolism, and discuss potential biological rules behind this interaction with integrative perspectives.
Collapse
Affiliation(s)
- Seonyoung Im
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Hawon Kim
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Myunghyun Jeong
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Hyeon Yang
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Jun Young Hong
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
39
|
Singh S, Sharma A, Guru B, Ahmad S, Gulzar F, Kumar P, Ahmad I, Tamrakar AK. Fructose-mediated NLRP3 activation induces inflammation and lipogenesis in adipose tissue. J Nutr Biochem 2022; 107:109080. [PMID: 35660098 DOI: 10.1016/j.jnutbio.2022.109080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/02/2022] [Accepted: 05/03/2022] [Indexed: 01/07/2023]
Abstract
Adipose tissue plays a crucial role in energy intake and regulation of metabolic homeostasis. Fructose consumption implicates in development and progression of metabolic dysfunctions. Fructose is a lipogenic sugar known to induce inflammatory response. However, the role of specific inflammatory signal such as nucleotide-binding and oligomerization domain-like receptor, leucine-rich repeat and pyrin domain containing protein 3 (NLRP3) in fructose-induced inflammatory response and its relevance to lipogenesis in adipose tissue are elusive. We assessed NLRP3 activation and its significance in inflammatory response and lipogenesis in epididymal adipose tissue of 60% fructose diet (HFrD)-fed rats. The long term consumption of HFrD led to impairment of glucose metabolism, development of visceral adiposity, insulin resistance, and elevation of serum triglycerides level, accompanied by activation of NLRP3 in adipose tissue. NLRP3 inflammasome activation in adipose tissue was associated with up-regulated expression of Nlrp3, Asc, and Caspase-1, and raised caspase-1 activity, which resulted in increased expression of IL-1β and IL-18 and secretion of IL-1β. Moreover, lipid accumulation and expression of transcription factors exacerbating accumulation of lipids were augmented in adipose tissue of HFrD-fed rats. Treatment with glyburide, quercetin or allopurinol corrected HFrD-induced dyslipidemia or hyperuricemia, and blocked NLRP3 activation, leading to mitigated inflammatory signalling and lipid accumulation in adipose tissue, improved glucose tolerance and insulin sensitivity in HFrD-fed rats. These data suggest the role of NLRP3 inflammasome to establish linkage among inflammation, lipid accumulation and insulin resistance in adipose tissue, and targeting NLRP3 inflammasome may be a plausible approach for prevention and management for fructose-induced metabolic impairments.
Collapse
Affiliation(s)
- Sushmita Singh
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Aditya Sharma
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Bhavimani Guru
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shadab Ahmad
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Farah Gulzar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Pawan Kumar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Ishbal Ahmad
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Akhilesh K Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
40
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
41
|
Bailin SS, Gabriel CL, Fan R, Ye F, Nair S, Terry JG, Carr JJ, Silver H, Wanjalla CN, Mashayekhi M, Lima M, Woodward B, Hannah L, Fuseini H, Ferguson JF, Kropski JA, Koethe JR. Relationship of Subcutaneous Adipose Tissue Inflammation-Related Gene Expression With Ectopic Lipid Deposition in Persons With HIV. J Acquir Immune Defic Syndr 2022; 90:175-183. [PMID: 35125474 PMCID: PMC9203874 DOI: 10.1097/qai.0000000000002926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Fat redistribution from subcutaneous adipose tissue (SAT) to the abdominal viscera, pericardium, liver, and skeletal muscle contributes to the rising burden of cardiometabolic disease among persons with HIV (PWH). Previous studies found SAT inflammation in PWH impairs lipid storage and persists despite plasma viral suppression on antiretroviral therapy (ART). In this study, we identified SAT immune-related genes associated with ectopic fat deposition in PWH on long-term ART. DESIGN AND METHODS A total of 92 PWH with well-controlled viremia underwent computed tomography imaging and abdominal SAT biopsy for gene expression analysis. SAT gene expression was measured using a NanoString panel of 255 immune-related genes. Associations between gene expression and computed tomography measurements of the volume and attenuation (radiodensity) of metabolically relevant ectopic fat depots were assessed using multivariable linear regression and network analysis. RESULTS Greater SAT volume was associated with higher visceral and pericardial adipose tissue volume, but lower skeletal muscle attenuation. Lower SAT attenuation, a measure of lipid content, was associated with lower visceral adipose tissue attenuation. Hierarchical clustering identified a subset of macrophage-related genes in SAT, including CCL2, CCL22, CCL13, CCR1, CD86, CD163, IL-6, IL-10, MRC1, and TREM2, which were associated with an increased lipid deposition in multiple ectopic depots. CONCLUSION Altered expression of macrophage-related genes in SAT is associated with differences in ectopic fat depot morphometrics among PWH on long-term ART, including in the pericardial and visceral compartments. These findings provide basis for future studies to assess host, virus, and treatment factors shaping the SAT immune environment and its effects on morphometric changes and metabolic comorbidities in PWH.
Collapse
Affiliation(s)
- Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L. Gabriel
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Run Fan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fei Ye
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sangeeta Nair
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James G. Terry
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heidi Silver
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Morgan Lima
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Beverly Woodward
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - LaToya Hannah
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hubaida Fuseini
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan A. Kropski
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
- Divison of Allergy, Pulmonary and Critical Care, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
42
|
Im S, Kim H, Jeong M, Yang H, Hong JY. Integrative understanding of immune-metabolic interaction. BMB Rep 2022; 55:259-266. [PMID: 35651325 PMCID: PMC9252895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/22/2022] [Accepted: 05/17/2022] [Indexed: 02/21/2025] Open
Abstract
Recent studies have revealed that the immune system plays a critical role in various physiological processes beyond its classical pathogen control activity. Even under a sterile condition, various cells and tissues can utilize the immune system to meet a specific demand for proper physiological functions. Particularly, a strong link between immunity and metabolism has been identified. Studies have identified the reciprocal regulation between these two systems. For example, immune signals can regulate metabolism, and metabolism (cellular or systemic) can regulate immunity. In this review, we will summarize recent findings on this reciprocal regulation between immunity and metabolism, and discuss potential biological rules behind this interaction with integrative perspectives. [BMB Reports 2022; 55(6): 259-266].
Collapse
Affiliation(s)
- Seonyoung Im
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Hawon Kim
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Myunghyun Jeong
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Hyeon Yang
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Jun Young Hong
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
43
|
Nutrition Interventions of Herbal Compounds on Cellular Senescence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1059257. [PMID: 35528514 PMCID: PMC9068308 DOI: 10.1155/2022/1059257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/01/2021] [Accepted: 04/02/2022] [Indexed: 01/10/2023]
Abstract
When cells undergo large-scale senescence, organ aging ensues, resulting in irreversible organ pathology and organismal aging. The study of senescence in cells provides an important avenue to understand the factors that influence aging and can be used as one of the useful tools for examining age-related human diseases. At present, many herbal compounds have shown effects on delaying cell senescence. This review summarizes the main characteristics and mechanisms of cell senescence, age-related diseases, and the recent progress on the natural products targeting cellular senescence, with the aim of providing insights to aid the clinical management of age-related diseases.
Collapse
|
44
|
Della Guardia L, Shin AC. White and brown adipose tissue functionality is impaired by fine particulate matter (PM2.5) exposure. J Mol Med (Berl) 2022; 100:665-676. [PMID: 35286401 PMCID: PMC9110515 DOI: 10.1007/s00109-022-02183-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 01/09/2022] [Accepted: 02/16/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università Degli Studi Di Milano, via Fratelli Cervi 93, 20090, Segrate, Milano, Italy.
| | - Andrew C Shin
- Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
45
|
Matacchione G, Perugini J, Di Mercurio E, Sabbatinelli J, Prattichizzo F, Senzacqua M, Storci G, Dani C, Lezoche G, Guerrieri M, Giordano A, Bonafè M, Olivieri F. Senescent macrophages in the human adipose tissue as a source of inflammaging. GeroScience 2022; 44:1941-1960. [PMID: 35247131 PMCID: PMC9616990 DOI: 10.1007/s11357-022-00536-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
Obesity is a major risk factor for type 2 diabetes and a trigger of chronic and systemic inflammation. Recent evidence suggests that an increased burden of senescent cells (SCs) in the adipose tissue of obese/diabetic animal models might underlie such pro-inflammatory phenotype. However, the role of macrophages as candidate SCs, their phenotype, the distribution of SCs among fat depots, and clinical relevance are debated. The senescence marker β-galactosidase and the macrophage marker CD68 were scored in visceral (vWAT) and subcutaneous (scWAT) adipose tissue from obese patients (n=17) undergoing bariatric surgery and control patients (n=4) subjected to cholecystectomy. A correlation was made between the number of SCs and BMI, serum insulin, and the insulin resistance (IR) index HOMA. The monocyte cell line (THP-1) was cultured in vitro in high glucose milieu (60 mM D-glucose) and subsequently co-cultured with human adipocytes (hMADS) to investigate the reciprocal inflammatory activation. In obese patients, a significantly higher number of SCs was observed in vWAT compared to scWAT; about 70% of these cells expressed the macrophage marker CD68; and the number of SCs in vWAT, but not in scWAT, positively correlated with BMI, HOMA-IR, and insulin. THP-1 cultured in vitro in high glucose milieu acquired a senescent-like phenotype (HgSMs), characterized by a polarization toward a mixed M1/M2-like secretory phenotype. Co-culturing HgSMs with hMADS elicited pro-inflammatory cytokine expression in both cell types, and defective insulin signaling in hMADS. In morbid obesity, expansion of visceral adipose depots involves an increased burden of macrophages with senescent-like phenotype that may promote a pro-inflammatory profile and impair insulin signaling in adipocytes, supporting a framework where senescent macrophages fuel obesity-induced systemic inflammation and possibly contribute to the development of IR.
Collapse
Affiliation(s)
- Giulia Matacchione
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy.
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - Eleonora Di Mercurio
- Department of Experimental and Clinical Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
| | | | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - Gianluca Storci
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Christian Dani
- Inserm, iBV, Faculté de Médecine, Université Côte d'Azur, CNRS, Nice Cedex, France
| | - Giovanni Lezoche
- Department of General Surgery, Università Politecnica delle Marche, Ancona, Italy
| | - Mario Guerrieri
- Department of General Surgery, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, Università di Bologna, Bologna, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
46
|
Chinta PK, Tambe S, Umrani D, Pal AK, Nandave M. Effect of parthenolide, an NLRP3 inflammasome inhibitor, on insulin resistance in high-fat diet-obese mice. Can J Physiol Pharmacol 2022; 100:272-281. [PMID: 35119950 DOI: 10.1139/cjpp-2021-0116] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The activation of Nod-like receptor proteins (NLRP3) containing the pyrin domain inflammasome is a hallmark of the pathogenesis of metabolic disorders. Inhibition of the NLRP3 inflammasome by phytoconstituents has been attempted as a strategy to mitigate these disorders. Therefore, the present study aimed to evaluate the efficacy of an NLRP3 inflammasome inhibitor, parthenolide (PN; 5 mg/kg i.p.) against inflammation and insulin resistance in high-fat diet (HFD) - obese mice. Treatment with PN and pioglitazone (PIO; 30 mg/kg p.o.) attenuated lipopolysaccharide (LPS; 1 ng/ml) - induced elevation of tumor necrosis factor-α and interleukin-1β in mouse peritoneal macrophages in a dose-dependent manner. Sixty days of PN and PIO treatment marginally reduced obesity-induced insulin resistance in HFD-obese mice. PN treatment also decreased blood glucose from 14th to 60th day, supporting the hypothesis of simultaneous attenuation of inflammation and insulin resistance in obese mice. Thus, PN treatment was also evident with significant improvement in glucose tolerance and peripheral insulin resistance validated through the respective tolerance tests. Therefore, the present study suggests that PN, an NLRP3 inflammasome inhibitor, could be a possible therapeutic agent for attenuating obesity-induced insulin resistance.
Collapse
Affiliation(s)
| | | | | | - Ajay Kumar Pal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi-110017, INDIA
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi-110017, INDIA
| |
Collapse
|
47
|
Hematological malignancies magnify the effect of body mass index on insulin resistance in cancer survivors. Blood Adv 2022; 6:1981-1990. [PMID: 35130338 PMCID: PMC9006273 DOI: 10.1182/bloodadvances.2021006241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/19/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer survivors have disproportionately greater insulin resistance with increasing BMI compared with controls without malignancies. Our findings indicate that interventions targeting obesity will be needed to prevent the sequelae of insulin resistance in cancer survivors.
Cancer survivors are at increased risk of type 2 diabetes, which usually develops from obesity and insulin resistance. Whether diabetes susceptibility is due to shared risk factors for cancer and insulin resistance or directly related to cancer and its treatment is unknown. We investigated effect modification between malignancy and body mass index (BMI) as determinants of insulin sensitivity in patients with hematologic malignancies and controls without cancer. In a cross-sectional study of 43 individuals without diabetes (20 patients with treated hematologic malignancies; 23 controls without malignancies), we measured insulin-stimulated whole-body glucose use (M) by hyperinsulinemic euglycemic clamp. Insulin sensitivity index (ISI) was calculated by dividing M over steady-state plasma insulin. Inflammatory cytokines were measured in plasma. Controls were more obese and included more non-White individuals and women vs patients with hematologic malignancies. Patients with cancer exhibited greater insulin sensitivity (median ISI, 42.4 mg/kg/min/[μU/mL]; interquartile range [IQR], 33.9-67.2 vs 23.4 mg/kg/min/[μU/mL]; IQR, 12.9-29.2; P < .001) and higher interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) concentrations vs controls. Patients with cancer demonstrated greater reduction in ISI with increasing BMI vs controls, which remained significant after adjustment for sex and race (β = −2.6 units; 95% confidence interval, −4.8 to −0.4; P interaction = .024). This interaction also remained significant after adjusting for log IL-6 (P interaction = .048) and log MCP-1 (P interaction = .021). Cancer survivors had disproportionately greater insulin resistance with increasing BMI vs controls without malignancies. Effect modification between cancer and BMI in determining insulin sensitivity implicated cancer-specific etiologies in glucose dysregulation and could partially explain excess diabetes diagnoses among oncology patients.
Collapse
|
48
|
Yang Y, Chen Z, Zhao X, Xie H, Du L, Gao H, Xie C. Mechanisms of Kaempferol in the treatment of diabetes: A comprehensive and latest review. Front Endocrinol (Lausanne) 2022; 13:990299. [PMID: 36157449 PMCID: PMC9490412 DOI: 10.3389/fendo.2022.990299] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023] Open
Abstract
Obesity-insulin resistance-β-cells apoptosis" is an important trilogy of the pathogenesis of type 2 diabetes. With the global pandemic of obesity and diabetes, continuous research and development of new drugs focuses on the prevention of the pathological progress of these diseases. According to a recent study, the natural product kaempferol has excellent antidiabetic effects. Therefore, this review comprehensively summarized the frontier studies and pharmacological mechanisms of kaempferol in the treatment of diabetes. The successful research and development of kaempferol may yield a significant leap in the treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Yan Yang
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Zhengtao Chen
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyan Zhao
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyan Xie
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Lian Du
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Gao
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Hong Gao, ; Chunguang Xie,
| | - Chunguang Xie
- Hospital of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Hong Gao, ; Chunguang Xie,
| |
Collapse
|
49
|
Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, Griffiths HR, Gao D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022; 13:873699. [PMID: 35909571 PMCID: PMC9329830 DOI: 10.3389/fendo.2022.873699] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic low-grade inflammation in adipose tissue (AT) is a hallmark of obesity and contributes to various metabolic disorders, such as type 2 diabetes and cardiovascular diseases. Inflammation in ATs is characterized by macrophage infiltration and the activation of inflammatory pathways mediated by NF-κB, JNK, and NLRP3 inflammasomes. Adipokines, hepatokines and myokines - proteins secreted from AT, the liver and skeletal muscle play regulatory roles in AT inflammation via endocrine, paracrine, and autocrine pathways. For example, obesity is associated with elevated levels of pro-inflammatory adipokines (e.g., leptin, resistin, chemerin, progranulin, RBP4, WISP1, FABP4, PAI-1, Follistatin-like1, MCP-1, SPARC, SPARCL1, and SAA) and reduced levels of anti-inflammatory adipokines such as adiponectin, omentin, ZAG, SFRP5, CTRP3, vaspin, and IL-10. Moreover, some hepatokines (Fetuin A, DPP4, FGF21, GDF15, and MANF) and myokines (irisin, IL-6, and DEL-1) also play pro- or anti-inflammatory roles in AT inflammation. This review aims to provide an updated understanding of these organokines and their role in AT inflammation and related metabolic abnormalities. It serves to highlight the molecular mechanisms underlying the effects of these organokines and their clinical significance. Insights into the roles and mechanisms of these organokines could provide novel and potential therapeutic targets for obesity-induced inflammation.
Collapse
Affiliation(s)
- Yakun Ren
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
| | - Hao Zhao
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xi Lan
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Litao Wu
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Xiaojuan Du
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Helen R. Griffiths
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Dan Gao
- Institute of Molecular and Translational Medicine, Xian Jiaotong University Health Science Center, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
- *Correspondence: Dan Gao,
| |
Collapse
|
50
|
Impact of Bariatric Surgery on Adipose Tissue Biology. J Clin Med 2021; 10:jcm10235516. [PMID: 34884217 PMCID: PMC8658722 DOI: 10.3390/jcm10235516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery (BS) procedures are actually the most effective intervention to help subjects with severe obesity achieve significant and sustained weight loss. White adipose tissue (WAT) is increasingly recognized as the largest endocrine organ. Unhealthy WAT expansion through adipocyte hypertrophy has pleiotropic effects on adipocyte function and promotes obesity-associated metabolic complications. WAT dysfunction in obesity encompasses an altered adipokine secretome, unresolved inflammation, dysregulated autophagy, inappropriate extracellular matrix remodeling and insufficient angiogenic potential. In the last 10 years, accumulating evidence suggests that BS can improve the WAT function beyond reducing the fat depot sizes. The causal relationships between improved WAT function and the health benefits of BS merits further investigation. This review summarizes the current knowledge on the short-, medium- and long-term outcomes of BS on the WAT composition and function.
Collapse
|