1
|
Zhao H, Lv J, Chen B, He F, Wang Q, Xie D, Koyama H, Zhang C, Cheng J. RAGE deficiency obstructs high uric acid-induced oxidative stress and inflammatory response. Int Immunopharmacol 2025; 151:114316. [PMID: 39987631 DOI: 10.1016/j.intimp.2025.114316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
Hyperuricemia is a metabolic disorder primarily associated with gout and implicated in various metabolic inflammatory diseases. While the role of monosodium urate crystals triggering inflammation has been well-documented, recent findings suggest that soluble high uric acid (HUA) also induces pro-inflammatory cytokine production in human monocytes. However, the comprehensive effects of HUA levels on macrophage dysfunction and the underlying mechanisms remain underexplored. This study employs urate oxidase knockout (UOX-KO) and receptor for advanced glycation end products deficiency (RAGE-/-) mouse models to elucidate macrophage function and its mechanistic pathways. Our results demonstrate that HUA promotes M1 polarization and migration of macrophages while impairing their phagocytic ability. This process is mediated through the high mobility group box 1 (HMGB1)-RAGE- ROS axis. Notably, RAGE deficiency in bone marrow-derived cells partially mediates these effects. Pathologically, elevated HMGB1 and monocyte chemoattractant protein 1 levels in pancreatic islets increases macrophage infiltration in UOX-KO mice. Treatment with the FPS-ZM1, as a pharmacological RAGE inhibitor, effectively decreases serum UA levels, ameliorates islet inflammation and insulin resistance. These findings suggest that soluble HUA serves as a pro-inflammatory trigger through the HMGB1-RAGE-ROS axis, and that RAGE inhibition may mitigate these effects by decreasing inflammatory macrophage infiltration in the islets. Additionally, the influence of UA on macrophages extends beyond gout, potentially contributing to the pathogenesis of other metabolic inflammatory conditions, such as atherosclerosis, non-alcoholic steatohepatitis, obesity, and hyperlipidemia.
Collapse
Affiliation(s)
- Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China; Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jiamin Lv
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Binyang Chen
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Furong He
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Qiang Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China.
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China; Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan.
| |
Collapse
|
2
|
Qu H, Yang Y, Xie Q, Ye L, Shao Y. Linear association of the dietary index for gut microbiota with insulin resistance and type 2 diabetes mellitus in U.S. adults: the mediating role of body mass index and inflammatory markers. Front Nutr 2025; 12:1557280. [PMID: 40191795 PMCID: PMC11968382 DOI: 10.3389/fnut.2025.1557280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Background Gut microbiota is reported to be related to the onset of insulin resistance (IR) and type 2 diabetes mellitus (T2DM). The dietary index for gut microbiota (DI-GM) is a novel index for reflecting gut microbiota diversity. We aimed to evaluate the association of DI-GM with T2DM and IR. Methods This cross-sectional research comprised 10,600 participants aged ≥20 from the National Health and Nutrition Examination Survey (NHANES) 2007-2018. We employed weighted multivariable linear and logistic regression models to examine the correlation of DI-GM with T2DM and IR. Linear or nonlinear relationships were examined by restricted cubic spline (RCS) regression. Additionally, subgroup and sensitivity analyses were performed to ensure the reliability of the results. Mediation analysis explored the roles of body mass index (BMI) and inflammatory factors in these associations. Results Higher DI-GM were inversely associated with T2DM (OR = 0.93, 95%CI: 0.89-0.98) and IR (OR = 0.95, 95%CI: 0.91-0.99) after adjusting for confounders. DI-GM ≥ 6 group showed significantly lower risks of T2DM (OR = 0.74, 95%CI: 0.60-0.91) and IR (OR = 0.77, 95%CI: 0.62-0.95). RCS demonstrated a linear relationship between DI-GM and T2DM, as well as IR. DI-GM was also inversely correlated with the risk markers of T2DM. Mediation analysis showed that BMI and the systemic inflammation response index partly mediated the association of DI-GM with T2DM and IR, while the systemic immune-inflammation index mediated only the association with T2DM. Conclusion DI-GM is inversely associated with T2DM and IR, with BMI and inflammatory markers partly mediating this association.
Collapse
Affiliation(s)
- Haoran Qu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyun Yang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qihang Xie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Ye
- Department of Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Shao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Correa-da-Silva F, Yi CX. Neuroglia in eating disorders (obesity, Prader-Willi syndrome and anorexia nervosa). HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:313-324. [PMID: 40148052 DOI: 10.1016/b978-0-443-19102-2.00019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The hypothalamus is widely recognized as one of the most extensively studied brain regions involved in the central regulation of energy homeostasis. Within the hypothalamus, peptidergic neurons play a crucial role in monitoring peripheral concentrations of metabolites and hormones, and they finely adjust the sensing of these factors, leading to the activation of either anorexigenic (appetite-suppressing) or orexigenic (appetite-stimulating) pathways. While cortical innervation of the hypothalamus does influence these processes, it is generally considered of secondary importance. Eating-related disorders, such as obesity and anorexia nervosa, are strongly associated with imbalances in energy intake and expenditure. The phenotypes of these disorders can be attributed to dysfunctions in the hypothalamus. Traditionally, it has been believed that hypothalamic dysfunction in these disorders primarily stems from defects in neural pathways. However, recent evidence challenges this perception, highlighting the active participation of neuroglial cells in shaping both physiologic and behavioral characteristics. This review aims to provide an overview of the latest insights into glial biology in three specific eating disorders: obesity, Prader-Willi syndrome, and anorexia. In these disorders, neural dysfunction coincides with glial malfunction, suggesting that neuroglia actively contribute to the development and progression of various neurologic disorders. These findings underscore the importance of glial cells and open up potential new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Felipe Correa-da-Silva
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, The Netherlands; Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Soedono S, Julietta V, Nawaz H, Cho KW. Dynamic Roles and Expanding Diversity of Adipose Tissue Macrophages in Obesity. J Obes Metab Syndr 2024; 33:193-212. [PMID: 39324219 PMCID: PMC11443328 DOI: 10.7570/jomes24030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024] Open
Abstract
Adipose tissue macrophages (ATMs) are key regulators of adipose tissue (AT) inflammation and insulin resistance in obesity, and the traditional M1/M2 characterization of ATMs is inadequate for capturing their diversity in obese conditions. Single-cell transcriptomic profiling has revealed heterogeneity among ATMs that goes beyond the old paradigm and identified new subsets with unique functions. Furthermore, explorations of their developmental origins suggest that multiple differentiation pathways contribute to ATM variety. These advances raise concerns about how to define ATM functions, how they are regulated, and how they orchestrate changes in AT. This review provides an overview of the current understanding of ATMs and their updated categorization in both mice and humans during obesity. Additionally, diverse ATM functions and contributions in the context of obesity are discussed. Finally, potential strategies for targeting ATM functions as therapeutic interventions for obesity-induced metabolic diseases are addressed.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Vivi Julietta
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Hadia Nawaz
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
5
|
Zhou X, Wang S, Yu D, Niu T. Investigating CR1 as an indicated Gene for mild cognitive impairment in type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:206. [PMID: 39182129 PMCID: PMC11344402 DOI: 10.1186/s13098-024-01449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) has beenis known as an important risk factor for cognitive impairment. Meanwhile, the liver plays a central role in the development of T2DM and insulin resistance. The present study attempted to identify and validate marker genes for mild cognitive impairment (MCI) in patients with T2DM. METHODS In this study, insulin resistance-related differentially expressed genes were identified from the liver tissues of individuals with T2DM and those with normal glucose tolerance using the Gene Expression Omnibus database and MCI-associated genes were identified using the GeneCards database. Next, enrichment analysis was performed with overlapping T2DM and MCI genes, followed by the identification of specific genes using the LASSO logistic regression and SVM-RFE algorithms. An important experiment involved the implementation of clinical and in vitro validation using real-time quantitative polymerase chain reaction (RT-qPCR). Finally, multiple linear regression, binary logistic regression, and receiver operating characteristic curve analyses were performed to investigate the relationship between the key gene and cognitive function in these patients. RESULT The present study identified 40 overlapping genes between MCI and T2DM, with subsequent enrichment analysis revealing their significant association with the roles of neuronal and glial projections. The marker gene complement receptor 1(CR1) was identified for both diseases using two regression algorithms. Based on RT-qPCR validation in 65 T2DM patients with MCI (MCI group) and 65 T2DM patients without MCI (NC group), a significant upregulation of CR1 mRNA in peripheral blood mononuclear cells was observed in the MCI group (P < 0.001). Furthermore, the CR1 gene level was significantly negatively associated with MoCA and MMSE scores, which reflect the overall cognitive function, and positively correlated with TMTB scores, which indicate the executive function. Finally, elevated CR1 mRNA levels were identified as an independent risk factor for MCI (OR = 1.481, P < 0.001). CONCLUSION These findings suggest that CR1 is an important predictor of MCI in patients with T2DM. Thus, CR1 has potential clinical significance, which may offer new ideas and directions for the management and treatment of T2DM. The identification and clinical validation of dysregulated marker genes in both T2DM and MCI can offer valuable insights into the intrinsic association between these two conditions. The current study insights may inspire the development of novel strategies for addressing the complicated issues related to cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Xueling Zhou
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Shaohua Wang
- School of Medicine, Southeast University, Nanjing, China.
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Dandan Yu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Tong Niu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
6
|
Wong A, Sun Q, Latif II, Karwi QG. Metabolic flux in macrophages in obesity and type-2 diabetes. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13210. [PMID: 38988822 PMCID: PMC11233469 DOI: 10.3389/jpps.2024.13210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
Recent literature extensively investigates the crucial role of energy metabolism in determining the inflammatory response and polarization status of macrophages. This rapidly expanding area of research highlights the importance of understanding the link between energy metabolism and macrophage function. The metabolic pathways in macrophages are intricate and interdependent, and they can affect the polarization of macrophages. Previous studies suggested that glucose flux through cytosolic glycolysis is necessary to trigger pro-inflammatory phenotypes of macrophages, and fatty acid oxidation is crucial to support anti-inflammatory responses. However, recent studies demonstrated that this understanding is oversimplified and that the metabolic control of macrophage polarization is highly complex and not fully understood yet. How the metabolic flux through different metabolic pathways (glycolysis, glucose oxidation, fatty acid oxidation, ketone oxidation, and amino acid oxidation) is altered by obesity- and type 2 diabetes (T2D)-associated insulin resistance is also not fully defined. This mini-review focuses on the impact of insulin resistance in obesity and T2D on the metabolic flux through the main metabolic pathways in macrophages, which might be linked to changes in their inflammatory responses. We closely evaluated the experimental studies and methodologies used in the published research and highlighted priority research areas for future investigations.
Collapse
Affiliation(s)
- Angela Wong
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Qiuyu Sun
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ismail Ibrahim Latif
- Department of Microbiology, College of Medicine, University of Diyala, Baqubaa, Diyala, Iraq
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, NL, Canada
| |
Collapse
|
7
|
Sun C, Zhao S, Pan Z, Li J, Wang Y, Kuang H. The Role Played by Mitochondria in Polycystic Ovary Syndrome. DNA Cell Biol 2024; 43:158-174. [PMID: 38588493 DOI: 10.1089/dna.2023.0345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) refers to an endocrine disorder syndrome that are correlated with multiple organs and systems. PCOS has an effect on women at all stages of their lives, and it has an incidence nearly ranging from 6% to 20% worldwide. Mitochondrial dysfunctions (e.g., oxidative stress, dynamic imbalance, and abnormal quality control system) have been identified in patients and animal models of PCOS, and the above processes may play a certain role in the development of PCOS and its associated complications. However, their specific pathogenic roles should be investigated in depth. In this review, recent studies on the mechanisms of action of mitochondrial dysfunction in PCOS and its associated clinical manifestations are summarized from the perspective of tissues and organs, and some studies on the treatment of the disease by improving mitochondrial function are reviewed to highlight key role of mitochondrial dysfunction in this syndrome.
Collapse
Affiliation(s)
- Chang Sun
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shanshan Zhao
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zimeng Pan
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Li
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yasong Wang
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongying Kuang
- Second Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Peng C, Chen J, Wu R, Jiang H, Li J. Unraveling the complex roles of macrophages in obese adipose tissue: an overview. Front Med 2024; 18:205-236. [PMID: 38165533 DOI: 10.1007/s11684-023-1033-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/15/2023] [Indexed: 01/03/2024]
Abstract
Macrophages, a heterogeneous population of innate immune cells, exhibit remarkable plasticity and play pivotal roles in coordinating immune responses and maintaining tissue homeostasis within the context of metabolic diseases. The activation of inflammatory macrophages in obese adipose tissue leads to detrimental effects, inducing insulin resistance through increased inflammation, impaired thermogenesis, and adipose tissue fibrosis. Meanwhile, adipose tissue macrophages also play a beneficial role in maintaining adipose tissue homeostasis by regulating angiogenesis, facilitating the clearance of dead adipocytes, and promoting mitochondrial transfer. Exploring the heterogeneity of macrophages in obese adipose tissue is crucial for unraveling the pathogenesis of obesity and holds significant potential for targeted therapeutic interventions. Recently, the dual effects and some potential regulatory mechanisms of macrophages in adipose tissue have been elucidated using single-cell technology. In this review, we present a comprehensive overview of the intricate activation mechanisms and diverse functions of macrophages in adipose tissue during obesity, as well as explore the potential of drug delivery systems targeting macrophages, aiming to enhance the understanding of current regulatory mechanisms that may be potentially targeted for treating obesity or metabolic diseases.
Collapse
Affiliation(s)
- Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Chen
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Rui Wu
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jia Li
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
9
|
Wang X, Li Y, Pu X, Liu G, Qin H, Wan W, Wang Y, Zhu Y, Yang J. Macrophage-related therapeutic strategies: Regulation of phenotypic switching and construction of drug delivery systems. Pharmacol Res 2024; 199:107022. [PMID: 38043691 DOI: 10.1016/j.phrs.2023.107022] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023]
Abstract
Macrophages, as highly phenotypic plastic immune cells, play diverse roles in different pathological conditions. Changing and controlling the phenotypes of macrophages is considered a novel potential therapeutic intervention. Meanwhile, specific transmembrane proteins anchoring on the surface of the macrophage membrane are relatively conserved, supporting its functional properties, such as inflammatory chemotaxis and tumor targeting. Thus, a series of drug delivery systems related to specific macrophage membrane proteins are commonly used to treat chronic inflammatory diseases. This review summarizes macrophages-based strategies for chronic diseases, discusses the regulation of macrophage phenotypes and their polarization processes, and presents how to design and apply the site-specific targeted drug delivery systems in vivo based on the macrophages and their derived membrane receptors. It aims to provide a better understanding of macrophages in immunoregulation and proposes macrophages-based targeted therapeutic approaches for chronic diseases.
Collapse
Affiliation(s)
- Xi Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Yixuan Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Xueyu Pu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Guiquan Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Honglin Qin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Weimin Wan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Yuying Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Yan Zhu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Jian Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
10
|
Takeuchi T, Kubota T, Nakanishi Y, Tsugawa H, Suda W, Kwon ATJ, Yazaki J, Ikeda K, Nemoto S, Mochizuki Y, Kitami T, Yugi K, Mizuno Y, Yamamichi N, Yamazaki T, Takamoto I, Kubota N, Kadowaki T, Arner E, Carninci P, Ohara O, Arita M, Hattori M, Koyasu S, Ohno H. Gut microbial carbohydrate metabolism contributes to insulin resistance. Nature 2023; 621:389-395. [PMID: 37648852 PMCID: PMC10499599 DOI: 10.1038/s41586-023-06466-x] [Citation(s) in RCA: 144] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes1,2. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance3-9. In particular, carbohydrate metabolism of commensals has been proposed to contribute up to 10% of the host's overall energy extraction10, thereby playing a role in the pathogenesis of obesity and prediabetes3,4,6. Nevertheless, the underlying mechanism remains unclear. Here we investigate this relationship using a comprehensive multi-omics strategy in humans. We combine unbiased faecal metabolomics with metagenomics, host metabolomics and transcriptomics data to profile the involvement of the microbiome in insulin resistance. These data reveal that faecal carbohydrates, particularly host-accessible monosaccharides, are increased in individuals with insulin resistance and are associated with microbial carbohydrate metabolisms and host inflammatory cytokines. We identify gut bacteria associated with insulin resistance and insulin sensitivity that show a distinct pattern of carbohydrate metabolism, and demonstrate that insulin-sensitivity-associated bacteria ameliorate host phenotypes of insulin resistance in a mouse model. Our study, which provides a comprehensive view of the host-microorganism relationships in insulin resistance, reveals the impact of carbohydrate metabolism by microbiota, suggesting a potential therapeutic target for ameliorating insulin resistance.
Collapse
Affiliation(s)
- Tadashi Takeuchi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Division of Diabetes and Metabolism, The Institute for Medical Science Asahi Life Foundation, Tokyo, Japan.
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan.
| | - Yumiko Nakanishi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
| | - Hiroshi Tsugawa
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science (CSRS), Yokohama, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Andrew Tae-Jun Kwon
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Junshi Yazaki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Yoshiki Mochizuki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Toshimori Kitami
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Katsuyuki Yugi
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Institute for Advanced Biosciences, Keio University, Fujisawa, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiko Mizuno
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
- Development Bank of Japan, Tokyo, Japan
| | - Nobutake Yamamichi
- Center for Epidemiology and Preventive Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | | | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Metabolism and Endocrinology, Tokyo Medical University Ibaraki Medical Center, Ami Town, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Erik Arner
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Fondazione Human Technopole, Milan, Italy
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Department of Applied Genomics, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Masahira Hattori
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.
- Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
11
|
Nakadate K, Kawakami K, Yamazaki N. Combined Ingestion of Tea Catechin and Citrus β-Cryptoxanthin Improves Liver Function via Adipokines in Chronic Obesity. Nutrients 2023; 15:3345. [PMID: 37571282 PMCID: PMC10421220 DOI: 10.3390/nu15153345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Recently, there has been an increase in the number of obese individuals, which has elevated the risk of related diseases. Although several studies have been performed to develop a definitive treatment for obesity, no solution has yet been achieved. Recent evidence suggests that tea catechins possess antiobesity effects; however, an impractical amount of catechin may be required to achieve antiobesity effects in humans. Moreover, studies are yet to elucidate the effects of the combined treatment of tea catechins with other substances. Here, we investigated the synergistic effects of catechins and β-cryptoxanthin in high-calorie diet-induced mice. Combined treatment with catechins and β-cryptoxanthin significantly suppressed obesity-induced weight gain and adipocyte size and area, restoring serum parameters to normal. Additionally, combined treatment with catechins and β-cryptoxanthin suppressed inflammatory responses in adipocytes, restored adiponectin levels to normal, protected the liver against obesity-induced damage, and restored normal liver function. Moreover, activin E level was restored to normal, possibly affecting the energy metabolism of brown adipocytes. Overall, these results suggest that the combined ingestion of tea catechins and β-cryptoxanthin was not only effective against obesity but may also help to prevent obesity-related diseases, such as diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
- Kazuhiko Nakadate
- Department of Basic Science, Educational and Research Center for Pharmacy, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan;
| | - Kiyoharu Kawakami
- Department of Basic Science, Educational and Research Center for Pharmacy, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan;
| | - Noriko Yamazaki
- Department of Community Health Care and Sciences, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan;
| |
Collapse
|
12
|
Nawaz A, Fujisaka S, Kado T, Jeelani I, Tobe K. Heterogeneity of adipose tissue-resident macrophages-beyond M1/M2 paradigm. Diabetol Int 2023; 14:125-133. [PMID: 37090127 PMCID: PMC10113418 DOI: 10.1007/s13340-023-00624-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Adipose tissue-resident macrophages (ATMs) are reported to be important for maintaining adipose tissue remodeling and homeostasis. ATMs were classified for the first time in 2007 into the M1 and M2 types. This theory suggests that in the non-obese adipose tissue, the anti-inflammatory, alternatively activated macrophages (AAMs) predominate, and regulate tissue homeostasis, remodeling, and insulin sensitivity. On the other hand, classically activated M1-type macrophages increase rapidly in obesity, secrete inflammatory cytokines, such as TNFα and IL-6, and induce insulin resistance. In recent years, experimental findings that cannot be explained by this theory have been clarified one after another and the theory is being reconsidered. In this review, based on recent findings, we summarize reports on the novel metabolic regulatory functions of ATMs beyond the M1/M2 paradigm.
Collapse
Affiliation(s)
- Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215 USA
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| | - Ishtiaq Jeelani
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, CA USA
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama-Shi, Toyama, 930-0194 Japan
| |
Collapse
|
13
|
Zhang M, Li X, Zhang Q, Yang J, Liu G. Roles of macrophages on ulcerative colitis and colitis-associated colorectal cancer. Front Immunol 2023; 14:1103617. [PMID: 37006260 PMCID: PMC10062481 DOI: 10.3389/fimmu.2023.1103617] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Colitis-associated colorectal cancer is the most serious complication of ulcerative colitis. Long-term chronic inflammation increases the incidence of CAC in UC patients. Compared with sporadic colorectal cancer, CAC means multiple lesions, worse pathological type and worse prognosis. Macrophage is a kind of innate immune cell, which play an important role both in inflammatory response and tumor immunity. Macrophages are polarized into two phenotypes under different conditions: M1 and M2. In UC, enhanced macrophage infiltration produces a large number of inflammatory cytokines, which promote tumorigenesis of UC. M1 polarization has an anti-tumor effect after CAC formation, whereas M2 polarization promotes tumor growth. M2 polarization plays a tumor-promoting role. Some drugs have been shown to that prevent and treat CAC effectively by targeting macrophages.
Collapse
|
14
|
Makhijani P, Basso PJ, Chan YT, Chen N, Baechle J, Khan S, Furman D, Tsai S, Winer DA. Regulation of the immune system by the insulin receptor in health and disease. Front Endocrinol (Lausanne) 2023; 14:1128622. [PMID: 36992811 PMCID: PMC10040865 DOI: 10.3389/fendo.2023.1128622] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
The signaling pathways downstream of the insulin receptor (InsR) are some of the most evolutionarily conserved pathways that regulate organism longevity and metabolism. InsR signaling is well characterized in metabolic tissues, such as liver, muscle, and fat, actively orchestrating cellular processes, including growth, survival, and nutrient metabolism. However, cells of the immune system also express the InsR and downstream signaling machinery, and there is increasing appreciation for the involvement of InsR signaling in shaping the immune response. Here, we summarize current understanding of InsR signaling pathways in different immune cell subsets and their impact on cellular metabolism, differentiation, and effector versus regulatory function. We also discuss mechanistic links between altered InsR signaling and immune dysfunction in various disease settings and conditions, with a focus on age related conditions, such as type 2 diabetes, cancer and infection vulnerability.
Collapse
Affiliation(s)
- Priya Makhijani
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
| | - Paulo José Basso
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yi Tao Chan
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nan Chen
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jordan Baechle
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Saad Khan
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - David Furman
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1, 000 Immunomes Project, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pilar, Argentina
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel A. Winer
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
15
|
Identification of serum metabolome signatures associated with retinal and renal complications of type 2 diabetes. COMMUNICATIONS MEDICINE 2023; 3:5. [PMID: 36624208 PMCID: PMC9829655 DOI: 10.1038/s43856-022-00231-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Type 2 diabetes is a common disease around the world and its major complications are diabetic retinopathy (DR) and diabetic kidney disease (DKD). Persons with type 2 diabetes with complications, especially who have both DR and DKD, have poorer prognoses than those without complications. Therefore, prevention and early identification of the complications of type 2 diabetes are necessary to improve the prognosis of persons with type 2 diabetes. The aim of this study is to identify factors associated with the development of multiple complications of type 2 diabetes. METHODS We profiled serum metabolites of persons with type 2 diabetes with both DR and DKD (N = 141) and without complications (N = 159) using a comprehensive non-targeted metabolomics approach with mass spectrometry. Based on the serum metabolite profiles, case-control comparisons and metabolite set enrichment analysis (MSEA) were performed. RESULTS Here we show that five metabolites (cyclohexylamine, P = 4.5 × 10-6; 1,2-distearoyl-glycero-3-phosphocholine, P = 7.3 × 10-6; piperidine, P = 4.8 × 10-4; N-acetylneuraminic acid, P = 5.1 × 10-4; stearoyl ethanolamide, P = 6.8 × 10-4) are significantly increased in those with the complications. MSEA identifies fatty acid biosynthesis as the type 2 diabetes complications-associated biological pathway (P = 0.0020). CONCLUSIONS Our metabolome analysis identifies the serum metabolite features of the persons with type 2 diabetes with multiple complications, which could potentially be used as biomarkers.
Collapse
|
16
|
Huang Z, Efthymiadou A, Liang N, Fan R, Treuter E. Antagonistic action of GPS2 and KDM1A at enhancers governs alternative macrophage activation by interleukin 4. Nucleic Acids Res 2023; 51:1067-1086. [PMID: 36610795 PMCID: PMC9943668 DOI: 10.1093/nar/gkac1230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/24/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
The Th2 cytokine interleukin 4 (IL4) promotes macrophage differentiation into alternative subtypes and plays important roles in physiology, in metabolic and inflammatory diseases, in cancer and in tissue regeneration. While the regulatory transcription factor networks governing IL4 signaling are already well-characterized, it is currently less understood which transcriptional coregulators are involved and how they operate mechanistically. In this study, we discover that G protein pathway suppressor 2 (GPS2), a core subunit of the HDAC3 corepressor complex assembled by SMRT and NCOR, represses IL4-dependent enhancer activation in mouse macrophages. Our genome-wide and gene-specific characterization revealed that, instead of directly repressing STAT6, chromatin-bound GPS2 cooperates with SMRT and NCOR to antagonize enhancer activation by lysine demethylase 1A (KDM1A, LSD1). Mechanistically, corepressor depletion increased KDM1A recruitment to enhancers linked to IL4-induced genes, accompanied by demethylation of the repressive histone marks H3K9me2/3 without affecting H3K4me1/2, the classic KDM1A substrates for demethylation in other cellular contexts. This in turn caused enhancer and gene activation already in the absence of IL4/STAT6 and sensitized the STAT6-dependent IL4 responsiveness of macrophages. Thus, our work identified with the antagonistic action of a GPS2-containing corepressor complex and the lysine demethylase KDM1A a hitherto unknown epigenetic corepressor-coactivator switching mechanism that governs alternative macrophage activation.
Collapse
Affiliation(s)
- Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Astradeni Efthymiadou
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Ning Liang
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Rongrong Fan
- Correspondence may also be addressed to Rongrong Fan. Tel: +46 8 524 81161;
| | - Eckardt Treuter
- To whom correspondence should be addressed. Tel: +46 8 524 81060;
| |
Collapse
|
17
|
Lucchinetti E, Lou PH, Holtzhauer G, Noureddine N, Wawrzyniak P, Hartling I, Lee M, Strachan E, Clemente-Casares X, Tsai S, Rogler G, Krämer SD, Hersberger M, Zaugg M. Novel lipid emulsion for total parenteral nutrition based on 18-carbon n-3 fatty acids elicits a superior immunometabolic phenotype in a murine model compared with standard lipid emulsions. Am J Clin Nutr 2022; 116:1805-1819. [PMID: 36166844 DOI: 10.1093/ajcn/nqac272] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/22/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND While lipid emulsions in modern formulations for total parenteral nutrition (TPN) provide essential fatty acids and dense calories, they also promote inflammation and immunometabolic disruptions. OBJECTIVES We aimed to develop a novel lipid emulsion for TPN use with superior immunometabolic actions compared with available standard lipid emulsions. METHODS A novel lipid emulsion [Vegaven (VV)] containing 30% of 18-carbon n-3 fatty acids (α-linolenic acid and stearidonic acid) was developed for TPN (VV-TPN) and compared with TPN containing soybean oil-based lipid emulsion (IL-TPN) and fish-oil-based lipid emulsion (OV-TPN). In vivo studies were performed in instrumented male C57BL/6 mice subjected to 7-d TPN prior to analysis of cytokines, indices of whole-body and hepatic glucose metabolism, immune cells, lipid mediators, and mucosal bowel microbiome. RESULTS IL-6 to IL-10 ratios were significantly lower in liver and skeletal muscle of VV-TPN mice when compared with IL-TPN or OV-TPN mice. VV-TPN and OV-TPN each increased hepatic insulin receptor abundance and resulted in similar HOMA-IR values, whereas only VV-TPN increased hepatic insulin receptor substrate 2 and maintained normal hepatic glycogen content, effects that were IL-10-dependent and mediated by glucokinase activation. The percentages of IFN-γ- and IL-17-expressing CD4+ T cells were increased in livers of VV-TPN mice, and liver macrophages exhibited primed phenotypes when compared with IL-TPN. This immunomodulation was associated with successful elimination of the microinvasive bacterium Akkermansia muciniphila from the bowel mucosa by VV-TPN as opposed to standard lipid emulsions. Assay of hepatic lipid mediators revealed a distinct profile with VV-TPN, including increases in 9(S)-hydroxy-octadecatrienoic acid. When co-administered with IL-TPN, hydroxy-octadecatrienoic acids mimicked the VV-TPN immunometabolic phenotype. CONCLUSIONS We here report the unique anti-inflammatory, insulin-sensitizing, and immunity-enhancing properties of a newly developed lipid emulsion designed for TPN use based on 18-carbon n-3 fatty acids.
Collapse
Affiliation(s)
- Eliana Lucchinetti
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Phing-How Lou
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | | | - Nazek Noureddine
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Paulina Wawrzyniak
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ivan Hartling
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Megan Lee
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Erin Strachan
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | | | - Sue Tsai
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Martin Hersberger
- Division of Clinical Chemistry and Biochemistry, Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Zaugg
- Department of Anesthesiology and Pain Medicine and Cardiovascular Research Centre, University of Alberta, Edmonton, Canada.,Department of Pharmacology, University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Ioannidis M, Mahata SK, van den Bogaart G. The immunomodulatory functions of chromogranin A-derived peptide pancreastatin. Peptides 2022; 158:170893. [PMID: 36244579 PMCID: PMC10760928 DOI: 10.1016/j.peptides.2022.170893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Chromogranin A (CgA) is a 439 amino acid protein secreted by neuroendocrine cells. Proteolytic processing of CgA results in the production of different bioactive peptides. These peptides have been associated with inflammatory bowel disease, diabetes, and cancer. One of the chromogranin A-derived peptides is ∼52 amino acid long Pancreastatin (PST: human (h)CgA250-301, murine (m)CgA263-314). PST is a glycogenolytic peptide that inhibits glucose-induced insulin secretion from pancreatic islet β-cells. In addition to this metabolic role, evidence is emerging that PST also has inflammatory properties. This review will discuss the immunomodulatory properties of PST and its possible mechanisms of action and regulation. Moreover, this review will discuss the potential translation to humans and how PST may be an interesting therapeutic target for treating inflammatory diseases.
Collapse
Affiliation(s)
- Melina Ioannidis
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Geert van den Bogaart
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands; Department of Medical Biology and Pathology, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
19
|
Nance SA, Muir L, Lumeng C. Adipose tissue macrophages: Regulators of adipose tissue immunometabolism during obesity. Mol Metab 2022; 66:101642. [PMID: 36402403 PMCID: PMC9703629 DOI: 10.1016/j.molmet.2022.101642] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Adipose tissue macrophages (ATMs) are a well characterized regulator of adipose tissue inflammatory tone. Previously defined by the M1 vs M2 classification, we now have a better understanding of ATM diversity that departs from the old paradigm and reports a spectrum of ATM function and phenotypes in both brown and white adipose tissue. SCOPE OF REVIEW This review provides an updated overview of ATM activation and function, ATM diversity in humans and rodents, and novel ATM functions that contribute to metabolic homeostasis and disease. MAJOR CONCLUSIONS While the paradigm that resident ATMs predominate in the lean state and obesity leads to the accumulation of lipid-associated and inflammatory ATMs still broadly remains rigorously supported, the details of this model continue to be refined and single cell data provide new insight into ATM subtypes and states.
Collapse
Affiliation(s)
- Sierra A. Nance
- Molecular & Integrative Physiology, University of Michigan Medical School, United States,Department of Pediatrics, University of Michigan Medical School, United States
| | - Lindsey Muir
- Computational Medicine and Bioinformatics, University of Michigan Medical School, United States
| | - Carey Lumeng
- Molecular & Integrative Physiology, University of Michigan Medical School, United States,Department of Pediatrics, University of Michigan Medical School, United States,Corresponding author. 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI 48109, United States.
| |
Collapse
|
20
|
Yang X, Xu Y, Gao W, Wang L, Zhao X, Liu G, Fan K, Liu S, Hao H, Qu S, Dong R, Ma X, Ma J. Hyperinsulinemia-induced microglial mitochondrial dynamic and metabolic alterations lead to neuroinflammation in vivo and in vitro. Front Neurosci 2022; 16:1036872. [DOI: 10.3389/fnins.2022.1036872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous studies have demonstrated that type 2 diabetes (T2D) is closely linked to the occurrence of Alzheimer’s disease (AD). Nevertheless, the underlying mechanisms for this association are still unknown. Insulin resistance (IR) hallmarked by hyperinsulinemia, as the earliest and longest-lasting pathological change in T2D, might play an important role in AD. Since hyperinsulinemia has an independent contribution to related disease progressions by promoting inflammation in the peripheral system, we hypothesized that hyperinsulinemia might have an effect on microglia which plays a crucial role in neuroinflammation of AD. In the present study, we fed 4-week-old male C57BL/6 mice with a high-fat diet (HFD) for 12 weeks to establish IR model, and the mice treated with standard diet (SD) were used as control. HFD led to obesity in mice with obvious glucose and lipid metabolism disorder, the higher insulin levels in both plasma and cerebrospinal fluid, and aberrant insulin signaling pathway in the whole brain. Meanwhile, IR mice appeared impairments of spatial learning and memory accompanied by neuroinflammation which was characterized by activated microglia and upregulated expression of pro-inflammatory factors in different brain regions. To clarify whether insulin contributes to microglial activation, we treated primary cultured microglia and BV2 cell lines with insulin in vitro to mimic hyperinsulinemia. We found that hyperinsulinemia not only increased microglial proliferation and promoted M1 polarization by enhancing the production of pro-inflammatory factors, but also impaired membrane translocation of glucose transporter 4 (GLUT4) serving as the insulin-responding glucose transporter in the processes of glucose up-taking, reduced ATP production and increased mitochondrial fission. Our study provides new perspectives and evidence for the mechanism underlying the association between T2D and AD.
Collapse
|
21
|
Lin CW, Hung CM, Chen WJ, Chen JC, Huang WY, Lu CS, Kuo ML, Chen SG. New Horizons of Macrophage Immunomodulation in the Healing of Diabetic Foot Ulcers. Pharmaceutics 2022; 14:pharmaceutics14102065. [PMID: 36297499 PMCID: PMC9606988 DOI: 10.3390/pharmaceutics14102065] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are one of the most costly and troublesome complications of diabetes mellitus. The wound chronicity of DFUs remains the main challenge in the current and future treatment of this condition. Persistent inflammation results in chronic wounds characterized by dysregulation of immune cells, such as M1 macrophages, and impairs the polarization of M2 macrophages and the subsequent healing process of DFUs. The interactive regulation of M1 and M2 macrophages during DFU healing is critical and seems manageable. This review details how cytokines and signalling pathways are co-ordinately regulated to control the functions of M1 and M2 macrophages in normal wound repair. DFUs are defective in the M1-to-M2 transition, which halts the whole wound-healing machinery. Many pre-clinical and clinical innovative approaches, including the application of topical insulin, CCL chemokines, micro RNAs, stem cells, stem-cell-derived exosomes, skin substitutes, antioxidants, and the most recent Phase III-approved ON101 topical cream, have been shown to modulate the activity of M1 and M2 macrophages in DFUs. ON101, the newest clinically approved product in this setting, is designed specifically to down-regulate M1 macrophages and further modulate the wound microenvironment to favour M2 emergence and expansion. Finally, the recent evolution of macrophage modulation therapies and techniques will improve the effectiveness of the treatment of diverse DFUs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming-Liang Kuo
- Microbio Co., Ltd., Taipei 115, Taiwan
- Correspondence: (M.-L.K.); or (S.-G.C.); Tel.: +886-2-27031298 (ext. 550) (M.-L.K.); +886-2-27031098 (ext. 551) (S.-G.C.)
| | - Shyi-Gen Chen
- Oneness Biotech Co., Ltd., Taipei 106, Taiwan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (M.-L.K.); or (S.-G.C.); Tel.: +886-2-27031298 (ext. 550) (M.-L.K.); +886-2-27031098 (ext. 551) (S.-G.C.)
| |
Collapse
|
22
|
Zhao H, Lu J, He F, Wang M, Yan Y, Chen B, Xie D, Xu C, Wang Q, Liu W, Yu W, Xi Y, Yu L, Yamamoto T, Koyama H, Wang W, Zhang C, Cheng J. Hyperuricemia contributes to glucose intolerance of hepatic inflammatory macrophages and impairs the insulin signaling pathway via IRS2-proteasome degradation. Front Immunol 2022; 13:931087. [PMID: 36177037 PMCID: PMC9513153 DOI: 10.3389/fimmu.2022.931087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/24/2022] [Indexed: 02/05/2023] Open
Abstract
AIM Numerous reports have demonstrated the key importance of macrophage-elicited metabolic inflammation in insulin resistance (IR). Our previous studies confirmed that hyperuricemia or high uric acid (HUA) treatment induced an IR state in several peripheral tissues to promote the development of type 2 diabetes mellitus (T2DM). However, the effect of HUA on glucose uptake and the insulin sensitivity of macrophages and its mechanism is unclear. METHODS To assess systemic IR, we generated hyperuricemic mice by urate oxidase knockout (UOX-KO). Then, glucose/insulin tolerance, the tissue uptake of 18F-fluorodeoxyglucose, body composition, and energy balance were assessed. Glucose uptake of circulating infiltrated macrophages in the liver was evaluated by glucose transporter type 4 (GLUT-4) staining. Insulin sensitivity and the insulin signaling pathway of macrophages were demonstrated using the 2-NBDG kit, immunoblotting, and immunofluorescence assays. The immunoprecipitation assay and LC-MS analysis were used to determine insulin receptor substrate 2 (IRS2) levels and its interacting protein enrichment under HUA conditions. RESULTS Compared to WT mice (10 weeks old), serum uric acid levels were higher in UOX-KO mice (WT, 182.3 ± 5.091 μM versus KO, 421.9 ± 45.47 μM). Hyperuricemic mice with metabolic disorders and systemic IR showed inflammatory macrophage recruitment and increased levels of circulating proinflammatory cytokines. HUA inhibited the nuclear translocation of GLUT-4 in hepatic macrophages, restrained insulin-induced glucose uptake and glucose tolerance, and blocked insulin IRS2/PI3K/AKT signaling. Meanwhile, HUA mediated the IRS2 protein degradation pathway and activated AMPK/mTOR in macrophages. LC-MS analysis showed that ubiquitination degradation could be involved in IRS2 and its interacting proteins to contribute to IR under HUA conditions. CONCLUSION The data suggest that HUA-induced glucose intolerance in hepatic macrophages contributed to insulin resistance and impaired the insulin signaling pathway via IRS2-proteasome degradation.
Collapse
Affiliation(s)
- Hairong Zhao
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
| | - Jiaming Lu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Furong He
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
| | - Yunbo Yan
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Binyang Chen
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - De Xie
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chenxi Xu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Qiang Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Weidong Liu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Wei Yu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Yuemei Xi
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Linqian Yu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Tetsuya Yamamoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Wei Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical Research and Development (R&D), College of Pharmacy, Dali University, Dali, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, Xiamen University, Xiamen, China
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
23
|
Yao J, Wu D, Qiu Y. Adipose tissue macrophage in obesity-associated metabolic diseases. Front Immunol 2022; 13:977485. [PMID: 36119080 PMCID: PMC9478335 DOI: 10.3389/fimmu.2022.977485] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue macrophage (ATM) has been appreciated for its critical contribution to obesity-associated metabolic diseases in recent years. Here, we discuss the regulation of ATM on both metabolic homeostatsis and dysfunction. In particular, the macrophage polarization and recruitment as well as the crosstalk between ATM and adipocyte in thermogenesis, obesity, insulin resistance and adipose tissue fibrosis have been reviewed. A better understanding of how ATM regulates adipose tissue remodeling may provide novel therapeutic strategies against obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Jingfei Yao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongmei Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- *Correspondence: Yifu Qiu,
| |
Collapse
|
24
|
Yin F, Wu MM, Wei XL, Ren RX, Liu MH, Chen CQ, Yang L, Xie RQ, Jiang SY, Wang XF, Wang H. Hepatic NCoR1 deletion exacerbates alcohol-induced liver injury in mice by promoting CCL2-mediated monocyte-derived macrophage infiltration. Acta Pharmacol Sin 2022; 43:2351-2361. [PMID: 35149852 PMCID: PMC9433401 DOI: 10.1038/s41401-022-00863-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023]
Abstract
Nuclear receptor corepressor 1 (NCoR1) is a corepressor of the epigenetic regulation of gene transcription that has important functions in metabolism and inflammation, but little is known about its role in alcohol-associated liver disease (ALD). In this study, we developed mice with hepatocyte-specific NCoR1 knockout (NCoR1Hep-/-) using the albumin-Cre/LoxP system and investigated the role of NCoR1 in the pathogenesis of ALD and the underlying mechanisms. The traditional alcohol feeding model and NIAAA model of ALD were both established in wild-type and NCoR1Hep-/- mice. We showed that after ALD was established, NCoR1Hep-/- mice had worse liver injury but less steatosis than wild-type mice. We demonstrated that hepatocyte-specific loss of NCoR1 attenuated liver steatosis by promoting fatty acid oxidation by upregulating BMAL1 (a circadian clock component that has been reported to promote peroxisome proliferator activated receptor alpha (PPARα)-mediated fatty β-oxidation by upregulating de novo lipid synthesis). On the other hand, hepatocyte-specific loss of NCoR1 exacerbated alcohol-induced liver inflammation and oxidative stress by recruiting monocyte-derived macrophages via C-C motif chemokine ligand 2 (CCL2). In the mouse hepatocyte line AML12, NCoR1 knockdown significantly increased ethanol-induced CCL2 release. These results suggest that hepatocyte NCoR1 plays distinct roles in controlling liver inflammation and steatosis, which provides new insights into the development of treatments for steatohepatitis induced by chronic alcohol consumption.
Collapse
Affiliation(s)
- Fan Yin
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Miao-Miao Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Li Wei
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Rui-Xue Ren
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Meng-Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Chong-Qing Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Liu Yang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Rui-Qian Xie
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Shan-Yue Jiang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Xue-Fu Wang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
25
|
The Essential Role of FoxO1 in the Regulation of Macrophage Function. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1068962. [PMID: 35993049 PMCID: PMC9388302 DOI: 10.1155/2022/1068962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
Macrophages are widely distributed in various tissues and organs. They not only participate in the regulation of innate and adaptive immune response, but also play an important role in tissue homeostasis. Dysregulation of macrophage function is closely related to the initiation, development and prognosis of multiple diseases, including infection and tumorigenesis. Forkhead box transcription factor O1 (FoxO1) is an important member among the forkhead box transcription factor family. Through directly binding to the promoter regions of downstream target genes, FoxO1 is implicated in cell proliferation, apoptosis, metabolic activities and other biological processes. In this review, we summarized the regulatory role of FoxO1 in macrophage phagocytosis, migration, differentiation and inflammatory activation. We also emphasized that macrophage reciprocally modulated FoxO1 activity via a post-translational modification (PTM) dominant manner.
Collapse
|
26
|
Iwamoto M, Kubota T, Sakurai Y, Wada N, Shioda S, Yamauchi T, Kadowaki T, Kubota N. The sodium-glucose co-transporter 2 inhibitor tofogliflozin suppresses atherosclerosis through glucose lowering in ApoE-deficient mice with streptozotocin-induced diabetes. Pharmacol Res Perspect 2022; 10:e00971. [PMID: 35707828 PMCID: PMC9201373 DOI: 10.1002/prp2.971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/11/2022] Open
Abstract
Epidemiological and animal studies have revealed that sodium-glucose cotransporter 2 (SGLT2) inhibitors suppress cardiovascular events in subjects with type 2 diabetes and atherosclerosis in animal models of diabetes. However, it still remains unclear if the anti-atherosclerotic effect of SGLT2 inhibitors is entirely dependent on their glucose-lowering effect. Tofogliflozin, a highly specific SGLT2 inhibitor, was administrated to apolipoprotein-E-deficient (ApoEKO) with streptozotocin (STZ)-induced diabetes and nondiabetic ApoEKO mice. After 6 weeks, samples were collected to investigate the histological changes and peritoneal macrophage inflammatory cytokine levels. Tofogliflozin suppressed atherosclerosis in the diabetic ApoEKO mice. The atherosclerosis lesion areas and accumulation of macrophages in these areas were reduced by tofogliflozin treatment. The expression levels of interleukin (IL)-1β and IL-6 in the peritoneal macrophages were significantly suppressed in the tofogliflozin-treated diabetic ApoEKO mice. Tofogliflozin treatment failed to inhibit atherosclerosis in the nondiabetic ApoEKO mice. No significant difference in the anti-atherosclerotic effects of insulin and tofogliflozin was observed between diabetic ApoEKO mice with equivalent degrees of glycemic control achieved with the two treatments. Insulin treatment significantly reduced the IL-1β and IL-6 expression levels in the peritoneal macrophages of the diabetic ApoEKO mice. Significant decrease of the LPS-stimulated IL-1β concentrations was also observed in the conditioned medium of the peritoneal macrophages collected from insulin- and tofogliflozin-treated diabetic ApoEKO mice. These results suggest that tofogliflozin suppresses atherosclerosis by improving glucose intolerance associated with inhibition of inflammation. Tofogliflozin suppresses atherosclerosis in ApoEKO mice with STZ-induced diabetes via its glucose-lowering effect.
Collapse
Affiliation(s)
- Masahiko Iwamoto
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
- Division of Diabetes and MetabolismThe Institute of Medical ScienceAsahi Life FoundationTokyoJapan
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
- Division of Diabetes and MetabolismThe Institute of Medical ScienceAsahi Life FoundationTokyoJapan
- Department of Clinical NutritionNational Institutes of Biomedical InnovationHealth and Nutrition (NIBIOHN)TokyoJapan
- Laboratory for Intestinal EcosystemRIKEN Center for Integrative Medical Sciences (IMS)KanagawaJapan
- Intestinal Microbiota ProjectKanagawa Institute of Industrial Science and Technology EbinaKanagawaJapan
- Division of Cardiovascular MedicineToho University Ohashi Medical CenterTokyoJapan
| | - Yoshitaka Sakurai
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Nobuhiro Wada
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
- Department of Clinical NutritionNational Institutes of Biomedical InnovationHealth and Nutrition (NIBIOHN)TokyoJapan
| | - Seiji Shioda
- Global Research Center for Innovative Life SciencePeptide Drug InnovationSchool of Pharmacy and Pharmaceutical SciencesHoshi UniversityTokyoJapan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
- Toranomon HospitalTokyoJapan
| | - Naoto Kubota
- Department of Diabetes and Metabolic DiseasesGraduate School of MedicineThe University of TokyoTokyoJapan
- Department of Clinical Nutrition TherapyThe University of TokyoTokyoJapan
| |
Collapse
|
27
|
Huang Z, Xiao L, Xiao Y, Chen C. The Modulatory Role of Growth Hormone in Inflammation and Macrophage Activation. Endocrinology 2022; 163:6607489. [PMID: 35695371 DOI: 10.1210/endocr/bqac088] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 11/19/2022]
Abstract
Inflammation is a body's response to remove harmful stimuli and heal tissue damage, which is involved in various physiology and pathophysiology conditions. If dysregulated, inflammation may lead to significant negative impacts. Growth hormone (GH) has been shown responsible for not only body growth but also critical in the modulation of inflammation. In this review, we summarize the current clinical and animal studies about the complex and critical role of GH in inflammation. Briefly, GH excess or deficiency may lead to pathological inflammatory status. In inflammatory diseases, GH may serve as an inflammatory modulator to control the disease progression and promote disease resolution. The detailed mechanisms and signaling pathways of GH on inflammation, with a focus on the modulation of macrophage polarization, are carefully discussed with potential direction for future investigations.
Collapse
Affiliation(s)
- Zhengxiang Huang
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD 4000, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), QUT, Brisbane, QLD 4000, Australia
| | - Lan Xiao
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD 4000, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), QUT, Brisbane, QLD 4000, Australia
| | - Yin Xiao
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD 4000, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), QUT, Brisbane, QLD 4000, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
28
|
Hydrolyzed chicken meat extract boosts the immunoregulatory effect by regulating M1/M2 Macrophage polarization. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
29
|
Macrophages, Low-Grade Inflammation, Insulin Resistance and Hyperinsulinemia: A Mutual Ambiguous Relationship in the Development of Metabolic Diseases. J Clin Med 2022; 11:jcm11154358. [PMID: 35955975 PMCID: PMC9369133 DOI: 10.3390/jcm11154358] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic derangement with poor glycemic control accompanying overweight and obesity is associated with chronic low-grade inflammation and hyperinsulinemia. Macrophages, which present a very heterogeneous population of cells, play a key role in the maintenance of normal tissue homeostasis, but functional alterations in the resident macrophage pool as well as newly recruited monocyte-derived macrophages are important drivers in the development of low-grade inflammation. While metabolic dysfunction, insulin resistance and tissue damage may trigger or advance pro-inflammatory responses in macrophages, the inflammation itself contributes to the development of insulin resistance and the resulting hyperinsulinemia. Macrophages express insulin receptors whose downstream signaling networks share a number of knots with the signaling pathways of pattern recognition and cytokine receptors, which shape macrophage polarity. The shared knots allow insulin to enhance or attenuate both pro-inflammatory and anti-inflammatory macrophage responses. This supposedly physiological function may be impaired by hyperinsulinemia or insulin resistance in macrophages. This review discusses the mutual ambiguous relationship of low-grade inflammation, insulin resistance, hyperinsulinemia and the insulin-dependent modulation of macrophage activity with a focus on adipose tissue and liver.
Collapse
|
30
|
Sun JX, Xu XH, Jin L. Effects of Metabolism on Macrophage Polarization Under Different Disease Backgrounds. Front Immunol 2022; 13:880286. [PMID: 35911719 PMCID: PMC9331907 DOI: 10.3389/fimmu.2022.880286] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
Macrophages are versatile immune cells associated with various diseases, and their phenotypes and functions change on the basis of the surrounding environments. Reprogramming of metabolism is required for the proper polarization of macrophages. This review will focus on basic metabolic pathways, the effects of key enzymes and specific products, relationships between cellular metabolism and macrophage polarization in different diseases and the potential prospect of therapy targeted key metabolic enzymes. In particular, the types and characteristics of macrophages at the maternal-fetal interface and their effects on a successful conception will be discussed.
Collapse
Affiliation(s)
| | | | - Liping Jin
- *Correspondence: Liping Jin, ; Xiang-Hong Xu,
| |
Collapse
|
31
|
Xing J, Chen C. Hyperinsulinemia: beneficial or harmful or both on glucose homeostasis. Am J Physiol Endocrinol Metab 2022; 323:E2-E7. [PMID: 35635329 DOI: 10.1152/ajpendo.00441.2021] [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] [Indexed: 11/22/2022]
Abstract
Insulin, a principal anabolic hormone produced by pancreatic β-cells, has a primary function of storage of nutrients following excessive energy intake. Pre- or early type 2 diabetes stages present hyperinsulinemia (β-cell dysfunction) and insulin resistance. Initiation of hyperinsulinemia is triggered by a loss of first-phase glucose-stimulated insulin secretion with altered membrane ion channel distribution. More factors, including insulin resistance and excessive proliferation of β-cells, deteriorate the hyperinsulinemia, whereas the hyperinsulinemia contributes to further development of insulin resistance and type 2 diabetes; to develop eventually late-stage diabetes with absolute insulin deficiency. In this mini-review, the major focus was put on the causes and pathophysiology of hyperinsulinemia, and the metabolic consequences and current treatment of hyperinsulinemia were discussed. The data used in this narrative review were collected mainly from relevant discoveries in the past 3 years.
Collapse
Affiliation(s)
- JingJing Xing
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
32
|
Chronic Central Leptin Infusion Promotes an Anti-Inflammatory Cytokine Profile Related to the Activation of Insulin Signaling in the Gastrocnemius of Male Rats. Biomedicines 2022; 10:biomedicines10071465. [PMID: 35884769 PMCID: PMC9312815 DOI: 10.3390/biomedicines10071465] [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: 05/30/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 11/16/2022] Open
Abstract
Leptin is involved in the modulation of insulin signaling in peripheral tissues, being closely associated with changes in lipid metabolism. This adipokine modifies inflammatory pathways that can interact with insulin targets in peripheral organs; however, the mechanisms remain unclear. Inflammatory and insulin signaling targets, cytokines, adiponectin, irisin and non-esterified fatty acid (NEFA) levels and enzymes of fatty acid anabolism were studied in the gastrocnemius of chronic centrally infused leptin (L), pair-fed and control rats. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) and c-Jun N-terminal kinase (JNK) was reduced in L rats (59% and 58%, respectively). The phosphorylation of the insulin receptor and Akt and adiponectin and irisin content was increased in L rats (154%, 157%, 308% and 329%, respectively). The levels of glucose-6-phosphate dehydrogenase, the mRNA content of acetyl Co-A carboxylase and NEFA concentrations were diminished in the muscles of L rats (59%, 50% and 61%, respectively). The activation of JNK correlated positively with STAT3 phosphorylation, tumoral necrosis factor-α and NEFA and negatively with irisin and Akt phosphorylation. These data suggest that the activation of insulin signaling targets and a decrease in NEFA content are associated with a reduction in muscle inflammation parameters, suggesting that leptin may integrate these pathways.
Collapse
|
33
|
Possible role of insulin resistance in activation of plasma xanthine oxidoreductase in health check-up examinees. Sci Rep 2022; 12:10281. [PMID: 35717455 PMCID: PMC9206666 DOI: 10.1038/s41598-022-11094-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
We previously found an association of insulin resistance (IR) with plasma xanthine oxidoreductase (XOR) activity in a cross-sectional study. However, whether IR induces increased XOR activity has not been elucidated. This retrospective longitudinal observational study included 347 participants (173 males, 174 females) who underwent annual health examinations and were medication naïve. Homeostasis model assessment of IR (HOMA-IR) index, and physical and laboratory measurements were determined at the baseline. At baseline and 12-month follow-up examinations, plasma XOR activity was determined using our novel assay based on [13C2,15N2] xanthine and liquid chromatography/triple quadrupole mass spectrometry. Subjects with IR, defined as HOMA-IR index ≥ 1.7 (n = 92), exhibited significantly (p < 0.001) higher plasma XOR activity levels than those without IR (n = 255), with an increase in that activity seen in 180 (51.9%) after 12 months. Multivariable linear and logistic regression analyses showed that IR, but not BMI or waist circumference, at baseline was significantly associated with plasma XOR activity (β = 0.094, p = 0.033) and increased plasma XOR activity over the 12-month period (odds ratio, 1.986; 95% confidence interval, 1.048–3.761; p = 0.035), after adjustments for various clinical parameters, including plasma XOR activity at baseline. These results suggest that IR induces increased plasma XOR activity in a manner independent of adiposity.
Collapse
|
34
|
Lee S, Usman TO, Yamauchi J, Chhetri G, Wang X, Coudriet GM, Zhu C, Gao J, McConnell R, Krantz K, Rajasundaram D, Singh S, Piganelli J, Ostrowska A, Soto-Gutierrez A, Monga SP, Singhi AD, Muzumdar RH, Tsung A, Dong HH. Myeloid FoxO1 depletion attenuates hepatic inflammation and prevents nonalcoholic steatohepatitis. J Clin Invest 2022; 132:154333. [PMID: 35700043 PMCID: PMC9282937 DOI: 10.1172/jci154333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis and fibrosis in mice and patients with NASH. Myeloid cell-conditional FoxO1 knockout skewed macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotypes, accompanied by the reduction of macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1 knockout mice on HFD. When fed a NASH-inducing diet, myeloid cell FoxO1 knockout mice were protected from developing NASH, culminating in the reduction of hepatic inflammation, steatosis and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward M1 signatures to perpetuate hepatic inflammation in NASH. FoxO1 appears as a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.
Collapse
Affiliation(s)
- Sojin Lee
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Taofeek O Usman
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jun Yamauchi
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Goma Chhetri
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Xingchun Wang
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Gina M Coudriet
- Department of Surgery, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Cuiling Zhu
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jingyang Gao
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Riley McConnell
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Kyler Krantz
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jon Piganelli
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alina Ostrowska
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Radhika H Muzumdar
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Allan Tsung
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, United States of America
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| |
Collapse
|
35
|
Zhao Q, Luo T, Gao F, Fu Y, Li B, Shao X, Chen H, Zhou Z, Guo S, Shen L, Jin L, Cen D, Zhou H, Lyu J, Fang H. GRP75 Regulates Mitochondrial-Supercomplex Turnover to Modulate Insulin Sensitivity. Diabetes 2022; 71:233-248. [PMID: 34810178 DOI: 10.2337/db21-0173] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022]
Abstract
GRP75 (75-kDA glucose-regulated protein), defined as a major component of both the mitochondrial quality control system and mitochondria-associated membrane, plays a key role in mitochondrial homeostasis. In this study, we assessed the roles of GRP75, other than as a component, in insulin action in both in vitro and in vivo models with insulin resistance. We found that GRP75 was downregulated in mice fed a high-fat diet (HFD) and that induction of Grp75 in mice could prevent HFD-induced obesity and insulin resistance. Mechanistically, GRP75 influenced insulin sensitivity by regulating mitochondrial function through its modulation of mitochondrial-supercomplex turnover rather than mitochondria-associated membrane communication: GRP75 was negatively associated with respiratory chain complex activity and was essential for mitochondrial-supercomplex assembly and stabilization. Moreover, mitochondrial dysfunction in Grp75-knockdown cells might further increase mitochondrial fragmentation, thus triggering cytosolic mtDNA release and activating the cGAS/STING-dependent proinflammatory response. Therefore, GRP75 can serve as a potential therapeutic target of insulin resistant-related diabetes or other metabolic diseases.
Collapse
Affiliation(s)
- Qiongya Zhao
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ting Luo
- Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Feng Gao
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yinxu Fu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Shao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haifeng Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuohua Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sihan Guo
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lijun Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liqin Jin
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dong Cen
- Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| | - Huaibin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianxin Lyu
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
36
|
Cruz-Pineda WD, Garibay-Cerdenares OL, Rodríguez-Ruíz HA, Matia-García I, Marino-Ortega LA, Espinoza-Rojo M, Reyes-Castillo Z, Castro-Alarcón N, Castañeda-Saucedo E, Illades-Aguiar B, Parra-Rojas I. Changes in the Expression of Insulin Pathway, Neutrophil Elastase and Alpha 1 Antitrypsin Genes from Leukocytes of Young Individuals with Insulin Resistance. Diabetes Metab Syndr Obes 2022; 15:1865-1876. [PMID: 35757193 PMCID: PMC9215908 DOI: 10.2147/dmso.s362881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chronic hyperinsulinemia is a hallmark of insulin resistance that affects a diversity of cells, including leukocytes modifying the expression of some genes involved in insulin signaling. PURPOSE The aim of this study was to evaluate how hyperinsulinemia affects the expression of genes involved in the proximal insulin signaling pathway in leukocytes from 45 young individuals grouped: normal weight with not insulin resistance (NIR), with insulin resistance (IR) and with obesity (OB-IR). METHODS qPCR was performed to analyze the expression of insulin receptor (INSR), insulin receptor substrate 1 and 2 (IRS-1 and IRS-2), neutrophil elastase (NE), alpha 1 antitrypsin (A1AT), glucose transporters 1, 3 and 4 (GLUT-1, GLUT-3 and GLUT-4) by the 2-ΔCt method, and the correlation between the genes was determined by Spearman's test. RESULTS The mRNA expression analysis of all genes between NIR and IR individuals revealed no differences. However, when comparing NIR and IR individuals with OB-IR, an increase in NE and A1AT expression and a clear trend towards a decrease in IRS-2 expression was observed, whereas the comparison of IR and OB-IR showed a decrease in GLUT-3 expression. Overall, the correlation analysis showed that in the IR group there was a positive correlation only between NE with IRS-1 (r = 0.72, p = 0.003), while in the OB-IR group, there was a positive correlation between the NE and A1AT with INSR (r = 0.62, p = 0.01 and r = 0.74, p = 0.002, respectively) and with IRS-2 (r = 0.74, p = 0.002 and r = 0.76, p = 0.001, respectively). CONCLUSION These results suggest that hyperinsulinemia and obesity are associated with changes in the expression of genes in leukocytes involved in the insulin pathway that are related to NE and A1AT.
Collapse
Affiliation(s)
- Walter David Cruz-Pineda
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Olga Lilia Garibay-Cerdenares
- CONACyT-Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Olga Lilia Garibay-Cerdenares, CONACyT-Universidad Autónoma de Guerrero, Avenida Lázaro Cárdenas S/N, Ciudad Universitaria, Chilpancingo, Guerrero, CP 39090, México, Tel/Fax +52 7474710901, Email
| | - Hugo Alberto Rodríguez-Ruíz
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Inés Matia-García
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Linda Anahí Marino-Ortega
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Mónica Espinoza-Rojo
- Laboratorio de Biología Molecular y Genómica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Zyanya Reyes-Castillo
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán, Jalisco, México
| | - Natividad Castro-Alarcón
- Laboratorio de Investigación en Microbiología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Investigación en Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Correspondence: Isela Parra-Rojas, Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Avenida Lázaro Cárdenas S/N, Ciudad Universitaria, Chilpancingo, Guerrero, CP 39090, México, Tel/Fax +52 7474719310, Email
| |
Collapse
|
37
|
Barrios V, Campillo-Calatayud A, Guerra-Cantera S, Canelles S, Martín-Rivada Á, Frago LM, Chowen JA, Argente J. Opposite Effects of Chronic Central Leptin Infusion on Activation of Insulin Signaling Pathways in Adipose Tissue and Liver Are Related to Changes in the Inflammatory Environment. Biomolecules 2021; 11:1734. [PMID: 34827732 PMCID: PMC8615824 DOI: 10.3390/biom11111734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Leptin modulates insulin signaling and this involves the Akt pathway, which is influenced by changes in the inflammatory environment and with leptin regulating cytokine synthesis. We evaluated the association between activation of the insulin-signaling pathway and alterations in pro- and anti-inflammatory cytokine levels in inguinal fat and liver of chronic central leptin infused (L), pair-fed (PF), and control rats. Signal transducer and activator of transcription 3 (STAT3) phosphorylation was increased in inguinal fat and reduced in liver of L rats. Phosphorylation of c-Jun N-terminal kinase (JNK) and nuclear factor kappa B (NFkB) was increased in inguinal fat of L rats, together with a pro-inflammatory cytokine profile, while in the liver activation of JNK and NFkB were reduced and an anti-inflammatory pattern was found. Phosphorylation of the insulin receptor, Akt and mechanistic target of rapamycin was decreased in inguinal fat and increased in liver of L rats. There was a direct relationship between pSTAT3 and JNK and a negative correlation of Akt with pSTAT3 and JNK in both tissues. These results indicate that the effects of chronically increased leptin on insulin-related signaling are tissue-specific and suggest that inflammation plays a relevant role in the crosstalk between leptin and insulin signaling.
Collapse
Affiliation(s)
- Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
| | - Ana Campillo-Calatayud
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
| | - Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
| | - Álvaro Martín-Rivada
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, E-28049 Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (A.C.-C.); (S.G.-C.); (S.C.); (Á.M.-R.); (L.M.F.); (J.A.C.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28009 Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, E-28029 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, E-28049 Madrid, Spain
| |
Collapse
|
38
|
Abstract
Acute Respiratory Distress Syndrome is a familiar and destructive clinical condition characterized by progressive, swift and impaired pulmonary state. It leads to mortality if not managed in a timely manner. Recently the role of imbalanced macrophage polarization has been reported in ARDS. Macrophages are known for their heterogeneity and plasticity. Under different microenvironmental stimuli, they (M0) can switch between classically activated macrophage (M1) and alternatively activated (M2) states. This switch is regulated by several signaling pathways and epigenetic changes. In this review, the importance of macrophage M1 and M2 has been discussed in the arena of ARDS citing the phase-wise impact of macrophage polarization. This will provide a further understanding of the molecular mechanism involved in ARDS and will help in developing novel therapeutic targets. Various biomarkers that are currently used concerning this pathophysiological feature have also been summarized.
Collapse
|
39
|
Hiraga Y, Kubota T, Katoh M, Horai Y, Suzuki H, Yamashita Y, Hirata R, Moroi M. AST-120 Treatment Alters the Gut Microbiota Composition and Suppresses Hepatic Triglyceride Levels in Obese Mice. Endocr Res 2021; 46:178-185. [PMID: 34060951 DOI: 10.1080/07435800.2021.1927074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: The prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing worldwide. The existence of a relationship between the microbiota and the pathology of hepatic steatosis is also becoming increasingly clear. AST-120, an oral spherical carbon adsorbent, has been shown to be useful for delaying dialysis initiation and improving uremic symptoms in patients with chronic kidney disease. However, little is known about the effect of AST-120 on fatty liver.Methods: AST-120 (5% w/w) was administrated to 6-week-old male db/db mice for 8 weeks. The body weight, blood glucose and food consumption were examined. Hepatic triglyceride (TG) levels, lipid droplets and epididymal fat cell size were measured. The gut microbiota compositions were investigated in feces and cecum.Results: Significant decreases of the hepatic weight and hepatic TG levels were observed in the AST-120-treated db/db mice. Furthermore, AST-120 treatment was also associated with a decrease of Bacteroidetes, increase of Firmicutes, and a reduced ratio of Bacteroidetes to Firmicutes (B/F ratio) in the feces in the db/db mice. The B/F ratio in the feces was correlated with the liver weight and area of the liver occupied by lipid droplets in the db/db mice.Conclusions: These data suggest that AST-120 treatment alters the composition of the fecal microbiota and suppresses hepatic TG levels in the db/db mice.
Collapse
Affiliation(s)
- Yuki Hiraga
- Department of Cardiovascular Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
- Sohyaku Project Planning & Management Department Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Tokyo, Japan
| | - Tetsuya Kubota
- Faculty of Medicine, Department of Internal Medicine, Division of Cardiovascular Medicine (Ohashi), Toho University, Tokyo, Japan
- Division of Diabetes and Metabolism, The Institute for Medical Science Asahi Life Foundation, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
- Analysis Tool Development Group, Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan
| | - Makoto Katoh
- Naka Kinen Clinic, Ibaraki, Japan
- Research Administration Center, Saitama Medical University, Saitama, Japan
- Department of Cardiology, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Yasushi Horai
- Research Unit/Frontier Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Hiroyuki Suzuki
- Medical Materials Laboratory, Medical Materials Research Laboratories, Kureha Corporation, Fukushima, Japan
| | - Yusuke Yamashita
- Medical Materials Laboratory, Medical Materials Research Laboratories, Kureha Corporation, Fukushima, Japan
| | - Rieko Hirata
- Medical Materials Laboratory, Medical Materials Research Laboratories, Kureha Corporation, Fukushima, Japan
| | - Masao Moroi
- Department of Cardiovascular Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
- Faculty of Medicine, Department of Internal Medicine, Division of Cardiovascular Medicine (Ohashi), Toho University, Tokyo, Japan
- Department of Cardiology, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Saranyutanon S, Acharya S, Deshmukh SK, Khan MA, Singh S, Singh AP. Nicotine causes alternative polarization of macrophages via Src-mediated STAT3 activation: Potential pathobiological implications. J Cell Physiol 2021; 237:1486-1497. [PMID: 34647621 DOI: 10.1002/jcp.30607] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Nicotine is an addictive ingredient of tobacco products and other noncigarette substitutes, including those being used for smoking cessation to relieve withdrawal symptoms. Earlier research, however, has associated nicotine with the risk and poorer outcome of several diseases, including cancer. Macrophages are an important component of the innate immune system and can have both pro-and anti-inflammatory functions depending upon their polarization state. Here, we investigated the effect of nicotine on macrophage polarization, growth, and invasion to understand its role in human physiology. We observed that nicotine induced M2 polarization of RAW264.7 and THP-1-derived macrophages in a dose-dependent manner. Cytokine profiling suggested a mixed M2a/d phenotype of nicotine-polarized macrophages associated with tissue repair and pro-angiogenic functions. Moreover, nicotine treatment also enhanced the growth, motility, and invasion of macrophages. Mechanistic studies revealed increased phosphorylation of STAT3 in nicotine-treated macrophages that was mediated through Src activation. Importantly, pretreatment of macrophages with either Src or STAT3 inhibitor abrogated nicotine-induced macrophage polarization, growth, and motility, suggesting a functional role of the Src-STAT3 signaling axis. Together, our findings reveal a novel role of nicotine in immunosuppression via causing M2 polarization of macrophages that could be implicated in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Sirin Saranyutanon
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Srijan Acharya
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Sachin Kumar Deshmukh
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Mohammad Aslam Khan
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Ajay Pratap Singh
- Department of Pathology, University of South Alabama, Mobile, Alabama, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
41
|
Ross EA, Devitt A, Johnson JR. Macrophages: The Good, the Bad, and the Gluttony. Front Immunol 2021; 12:708186. [PMID: 34456917 PMCID: PMC8397413 DOI: 10.3389/fimmu.2021.708186] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Macrophages are dynamic cells that play critical roles in the induction and resolution of sterile inflammation. In this review, we will compile and interpret recent findings on the plasticity of macrophages and how these cells contribute to the development of non-infectious inflammatory diseases, with a particular focus on allergic and autoimmune disorders. The critical roles of macrophages in the resolution of inflammation will then be examined, emphasizing the ability of macrophages to clear apoptotic immune cells. Rheumatoid arthritis (RA) is a chronic autoimmune-driven spectrum of diseases where persistent inflammation results in synovial hyperplasia and excessive immune cell accumulation, leading to remodeling and reduced function in affected joints. Macrophages are central to the pathophysiology of RA, driving episodic cycles of chronic inflammation and tissue destruction. RA patients have increased numbers of active M1 polarized pro-inflammatory macrophages and few or inactive M2 type cells. This imbalance in macrophage homeostasis is a main contributor to pro-inflammatory mediators in RA, resulting in continual activation of immune and stromal populations and accelerated tissue remodeling. Modulation of macrophage phenotype and function remains a key therapeutic goal for the treatment of this disease. Intriguingly, therapeutic intervention with glucocorticoids or other DMARDs promotes the re-polarization of M1 macrophages to an anti-inflammatory M2 phenotype; this reprogramming is dependent on metabolic changes to promote phenotypic switching. Allergic asthma is associated with Th2-polarised airway inflammation, structural remodeling of the large airways, and airway hyperresponsiveness. Macrophage polarization has a profound impact on asthma pathogenesis, as the response to allergen exposure is regulated by an intricate interplay between local immune factors including cytokines, chemokines and danger signals from neighboring cells. In the Th2-polarized environment characteristic of allergic asthma, high levels of IL-4 produced by locally infiltrating innate lymphoid cells and helper T cells promote the acquisition of an alternatively activated M2a phenotype in macrophages, with myriad effects on the local immune response and airway structure. Targeting regulators of macrophage plasticity is currently being pursued in the treatment of allergic asthma and other allergic diseases. Macrophages promote the re-balancing of pro-inflammatory responses towards pro-resolution responses and are thus central to the success of an inflammatory response. It has long been established that apoptosis supports monocyte and macrophage recruitment to sites of inflammation, facilitating subsequent corpse clearance. This drives resolution responses and mediates a phenotypic switch in the polarity of macrophages. However, the role of apoptotic cell-derived extracellular vesicles (ACdEV) in the recruitment and control of macrophage phenotype has received remarkably little attention. ACdEV are powerful mediators of intercellular communication, carrying a wealth of lipid and protein mediators that may modulate macrophage phenotype, including a cargo of active immune-modulating enzymes. The impact of such interactions may result in repair or disease in different contexts. In this review, we will discuss the origin, characterization, and activity of macrophages in sterile inflammatory diseases and the underlying mechanisms of macrophage polarization via ACdEV and apoptotic cell clearance, in order to provide new insights into therapeutic strategies that could exploit the capabilities of these agile and responsive cells.
Collapse
Affiliation(s)
- Ewan A Ross
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Andrew Devitt
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Jill R Johnson
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
42
|
Zhen Y, Shu W, Hou X, Wang Y. Innate Immune System Orchestrates Metabolic Homeostasis and Dysfunction in Visceral Adipose Tissue During Obesity. Front Immunol 2021; 12:702835. [PMID: 34421909 PMCID: PMC8377368 DOI: 10.3389/fimmu.2021.702835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Arising incidence of metabolic disorders and related diseases caused by obesity is a global health concern. Elucidating the role of the immune system in this process will help to understand the related mechanisms and develop treatment strategies. Here, we have focused on innate immune cells in visceral adipose tissue (VAT) and summarized the roles of these cells in maintaining the homeostasis of VAT. Furthermore, this review reveals the importance of quantitative and functional changes of innate immune cells when the metabolic microenvironment changes due to obesity or excess lipids, and confirms that these changes eventually lead to the occurrence of chronic inflammation and metabolic diseases of VAT. Two perspectives are reviewed, which include sequential changes in various innate immune cells in the steady state of VAT and its imbalance during obesity. Cross-sectional interactions between various innate immune cells at the same time point are also reviewed. Through delineation of a comprehensive perspective of VAT homeostasis in obesity-induced chronic inflammation, and ultimately metabolic dysfunction and disease, we expect to clarify the complex interactive networks among distinct cell populations and propose that these interactions should be taken into account in the development of biotherapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhen
- Department of Dermatology, The First Hospital of Jilin University, Changchun, China
| | - Wentao Shu
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hou
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China.,Institute of Immunology, Jilin University, Changchun, China
| | - Yinan Wang
- Department of Biobank, Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Tourki B, Halade GV. Heart Failure Syndrome With Preserved Ejection Fraction Is a Metabolic Cluster of Non-resolving Inflammation in Obesity. Front Cardiovasc Med 2021; 8:695952. [PMID: 34409075 PMCID: PMC8367012 DOI: 10.3389/fcvm.2021.695952] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an emerging disease with signs of nonresolving inflammation, endothelial dysfunction, and multiorgan defects. Moreover, based on the clinical signs and symptoms and the rise of the obesity epidemic, the number of patients developing HFpEF is increasing. From recent molecular and cellular studies, it becomes evident that HFpEF is not a single and homogenous disease but a cluster of heterogeneous pathophysiology with aging at the base of the pyramid. Obesity superimposed on aging drives the number of inflammatory pathways that intersect with metabolic dysfunction and suboptimal inflammation. Here, we compiled information on obesity-directed macrophage dysfunction that coincide with metabolic defects. Obesity-associated proinflammatory stimuli facilitates heart and interorgan inflammation in HFpEF. Furthermore, diversified mechanisms that drive heart failure urge the need of studying pervasive and unresolved inflammation in animal models to understand HFpEF. A broad and system-based approach will help to study major translational aspects of HFpEF, since no single animal model recapitulates all signs of differential HFpEF stages in the clinical setting. Here, we covered experimental models that target HFpEF and emphasized the advances observed with formyl peptide 2 (FPR2) receptor, a prime sensor that is important in inflammation-resolution signaling. Dysfunction of FPR2 led to the development of spontaneous obesity, impaired macrophage function, and triggered kidney fibrosis, providing evidence of multiorgan defects in HFpEF in an obesogenic aging experimental model.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| |
Collapse
|
44
|
Hayashi T, Kubota T, Mariko I, Takamoto I, Aihara M, Sakurai Y, Wada N, Miki T, Yamauchi T, Kubota N, Kadowaki T. Lack of Brain Insulin Receptor Substrate-1 Causes Growth Retardation, With Decreased Expression of Growth Hormone-Releasing Hormone in the Hypothalamus. Diabetes 2021; 70:1640-1653. [PMID: 33980693 DOI: 10.2337/db20-0482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/07/2021] [Indexed: 11/13/2022]
Abstract
Insulin receptor substrate-1 (Irs1) is one of the major substrates for insulin receptor and insulin-like growth factor-1 (IGF-1) receptor tyrosine kinases. Systemic Irs1-deficient mice show growth retardation, with resistance to insulin and IGF-1, although the underlying mechanisms remain poorly understood. For this study, we generated mice with brain-specific deletion of Irs1 (NIrs1KO mice). The NIrs1KO mice exhibited lower body weights, shorter bodies and bone lengths, and decreased bone density. Moreover, the NIrs1KO mice exhibited increased insulin sensitivity and glucose utilization in the skeletal muscle. Although the ability of the pituitary to secrete growth hormone (GH) remained intact, the amount of hypothalamic growth hormone-releasing hormone (GHRH) was significantly decreased and, accordingly, the pituitary GH mRNA expression levels were impaired in these mice. Plasma GH and IGF-1 levels were also lower in the NIrs1KO mice. The expression levels of GHRH protein in the median eminence, where Irs1 antibody staining is observed, were markedly decreased in the NIrs1KO mice. In vitro, neurite elongation after IGF-1 stimulation was significantly impaired by Irs1 downregulation in the cultured N-38 hypothalamic neurons. In conclusion, brain Irs1 plays important roles in the regulation of neurite outgrowth of GHRH neurons, somatic growth, and glucose homeostasis.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
- Division of Cardiovascular Medicine, Toho University, Ohashi Hospital, Tokyo, Japan
| | - Inoue Mariko
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masakazu Aihara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Sakurai
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Wada
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
| | - Takashi Miki
- Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Tokyo, Japan
- Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Prevention of Diabetes and Lifestyle-Related Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Luo W, Zhou Y, Tang Q, Ai L, Zhang Y. Modulation of TRIB3 and Macrophage Phenotype to Attenuate Insulin Resistance After Downhill Running in Mice. Front Physiol 2021; 12:637432. [PMID: 34177606 PMCID: PMC8220223 DOI: 10.3389/fphys.2021.637432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
Eccentric exercise training accompanied by a low-fat diet can prevent insulin resistance (IR) and is currently an effective method for the treatment of IR induced by high-fat diet (HFD)-associated obesity. However, the molecular mechanisms underlying this improvement of IR in adipose tissue are still not completely clear. In this study, 5–6-week-old male mice were randomly divided into a standard control diet (SCD) group (SC, n = 12) and a HFD group (HF, n = 72). After 12 weeks, 12 mice in each group were randomly sacrificed. The remaining mice in the HF group were randomly submitted to one of the following experimental protocols for 8 weeks: obesity-HFD-sedentary (OHF-Sed, n = 14), obesity-HFD-exercise (OHF-Ex, n = 16), obesity-SCD-sedentary (OSC-Sed, n = 14), and obesity-SCD-exercise (OSC-Ex, n = 16). All obese mice in the exercise group were subjected to downhill running. Half of the mice in each group received an insulin injection (0.75 U/kg) before sample collection. Epididymal fat was removed and weighed. Adipocyte size and inflammatory cell infiltration were observed by H&E staining. Both basal and insulin-stimulated GLUT4 fluorescence and protein contents were detected by immunofluorescence and Western blot. Levels of IL-1β and IL-10 were detected by ELISA. Protein contents of iNOS, Arg-1, TRIB3, p-AKT, and AKT were determined by Western blot. CD86 and CD206 fluorescence were determined by immunofluorescence. The results showed that a HFD for 12 weeks induced IR accompanied by adipose tissue macrophages M1 polarization (increased iNOS protein content and CD86 fluorescence) and TRIB3-AKT activation. Downhill running accompanied by a low-fat diet attenuated IR (p < 0.01), reduced inflammation levels (increased IL-10 protein content and decreased IL-1β protein content), inhibited adipose tissue macrophages M1 polarization (decreased iNOS protein content and CD86 fluorescence) and promoted M2 polarization (increased Arg-1 protein content and CD206 fluorescence), and suppressed TRIB3-AKT signaling. We concluded that downhill running accompanied by dietary fat regulation attenuates HFD-related IR in mice, which may be associated with reduced TRIB3-AKT signaling and activated M2 macrophages in adipose tissue.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China.,Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yue Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Qiang Tang
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| | - Lei Ai
- Jiangsu Research Institute of Sports Science, Nanjing, China
| | - Yuan Zhang
- Department of Sports and Health Sciences, Nanjing Sport Institute, Nanjing, China
| |
Collapse
|
46
|
High-Protein, Low-Glycaemic Meal Replacement Decreases Fasting Insulin and Inflammation Markers-A 12-Month Subanalysis of the ACOORH Trial. Nutrients 2021; 13:nu13051433. [PMID: 33922802 PMCID: PMC8145939 DOI: 10.3390/nu13051433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
Lifestyle interventions, including meal replacement, are effective in the prevention and treatment of type-2-diabetes and obesity. Since insulin is the key weight regulator, we hypothesised that the addition of meal replacement to a lifestyle intervention reduces insulin levels more effectively than lifestyle intervention alone. In the international multicentre randomised controlled ACOORH (Almased Concept against Overweight and Obesity and Related Health Risk) trial, overweight or obese persons who meet the criteria for metabolic syndrome (n = 463) were randomised into two groups. Both groups received nutritional advice focusing on carbohydrate restriction and the use of telemonitoring devices. The intervention group substituted all three main meals per day in week 1, two meals per day in weeks 2–4, and one meal per day in weeks 5–26 with a protein-rich, low-glycaemic meal replacement. Data were collected at baseline and after 1, 3, 6 and 12 months. All datasets providing insulin data (n = 446) were included in this predefined subanalysis. Significantly higher reductions in insulin (−3.3 ± 8.7 µU/mL vs. −1.6 ± 9.8 µU/mL), weight (−6.1 ± 5.2 kg vs. −3.2 ± 4.6 kg), and inflammation markers were observed in the intervention group. Insulin reduction correlated with weight reduction and the highest amount of weight loss (−7.6 ± 4.9 kg) was observed in those participants with an insulin decrease > 2 µU/mL. These results underline the potential for meal replacement-based lifestyle interventions in diabetes prevention, and measurement of insulin levels may serve as an indicator for adherence to carbohydrate restriction.
Collapse
|
47
|
Thibaut R, Gage MC, Pineda-Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non-alcoholic fatty liver disease. FEBS J 2021; 289:3024-3057. [PMID: 33860630 PMCID: PMC9290065 DOI: 10.1111/febs.15877] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/05/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
Non‐alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome, being a common comorbidity of type 2 diabetes and with important links to inflammation and insulin resistance. NAFLD represents a spectrum of liver conditions ranging from steatosis in the form of ectopic lipid storage, to inflammation and fibrosis in nonalcoholic steatohepatitis (NASH). Macrophages that populate the liver play important roles in maintaining liver homeostasis under normal physiology and in promoting inflammation and mediating fibrosis in the progression of NAFLD toward to NASH. Liver macrophages are a heterogenous group of innate immune cells, originating from the yolk sac or from circulating monocytes, that are required to maintain immune tolerance while being exposed portal and pancreatic blood flow rich in nutrients and hormones. Yet, liver macrophages retain a limited capacity to raise the alarm in response to danger signals. We now know that macrophages in the liver play both inflammatory and noninflammatory roles throughout the progression of NAFLD. Macrophage responses are mediated first at the level of cell surface receptors that integrate environmental stimuli, signals are transduced through multiple levels of regulation in the cell, and specific transcriptional programmes dictate effector functions. These effector functions play paramount roles in determining the course of disease in NAFLD and even more so in the progression towards NASH. The current review covers recent reports in the physiological and pathophysiological roles of liver macrophages in NAFLD. We emphasise the responses of liver macrophages to insulin resistance and the transcriptional machinery that dictates liver macrophage function.
Collapse
Affiliation(s)
- Ronan Thibaut
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| | - Matthew C Gage
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Inès Pineda-Torra
- Department of Medicine, Centre for Cardiometabolic and Vascular Science, University College London, UK
| | - Gwladys Chabrier
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| | - Fawaz Alzaid
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, France
| |
Collapse
|
48
|
Lange C, Machado Weber A, Schmidt R, Schroeder C, Strowitzki T, Germeyer A. Changes in protein expression due to metformin treatment and hyperinsulinemia in a human endometrial cancer cell line. PLoS One 2021; 16:e0248103. [PMID: 33690729 PMCID: PMC7943011 DOI: 10.1371/journal.pone.0248103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/21/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of endometrial cancer (EC) has increased over the past years and mainly affects women above the age of 45 years. Metabolic diseases such as obesity and type II diabetes mellitus as well as associated conditions like polycystic ovary syndrome (PCOS), insulin resistance and hyperinsulinemia lead to elevated levels of circulating estrogens. Increased estrogen concentrations, in turn, further trigger the proliferation of endometrial cells and thus promote EC development and progression, especially in the absence of progesterone as seen in postmenopausal women. Elevated blood glucose levels in diabetic patients further contribute to the risk of EC development. Metformin is an insulin-sensitizing biguanide drug, commonly used in the treatment of type II diabetes mellitus, especially in obese patients. Besides its effects on glucose metabolism, metformin displayed anti-cancer effects in various cancer types, including EC. Direct anti-cancer effects of metformin target signaling pathways that are involved in cellular growth and proliferation, e.g. the AKT/PKB/mTOR pathway. Further proteins and pathways have been suggested as potential targets, but the underlying mechanism of action of metformin's anti-cancer activity is still not completely understood. In the present study, the effects of metformin on protein expression were investigated in the human EC cell line HEC-1A using an affinity proteomic approach. Cells were treated with 0.5 mmol/L metformin over a period of 7 days and changes in the expression pattern of 1,300 different proteins were compared to the expression in untreated control cells as well as insulin-treated cells. Insulin treatment (100 ng/mL) was incorporated into the study in order to implement a model for insulin resistance and associated hyperinsulinemia, conditions that are often observed in obese and diabetic patients. Furthermore, the culture medium was supplemented with 10 nmol/L ß-estradiol (E2) during treatments to mimic increased estrogen levels, a common risk factor for EC development. Based on the most prominent and significant changes in expression, a set of 80 proteins was selected and subjected to a more detailed analysis. The data revealed that metformin and insulin targeted similar pathways in the present study and mostly acted on proteins related to proliferation, migration and tumor immune response. These pathways may be affected in a tumor-promoting as well as a tumor-suppressing way by either metformin treatment or insulin supplementation. The consequences for the cells resulting from the detected expression changes were discussed in detail for several proteins. The presented data helps identify potential targets affected by metformin treatment in EC and allows for a better understanding of the mechanism of action of the biguanide drug's anti-cancer activity. However, further investigations are necessary to confirm the observations and conclusions drawn from the presented data after metformin administration, especially for proteins that were regulated in a favorable way, i.e. AKT3, CCND2, CD63, CD81, GFAP, IL5, IL17A, IRF4, PI3, and VTCN1. Further proteins might be of interest, where metformin counteracted unfavorable effects that have been induced by hyperinsulinemia.
Collapse
Affiliation(s)
- Carsten Lange
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Amanda Machado Weber
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | | | | | - Thomas Strowitzki
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Ariane Germeyer
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
49
|
Ratter JM, van Heck JIP, Rooijackers HMM, Jansen HJ, van Poppel PCM, Tack CJ, Stienstra R. Insulin acutely activates metabolism of primary human monocytes and promotes a proinflammatory phenotype. J Leukoc Biol 2021; 110:885-891. [PMID: 33477205 DOI: 10.1002/jlb.3ab0120-019rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022] Open
Abstract
Increased glycolysis is a metabolic trait of activated innate immune cells and supports functional changes including cytokine production. Insulin drives glycolysis in nonimmune cells, yet its metabolic effects on human innate immune cells remain unexplored. Potential effects of insulin on immune cell metabolism may occur acutely after a postprandial increase in plasma insulin levels or as a consequence of chronically elevated insulin levels as observed in obese insulin-resistant individuals and patients with diabetes. Here, we investigated the effects of acute and chronic exposure to insulin on metabolism and function of primary human monocytes. Insulin acutely activated the PI3K/Akt/mTOR pathway in monocytes and increased both oxygen consumption and glycolytic rates. Functionally, acute exposure to insulin increased LPS-induced IL-6 secretion and reactive oxygen species production. To model chronically elevated insulin levels in patients with diabetes, we exposed monocytes from healthy individuals for 24 h to insulin. Although we did not find any changes in expression of metabolic genes that are regulated by insulin in non-immune cells, chronic exposure to insulin increased LPS-induced TNFα production and enhanced MCP-1-directed migration. Supporting this observation, we identified a positive correlation between plasma insulin levels and macrophage numbers in adipose tissue of overweight individuals. Altogether, insulin acutely activates metabolism of human monocytes and induces a shift toward a more proinflammatory phenotype, which may contribute to chronic inflammation in patients with diabetes.
Collapse
Affiliation(s)
- Jacqueline M Ratter
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Julia I P van Heck
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hanne M M Rooijackers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henry J Jansen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pleun C M van Poppel
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
50
|
Liu XL, Pan Q, Cao HX, Xin FZ, Zhao ZH, Yang RX, Zeng J, Zhou H, Fan JG. Lipotoxic Hepatocyte-Derived Exosomal MicroRNA 192-5p Activates Macrophages Through Rictor/Akt/Forkhead Box Transcription Factor O1 Signaling in Nonalcoholic Fatty Liver Disease. Hepatology 2020; 72:454-469. [PMID: 31782176 PMCID: PMC10465073 DOI: 10.1002/hep.31050] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Hepatic macrophages can be activated by many factors such as gut-derived bacterial components and factors released from damaged hepatocytes. Macrophage polarization toward a proinflammatory phenotype (M1) represents an important event in the disease progression of nonalcoholic fatty liver disease (NAFLD). However, the underlying molecular mechanisms remain incompletely understood. Exosomes have been identified as important mediators for cell-cell communication by transferring various biological components such as microRNAs (miRs), proteins, and lipids. The role of exosomes in crosstalk between hepatocytes and macrophages in disease progression of NAFLD is yet to be explored. APPROACH AND RESULTS In the present study, we reported that lipotoxic injury-induced release of hepatocyte exosomes enriched with miR-192-5p played a critical role in the activation of M1 macrophages and hepatic inflammation. Serum miR-192-5p levels in patients with NAFLD positively correlated with hepatic inflammatory activity score and disease progression. Similarly, the serum miR-192-5p level and the number of M1 macrophages, as well as the expression levels of the hepatic proinflammatory mediators, were correlated with disease progression in high-fat high-cholesterol diet-fed rat models. Lipotoxic hepatocytes released more miR-192-5p-enriched exosomes than controls, which induced M1 macrophage (cluster of differentiation 11b-positive [CD11b+ ]/CD86+ ) activation and increase of inducible nitric oxide synthase, interleukin 6, and tumor necrosis factor alpha expression. Furthermore, hepatocyte-derived exosomal miR-192-5p inhibited the protein expression of the rapamycin-insensitive companion of mammalian target of rapamycin (Rictor), which further inhibited the phosphorylation levels of Akt and forkhead box transcription factor O1 (FoxO1) and resulted in activation of FoxO1 and subsequent induction of the inflammatory response. CONCLUSIONS Hepatocyte-derived exosomal miR-192-5p plays a critical role in the activation of proinflammatory macrophages and disease progression of NAFLD through modulating Rictor/Akt/FoxO1 signaling. Serum exosomal miR-192-5p represents a potential noninvasive biomarker and therapeutic target for nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Xiao-Lin Liu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qin Pan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hai-Xia Cao
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Feng-Zhi Xin
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ze-Hua Zhao
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Zeng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huiping Zhou
- Department of Microbiology and Immunology, Medical College of Virginia, Virginia Commonwealth University; McGuire VA Medical Center, Richmond, VA, 23298, USA
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai Key Laboratory of Children’s Digestion and Nutrition, Shanghai 200092, China
| |
Collapse
|