1
|
Chu C, Huang Y, Cao L, Ji S, Zhu B, Shen Q. Role of macrophages in peritoneal dialysis-associated peritoneal fibrosis. Ren Fail 2025; 47:2474203. [PMID: 40044628 PMCID: PMC11884102 DOI: 10.1080/0886022x.2025.2474203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Peritoneal dialysis (PD) can be used as renal replacement therapy when chronic kidney disease (CKD) progresses to end-stage renal disease. However, peritoneal fibrosis (PF) is a major cause of PD failure. Studies have demonstrated that PD fluid contains a significantly larger numbers of macrophages compared with the healthy individuals. During PD, macrophages can secrete cytokines to keep peritoneal tissue in sustained low-grade inflammation, and participate in the regulation of fibrosis-related signaling pathways, such as NF-κB, TGF-β/Smad, IL4/STAT6, and PI3K/AKT. A series of basic pathological changes occurs in peritoneal tissues, including epithelial mesenchymal transformation, overgeneration of neovasculature, and abnormal deposition of extracellular matrix. This review focuses on the role of macrophages in promoting PF during PD, summarizes the targets of macrophage-related inhibition of fibrosis, and provides new ideas for clinical research on delaying PF, maintaining the function and integrity of peritoneum, prolonging duration of PD as a renal replacement modality, and achieving longer survival in CKD patients.
Collapse
Affiliation(s)
- Chenling Chu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Huang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Public Health and Preventive Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luxi Cao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Shuiyu Ji
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Quanquan Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
| |
Collapse
|
2
|
He H, Zhu Y, Ji X, Pu S, Zheng H. The miR-22-5p/Clec4e axis has diagnostic potential in fructose-induced nonalcoholic fatty liver disease. Biochem Biophys Res Commun 2025; 753:151496. [PMID: 39978254 DOI: 10.1016/j.bbrc.2025.151496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/29/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is significantly influenced by microRNAs in its development and progression. This study aimed to identify microRNA profiles and RNA regulatory networks for NAFLD intervention. Mice were fed a high-fructose diet (HFrD) to induce NAFLD. Small RNA-seq and mRNA-seq were used to analyze liver microRNA and mRNA profiles of HFrD-fed versus normal chow-fed (Chow) mice. The differentially expressed genes (DEGs) and miRNAs (DE-miRNAs) were identified, followed by enrichment analysis. A protein‒protein interaction network of overlapping DEGs and DE-miRNA targets was constructed, along with a competing endogenous RNA (ceRNA) network. Mendelian randomization (MR) was performed to verify the causal relationship between top DEGs and NAFLD. The study identified 13 DE-miRNAs and 854 DEGs in the liver between HFrD mice and Chow mice. A Venn diagram revealed that 58 of the predicted target genes of the 13 DE-miRNAs were shared with the DEGs. Finally, 6 DE-miRNAs, 34 DEGs, and 20 predicted lncRNAs were selected to construct the ceRNA regulatory network. The upregulated DEG Clec4e, a target gene of miR-22-5p, was significantly correlated with the risk of NAFLD (OR: 1.41, 95 % CI: 1.04-1.92, P = 0.029) in the MR analysis, and RT-qPCR was applied to validate Clec4e expression in the livers of HFrD mice. Further, the dual-luciferase reporter assay confirmed that miR-22-5p could directly inhibit Clec4e expression by targeting its 3'-UTR. This study identified several novel miRNAs and genes as potential biomarkers of NAFLD. In particular, the miR-22-5p/Clec4e axis is a potential diagnostic target for NAFLD.
Collapse
Affiliation(s)
- Haidong He
- Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Yifan Zhu
- Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Xiaoguo Ji
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Suying Pu
- Shanghai Xuhui District Dahua Hospital, Shanghai, 200237, China.
| | - Hui Zheng
- Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
3
|
Jang JH, Sung JH, Huh JY. Diverse Functions of Macrophages in Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease: Bridging Inflammation and Metabolism. Immune Netw 2025; 25:e12. [PMID: 40078789 PMCID: PMC11896663 DOI: 10.4110/in.2025.25.e12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Macrophages play crucial roles in immune response and tissue homeostasis, with their functions becoming increasingly complex in obesity-mediated metabolic disorders. This review explores the extensive range of macrophage activities within adipose and liver tissues, emphasizing their contribution to the pathogenesis and progression of obesity and its related metabolic dysfunction-associated steatotic liver disease (MASLD). In the context of obesity, macrophages respond adaptively to lipid overloads and inflammatory cues in adipose tissue, profoundly influencing insulin resistance and metabolic homeostasis. Concurrently, their role in the liver extends to moderating inflammation and orchestrating fibrotic responses, integral to the development of MASLD. Highlighting the spectrum of macrophage phenotypes across these metabolic landscapes, we summarize their diverse roles in linking inflammatory processes with metabolic functions. This review advocates for a deeper understanding of macrophage subsets in metabolic tissues, proposing targeted research to harness their therapeutic potential in mitigating MASLD and other metabolic disorders.
Collapse
Affiliation(s)
- Jun Hee Jang
- Department of Life Science, Sogang University, Seoul 04107, Korea
- Center for Nano Materials, Sogang University, Seoul 04107, Korea
| | - Jin Hyun Sung
- Department of Life Science, Sogang University, Seoul 04107, Korea
- Center for Nano Materials, Sogang University, Seoul 04107, Korea
| | - Jin Young Huh
- Department of Life Science, Sogang University, Seoul 04107, Korea
- Center for Nano Materials, Sogang University, Seoul 04107, Korea
| |
Collapse
|
4
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024; 25:5239-5264. [PMID: 39482490 PMCID: PMC11624271 DOI: 10.1038/s44319-024-00296-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
5
|
Li L, Ren J, Guo M, An Z, Duan W, Lv J, Tan Z, Yang J, Zhu Y, Yang H, Liu Y, Ma Y, Guo H. SAP130 mediates crosstalk between hepatocyte ferroptosis and M1 macrophage polarization in PFOS-induced hepatotoxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175612. [PMID: 39163934 DOI: 10.1016/j.scitotenv.2024.175612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant widely utilized in industrial manufacturing and daily life, leading to significant environmental accumulation and various public health issues. This study aims to characterize spliceosome-associated protein 130 (SAP130) as a key mediator of crosstalk between hepatocytes and macrophages, elucidating its role in PFOS-induced liver inflammation. The data demonstrate that PFOS exposure induces ferroptosis in mouse liver and AML12 cells. During ferroptosis, SAP130 is released from injured hepatocytes into the microenvironment, binding to macrophage-inducible C-type lectin (Mincle) and activating the Mincle/Syk signaling pathway in macrophages, ultimately promoting M1 polarization and exacerbating liver injury. Treatment with the ferroptosis inhibitor Ferrostatin-1 reduces SAP130 release, inhibits Mincle/Syk signaling activation, and mitigates inflammatory response. Furthermore, siSAP130 suppresses the activation of the Mincle signaling pathway and M1 polarization in BMDM cells. Conversely, treatment with the ferroptosis agonist Erastin enhances paracrine secretion of SAP130 and exacerbates inflammation. These findings emphasize the significance of hepatocyte-macrophage crosstalk as a critical pathway for PFOS-induced liver injury in mice while highlighting SAP130 as a pivotal regulator of ferroptosis and inflammation, thereby elucidating the potential mechanism of PFOS-induced liver injury.
Collapse
Affiliation(s)
- Longfei Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Mingmei Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Ziwen An
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Wenjing Duan
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Junli Lv
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Zhenzhen Tan
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jing Yang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yiming Zhu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Huiling Yang
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China
| | - Yi Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China.
| | - Huicai Guo
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China.
| |
Collapse
|
6
|
Ishihara T, Kato K, Matsumoto K, Tanaka M, Hara A, Shiraki Y, Morisaki H, Urano Y, Ando R, Ito K, Mii S, Esaki N, Furuhashi K, Takefuji M, Suganami T, Murohara T, Enomoto A. Meflin/ISLR is a marker of adipose stem and progenitor cells in mice and humans that suppresses white adipose tissue remodeling and fibrosis. Genes Cells 2024; 29:902-920. [PMID: 39136356 PMCID: PMC11555626 DOI: 10.1111/gtc.13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024]
Abstract
Identifying specific markers of adipose stem and progenitor cells (ASPCs) in vivo is crucial for understanding the biology of white adipose tissues (WAT). PDGFRα-positive perivascular stromal cells represent the best candidates for ASPCs. This cell lineage differentiates into myofibroblasts that contribute to the impairment of WAT function. However, ASPC marker protein(s) that are functionally crucial for maintaining WAT homeostasis are unknown. We previously identified Meflin as a marker of mesenchymal stem cells (MSCs) in bone marrow and tissue-resident perivascular fibroblasts in various tissues. We also demonstrated that Meflin maintains the undifferentiated status of MSCs/fibroblasts. Here, we show that Meflin is expressed in WAT ASPCs. A lineage-tracing experiment showed that Meflin+ ASPCs proliferate in the WAT of obese mice induced by a high-fat diet (HFD), while some of them differentiate into myofibroblasts or mature adipocytes. Meflin knockout mice fed an HFD exhibited a significant fibrotic response as well as increases in adipocyte cell size and the number of crown-like structures in WAT, accompanied by impaired glucose tolerance. These data suggested that Meflin expressed by ASPCs may have a role in reducing disease progression associated with WAT dysfunction.
Collapse
Grants
- 22ck0106779h0001 Japan Agency for Medical Research and Development
- 23gm1210009s0105 Japan Agency for Medical Research and Development
- 20H03467 Ministry of Education, Culture, Sports, Science and Technology
- 22H02848 Ministry of Education, Culture, Sports, Science and Technology
- 22K18390 Ministry of Education, Culture, Sports, Science and Technology
- Japan Agency for Medical Research and Development
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Toshikazu Ishihara
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Katsuhiro Kato
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kotaro Matsumoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Miyako Tanaka
- Department of ImmunometabolismNagoya University Graduate School of MedicineNagoyaJapan
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental Medicine, Nagoya UniversityNagoyaJapan
| | - Akitoshi Hara
- Center for Cardiovascular Research, University of Hawaii at ManoaHonoluluHawaiiUSA
| | - Yukihiro Shiraki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Hidenori Morisaki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yuya Urano
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Ryota Ando
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kisuke Ito
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Shinji Mii
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Nobutoshi Esaki
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Kazuhiro Furuhashi
- Department of NephrologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Mikito Takefuji
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Takayoshi Suganami
- Department of ImmunometabolismNagoya University Graduate School of MedicineNagoyaJapan
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental Medicine, Nagoya UniversityNagoyaJapan
| | - Toyoaki Murohara
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced StudyGifuJapan
| |
Collapse
|
7
|
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
|
8
|
Adhikary K, Sarkar R, Maity S, Sadhukhan I, Sarkar R, Ganguly K, Barman S, Maiti R, Chakraborty S, Chakraborty TR, Bagchi D, Banerjee P. Immunomodulation of Macrophages in Diabetic Wound Individuals by Structurally Diverse Bioactive Phytochemicals. Pharmaceuticals (Basel) 2024; 17:1294. [PMID: 39458935 PMCID: PMC11510503 DOI: 10.3390/ph17101294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetes-related ulcers and slow-healing wounds pose a significant health risk to individuals due to their uncertain causes. Mortality rates for diabetes foot ulcers (DFUs) range from 10% after 16 months to 24% after five years. The use of bioactive phytochemicals can play a key role in healing wounds in a predictable time. Recent literature has demonstrated that various natural substances, including flavonoids, saponins, phenolic compounds, and polysaccharides, play key roles at different stages of the wound-healing process through diverse mechanisms. These studies have categorized the compounds according to their characteristics, bioactivities, and modes of action. In this study, we evaluated the role of natural compounds derived from plant sources that have been shown to play a crucial role in immunomodulation. Macrophages are closely involved in immunomodulation within the wound microenvironment and are key players in efferocytosis, inflammation resolution, and tissue regeneration, all of which contribute to successful wound healing. Phytochemicals and their derivatives have shown capabilities in immune regulation, including macrophage migration, nitric oxide synthase inhibition, lymphocyte and T-cell stimulation, cytokine activation, natural killer cell enhancement, and the regulation of NF-κβ, TNF-α, and apoptosis. In this review, we have studied the role of phytochemicals in immunomodulation for the resolution of diabetic wound inflammation.
Collapse
Affiliation(s)
- Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Khurda 752050, Odisha, India;
| | - Riya Sarkar
- Department of Medical Lab Technology, Dr. B. C. Roy Academy of Professional Courses, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Sriparna Maity
- Department of Medical Lab Technology, Dr. B. C. Roy Academy of Professional Courses, Bidhannagar, Durgapur 713212, West Bengal, India
| | - Ishani Sadhukhan
- Department of Food Processing, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, West Bengal, India
| | - Riya Sarkar
- Department of Medical Lab Technology & Biotechnology, Paramedical College Durgapur, Durgapur 713212, West Bengal, India
| | - Krishnendu Ganguly
- Department of Medical Lab Technology & Biotechnology, Paramedical College Durgapur, Durgapur 713212, West Bengal, India
| | - Saurav Barman
- Department of Soil Science, Centurion University of Technology and Management, Paralakhemundi 761211, Odisha, India
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura 722101, West Bengal, India;
| | - Sanjoy Chakraborty
- Department of Biological Sciences, New York City College of Technology, City University of New York (CUNY), Brooklyn, NY 11201, USA
| | - Tandra R. Chakraborty
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, NY 11530, USA
| | - Debasis Bagchi
- Department of Biology, College of Arts and Sciences, Adelphi University, Garden City, NY 11530, USA
- Department of Psychology, Gordon F. Derner School of Psychology, Adelphi University, Garden City, NY 11530, USA
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Pradipta Banerjee
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
Zou Y, Li J, Su H, Dechsupa N, Liu J, Wang L. Mincle as a potential intervention target for the prevention of inflammation and fibrosis (Review). Mol Med Rep 2024; 29:103. [PMID: 38639174 PMCID: PMC11058355 DOI: 10.3892/mmr.2024.13227] [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: 11/23/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024] Open
Abstract
Macrophage‑inducible C‑type lectin receptor (Mincle) is predominantly found on antigen‑presenting cells. It can recognize specific ligands when stimulated by certain pathogens such as fungi and Mycobacterium tuberculosis. This recognition triggers the activation of the nuclear factor‑κB pathway, leading to the production of inflammatory factors and contributing to the innate immune response of the host. Moreover, Mincle identifies lipid damage‑related molecules discharged by injured cells, such as Sin3‑associated protein 130, which triggers aseptic inflammation and ultimately hastens the advancement of renal damage, autoimmune disorders and malignancies by fostering tissue inflammation. Presently, research on the functioning of the Mincle receptor in different inflammatory and fibrosis‑associated conditions has emerged as a popular topic. Nevertheless, there remains a lack of research on the impact of Mincle in promoting long‑lasting inflammatory reactions and fibrosis. Additional investigation is required into the function of Mincle receptors in chronological inflammatory reactions and fibrosis of organ systems, including the progression from inflammation to fibrosis. Hence, the present study showed an overview of the primary roles and potential mechanism of Mincle in inflammation, fibrosis, as well as the progression of inflammation to fibrosis. The aim of the present study was to clarify the potential mechanism of Mincle in inflammation and fibrosis and to offer perspectives for the development of drugs that target Mincle.
Collapse
Affiliation(s)
- Yuanxia Zou
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50000, Thailand
- Department of Newborn Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jianchun Li
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50000, Thailand
| | - Hongwei Su
- Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50000, Thailand
| | - Jian Liu
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Wang
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
10
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
11
|
Engin AB. Message Transmission Between Adipocyte and Macrophage in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:273-295. [PMID: 39287855 DOI: 10.1007/978-3-031-63657-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity is characterized by the chronic low-grade activation of the innate immune system. In this respect, macrophage-elicited metabolic inflammation and adipocyte-macrophage interaction have primary importance in obesity. Large quantity of macrophages is accumulated by different mechanisms in obese adipose tissue. Hypertrophic adipocyte-derived chemotactic monocyte chemoattractant protein-1 (MCP-1)/C-C chemokine receptor 2 (CCR2) pathway promotes more macrophage accumulation into the obese adipose tissue. However, obesity-induced changes in adipose tissue macrophage density are mainly dependent on increases in the triple-positive cluster of differentiation (CD)11b+ F4/80+ CD11c+ adipose tissue macrophage subpopulation. As epigenetic regulators, microRNAs (miRNAs) are one of the most important mediators of obesity. miRNAs are expressed by adipocytes as well as macrophages and regulate inflammation with the expression of target genes. A paracrine loop involving free fatty acids and tumor necrosis factor-alpha (TNF-α) between adipocytes and macrophages establishes a vicious cycle that aggravates inflammatory changes in the adipose tissue. Adipocyte-specific caspase-1 and production of interleukin-1beta (IL-1β) by macrophages; both adipocyte and macrophage induction by toll-like receptor-4 (TLR4) through nuclear factor-kappaB (NF-κB) activation; free fatty acid-induced and TLR-mediated activation of c-Jun N-terminal kinase (JNK)-related pro-inflammatory pathways in CD11c+ immune cells; are effective in mutual message transmission between adipocyte and macrophage and in the development of adipose tissue inflammation. Thus, the metabolic status of adipocytes and their released exosomes are important determinants of macrophage inflammatory output. However, old adipocytes are removed by macrophages through trogocytosis or sending an "eat me" signal. As a single miRNA can be able to regulate a variety of target genes and signaling pathways, reciprocal transfer of miRNAs between adipocytes and macrophages via miRNA-loaded exosomes reorganizes the different stages of obesity. Changes in the expression of circulating miRNAs because of obesity progression or anti-obesity treatment indicate that miRNAs could be used as potential biomarkers. Therefore, it is believed that targeting macrophage-associated miRNAs with anti-obesity miRNA-loaded nano-carriers may be successful in the attenuation of both obesity and adipose tissue inflammation in clinical practice. Moreover, miRNA-containing exosomes and transferable mitochondria between the adipocyte and macrophage are investigated as new therapeutic targets for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
12
|
Hatano M, Akiyama Y, Shimada S, Yagi K, Akahoshi K, Itoh M, Tanabe M, Ogawa Y, Tanaka S. Loss of KDM6B epigenetically confers resistance to lipotoxicity in nonalcoholic fatty liver disease-related HCC. Hepatol Commun 2023; 7:e0277. [PMID: 37782459 PMCID: PMC10545410 DOI: 10.1097/hc9.0000000000000277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/09/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND NAFLD caused by abnormalities in hepatic lipid metabolism is associated with an increased risk of developing HCC. The molecular mechanisms underlying the progression of NAFLD-related HCC are not fully understood. We investigated the molecular mechanism and role of KDM6B downregulation in NAFLD-related HCC after the KDM6B gene was identified using microarray analysis as commonly downregulated in mouse NAFLD-related HCC and human nonhepatitis B and nonhepatitis C viral-HCC. METHODS The 5-hydroxymethylcytosine levels of KDM6B in HCC cells were determined using glycosylated hydroxymethyl-sensitive PCR. Microarray and chromatin immunoprecipitation analyses using KDM6B-knockout (KO) cells were used to identify KDM6B target genes. Lipotoxicity was assessed using a palmitate-treated cell proliferation assay. Immunohistochemistry was used to evaluate KDM6B expression in human HCC tissues. RESULTS KDM6B expression levels in HCC cells correlated with the 5-hydroxymethylcytosine levels in the KDM6B gene body region. Gene set enrichment analysis revealed that the lipid metabolism pathway was suppressed in KDM6B-KO cells. KDM6B-KO cells acquired resistance to lipotoxicity (p < 0.01) and downregulated the expression of G0S2, an adipose triglyceride lipase/patatin like phospholipase domain containing 2 (ATGL/PNPLA2) inhibitor, through increased histone H3 lysine-27 trimethylation levels. G0S2 knockdown in KDM6B-expressed HCC cells conferred lipotoxicity resistance, whereas ATGL/PNPLA2 inhibition in the KDM6B-KO cells reduced these effects. Immunohistochemistry revealed that KDM6B expression was decreased in human NAFLD-related HCC tissues (p < 0.001), which was significantly associated with decreased G0S2 expression (p = 0.032). CONCLUSIONS KDM6B-disrupted HCC acquires resistance to lipotoxicity via ATGL/PNPLA2 activation caused by epigenetic downregulation of G0S2 expression. Reduced KDM6B and G0S2 expression levels are common in NAFLD-related HCC. Targeting the KDM6B-G0S2-ATGL/PNPLA2 pathway may be a useful therapeutic strategy for NAFLD-related HCC.
Collapse
Affiliation(s)
- Megumi Hatano
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohei Yagi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichi Akahoshi
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiko Itoh
- Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
13
|
Valenzuela PL, Carrera-Bastos P, Castillo-García A, Lieberman DE, Santos-Lozano A, Lucia A. Obesity and the risk of cardiometabolic diseases. Nat Rev Cardiol 2023; 20:475-494. [PMID: 36927772 DOI: 10.1038/s41569-023-00847-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/18/2023]
Abstract
The prevalence of obesity has reached pandemic proportions, and now approximately 25% of adults in Westernized countries have obesity. Recognized as a major health concern, obesity is associated with multiple comorbidities, particularly cardiometabolic disorders. In this Review, we present obesity as an evolutionarily novel condition, summarize the epidemiological evidence on its detrimental cardiometabolic consequences and discuss the major mechanisms involved in the association between obesity and the risk of cardiometabolic diseases. We also examine the role of potential moderators of this association, with evidence for and against the so-called 'metabolically healthy obesity phenotype', the 'fatness but fitness' paradox or the 'obesity paradox'. Although maintenance of optimal cardiometabolic status should be a primary goal in individuals with obesity, losing body weight and, particularly, excess visceral adiposity seems to be necessary to minimize the risk of cardiometabolic diseases.
Collapse
Affiliation(s)
- Pedro L Valenzuela
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain.
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain.
| | - Pedro Carrera-Bastos
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Alejandro Santos-Lozano
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain
- Department of Health Sciences, European University Miguel de Cervantes, Valladolid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
| |
Collapse
|
14
|
Mikami T, Furuhashi M, Numaguchi R, Hosaka I, Sakai A, Tanaka M, Ito T, Maeda T, Sakurada T, Muraki S, Yanase Y, Sato H, Fukada J, Tamiya Y, Iba Y, Kawaharada N. Comparison of Phenotypes in Subcutaneous Fat and Perivascular Adipose Tissue Surrounding the Saphenous Vein in Coronary Artery Bypass Grafting. Circ J 2023; 87:791-798. [PMID: 36740256 DOI: 10.1253/circj.cj-22-0740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The saphenous vein (SV) is used as an essential conduit in coronary artery bypass grafting (CABG), but the long-term patency of SV grafts is a crucial issue. The use of the novel "no-touch" technique of harvesting the SV together with its surrounding tissue has been reported to result in good long-term graft patency of SV grafts. We recently showed that perivascular adipose tissue (PVAT) surrounding the SV (SV-PVAT) had lower levels of metaflammation and consecutive adipose tissue remodeling than did PVAT surrounding the coronary artery. However, the difference between SV-PVAT and subcutaneous adipose tissue (SCAT) remains unclear. METHODS AND RESULTS Fat pads were sampled from 55 patients (38 men, 17 women; mean [±SD] age 71±8 years) with coronary artery disease who underwent elective CABG. Adipocyte size was significantly larger in SV-PVAT than SCAT. The extent of fibrosis was smaller in SV-PVAT than SCAT. There were no significant differences between SCAT and SV-PVAT in macrophage infiltration area, quantified by antibodies for CD68, CD11c, and CD206, or in gene expression levels of metaflammation-related markers. Expression patterns of adipocyte developmental and pattern-forming genes differed between SCAT and SV-PVAT. CONCLUSIONS The properties of SV-PVAT are close to, but not the same as, those of SCAT, possibly resulting from inherent differences in adipocytes. SV-PVAT has healthy expansion with less fibrosis in fat than SCAT.
Collapse
Affiliation(s)
- Takuma Mikami
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
- Department of Cardiovascular Surgery, National Hospital Organization, Obihiro Hospital
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Ryosuke Numaguchi
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
| | - Itaru Hosaka
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
| | - Akiko Sakai
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Toshiro Ito
- Department of Cardiovascular Surgery, Hokkaido Ohno Memorial Hospital
| | - Toshiyuki Maeda
- Department of Cardiovascular Surgery, Sapporo Central Hospital
| | - Taku Sakurada
- Department of Cardiovascular Surgery, Sapporo Central Hospital
| | - Satoshi Muraki
- Department of Cardiovascular Surgery, Sapporo Central Hospital
| | - Yousuke Yanase
- Department of Cardiovascular Surgery, Teine Keijinkai Hospital
| | - Hiroshi Sato
- Department of Cardiovascular Surgery, Otaru City General Hospital
| | - Joji Fukada
- Department of Cardiovascular Surgery, Otaru City General Hospital
| | - Yukihiko Tamiya
- Department of Cardiovascular Surgery, Otaru City General Hospital
| | - Yutaka Iba
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
| | - Nobuyoshi Kawaharada
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
| |
Collapse
|
15
|
Tan RZ, Zhong X, Han RY, Xie KH, Jia J, Yang Y, Cheng M, Yang CY, Lan HY, Wang L. Macrophages mediate psoriasis via Mincle-dependent mechanism in mice. Cell Death Discov 2023; 9:140. [PMID: 37117184 PMCID: PMC10147944 DOI: 10.1038/s41420-023-01444-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023] Open
Abstract
Psoriasis is currently considered to be an immune and inflammatory disease characterized by massive immune cells infiltration including macrophages. It has been reported that macrophage-inducible C-type lectin (Mincle) is essential to maintain the pro-inflammatory phenotype of M1 macrophages, however, its role and mechanisms in psoriasis remain largely unknown. A model of psoriasis was induced in mice by a daily topical application of imiquimod for 7 days. Role and mechanisms of Mincle in macrophage-mediated psoriasis were investigated in clodronate liposomes induced macrophage depletion mice followed by adoptively transferring with Mincle-expressing or -knockout (KO) macrophages, and in macrophage specific Mincle knockout mice (Mincleloxp/loxp/Lyz2-cre+/+). Finally, a Mincle neutralizing antibody was employed to the psoriasis mice to reveal the therapeutic potential for psoriasis by targeting Mincle. Mincle was highly expressed by M1 macrophages in the skin lesions of patients and mice with psoriasis. Clodronate liposomes-induced macrophage depletion inhibited psoriasis in mice, which was restored by adoptive transfer with Mincle-expressing macrophages but not by Mincle-KO macrophages. This was further confirmed in macrophage-specific Mincle-KO mice. Mechanistically, macrophages mediated psoriasis via the Mincle-Syk-NF-κB pathway as blocking macrophage Mincle inhibited Syk/NF-κB-driven skin lesions and epidermal injury in vivo and in vitro. We also found that LPS induced Mincle expression by M1 macrophages via the PU.1-dependent mechanism. Most importantly, we revealed that targeting Mincle with a neutralizing antibody significantly improved psoriasis in mice. In summary, our findings demonstrated that macrophages mediate psoriasis in mice via the Mincle-dependent mechanism, targeting Mincle may represent as a novel therapy for psoriasis. A simplified pathway model of Mincle in macrophage-mediated psoriasis.
Collapse
Affiliation(s)
- Rui-Zhi Tan
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Xia Zhong
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Rang-Yue Han
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ke-Huan Xie
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jian Jia
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ye Yang
- Department of Orthopaedics, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Mei Cheng
- Dermatological Department, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Chun-Yan Yang
- Dermatological Department, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China.
| |
Collapse
|
16
|
Meng C, Song Z, Zhang L, Geng Y, Sun J, Miao G, Liu P. Effects of losartan in patients with NAFLD: A meta-analysis of randomized controlled trial. Open Life Sci 2023; 18:20220583. [PMID: 36970603 PMCID: PMC10031500 DOI: 10.1515/biol-2022-0583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 03/24/2023] Open
Abstract
Losartan has become a hot spot in the treatment of non-alcoholic fatty liver disease (NAFLD) among angiotensin receptor blocker drugs. We sought to conduct a systematic examination and meta-analysis to examine the effects of losartan on patients with NAFLD. We searched for potentially randomized controlled trials in PubMed, Embase, China National Knowledge Infrastructure, Wanfang, and the Cochrane database up to October 09, 2022. We used the Cochrane risk of bias tool to evaluate the study quality. Analysis of subgroups, sensitivity analysis, and publishing bias were explored. The quality of the included studies was moderate to high. Six trials involving 408 patients were included. The meta-analysis demonstrated that aspartate transaminase was significantly affected by losartan therapy (mean difference [MD] = −5.34, 95% confidence interval [CI] [−6.54, −4.13], Z = 8.70, P < 0.01). The meta-analysis subgroup showed that losartan 50 mg once daily could lower the level of alanine aminotransferase (MD = −18.92, 95% CI [−21.18, −16.66], Z = 16.41, P < 0.01). There was no statistically significant difference in serum total cholesterol, triglyceride, low-density lipoprotein, and high-density lipoprotein.
Collapse
Affiliation(s)
- Chang Meng
- Department of Emergency, Emergency General Hospital, XiBaHe South Road 29, Chaoyang District, Beijing, 100028, PR China
| | - Zejun Song
- Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, PR China
| | - Lingnan Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Hebei University, 212 Yuhua East Road, Lianchi District, Baoding City, 071000, PR China
| | - Yu Geng
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, NO. 168 Litang Road, Changping District, Beijing102218, P. R. China
| | - Jing Sun
- Department of Critical Care Medicine, Emergency General Hospital, XiBaHe South Road 29, Chaoyang District, Beijing, 100028, PR China
| | - Guobin Miao
- Department of Emergency, Emergency General Hospital, XiBaHe South Road 29, Chaoyang District, Beijing, 100028, PR China
| | - Peng Liu
- Department of Cardiology, Ordos Central Hospital, Ordos School of Clinical Medicine, Inner Mongolia Medical University, 23 Yijinhuoluo West Street, Dongsheng District, Inner Mongolia, 017000, PR China
| |
Collapse
|
17
|
The Role of IL-13 and IL-4 in Adipose Tissue Fibrosis. Int J Mol Sci 2023; 24:ijms24065672. [PMID: 36982747 PMCID: PMC10051142 DOI: 10.3390/ijms24065672] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
White adipose tissue (WAT) fibrosis, characterized by an excess of extracellular (ECM) matrix components, is strongly associated with WAT inflammation and dysfunction due to obesity. Interleukin (IL)-13 and IL-4 were recently identified as critical mediators in the pathogenesis of fibrotic diseases. However, their role in WAT fibrosis is still ill-defined. We therefore established an ex vivo WAT organotypic culture system and demonstrated an upregulation of fibrosis-related genes and an increase of α-smooth muscle actin (αSMA) and fibronectin abundance upon dose-dependent stimulation with IL-13/IL-4. These fibrotic effects were lost in WAT lacking il4ra, which encodes for the underlying receptor controlling this process. Adipose tissue macrophages were found to play a key role in mediating IL-13/IL-4 effects in WAT fibrosis as their depletion through clodronate dramatically decreased the fibrotic phenotype. IL-4-induced WAT fibrosis was partly confirmed in mice injected intraperitoneally with IL-4. Furthermore, gene correlation analyses of human WAT samples revealed a strong positive correlation of fibrosis markers with IL-13/IL-4 receptors, whereas IL13 and IL4 correlations failed to confirm this association. In conclusion, IL-13 and IL-4 can induce WAT fibrosis ex vivo and partly in vivo, but their role in human WAT remains to be further elucidated.
Collapse
|
18
|
Natsui K, Tsuchiya A, Imamiya R, Osada-Oka M, Ishii Y, Koseki Y, Takeda N, Tomiyoshi K, Yamazaki F, Yoshida Y, Ohashi R, Ling Y, Ueda K, Moritoki N, Sato K, Nakajima T, Hasegawa Y, Okuda S, Shibata S, Terai S. Escherichia coli-derived outer-membrane vesicles induce immune activation and progression of cirrhosis in mice and humans. Liver Int 2023; 43:1126-1140. [PMID: 36751961 DOI: 10.1111/liv.15539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/14/2023] [Accepted: 02/05/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND AND AIMS Decompensated cirrhosis with fibrosis progression causes portal hypertension followed by an oedematous intestinal tract. These conditions weaken the barrier function against bacteria in the intestinal tract, a condition called leaky gut, resulting in invasion by bacteria and bacterial components. Here, we investigated the role of outer-membrane vesicles (OMVs) of Escherichia coli, which is the representative pathogenic gut-derived bacteria in patients with cirrhosis in the pathogenesis of cirrhosis. METHODS We investigated the involvement of OMVs in humans using human serum and ascites samples and also investigated the involvement of OMVs from E. coli in mice using mouse liver-derived cells and a mouse cirrhosis model. RESULTS In vitro, OMVs induced inflammatory responses to macrophages and neutrophils, including the upregulation of C-type lectin domain family 4 member E (Clec4e), and induced the suppression of albumin production in hepatocytes but had a relatively little direct effect on hepatic stellate cells. In a mouse cirrhosis model, administration of OMVs led to increased liver inflammation, especially affecting the activation of macrophages, worsening fibrosis and decreasing albumin production. Albumin administration weakened these inflammatory changes. In addition, multiple antibodies against bacterial components were increased with a progressing Child-Pugh grade, and OMVs were detected in ascites of patients with decompensated cirrhosis. CONCLUSIONS In conclusion, OMVs induce inflammation, fibrosis and suppression of albumin production, affecting the pathogenesis of cirrhosis. We believe that our study paves the way for the future prevention and treatment of cirrhosis.
Collapse
Affiliation(s)
- Kazuki Natsui
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Future Medical Research Center for Exosome and Designer Cell (F-DEC), Niigata University, Niigata, Japan
| | - Risa Imamiya
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Yui Ishii
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yohei Koseki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Nobutaka Takeda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kei Tomiyoshi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Fusako Yamazaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuki Yoshida
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Riuko Ohashi
- Histopathology Core Facility, Niigata University Faculty of Medicine, Niigata, Japan
| | - Yiwei Ling
- Medical AI Center, Niigata University School of Medicine, Niigata, Japan
| | - Koji Ueda
- Project for Realization of Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Sato
- Laboratory of Clinical Omics Research, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Takahiro Nakajima
- Laboratory of Medical Omics Research, KAZUSA DNA Research Institute, Chiba, Japan
| | - Yoshinori Hasegawa
- Laboratory of Clinical Omics Research, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Shujiro Okuda
- Medical AI Center, Niigata University School of Medicine, Niigata, Japan
| | - Shinsuke Shibata
- Future Medical Research Center for Exosome and Designer Cell (F-DEC), Niigata University, Niigata, Japan.,Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan.,Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Future Medical Research Center for Exosome and Designer Cell (F-DEC), Niigata University, Niigata, Japan
| |
Collapse
|
19
|
Research progress on Mincle as a multifunctional receptor. Int Immunopharmacol 2023; 114:109467. [PMID: 36436471 DOI: 10.1016/j.intimp.2022.109467] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 11/13/2022] [Indexed: 11/25/2022]
Abstract
Macrophage-induced C-type lectin (Mincle), a lipopolysaccharide-induced protein, is widely expressed on antigen-presenting cells. Mincle acts as a pattern recognition receptor that recognizes pathogen-associated molecular patterns of pathogens such as bacteria and fungi, mainly glycolipids, which induces an acquired immune response against microbial infection. Interestingly, Mincle can also identify patterns of lipid damage-associated molecule patterns released by injured cells, such as Sin3-associated protein 130 and β-glucosylceramides, which induces sterile inflammation and ultimately accelerates the progression of stroke, obesity, hepatitis, kidney injury, autoimmune diseases and tumors by promoting tissue inflammation. This article will review the various functions of Mincle, such as mediating sterile inflammation of tissues to accelerate disease progression, initiating immune responses to fight infection and promoting tumor progression.
Collapse
|
20
|
Sano M, Hirakawa S, Sasaki T, Inuzuka K, Katahashi K, Kayama T, Yamanaka Y, Tsuyuki H, Endo Y, Naruse E, Yokoyama Y, Sato K, Yamauchi K, Takeuchi H, Unno N. Role of Subcutaneous Adipose Tissues in the Pathophysiology of Secondary Lymphedema. Lymphat Res Biol 2022; 20:593-599. [PMID: 35394362 DOI: 10.1089/lrb.2021.0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Secondary lymphedema (LE) occurs due to the disruption of lymphatic circulation. Lymphatic fluid accumulation in subcutaneous tissues induces adipocyte proliferation. Obesity is an important risk factor for the occurrence and deterioration of LE. Although the relationship between LE and subcutaneous adipose tissue increase has been reported clinically, their pathophysiological relationship remains unknown. Thus, we aimed to verify whether subcutaneous adipose tissue increase is involved in the pathophysiology of secondary LE. Methods and Results: The hindlimb model of secondary LE was created using male Sprague-Dawley rats (control and LE groups; n = 5 each). Skin samples were obtained on postoperative day 168. Histological examination and quantitative real-time polymerase chain reaction analysis of inflammatory adipokines, tumor necrosis factor-alpha (Tnf-α), C-C chemokine ligand 2 (Ccl2), and interleukin-6 (Il-6) were performed. Limb volume and subcutaneous adipose tissues significantly increased in the LE group compared with those in the control. Macrophages aggregated in the augmented adipose tissues, around the adipocytes, and formed crown-like structures (CLSs). The number of CLSs significantly increased in the LE group. These macrophages expressed transforming growth factor-beta 1 (TGF-β1). Inflammatory adipokine secretion was not observed. Although Il-6 expression increased in the LE group, IL-6 was expressed in subcutaneous myofibroblasts but not in subcutaneous adipocytes. Conclusion: As TGF-β1 derived from subcutaneous myofibroblasts is involved in skin fibrosis during LE, TGF-β1 derived from adipose tissues may also play a similar role. Drug treatment for subcutaneous adipose tissue reduction may improve the skin condition in secondary LE and may be a new therapeutic strategy.
Collapse
Affiliation(s)
- Masaki Sano
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Hirakawa
- Preeminent Medical Photonics Education and Research Center Institute for NanoSuit Research, Departments of Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takeshi Sasaki
- Anatomy and Neuroscience and Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazunori Inuzuka
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuto Katahashi
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Kayama
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuta Yamanaka
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hajime Tsuyuki
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yusuke Endo
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ena Naruse
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yurina Yokoyama
- Rehabilitation, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kohji Sato
- Anatomy and Neuroscience and Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Katsuya Yamauchi
- Rehabilitation, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroya Takeuchi
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Unno
- Division of Vascular Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
21
|
Abstract
Adipose tissue is a complex heterogeneous tissue composed of adipocytes along with several non-adipocyte populations, including blood, stromal, endothelial, and progenitor cells, as well as extracellular matrix (ECM) components. As obesity progresses, the adipose tissue expands dynamically through adipocyte hypertrophy and/or hyperplasia. This expansion requires continuous ECM remodeling to properly accommodate the size increase as well as functional changes. Upon reaching a hypertrophic threshold beyond the adipocyte buffering capacity, excess ECM components are deposited, causing fibrosis and ultimately resulting in unhealthy metabolic maladaptation. These complex ECM remodeling processes in adipose tissues are regulated by the local environment, several key mediators, and genetic factors that are closely linked to insulin sensitivity. It is crucial to understand how adipocytes interact with nonadipocyte populations and various mediators (i.e., immune cells, ECM components, and adipokines) during these processes. This mini-review provides an overview of the latest research into the biology of obesity-induced adipose tissue fibrosis and its related clinical manifestations, providing insight for further studies aimed at controlling metabolic syndrome and its comorbidities.
Collapse
Affiliation(s)
- Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
22
|
Liang W, Qi Y, Yi H, Mao C, Meng Q, Wang H, Zheng C. The Roles of Adipose Tissue Macrophages in Human Disease. Front Immunol 2022; 13:908749. [PMID: 35757707 PMCID: PMC9222901 DOI: 10.3389/fimmu.2022.908749] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Macrophages are a population of immune cells functioning in antigen presentation and inflammatory response. Research has demonstrated that macrophages belong to a cell lineage with strong plasticity and heterogeneity and can be polarized into different phenotypes under different microenvironments or stimuli. Many macrophages can be recruited by various cytokines secreted by adipose tissue. The recruited macrophages further secrete various inflammatory factors to act on adipocytes, and the interaction between the two leads to chronic inflammation. Previous studies have indicated that adipose tissue macrophages (ATMs) are closely related to metabolic diseases like obesity and diabetes. Here, we will not only conclude the current progress of factors affecting the polarization of adipose tissue macrophages but also elucidate the relationship between ATMs and human diseases. Furthermore, we will highlight its potential in preventing and treating metabolic diseases as immunotherapy targets.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China.,Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yanxu Qi
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Chenyu Mao
- School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Qingxue Meng
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Hao Wang
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
23
|
Inokuchi JI, Kanoh H. Pathophysiological Significance of GM3 Ganglioside Molecular Species With a Particular Attention to the Metabolic Syndrome Focusing on Toll-Like Receptor 4 Binding. Front Mol Biosci 2022; 9:918346. [PMID: 35712350 PMCID: PMC9196240 DOI: 10.3389/fmolb.2022.918346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
GM3 ganglioside, the first molecule in ganglioside family biosynthesis, is formed by transfer of sialic acid to lactosylceramide. Several dozen GM3 molecular species exist, based on diversity of ceramide structures. Among ceramide structures composed of sphingosine and fatty acids, there is a great diversity resulting from different combinations of chain length, hydroxylation, and unsaturation of fatty acid chains. Expression patterns of GM3 species in serum vary during pathogenesis of metabolic syndrome. Physiological activity of each species, and significance of the variability, are poorly understood. Our studies revealed that GM3 species with differing fatty acid structures act as pro- or anti-inflammatory endogenous Toll-like receptor 4 (TLR4) ligands. Very long-chain fatty acid (VLCFA) and α-hydroxyl VLCFA GM3 variants strongly enhanced TLR4 activation. In contrast, long-chain fatty acid (LCFA) and ω-9 unsaturated VLCFA GM3 variants suppressed TLR4 activation. GM3 interacted with extracellular TLR4/myeloid differentiation factor 2 (MD-2) complex, thereby promoting dimerization/oligomerization. In obesity and metabolic syndrome, VLCFA-variant GM3 species were elevated in serum and adipose tissue, whereas LCFA-variant species were reduced, and such imbalances were correlated with disease progression. Our findings summarized in this review demonstrate that GM3 molecular species are disease-related endogenous TLR4 ligands and modulate homeostatic and pathogenic innate immune responses.
Collapse
Affiliation(s)
- Jin-ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembranes and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Osaka, Japan
- *Correspondence: Jin-ichi Inokuchi,
| | - Hirotaka Kanoh
- Division of Glycopathology, Institute of Molecular Biomembranes and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
24
|
Hachiya R, Tanaka M, Itoh M, Suganami T. Molecular mechanism of crosstalk between immune and metabolic systems in metabolic syndrome. Inflamm Regen 2022; 42:13. [PMID: 35490239 PMCID: PMC9057063 DOI: 10.1186/s41232-022-00198-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is currently considered as a molecular basis of metabolic syndrome. Particularly, obesity-induced inflammation in adipose tissue is the origin of chronic inflammation of metabolic syndrome. Adipose tissue contains not only mature adipocytes with large lipid droplets, but also a variety of stromal cells including adipocyte precursors, vascular component cells, immune cells, and fibroblasts. However, crosstalk between those various cell types in adipose tissue in obesity still remains to be fully understood. We focus on two innate immune receptors, Toll-like receptor 4 (TLR4) and macrophage-inducible C-type lectin (Mincle). We provided evidence that adipocyte-derived saturated fatty acids (SFAs) activate macrophage TLR4 signaling pathway, thereby forming a vicious cycle of inflammatory responses during the development of obesity. Intriguingly, the TLR4 signaling pathway is modulated metabolically and epigenetically: SFAs augment TLR4 signaling through the integrated stress response and chromatin remodeling, such as histone methylation, regulates dynamic transcription patterns downstream of TLR4 signaling. Another innate immune receptor Mincle senses cell death, which is a trigger of chronic inflammatory diseases including obesity. Macrophages form a histological structure termed “crown-like structure (CLS)”, in which macrophages surround dead adipocytes to engulf cell debris and residual lipids. Mincle is exclusively expressed in macrophages forming the CLS in obese adipose tissue and regulates adipocyte death-triggered adipose tissue fibrosis. In addition to adipose tissue, we found a structure similar to CLS in the liver of nonalcoholic steatohepatitis (NASH) and the kidney after acute kidney injury. This review article highlights the recent progress of the crosstalk between immune and metabolic systems in metabolic syndrome, with a focus on innate immune receptors.
Collapse
Affiliation(s)
- Rumi Hachiya
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Department of Pediatrics, Tokyo Dental College Ichikawa General Hospital, Chiba, Japan
| | - Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michiko Itoh
- Department of Metabolic Syndrome and Nutritional Science, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan. .,Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
25
|
Innate-Immunity Genes in Obesity. J Pers Med 2021; 11:jpm11111201. [PMID: 34834553 PMCID: PMC8623883 DOI: 10.3390/jpm11111201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023] Open
Abstract
The main functions of adipose tissue are thought to be storage and mobilization of the body’s energy reserves, active and passive thermoregulation, participation in the spatial organization of internal organs, protection of the body from lipotoxicity, and ectopic lipid deposition. After the discovery of adipokines, the endocrine function was added to the above list, and after the identification of crosstalk between adipocytes and immune cells, an immune function was suggested. Nonetheless, it turned out that the mechanisms underlying mutual regulatory relations of adipocytes, preadipocytes, immune cells, and their microenvironment are complex and redundant at many levels. One possible way to elucidate the picture of adipose-tissue regulation is to determine genetic variants correlating with obesity. In this review, we examine various aspects of adipose-tissue involvement in innate immune responses as well as variants of immune-response genes associated with obesity.
Collapse
|
26
|
van der Zande HJP, Nitsche D, Schlautmann L, Guigas B, Burgdorf S. The Mannose Receptor: From Endocytic Receptor and Biomarker to Regulator of (Meta)Inflammation. Front Immunol 2021; 12:765034. [PMID: 34721436 PMCID: PMC8551360 DOI: 10.3389/fimmu.2021.765034] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/27/2021] [Indexed: 01/27/2023] Open
Abstract
The mannose receptor is a member of the C-type lectin (CLEC) family, which can bind and internalize a variety of endogenous and pathogen-associated ligands. Because of these properties, its role in endocytosis as well as antigen processing and presentation has been studied intensively. Recently, it became clear that the mannose receptor can directly influence the activation of various immune cells. Cell-bound mannose receptor expressed by antigen-presenting cells was indeed shown to drive activated T cells towards a tolerogenic phenotype. On the other hand, serum concentrations of a soluble form of the mannose receptor have been reported to be increased in patients suffering from a variety of inflammatory diseases and to correlate with severity of disease. Interestingly, we recently demonstrated that the soluble mannose receptor directly promotes macrophage proinflammatory activation and trigger metaflammation. In this review, we highlight the role of the mannose receptor and other CLECs in regulating the activation of immune cells and in shaping inflammatory responses.
Collapse
Affiliation(s)
| | - Dominik Nitsche
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Laura Schlautmann
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Sven Burgdorf
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
27
|
Han Q, Wang X, Ding X, He J, Cai G, Zhu H. Immunomodulatory Effects of Mesenchymal Stem Cells on Drug-Induced Acute Kidney Injury. Front Immunol 2021; 12:683003. [PMID: 34149721 PMCID: PMC8213363 DOI: 10.3389/fimmu.2021.683003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/10/2021] [Indexed: 12/29/2022] Open
Abstract
Drug-induced nephrotoxicity is an important and increasing cause of acute kidney injury (AKI), which accounts for approximately 20% of hospitalized patients. Previous reviews studies on immunity and AKI focused mainly on ischemia-reperfusion (IR), whereas no systematic review addressing drug-induced AKI and its related immune mechanisms is available. Recent studies have provided a deeper understanding on the mechanisms of drug-induced AKI, among which acute tubular interstitial injury induced by the breakdown of innate immunity was reported to play an important role. Emerging research on mesenchymal stem cell (MSC) therapy has revealed its potential as treatment for drug-induced AKI. MSCs can inhibit kidney damage by regulating the innate immune balance, promoting kidney repair, and preventing kidney fibrosis. However, it is important to note that there are various sources of MSCs, which impacts on the immunomodulatory ability of the cells. This review aims to address the immune pathogenesis of drug-induced AKI versus that of IR-induced AKI, and to explore the immunomodulatory effects and therapeutic potential of MSCs for drug-induced AKI.
Collapse
Affiliation(s)
- Qiuxia Han
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Jun He
- Department of Genetics, Changsha Hospital for Maternal and Child Health Care, Hunan, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
28
|
Expression and regulation of macrophage-inducible C-type lectin in human synovial macrophages. Cent Eur J Immunol 2021; 45:377-381. [PMID: 33613092 PMCID: PMC7882416 DOI: 10.5114/ceji.2020.103411] [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] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/25/2018] [Indexed: 11/17/2022] Open
Abstract
Recent evidence suggests that synovial macrophage activation may be involved in cartilage destruction and pain in osteoarthritis (OA). The macrophage-inducible C-type lectin (Mincle) Clec4e is expressed in macrophages and is regulated in inflammatory conditions. Given that the regulation of Mincle in synovial macrophages has not been elucidated, we investigated the expression and regulation of Mincle in human synovial tissue (ST) harvested from patients with radiographic knee OA during total knee arthroplasty. Immunohistochemical and flow cytometric analyses were used to identify cells with Mincle expression in resected tissues. CD14-positive (CD14+; macrophage-rich cell fraction) and CD14-negative (CD14-; fibroblast-rich cell fraction) cells were extracted from the ST and used to assess MINCLE mRNA expression levels. To determine the role of tumor necrosis factor alpha (TNF-α) in the regulation of MINCLE expression, TNF-α was used to stimulate cultured CD14+ cells. Immunohistochemical staining revealed Mincle-positive cells in the synovial lining layer. Flow cytometric analysis showed that CD45+CD14+ cells were Mincle positive while CD45–/CD14– cells were Mincle negative. MINCLE expression was significantly higher in CD14+ cells than in CD14– cells. Stimulation of cultured CD14+ macrophages with TNF-α significantly increased MINCLE mRNA expression, while stimulation with TNF-α neutralizing antibody significantly decreased expression. That Mincle expression was observed in synovial macrophages and its expression was induced by TNF-α suggests that Mincle might have a key role in synovial inflammation in the osteoarthritic synovium.
Collapse
|
29
|
Liao Y, Tan RZ, Li JC, Liu TT, Zhong X, Yan Y, Yang JK, Lin X, Fan JM, Wang L. Isoliquiritigenin Attenuates UUO-Induced Renal Inflammation and Fibrosis by Inhibiting Mincle/Syk/NF-Kappa B Signaling Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1455-1468. [PMID: 32341639 PMCID: PMC7166058 DOI: 10.2147/dddt.s243420] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
Purpose Chronic kidney disease (CKD) is a global nephrotic syndrome characterized by chronic inflammation, oxidative stress and fibrosis in the kidney. Isoliquiritigenin (ISL), a flavonoid from licorice, has historically been reported to inhibit innate immune responses to inflammation and fibrosis in vivo. However, the effect of ISL on CKD progression is largely unknown. Materials and Methods In this study, we employed the inflammatory and fibrotic models of LPS/TGF-β-induced bone marrow-derived macrophages (BMDM) in vitro and unilateral ureteral obstruction (UUO) model in vivo to explore the potential effects and mechanism of ISL on renal inflammation and fibrosis. Results Our results manifest that ISL improved UUO-induced renal dysfunction and reduced tubular damage with a significantly downregulated mRNA expression and secretion of IL-1β, IL-6, TNF-α and MCP-1 in vitro and in vivo. It is worth noting that ISL can strongly inhibit the mRNA and protein expression of Mincle (macrophage-induced c-type lectin) in BMDM and UUO. ISL inhibited the phosphorylation of Syk and NF-kappa B and simultaneously reduced the expression of α-SMA and Col III in vivo and in vitro. More interestingly, when dealing with TDB, a ligand of Mincle, it revealed significant reversal of protein expression levels as that observed with ISL. The expressions of IL-1β, IL-6, TNF-α, iNOS, p-Syk, p-NF-kappa B, α-SMA and FN in BMDM inflammatory model were significantly upregulated with TDB treatment. This confirms that ISL inhibits inflammation and fibrosis of macrophage by suppressing Mincle/Syk/NF-kappa B signaling pathway. Conclusion To conclude, ISL protects UUO-induced CKD by inhibiting Mincle-induced inflammation and suppressing renal fibrosis, which might be a specific renal protective mechanism of ISL, making it a novel drug to ameliorate CKD.
Collapse
Affiliation(s)
- Yuan Liao
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Rui-Zhi Tan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Jian-Chun Li
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Tong-Tong Liu
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Xia Zhong
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Ying Yan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Jie-Ke Yang
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Xiao Lin
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, Sichuan 610000, People's Republic of China
| | - Li Wang
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
30
|
Tanaka M. Molecular mechanism of obesity-induced adipose tissue inflammation; the role of Mincle in adipose tissue fibrosis and ectopic lipid accumulation. Endocr J 2020; 67:107-111. [PMID: 31852849 DOI: 10.1507/endocrj.ej19-0417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Metabolic syndrome is a common metabolic disorder that involves multiple organs and is predominantly influenced by obesity, especially the accumulation of visceral fat. It is also known that macrophages that infiltrate obese adipose tissue play an important role in inflammation of the adipose tissue. Macrophage-inducible C-type lectin (Mincle), a new inflammatory regulator found in obese adipose tissue, is expressed in pro-inflammatory M1 macrophages in adipose tissue. In addition, Mincle is expressed in macrophages that form a crown-like structure, where dead or dying adipocytes are surrounded by pro-inflammatory M1 macrophages; within this crown-like structure, adipocyte-macrophage crosstalk may occur in close proximity. Although there is no significant difference in body weight between Mincle-deficient and wild-type mice under high-fat diet, the epididymal fat weight is significantly higher and the liver weight is significantly lower in Mincle-deficient mice than those in wild-type mice. It has been shown that adipose tissue inflammation and fibrosis are attenuated in Mincle-deficient mice when compared with wild-type mice. In addition, Mincle-deficient mice have reduced hepatic lipid accumulation and better glucose metabolism. These results suggest that Mincle signaling in adipose tissue macrophages activates adipose tissue fibroblasts, which leads to adipose tissue fibrosis.
Collapse
Affiliation(s)
- Miyako Tanaka
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
31
|
Drouin M, Saenz J, Chiffoleau E. C-Type Lectin-Like Receptors: Head or Tail in Cell Death Immunity. Front Immunol 2020; 11:251. [PMID: 32133013 PMCID: PMC7040094 DOI: 10.3389/fimmu.2020.00251] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
C-type lectin-like receptors (CLRs) represent a family of transmembrane pattern recognition receptors, expressed primarily by myeloid cells. They recognize not only pathogen moieties for host defense, but also modified self-antigens such as damage-associated molecular patterns released from dead cells. Upon ligation, CLR signaling leads to the production of inflammatory mediators to shape amplitude, duration and outcome of the immune response. Thus, following excessive injury, dysregulation of these receptors leads to the development of inflammatory diseases. Herein, we will focus on four CLRs of the "Dectin family," shown to decode the immunogenicity of cell death. CLEC9A on dendritic cells links F-actin exposed by dying cells to favor cross-presentation of dead-cell associated antigens to CD8+ T cells. Nevertheless, CLEC9A exerts also feedback mechanisms to temper neutrophil recruitment and prevent additional tissue damage. MINCLE expressed by macrophages binds nuclear SAP130 released by necrotic cells to potentiate pro-inflammatory responses. However, the consequent inflammation can exacerbate pathogenesis of inflammatory diseases. Moreover, in a tumor microenvironment, MINCLE induces macrophage-induced immune suppression and cancer progression. Similarly, triggering of LOX-1 by oxidized LDL, amplifies pro-inflammatory response but promotes tumor immune escape and metastasis. Finally, CLEC12A that recognizes monosodium urate crystals formed during cell death, inhibits activating signals to prevent detrimental inflammation. Interestingly, CLEC12A also sustains type-I IFN response to finely tune immune responses in case of viral-induced collateral damage. Therefore, CLRs acting in concert as sensors of injury, could be used in a targeted way to treat numerous diseases such as allergies, obesity, tumors, and autoimmunity.
Collapse
Affiliation(s)
- Marion Drouin
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Javier Saenz
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Elise Chiffoleau
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
32
|
Numaguchi R, Furuhashi M, Matsumoto M, Sato H, Yanase Y, Kuroda Y, Harada R, Ito T, Higashiura Y, Koyama M, Tanaka M, Moniwa N, Nakamura M, Doi H, Miura T, Kawaharada N. Differential Phenotypes in Perivascular Adipose Tissue Surrounding the Internal Thoracic Artery and Diseased Coronary Artery. J Am Heart Assoc 2020; 8:e011147. [PMID: 30638109 PMCID: PMC6497339 DOI: 10.1161/jaha.118.011147] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Perivascular adipose tissue (PVAT) is causally associated with vascular function and the pathogenesis of vascular disease in association with metabolically driven chronic inflammation called metaflammation. However, the difference in PVAT surrounding the coronary artery (CA‐PVAT) and that surrounding the internal thoracic artery (ITA‐PVAT), a vessel resistant to atherosclerosis, remains unclear. Herein, we investigated whether CA‐PVAT, ITA‐PVAT, and subcutaneous adipose tissue (SCAT) have distinct phenotypes. Methods and Results Fat pads were sampled from 44 patients (men/women, 36:8; age, 67±13 years) with CA disease who underwent elective CA bypass grafting. Adipocyte size in ITA‐PVAT and that in CA‐PVAT were significantly smaller than that in SCAT. A greater extent of fibrosis and increased gene expression levels of fibrosis‐related molecules were observed in CA‐PVAT than those in SCAT and those in ITA‐PVAT. CA‐PVAT exhibited more pronounced metaflammation, as indicated by a significantly larger extent of CD68‐positive and CD11c‐positive M1 macrophages, a lower ratio of CD206‐positive M2 to CD11c‐positive M1 macrophages, a lower gene expression level of adiponectin, and higher gene expression levels of inflammatory cytokines and inflammasome‐ and endoplasmic reticulum stress–related molecules, than did ITA‐PVAT and SCAT. Expression patterns of adipocyte developmental and pattern‐forming genes were totally different among SCAT, ITA‐PVAT, and CA‐PVAT. Conclusions The phenotype of ITA‐PVAT is closer to that of SCAT than that of CA‐PVAT, which may result from inherent differences in adipocytes. ITA‐PVAT appears to be protected from metaflammation and consecutive adipose tissue remodeling, which may contribute to the decreased atherosclerotic plaque burden in the ITA.
Collapse
Affiliation(s)
- Ryosuke Numaguchi
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Masato Furuhashi
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Megumi Matsumoto
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Hiroshi Sato
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Yosuke Yanase
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Yosuke Kuroda
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Ryo Harada
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Toshiro Ito
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| | - Yukimura Higashiura
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Masayuki Koyama
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Marenao Tanaka
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Norihito Moniwa
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Masanori Nakamura
- 3 Department of Cardiovascular Surgery Sapporo City General Hospital Sapporo Japan
| | - Hirosato Doi
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan.,4 Department of Cardiovascular Surgery Sapporo Cardiovascular Clinic Sapporo Japan
| | - Tetsuji Miura
- 2 Department of Cardiovascular, Renal and Metabolic Medicine Sapporo Medical University School of Medicine Sapporo Japan
| | - Nobuyoshi Kawaharada
- 1 Department of Cardiovascular Surgery Sapporo Medical University School of Medicine Sapporo Japan
| |
Collapse
|
33
|
Thiem K, Hoeke G, Zhou E, Hijmans A, Houben T, Boels MG, Mol IM, Lutgens E, Shiri-Sverdlov R, Bussink J, Kanneganti TD, Boon MR, Stienstra R, Tack CJ, Rensen PCN, Netea MG, Berbée JFP, van Diepen JA. Deletion of haematopoietic Dectin-2 or CARD9 does not protect from atherosclerosis development under hyperglycaemic conditions. Diab Vasc Dis Res 2020; 17:1479164119892140. [PMID: 31868000 PMCID: PMC7510497 DOI: 10.1177/1479164119892140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND C-type lectin receptors, including Dectin-2, are pattern recognition receptors on monocytes and macrophages that mainly recognize sugars and sugar-like structures present on fungi. Activation of C-type lectin receptors induces downstream CARD9 signalling, leading to the production of cytokines. We hypothesized that under hyperglycaemic conditions, as is the case in diabetes mellitus, glycosylated protein (sugar-like) structures activate C-type lectin receptors, leading to immune cell activation and increased atherosclerosis development. METHODS Low-density lipoprotein receptor-deficient mice were lethally irradiated and transplanted with bone marrow from control wild-type, Dectin-2-/- or Card9-/- mice. After 6 weeks of recovery, mice received streptozotocin injections (50 mg/g BW; 5 days) to induce hyperglycaemia. After an additional 2 weeks, mice were fed a Western-type diet (0.1% cholesterol) for 10 weeks. RESULTS AND CONCLUSION Deletion of haematopoietic Dectin-2 reduced the number of circulating Ly6Chi monocytes, increased pro-inflammatory cytokine production, but did not affect atherosclerosis development. Deletion of haematopoietic CARD9 tended to reduce macrophage and collagen content in atherosclerotic lesions, again without influencing the lesion size. Deletion of haematopoietic Dectin-2 did not influence atherosclerosis development under hyperglycaemic conditions, despite some minor effects on inflammation. Deletion of haematopoietic CARD9 induced minor alterations in plaque composition under hyperglycaemic conditions, without affecting lesion size.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/etiology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Biomarkers/blood
- Blood Glucose/metabolism
- Bone Marrow Transplantation
- CARD Signaling Adaptor Proteins/deficiency
- CARD Signaling Adaptor Proteins/genetics
- Cells, Cultured
- Collagen/metabolism
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diet, Western
- Gene Deletion
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/metabolism
- Monocytes/pathology
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Kathrin Thiem
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Kathrin Thiem, Department of Internal
Medicine and Radboud Institute for Molecular Life Sciences, Radboud University
Medical Center, 463, Geert Grooteplein zuid 8, 6525 GA Nijmegen, The
Netherlands.
| | - Geerte Hoeke
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Enchen Zhou
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Anneke Hijmans
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| | - Tom Houben
- Departments of Molecular Genetics, Human
Biology and Surgery, School of Nutrition and Translational Research in Metabolism
(NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Margien G Boels
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Division of Nephrology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Isabel M Mol
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Lutgens
- Division of Experimental Vascular
Biology, Department of Medical Biochemistry, Academic Medical Center, University of
Amsterdam, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention,
Ludwig Maximilians University of Munich, Munich, Germany
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics, Human
Biology and Surgery, School of Nutrition and Translational Research in Metabolism
(NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Johan Bussink
- Department of Radiation Oncology,
Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Mariëtte R Boon
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Division of Human Nutrition,
Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| | - Patrick CN Rensen
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Department for Genomics and
Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn,
Bonn, Germany
| | - Jimmy FP Berbée
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Janna A van Diepen
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| |
Collapse
|
34
|
Miyake Y, Yamasaki S. Immune Recognition of Pathogen-Derived Glycolipids Through Mincle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:31-56. [DOI: 10.1007/978-981-15-1580-4_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Chen C, Perry TL, Chitko-McKown CG, Smith AD, Cheung L, Beshah E, Urban JF, Dawson HD. The regulatory actions of retinoic acid on M2 polarization of porcine macrophages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 98:20-33. [PMID: 30974109 DOI: 10.1016/j.dci.2019.03.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/25/2019] [Accepted: 03/30/2019] [Indexed: 06/09/2023]
Abstract
We previously demonstrated that the most bioactive vitamin A metabolite, all-trans retinoic acid (ATRA), increased T helper 2-associated responses induced in pigs by infection with the parasitic nematode Ascaris suum We also showed that ATRA potentiated the mRNA expression of several IL-4 induced chemokines (chemokine (CC motif) ligand 11 [(CCL11), CCL17, CCL22 and CCL26] associated with alternative activation (M2a) in porcine macrophages in vitro. Herein, several mechanisms whereby ATRA affects IL-4 signaling are profiled using large-scale real time PCR and RNA-Seq analysis. Twenty-three genes associated with M2a markers in other species were independently upregulated by both IL-4 and ATRA, including the adenosine receptor A2B (ADORA2B), cysteinyl leukotriene receptor 2 (CYSLTR2) and the vitamin D receptor (VDR). ATRA synergistically enhanced IL-4 up-regulation of Hepatitis A virus cellular receptor 2 (HAVCR2) and transglutaminase 2 (TGM2) and further repressed IL-4 down-regulated CD163 and Cytochrome b-245, beta polypeptide (CYBB) mRNA. Macrophages treated with ATRA exhibited a dose-dependent reduction in phagocytosis of opsonized Staphylococcus aureus. In addition, the combination of IL-4 and ATRA up-regulated the anti-inflammatory protein, IL-1R antagonist (IL1RN) and TGM2. These data indicate that ATRA induces a state of partial alternative activation in porcine macrophages, and amplifies certain aspects of M2a activation induced by IL-4. Given the prevalence of allergic and parasitic diseases worldwide and the close similarities in the porcine and human immune responses, these findings have important implications for the nutritional regulation of allergic inflammation at mucosal surfaces.
Collapse
Affiliation(s)
- Celine Chen
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | | | - Carol G Chitko-McKown
- Genetics, Breeding, and Animal Health Research Unit, USMARC, Clay Center, NE, 68933, USA
| | - Allen D Smith
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | - Lumei Cheung
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | - Ethiopia Beshah
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | - Joseph F Urban
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | - Harry D Dawson
- U.S. Department of Agriculture - Agriculture Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA.
| |
Collapse
|
36
|
TNF-α in Combination with Palmitate Enhances IL-8 Production via The MyD88- Independent TLR4 Signaling Pathway: Potential Relevance to Metabolic Inflammation. Int J Mol Sci 2019; 20:ijms20174112. [PMID: 31443599 PMCID: PMC6747275 DOI: 10.3390/ijms20174112] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 01/08/2023] Open
Abstract
Elevated levels of IL-8 (CXCL8) in obesity have been linked with insulin resistance and type 2 diabetes (T2D). The mechanisms that lead to the profound production of IL-8 in obesity remains to be understood. TNF-α and saturated free fatty acids (FFAs) are increased in obese humans and correlate with insulin resistance. Hence, we sought to investigate whether the cooccurrence of TNF-α and FFAs led to increase the production of IL-8 by human monocytes. We found that co-stimulation of human monocytes with palmitate and TNF-α led to increased IL-8 production as compared to those stimulated with palmitate or TNF-α alone. The synergistic production of IL-8 by TNF-α/palmitate was suppressed by neutralizing anti- Toll like receptor 4 (TLR4) antibody and by genetic silencing of TLR4. Both MyD88-deficient and MyD88-competent cells responded comparably to TNF-α/Palmitate. However, TIR-domain-containing adapter-inducing interferon (TRIF) inhibition or interferon regulatory transcription factor 3 (IRF3) knockdown partly blocked the synergistic production of IL-8. Our human data show that increased adipose tissue TNF-α expression correlated positively with IL-8 expression (r = 0.49, P = 0.001). IL-8 and TNF-α correlated positively with macrophage markers including CD68, CD163 and CD86 in adipose tissue. These findings suggest that the signaling cross-talk between saturated fatty acid palmitate and TNF-α may be a key driver in obesity-associated chronic inflammation via an excessive production of IL-8.
Collapse
|
37
|
Engin AB, Engin A, Gonul II. The effect of adipocyte-macrophage crosstalk in obesity-related breast cancer. J Mol Endocrinol 2019; 62:R201-R222. [PMID: 30620711 DOI: 10.1530/jme-18-0252] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022]
Abstract
Adipose tissue is the primary source of many pro-inflammatory cytokines in obesity. Macrophage numbers and pro-inflammatory gene expression are positively associated with adipocyte size. Free fatty acid and tumor necrosis factor-α involve in a vicious cycle between adipocytes and macrophages aggravating inflammatory changes. Thereby, M1 macrophages form a characteristic 'crown-like structure (CLS)' around necrotic adipocytes in obese adipose tissue. In obese women, CLSs of breast adipose tissue are responsible for both increase in local aromatase activity and aggressive behavior of breast cancer cells. Interlinked molecular mechanisms between adipocyte-macrophage-breast cancer cells in obesity involve seven consecutive processes: Excessive release of adipocyte- and macrophage-derived inflammatory cytokines, TSC1-TSC2 complex-mTOR crosstalk, insulin resistance, endoplasmic reticulum (ER) stress and excessive oxidative stress generation, uncoupled respiration and hypoxia, SIRT1 controversy, the increased levels of aromatase activity and estrogen production. Considering elevated risks of estrogen receptor (E2R)-positive postmenopausal breast cancer growth in obesity, adipocyte-macrophage crosstalk is important in the aforementioned issues. Increased mTORC1 signaling in obesity ensures the strong activation of oncogenic signaling in E2Rα-positive breast cancer cells. Since insulin and insulin-like growth factors have been identified as tumor promoters, hyperinsulinemia is an independent risk factor for poor prognosis in breast cancer despite peripheral insulin resistance. The unpredictable effects of adipocyte-derived leptin-estrogen-macrophage axis, and sirtuin 1 (SIRT1)-adipose-resident macrophage axis in obese postmenopausal patients with breast cancer are unresolved mechanistic gaps in the molecular links between the tumor growth and adipocytokines.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ipek Isik Gonul
- Department of Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
38
|
Lim R, Lappas M. Expression and function of macrophage-inducible C-type lectin (Mincle) in inflammation driven parturition in fetal membranes and myometrium. Clin Exp Immunol 2019; 197:95-110. [PMID: 30793298 DOI: 10.1111/cei.13281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2019] [Indexed: 12/31/2022] Open
Abstract
The pivotal role of inflammatory processes in human parturition is well known, but not completely understood. We have performed a study to examine the role of macrophage-inducible C-type lectin (Mincle) in inflammation-associated parturition. Using human samples, we show that spontaneous labour is associated with up-regulated Mincle expression in the myometrium and fetal membranes. Mincle expression was also increased in fetal membranes and myometrium in the presence of pro-labour mediators, the proinflammatory cytokines interleukin (IL)-1B and tumour necrosis factor (TNF), and Toll-like receptor (TLR) ligands fsl-1, poly(I:C), lipopolysaccharide (LPS) and flagellin. These clinical studies are supported by mouse studies, where an inflammatory challenge in a mouse model of preterm birth increased Mincle expression in the uterus. Importantly, elimination of Mincle decreased the effectiveness of proinflammatory cytokines and TLR ligands to induce the expression of pro-labour mediators; namely, proinflammatory cytokines and chemokines, contraction-associated proteins and prostaglandins, and extracellular matrix remodelling enzymes, matrix metalloproteinases. The data presented in this study suggest that Mincle is required when inflammatory activation precipitates parturition.
Collapse
Affiliation(s)
- R Lim
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - M Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Kim SY, Nair MG. Macrophages in wound healing: activation and plasticity. Immunol Cell Biol 2019; 97:258-267. [PMID: 30746824 PMCID: PMC6426672 DOI: 10.1111/imcb.12236] [Citation(s) in RCA: 344] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/31/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022]
Abstract
Macrophages are critically involved in wound healing, from dampening inflammation to clearing cell debris and coordinating tissue repair. Within the wound, the complexity of macrophage function is increasingly recognized, with adverse outcomes when macrophages are inappropriately activated, such as in fibrosis or chronic non-healing wounds. Recent advances in in vivo and translational wound models, macrophage-specific deletions and new technologies to distinguish macrophage subsets, have uncovered the vast spectrum of macrophage activation and effector functions. Here, we summarize the main players in wound-healing macrophage activation and function, including cytokines, apoptotic cells, nucleotides and mechanical stimuli. We highlight recent studies demonstrating cooperation between these factors for optimal wound healing. Next, we describe recent technologies such as cell tracking and single-cell RNA-seq, which have uncovered remarkable plasticity and heterogeneity in blood-derived or tissue-resident macrophages and discuss the implications for wound healing. Lastly, we evaluate macrophage dysfunction in aberrant wound healing that occurs in aging, diabetes and fibrosis. A better understanding of the longevity and plasticity of wound-healing macrophages, and identification of unique macrophage subsets or specific effector molecules in wound healing, would shed light on the therapeutic potential of manipulating macrophage function for optimal wound healing.
Collapse
Affiliation(s)
- Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
40
|
Ahmad R, Al-Roub A, Kochumon S, Akther N, Thomas R, Kumari M, Koshy MS, Tiss A, Hannun YA, Tuomilehto J, Sindhu S, Rosen ED. The Synergy between Palmitate and TNF-α for CCL2 Production Is Dependent on the TRIF/IRF3 Pathway: Implications for Metabolic Inflammation. THE JOURNAL OF IMMUNOLOGY 2018; 200:3599-3611. [PMID: 29632147 DOI: 10.4049/jimmunol.1701552] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/16/2018] [Indexed: 12/24/2022]
Abstract
The chemokine CCL2 (also known as MCP-1) is a key regulator of monocyte infiltration into adipose tissue, which plays a central role in the pathophysiology of obesity-associated inflammation and insulin resistance. It remains unclear how CCL2 production is upregulated in obese humans and rodents. Because elevated levels of the free fatty acid (FFA) palmitate and TNF-α have been reported in obesity, we studied whether these agents interact to trigger CCL2 production. Our data show that treatment of THP-1 and primary human monocytic cells with palmitate and TNF-α led to a marked increase in CCL2 production compared with either treatment alone. Mechanistically, we found that cooperative production of CCL2 by palmitate and TNF-α did not require MyD88, but it was attenuated by blocking TLR4 or TRIF. IRF3-deficient cells did not show synergistic CCL2 production in response to palmitate/TNF-α. Moreover, IRF3 activation by polyinosinic-polycytidylic acid augmented TNF-α-induced CCL2 secretion. Interestingly, elevated NF-κB/AP-1 activity resulting from palmitate/TNF-α costimulation was attenuated by TRIF/IRF3 inhibition. Diet-induced C57BL/6 obese mice with high FFAs levels showed a strong correlation between TNF-α and CCL2 in plasma and adipose tissue and, as expected, also showed increased adipose tissue macrophage accumulation compared with lean mice. Similar results were observed in the adipose tissue samples from obese humans. Overall, our findings support a model in which elevated FFAs in obesity create a milieu for TNF-α to trigger CCL2 production via the TLR4/TRIF/IRF3 signaling cascade, representing a potential contribution of FFAs to metabolic inflammation.
Collapse
Affiliation(s)
- Rasheed Ahmad
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait;
| | - Areej Al-Roub
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Shihab Kochumon
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Nadeem Akther
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Reeby Thomas
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Manju Kumari
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard School of Medicine, Boston, MA 02215
| | - Merin S Koshy
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Ali Tiss
- Proteomics Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait; and
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794
| | - Jaakko Tuomilehto
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Sardar Sindhu
- Immunology Unit, Dasman Diabetes Institute, Kuwait City 15462, Kuwait
| | - Evan D Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard School of Medicine, Boston, MA 02215
| |
Collapse
|
41
|
Ann SJ, Kim KK, Cheon EJ, Noh HM, Hwang I, Yu JW, Park S, Kang SM, Manabe I, Miller YI, Kim S, Lee SH. Palmitate and minimally-modified low-density lipoprotein cooperatively promote inflammatory responses in macrophages. PLoS One 2018. [PMID: 29518116 PMCID: PMC5843266 DOI: 10.1371/journal.pone.0193649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Increased consumption of Western-type diets and environmental insults lead to wide-spread increases in the plasma levels of saturated fatty acids and lipoprotein oxidation. The aim of this study is to examine whether palmitate and minimally modified low-density lipoprotein (mmLDL) exert an additive effect on macrophage activation. We found that CXCL2 and TNF-α secretion as well as ERK and p38 phosphorylation were additively increased by co-treatment of J774 macrophages with palmitate and mmLDL in the presence of lipopolysaccharide (LPS). Furthermore, the analysis of differentially expressed genes using the KEGG database revealed that several pathways, including cytokine-cytokine receptor interaction, and genes were significantly altered. These results were validated with real-time PCR, showing upregulation of Il-6, Csf3, Il-1β, and Clec4d. The present study demonstrated that palmitate and mmLDL additively potentiate the LPS-induced activation of macrophages. These results suggest the existence of synergistic mechanisms by which saturated fatty acids and oxidized lipoproteins activate immune cells.
Collapse
Affiliation(s)
- Soo-jin Ann
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ka-Kyung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jeong Cheon
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Min Noh
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Min Kang
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, La Jolla, United States of America
| | - Sangwoo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (SH Lee); (S Kim)
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (SH Lee); (S Kim)
| |
Collapse
|
42
|
Tanaka M, Itoh M, Ogawa Y, Suganami T. Molecular mechanism of obesity-induced 'metabolic' tissue remodeling. J Diabetes Investig 2018; 9:256-261. [PMID: 29086488 PMCID: PMC5835451 DOI: 10.1111/jdi.12769] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic inflammation is a common molecular basis underlying a variety of chronic diseases. Accumulating evidence has also suggested that chronic inflammation contributes to the pathogenesis of obesity and diabetes, which have been considered as metabolic diseases. For the past several decades, there has been dramatic progress in understanding the underlying mechanism of adipose tissue dysfunction induced by obesity. Tissue remodeling is one of the histological features of chronic inflammation, in which stromal cells dramatically change in number and cell type. Indeed, adipose tissue remodeling is induced by various stromal cells, and results in the impairment of adipose tissue function, such as adipocytokine production and lipid storage, which leads to systemic metabolic disorder. In addition to adipose tissue, the liver is another example of obesity-induced tissue remodeling. In the present review, we discuss how obesity induces interstitial fibrosis in adipose tissue and the liver, particularly focusing on the role of macrophages.
Collapse
Affiliation(s)
- Miyako Tanaka
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
| | - Michiko Itoh
- Department of Organ Network and MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Yoshihiro Ogawa
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Department of Molecular Endocrinology and MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Department of Molecular and Cellular MetabolismGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Department of Medicine and Bioregulatory ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Japan Agency for Medical Research and DevelopmentCRESTTokyoJapan
| | - Takayoshi Suganami
- Department of Molecular Medicine and MetabolismResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
| |
Collapse
|
43
|
Itoh M, Suganami T, Kato H, Kanai S, Shirakawa I, Sakai T, Goto T, Asakawa M, Hidaka I, Sakugawa H, Ohnishi K, Komohara Y, Asano K, Sakaida I, Tanaka M, Ogawa Y. CD11c+ resident macrophages drive hepatocyte death-triggered liver fibrosis in a murine model of nonalcoholic steatohepatitis. JCI Insight 2017; 2:92902. [PMID: 29202448 DOI: 10.1172/jci.insight.92902] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022] Open
Abstract
Although recent evidence has pointed to the role of organ- and pathogenesis-specific macrophage subsets, it is still unclear which subsets are critically involved in the pathogenesis of nonalcoholic steatohepatitis (NASH). Using melanocortin-4 receptor-deficient (MC4R-KO) mice fed Western diet (WD), which exhibit liver phenotypes similar to those of human NASH, we found a histological structure, termed hepatic crown-like structure (hCLS), in which CD11c+ macrophages surround dead/dying hepatocytes, a prominent feature of NASH. Here, we demonstrate that hCLS-constituting macrophages could be a novel macrophage subset that drives hepatocyte death-triggered liver fibrosis. In an "inducible NASH model," hepatocyte death induces hCLS formation and liver fibrosis sequentially in the short term. In combination with the long-term WD feeding model, we also showed that resident macrophages are a major cellular source of CD11c+ macrophages constituting hCLS, which exhibited gene expression profiles distinct from CD11c- macrophages scattered in the liver. Moreover, depletion of CD11c+ macrophages abolished hCLS formation and fibrogenesis in NASH. Our clinical data suggest the role of CD11c+ macrophages in the disease progression from simple steatosis to NASH. This study sheds light on the role of resident macrophages, in addition to recruited macrophages, in the pathogenesis of NASH.
Collapse
Affiliation(s)
| | - Takayoshi Suganami
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Hideaki Kato
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Drug Discovery & Disease Research Laboratory, Shionogi & Co. Ltd., Osaka, Japan
| | - Sayaka Kanai
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Takeru Sakai
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiro Goto
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahiro Asakawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Isao Hidaka
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | - Koji Ohnishi
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Asano
- Laboratory of Immune regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Masato Tanaka
- Laboratory of Immune regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Japan Agency for Medical Research and Development, CREST, Tokyo, Japan
| |
Collapse
|
44
|
Abstract
Lectins recognize a diverse array of carbohydrate structures and perform numerous essential biological functions. Here we focus on only two families of lectins, the Siglecs and C-type lectins. Triggering of intracellular signaling cascades following ligand recognition by these receptors can have profound effects on the induction and modulation of immunity. In this chapter, we provide a brief overview of each family and then focus on selected examples that highlight how these lectins can influence myeloid cell functioning in health and disease. Receptors that are discussed include Sn (Siglec-1), CD33 (Siglec-3), and Siglec-5, -7, -8, -9, -10, -11, -14, -15, -E, -F, and -G as well as Dectin-1, MICL, Dectin-2, Mincle/MCL, and the macrophage mannose receptor.
Collapse
|
45
|
Adipocyte-Macrophage Cross-Talk in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:327-343. [DOI: 10.1007/978-3-319-48382-5_14] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Zhou L, Zhuo H, Ouyang H, Liu Y, Yuan F, Sun L, Liu F, Liu H. Glycoprotein non-metastatic melanoma protein b (Gpnmb) is highly expressed in macrophages of acute injured kidney and promotes M2 macrophages polarization. Cell Immunol 2017; 316:53-60. [PMID: 28433199 DOI: 10.1016/j.cellimm.2017.03.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/23/2017] [Accepted: 03/29/2017] [Indexed: 01/22/2023]
Abstract
Acute kidney injury (AKI) is an increasingly common disorder that is strongly linked to short- and long-term morbidity and mortality. During AKI process, macrophages, one of the important immune response cells, can polarize into M1 and M2 subtype from M0 subtype. It is well-known that M1 macrophages play a pro inflammatory role while M2 macrophages play an anti-inflammatory role. Glycoprotein non-metastatic melanoma protein b (Gpnmb) is a glycosylated transmembrane protein highly expressed in numerous cells, including osteoblasts, dendritic cells and macrophages. Gpnmb serves as a negative regulator of inflammation in macrophages and has a protective effect on injuries. In acute kidney injury, the macrophage has been shown diverse roles depending on different phenotype. This study provided gene expression and protein expression evidence that Gpnmb was highly expressed in M2 macrophages in the damaged areas of kidney after ischemia-reperfusion injury. Then, we successful isolated and culture mouse bone marrow-derived macrophages (BMMφ) and found that Gpnmb showed different expression levels in M0, M1 and M2 BMMφ: lowest in M1, highest in M2. After knocking down Gpnmb with si-Gpnmb, BMMφ M2 polarization and secretion of anti-inflammatory cytokines IL-10 and TGF-β were inhibited, while M1 polarization and secretion of proinflammatory cytokines IL-1β and TNF-α were promoted. Moreover, IL-4-STAT6 pathway was involved in the promotion of M2 polarization by Gpnmb. Taken together, Gpnmb may serve as a potential biomarker of AKI and play a protective role against the AKI by modulating the polarization of macrophage.
Collapse
Affiliation(s)
- Letian Zhou
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Hui Zhuo
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Huiyu Ouyang
- Nephrology Department, The 331 Hospital of Zhuzhou, Zhuzhou 412200, PR China
| | - Yexin Liu
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Fang Yuan
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Lin Sun
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Fuyou Liu
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China
| | - Hong Liu
- Nephrology Department, The Second Xiangya Hospital, Central South University, Key Lab of Kidney Disease and Blood Purification in Hunan, Changsha 410011, PR China.
| |
Collapse
|
47
|
The pattern recognition receptor, Mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation. Kidney Int 2017; 91:587-602. [DOI: 10.1016/j.kint.2016.10.020] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/29/2016] [Accepted: 10/13/2016] [Indexed: 01/23/2023]
|
48
|
The Role of Tissue Macrophage-Mediated Inflammation on NAFLD Pathogenesis and Its Clinical Implications. Mediators Inflamm 2017; 2017:8162421. [PMID: 28115795 PMCID: PMC5237469 DOI: 10.1155/2017/8162421] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/22/2016] [Accepted: 12/04/2016] [Indexed: 02/06/2023] Open
Abstract
The obese phenotype is characterized by a state of chronic low-grade systemic inflammation that contributes to the development of comorbidities, including nonalcoholic fatty liver disease (NAFLD). In fact, NAFLD is often associated with adipocyte enlargement and consequent macrophage recruitment and inflammation. Macrophage polarization is often associated with the proinflammatory state in adipose tissue. In particular, an increase of M1 macrophages number or of M1/M2 ratio triggers the production and secretion of various proinflammatory signals (i.e., adipocytokines). Next, these inflammatory factors may reach the liver leading to local M1/M2 macrophage polarization and consequent onset of the histological damage characteristic of NAFLD. Thus, the role of macrophage polarization and inflammatory signals appears to be central for pathogenesis and progression of NAFLD, even if the heterogeneity of macrophages and molecular mechanisms that govern their phenotype switch remain incompletely understood. In this review, we discuss the role of adipose and liver tissue macrophage-mediated inflammation in experimental and human NAFLD. This focus is relevant because it may help researchers that approach clinical and experimental studies on this disease advancing the knowledge of mechanisms that could be targeted in order to revert NAFLD-related fibrosis.
Collapse
|
49
|
Takeda Y, Shimomura T, Asao H, Wakabayashi I. Relationship between Immunological Abnormalities in Rat Models of Diabetes Mellitus and the Amplification Circuits for Diabetes. J Diabetes Res 2017; 2017:4275851. [PMID: 28299342 PMCID: PMC5337356 DOI: 10.1155/2017/4275851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/13/2016] [Accepted: 01/26/2017] [Indexed: 12/12/2022] Open
Abstract
A better understanding of pathogenic mechanisms is required in order to treat diseases. However, the mechanisms of diabetes mellitus and diabetic complications are extremely complex. Immune reactions are involved in the pathogenesis of diabetes and its complications, while diabetes influences immune reactions. Furthermore, both diabetes and immune reactions are influenced by genetic and environmental factors. To address these issues, animal models are useful tools. So far, various animal models of diabetes have been developed in rats, which have advantages over mice models in terms of the larger volume of tissue samples and the variety of type 2 diabetes models. In this review, we introduce rat models of diabetes and summarize the immune reactions in diabetic rat models. Finally, we speculate on the relationship between immune reactions and diabetic episodes. For example, diabetes-prone Biobreeding rats, type 1 diabetes model rats, exhibit increased autoreactive cellular and inflammatory immune reactions, while Goto-Kakizaki rats, type 2 diabetes model rats, exhibit increased Th2 reactions and attenuation of phagocytic activity. Investigation of immunological abnormalities in various diabetic rat models is useful for elucidating complicated mechanisms in the pathophysiology of diabetes. Studying immunological alterations, such as predominance of Th1/17 or Th2 cells, humoral immunity, and innate immune reactions, may improve understanding the structure of amplification circuits for diabetes in future studies.
Collapse
Affiliation(s)
- Yuji Takeda
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Nishinomiya, Japan
- Department of Immunology, Faculty of Medicine, Yamagata University, Yamagata, Japan
- *Yuji Takeda:
| | - Tomoko Shimomura
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hironobu Asao
- Department of Immunology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Ichiro Wakabayashi
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
50
|
The Pathogenesis of Obesity-Associated Adipose Tissue Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:221-245. [PMID: 28585201 DOI: 10.1007/978-3-319-48382-5_9] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|