1
|
Qiu X, Wen R, Wu F, Mao J, Azad T, Wang Y, Zhu J, Zhou X, Xie H, Hong K, Li B, Zhang L, Wen C. The role of double-negative B cells in the pathogenesis of systemic lupus erythematosus. Autoimmun Rev 2025; 24:103821. [PMID: 40274006 DOI: 10.1016/j.autrev.2025.103821] [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: 02/10/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
B cells are essential to the pathophysiology of systemic lupus erythematosus (SLE), a chronic autoimmune illness. IgD-CD27-double negative B cells (DNB cells) are one of the aberrant B cell subsets linked to SLE that have attracted much scientific interest. There is growing evidence that DNB cells play a significant role in the development of the disease and are strongly linked to the activity of lupus. These cells play a pivotal role in the pathogenesis of SLE by producing a diverse array of autoantibodies, which form immune complexes that drive target organ damage. A comprehensive understanding of SLE pathophysiology necessitates in-depth investigation into DNB cells, not only to elucidate their mechanistic contributions but also to uncover novel therapeutic strategies. According to available data, treatments that target B cells have proven effective in managing SLE; nevertheless, a significant breakthrough in precision medicine for SLE may come from targeting DNB cells specifically. Despite growing interest in DNB cells, their precise characteristics, developmental trajectories, and regulatory mechanisms remain incompletely defined, posing significant challenges to the field. A comprehensive investigation of the regulatory mechanisms governing DNB cell differentiation and expansion in SLE may facilitate novel therapeutic discoveries. This review aims to provide an updated synthesis of current research on DNB cells, with a focus on their origins, developmental trajectories in SLE, and potential as precision therapeutic targets.
Collapse
Affiliation(s)
- Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China; The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China
| | - RuiFan Wen
- Medical School, Hunan University of Chinese Medicine, No.300 Xueshi Road, Hanpu Science & Education District, Changsha, Hunan 410208, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Tasnim Azad
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Liang Zhang
- The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China; Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha 410007, Hunan, China.
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
2
|
Harirah HAA, Mohammed MH, Basha SAZ, Uthirapathy S, Ganesan S, Shankhyan A, Sharma GC, Devi A, Kadhim AJ, S NH. Targeting EZH2 in autoimmune diseases: unraveling epigenetic regulation and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04127-6. [PMID: 40198399 DOI: 10.1007/s00210-025-04127-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/29/2025] [Indexed: 04/10/2025]
Abstract
Approximately 8-10% of the global population is affected by autoimmune diseases (ADs), which encompass a wide array of idiopathic conditions resulting from dysregulated immune responses. The enzymatic component of the polycomb-repressive complex 2 (PRC2), enhancer of zeste homolog 2 (EZH2, also referred to as KMT6), functions as a methyltransferase possessing a SET domain that plays crucial roles in epigenetic regulation, explicitly facilitating the methylation of histone H3 at lysine 27. Notably, EZH2 is catalytically inactive and requires association with EED and SUZ12 to form an active PRC2 complex. Hyperactivation of EZH2 has been implicated in various malignancies, prompting the development of EZH2 inhibitors as therapeutic agents for several cancers, including lymphoma, prostate, breast, and colon cancer. The application of EZH2-targeting therapies has also been explored in the context of autoimmune diseases. While there have been advancements in certain ADs, responses can vary significantly, as evidenced by mixed outcomes in cases such as inflammatory bowel disease. Consequently, the dual role of EZH2 and the therapeutic potential of its inhibitors in the treatment of ADs remain nascent fields of study. This review will elucidate the interplay between EZH2 and autoimmune diseases, highlighting emerging insights and therapeutic avenues.
Collapse
Affiliation(s)
- Hashem Ahmed Abu Harirah
- Medical Laboratory Department, Faculty of Allied Medical Sciences, Zarqa University, Zarqa, Jordan.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | - Sami Ahmed Zaher Basha
- Physical Therapy Department, Faculty of Allied Medical Sciences, Zarqa University, Zarqa, Jordan
- Department of Cardiovascular Pulmonary and Geriatrics, Faculty of Physical Therapy, Pharos University, Alexandria, Egypt
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Kurdistan Region, Erbil, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Naher H S
- Laboratories Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
3
|
Gao J, Li C, Lin X, Zhuang Y, Wang M, Lin H, Zhu X. Role of IRF4 in mediating plasmablast differentiation in diffuse large B-cell lymphomas via mTORC1 pathway. Ann Hematol 2025; 104:2449-2459. [PMID: 40204935 PMCID: PMC12053343 DOI: 10.1007/s00277-025-06273-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/17/2025] [Indexed: 04/11/2025]
Abstract
Autoimmune haemolytic anaemia (AIHA) is common secondary to diffuse large B-cell lymphoma (DLBCL). However, there are no reports on tumour B-cells differentiating into plasmablasts in DLBCL secondary AIHA. To state impact of the interferon regulatory factor 4 (IRF4) on DLBCL and explore the mechanism of IRF4 on plasmablast differentiation.We analysed the expression of immunity-related genes from the Gene Expression Omnibus and correlated predictors from clinical and laboratory data using R package and various statistical tools. Western blotting (WB) was used to detect protein levels in DLBCL cell lines of different subtypes to investigate the plasmablast and activation of mTORC1. To furtherly validate mTOR regulation of plasmablast differentiation, mTOR-activated and -inhibited cell models were constructed by CCK8 and flow cytometry (FCM) was used to assess the proportion of CD38 positive cells. We found that IRF4 was highly expressed in activated B-cell-like (ABC) DLBCL vs. germinal centre B-cell-like (GCB) DLBCL. Positive MUM-1and low haemoglobin values were corrected to non-CGB patients. Plasmablast indictors (BLIMP-1, ARF4, IRE1α, and IRF4) and mTORC1 pathway indictors (mTOR, p70S6K and phosphorylated-p70S6K) were different level in ABC cell lines. After successfully constructing cell models, the proportion of CD38+ cells changed in mTOR-activated and -inhibited ABC-DLBCL cell models. We first pointed out that the role of the IRF4 invovling in DLBCL cell plasmablast differentiation via mTORC1 pathway. These findings could be extended to provide experimental evidence for novel treatments of secondary AIHA in ABC-DLBCL.
Collapse
Affiliation(s)
- Jingjing Gao
- Department of Blood transfusion, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Chuntuan Li
- Department of Hematology, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Xingzhi Lin
- Department of Blood transfusion, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Yanling Zhuang
- Department of Blood transfusion, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Mingquan Wang
- Department of Blood transfusion, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China
| | - Hongjun Lin
- Medical College of Huaqiao University, Quanzhou, Fujian, 362000, China
| | - Xiongpeng Zhu
- Department of Hematology, Quanzhou First Hospital affiliated to Fujian Medical University, Quanzhou, Fujian, 362000, China.
| |
Collapse
|
4
|
Yu P, Satyaraj E. Effect of Bovine Colostrum on Canine Immune Health. Animals (Basel) 2025; 15:185. [PMID: 39858185 PMCID: PMC11759141 DOI: 10.3390/ani15020185] [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: 11/15/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Colostrum, the first fluid secreted by the mammary glands of mammalian mothers, contains essential nutrients for the health and survival of newborns. Bovine colostrum (BC) is notable for its high concentrations of bioactive components, such as immunoglobulins and lactoferrin. Despite dogs being the world's most popular companion animals, there is limited research on their immune systems compared to humans. This summary aims to consolidate published studies that explore the immune benefits of BC, focusing specifically on its implications for dogs.
Collapse
Affiliation(s)
- Ping Yu
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| | - Ebenezer Satyaraj
- Nestlé Purina Research, One Checkerboard Square, St. Louis, MO 63164, USA
| |
Collapse
|
5
|
Ueno M, Iwata S, Yamagata K, Todoroki Y, Sonomoto K, Nagayasu A, Miyagawa I, Kubo S, Miyazaki Y, Miyata H, Kanda R, Aritomi T, Nakayamada S, Tanaka Y. Induction of interleukin 21 receptor expression via enhanced intracellular metabolism in B cells and its relevance to the disease activity in systemic lupus erythematosus. RMD Open 2024; 10:e004567. [PMID: 39740932 PMCID: PMC11749818 DOI: 10.1136/rmdopen-2024-004567] [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: 05/25/2024] [Accepted: 11/26/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE To elucidate the association between the changes in intracellular metabolism in the early stage of B cell activation and systemic lupus erythematosus (SLE) pathogenesis. METHODS CD19+ or CD19+CD27- (naïve) cells from the peripheral blood of healthy controls and lupus patients were cultured under different stimuli. The changes in intracellular metabolism and signalling pathways in these cells were evaluated. RESULTS Stimulation with CpG (Toll-like receptor 9 (TLR9) ligand) in vitro induced enhanced interleukin 21 (IL-21) receptor expression in CD19+CD27- cells after 24 hours. The addition of IL-21 to the CpG stimulation enhanced the extracellular acidification rate, which indicates glycolysis, within 30 min. IL-21 receptor (IL-21R) expression induced by CpG stimulation was selectively inhibited by 2-deoxy-D-glucose (hexokinase 2 (HK2) inhibitor) and heptelidic acid (glyceraldehyde 3-phosphate dehydrogenase (GAPDH) inhibitor). RNA immunoprecipitation with anti-GAPDH antibody revealed that CpG stimulation dissociated the binding between IL-21R messenger RNA (mRNA) and GAPDH under no stimulation. HK2 and GAPDH expression were higher in CD19+CD27- cells of lupus patients than in those of healthy controls, and GAPDH expression was correlated with the plasmocyte count and disease activity score. CONCLUSION IL-21R mRNA-GAPDH binding dissociation associated with rapid glycolytic enhancement by the TLR9 ligand in B cells may induce plasmocyte differentiation through IL-21 signals and be involved in exacerbating SLE.
Collapse
Affiliation(s)
- Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shigeru Iwata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
| | - Kaoru Yamagata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Clinical Nursing, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Atsushi Nagayasu
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Ippei Miyagawa
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yusuke Miyazaki
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Takafumi Aritomi
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| |
Collapse
|
6
|
Yang Y, Li M, Ding L, Zhang Y, Liu K, Liu M, Li Y, Luo H, Zuo X, Zhang H, Guo M. EZH2 promotes B-cell autoimmunity in primary Sjogren's syndrome via METTL3-mediated m6A modification. J Autoimmun 2024; 149:103341. [PMID: 39577129 DOI: 10.1016/j.jaut.2024.103341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE Enhancer of zeste homologue 2 (EZH2) plays an important role in promoting B-cell activation and differentiation. This study aimed to elucidate the role of EZH2 in the B-cell autoimmune response in primary Sjögren's syndrome (pSS) and to explore the therapeutic potential of inhibiting EZH2 in pSS. METHODS Single-cell RNA sequencing analysis of B cells in peripheral blood from pSS patients was conducted to identify abnormal expression of EZH2 and METTL3 in B-cell subsets. The levels of EZH2 were further validated across multiple B-cell subsets and the salivary glands (SGs) of pSS patients, as well as three different mouse models of Sjögren's syndrome (SS). Correlation analyses were performed to explore the relationship between the expression of EZH2 and clinical features of pSS patients. Following EZH2 inhibition, SS-like signs and antibody production were assessed in an experimental Sjögren syndrome (ESS) mouse model. RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) data post-EZH2 inhibition were bioinformatically analyzed to identify the EZH2 targets in pSS. ChIP-qPCR was performed to validate the binding of H3K27me3 to the CDKN1A promoter. Flow cytometric apoptosis analysis and Carboxy Fluorescein Succinimidyl Ester (CFSE) assay were used to assess the impact of an EZH2 inhibitor on B-cell apoptosis and proliferation. Additionally, METTL3 expression and its correlation with disease activity were analyzed in pSS patients. EZH2 expression was examined after METTL3 knockdown. METTL3-RNA immunoprecipitation (RIP) and actinomycin D assays were conducted to confirm the direct binding of METTL3 to EZH2 mRNA and its impact on mRNA stability. M6A-RIP-qPCR was performed to validate the presence of m6A modifications on EZH2 mRNA. RESULTS EZH2 was found upregulated in multiple B-cell subsets from the peripheral blood and SGs of pSS patients, as well as in three different animal models of SS. The expression of EZH2 in B cells was positively correlated with the ESSDAI score, which is a measure of disease activity. With treatment of EZH2 inhibitor, SS-like signs alleviated and autoantibody production reduced in ESS mice. Similarly, in pSS patients, METTL3 expression was increased in the SGs and peripheral blood CD19+ B cells, also showing a positively correlated with the ESSDAI score. With knockdown of METTL3, the expression of EZH2 reduced. Mechanistically, EZH2 inhibited B-cell apoptosis and promoted B-cell proliferation by catalyzing H3K27me3 modification at the CDKN1A locus. Furthermore, METTL3 bound to EZH2 mRNA and increased m6A modification on EZH2 mRNA, enhancing its stability and promoting EZH2 expression. CONCLUSIONS The upregulation of EZH2 mediated by METTL3 is implicated in the B-cell autoimmune response in pSS. Inhibition of EZH2 presents a promising therapeutic strategy for pSS treatment.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China; Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Muyuan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liqing Ding
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Zhang
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Yisha Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China.
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Zheng J, Bu X, Wei X, Ma X, Zhao P. The role of FoxM1 in immune cells. Clin Exp Med 2023; 23:1973-1979. [PMID: 36913035 DOI: 10.1007/s10238-023-01037-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/01/2023] [Indexed: 03/14/2023]
Abstract
Forkhead box M1 (FoxM1), a proliferation specific transcriptional modulator, plays a principal role in many physiological and pathological processes. FoxM1-mediated oncogenic processes have been well addressed. However, functions of FoxM1 in immune cells are less summarized. The literatures about the expression of FoxM1 and its regulation on immune cells were searched on PubMed and Google Scholar. In this review, we provide an overview on the roles of FoxM1 in regulating functions of immune cells, including T cells, B cells, monocytes, macrophages, and dendritic cells, and discuss their contributions to diseases.
Collapse
Affiliation(s)
- Jinju Zheng
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Xiaocui Bu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Xiaofang Wei
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Xuezhen Ma
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China.
| | - Peng Zhao
- Biotherapy Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
8
|
Zheng X, Dozmorov MG, Strohlein CE, Bastacky S, Sawalha AH. Ezh2 Knockout in B Cells Impairs Plasmablast Differentiation and Ameliorates Lupus-like Disease in MRL/lpr Mice. Arthritis Rheumatol 2023; 75:1395-1406. [PMID: 36897808 PMCID: PMC10492897 DOI: 10.1002/art.42492] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/25/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVES EZH2 regulates B cell development and differentiation. We previously demonstrated increased EZH2 expression in peripheral blood mononuclear cells from lupus patients. The goal of this study was to evaluate the role of EZH2 expression in B cells in the pathogenesis of lupus. METHODS We generated an MRL/lpr mouse with floxed Ezh2, which was crossed with CD19-Cre mice to examine the effect of B cell EZH2 deficiency in MRL/lpr lupus-prone mice. Differentiation of B cells was assessed using flow cytometry. Single-cell RNA sequencing and single-cell B cell receptor sequencing were performed. In vitro B cell culture with an X-box binding protein 1 (XBP1) inhibitor was performed. EZH2 and XBP1 messenger RNA levels in CD19+ B cells isolated from lupus patients and healthy controls were analyzed. RESULTS We show that Ezh2 deletion in B cells significantly decreased autoantibody production and improved glomerulonephritis. B cell development was altered in the bone marrow and spleen of EZH2-deficient mice. Differentiation of germinal center B cells and plasmablasts was impaired. Single-cell RNA sequencing showed that XBP1, a key transcription factor in B cell development, is down-regulated in the absence of EZH2. Inhibiting XBP1 in vitro impairs plasmablast development similar to EZH2 deficiency in mice. Single-cell B cell receptor RNA sequencing revealed defective immunoglobulin class-switch recombination in EZH2-deficient mice. In human lupus B cells, we observed a strong correlation between EZH2 and XBP1 messenger RNA expression levels. CONCLUSION EZH2 overexpression in B cells contributes to disease pathogenesis in lupus.
Collapse
Affiliation(s)
- Xiaoqing Zheng
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Colleen E Strohlein
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amr H Sawalha
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Lupus Center of Excellence, and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Feng L, Chen X, Huang Y, Zhang X, Zheng S, Xie N. Immunometabolism changes in fibrosis: from mechanisms to therapeutic strategies. Front Pharmacol 2023; 14:1243675. [PMID: 37576819 PMCID: PMC10412938 DOI: 10.3389/fphar.2023.1243675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Immune cells are essential for initiating and developing the fibrotic process by releasing cytokines and growth factors that activate fibroblasts and promote extracellular matrix deposition. Immunometabolism describes how metabolic alterations affect the function of immune cells and how inflammation and immune responses regulate systemic metabolism. The disturbed immune cell function and their interactions with other cells in the tissue microenvironment lead to the origin and advancement of fibrosis. Understanding the dysregulated metabolic alterations and interactions between fibroblasts and the immune cells is critical for providing new therapeutic targets for fibrosis. This review provides an overview of recent advances in the pathophysiology of fibrosis from the immunometabolism aspect, highlighting the altered metabolic pathways in critical immune cell populations and the impact of inflammation on fibroblast metabolism during the development of fibrosis. We also discuss how this knowledge could be leveraged to develop novel therapeutic strategies for treating fibrotic diseases.
Collapse
Affiliation(s)
- Lixiang Feng
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xingyu Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yujing Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaodian Zhang
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shaojiang Zheng
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
10
|
Tsai CY, Li KJ, Shen CY, Lu CH, Lee HT, Wu TH, Ng YY, Tsao YP, Hsieh SC, Yu CL. Decipher the Immunopathological Mechanisms and Set Up Potential Therapeutic Strategies for Patients with Lupus Nephritis. Int J Mol Sci 2023; 24:10066. [PMID: 37373215 PMCID: PMC10298725 DOI: 10.3390/ijms241210066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Lupus nephritis (LN) is one of the most severe complications in patients with systemic lupus erythematosus (SLE). Traditionally, LN is regarded as an immune complex (IC) deposition disease led by dsDNA-anti-dsDNA-complement interactions in the subendothelial and/or subepithelial basement membrane of glomeruli to cause inflammation. The activated complements in the IC act as chemoattractants to chemically attract both innate and adaptive immune cells to the kidney tissues, causing inflammatory reactions. However, recent investigations have unveiled that not only the infiltrating immune-related cells, but resident kidney cells, including glomerular mesangial cells, podocytes, macrophage-like cells, tubular epithelial cells and endothelial cells, may also actively participate in the inflammatory and immunological reactions in the kidney. Furthermore, the adaptive immune cells that are infiltrated are genetically restricted to autoimmune predilection. The autoantibodies commonly found in SLE, including anti-dsDNA, are cross-reacting with not only a broad spectrum of chromatin substances, but also extracellular matrix components, including α-actinin, annexin II, laminin, collagen III and IV, and heparan sulfate proteoglycan. Besides, the glycosylation on the Fab portion of IgG anti-dsDNA antibodies can also affect the pathogenic properties of the autoantibodies in that α-2,6-sialylation alleviates, whereas fucosylation aggravates their nephritogenic activity. Some of the coexisting autoantibodies, including anti-cardiolipin, anti-C1q, anti-ribosomal P autoantibodies, may also enhance the pathogenic role of anti-dsDNA antibodies. In clinical practice, the identification of useful biomarkers for diagnosing, monitoring, and following up on LN is quite important for its treatments. The development of a more specific therapeutic strategy to target the pathogenic factors of LN is also critical. We will discuss these issues in detail in the present article.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Immunology & Rheumatology, Department of Medicine, Fu Jen Catholic University Hospital & College of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan
| | - Ko-Jen Li
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Chieh-Yu Shen
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Cheng-Hsun Lu
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Hui-Ting Lee
- MacKay Memorial Hospital & MacKay Medical College, New Taipei City 25245, Taiwan;
| | - Tsai-Hung Wu
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital and Faculty of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112304, Taiwan;
| | - Yee-Yung Ng
- Department of Medicine, Fu Jen Catholic University Hospital & College of Medicine, Fu Jen Catholic University, New Taipei City 24352, Taiwan;
| | - Yen-Po Tsao
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital and Faculty of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112304, Taiwan;
| | - Song-Chou Hsieh
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| | - Chia-Li Yu
- Division of Rheumatology, Immunology & Allergy, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 106319, Taiwan; (K.-J.L.); (C.-Y.S.); (C.-H.L.); (S.-C.H.)
| |
Collapse
|
11
|
Iwata S, Hajime Sumikawa M, Tanaka Y. B cell activation via immunometabolism in systemic lupus erythematosus. Front Immunol 2023; 14:1155421. [PMID: 37256149 PMCID: PMC10225689 DOI: 10.3389/fimmu.2023.1155421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease involving multiple organs in which B cells perform important functions such as antibody and cytokine production and antigen presentation. B cells are activated and differentiated by the primary B cell receptor, co-stimulatory molecule signals-such as CD40/CD40L-, the Toll-like receptors 7,9, and various cytokine signals. The importance of immunometabolism in the activation, differentiation, and exerting functions of B cells and other immune cells has been widely reported in recent years. However, the regulatory mechanism of immunometabolism in B cells and its involvement in SLE pathogenesis remain elusive. Similarly, the importance of the PI3K-Akt-mTOR signaling pathway, glycolytic system, and oxidative phosphorylation has been demonstrated in the mechanisms of B cell immunometabolic activation, mainly in mouse studies. However, the activation of the mTOR pathway in B cells in patients with SLE, the induction of plasmablast differentiation through metabolic and transcription factor regulation by mTOR, and the involvement of this phenomenon in SLE pathogenesis are unclear. In our studies using activated B cells derived from healthy donors and from patients with SLE, we observed that methionine, an essential amino acid, is important for mTORC1 activation. Further, we observed that splenic tyrosine kinase and mTORC1 activation synergistically induce EZH2 expression and plasmablasts by suppressing BACH2 expression through epigenomic modification. Additionally, we identified another mechanism by which the glutaminolysis-induced enhancement of mitochondrial function promotes plasmablast differentiation in SLE. In this review, we focused on the SLE exacerbation mechanisms related to the activation of immune cells-especially B cells-and immunometabolism and reported the latest findings in the field.
Collapse
Affiliation(s)
- Shigeru Iwata
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Maiko Hajime Sumikawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
12
|
Wu Y, Zhao M, Gong N, Zhang F, Chen W, Liu Y. Immunometabolomics provides a new perspective for studying systemic lupus erythematosus. Int Immunopharmacol 2023; 118:109946. [PMID: 36931174 DOI: 10.1016/j.intimp.2023.109946] [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: 12/13/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multi-organ autoimmune disease characterized by clinical heterogeneity, unpredictable progression, and flare ups. Due to the heterogeneous nature of lupus, it has been challenging to identify sensitive and specific biomarkers for its diagnosis and monitoring. Despite the fact that the mechanism of SLE remains unknown, impressive progress has been made over the last decade towards understanding how different immune cells contribute to its pathogenesis. Research suggests that cellular metabolic programs could affect the immune response by regulating the activation, proliferation, and differentiation of innate and adaptive immune cells. Many studies have shown that the dysregulation of the immune system is associated with changes to metabolite profiles. The study of metabolite profiling may provide a means for mechanism exploration and novel biomarker discovery for disease diagnostic, classification, and monitoring. Here we review the latest advancements in understanding the role of immunometabolism in SLE, as well as the systemic metabolite profiling of this disease along with possible clinical application.
Collapse
Affiliation(s)
- Yuxian Wu
- College of Basic Medicine, Naval Medical University, Shanghai, China
| | - Mengpei Zhao
- Department of Pharmacy, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Na Gong
- College of Basic Medicine, Naval Medical University, Shanghai, China
| | - Feng Zhang
- Department of Pharmacy, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Yaoyang Liu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
13
|
Xiao F, Rui K, Shi X, Wu H, Cai X, Lui KO, Lu Q, Ballestar E, Tian J, Zou H, Lu L. Epigenetic regulation of B cells and its role in autoimmune pathogenesis. Cell Mol Immunol 2022; 19:1215-1234. [PMID: 36220996 PMCID: PMC9622816 DOI: 10.1038/s41423-022-00933-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
B cells play a pivotal role in the pathogenesis of autoimmune diseases. Although previous studies have shown many genetic polymorphisms associated with B-cell activation in patients with various autoimmune disorders, progress in epigenetic research has revealed new mechanisms leading to B-cell hyperactivation. Epigenetic mechanisms, including those involving histone modifications, DNA methylation, and noncoding RNAs, regulate B-cell responses, and their dysregulation can contribute to the pathogenesis of autoimmune diseases. Patients with autoimmune diseases show epigenetic alterations that lead to the initiation and perpetuation of autoimmune inflammation. Moreover, many clinical and animal model studies have shown the promising potential of epigenetic therapies for patients. In this review, we present an up-to-date overview of epigenetic mechanisms with a focus on their roles in regulating functional B-cell subsets. Furthermore, we discuss epigenetic dysregulation in B cells and highlight its contribution to the development of autoimmune diseases. Based on clinical and preclinical evidence, we discuss novel epigenetic biomarkers and therapies for patients with autoimmune disorders.
Collapse
Affiliation(s)
- Fan Xiao
- Department of Pathology, Shenzhen Institute of Research and Innovation and Shenzhen Hospital, The University of Hong Kong, Hong Kong; Chongqing International Institute for Immunology, Chongqing, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital and School of Medicine, Henan University of Science and Technology, Luoyang, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Kathy O Lui
- Department of Chemical Pathology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute, Badalona, 08916, Barcelona, Spain
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center, East China Normal University, Shanghai, China
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Liwei Lu
- Department of Pathology, Shenzhen Institute of Research and Innovation and Shenzhen Hospital, The University of Hong Kong, Hong Kong; Chongqing International Institute for Immunology, Chongqing, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
14
|
Yang Y, Liu K, Liu M, Zhang H, Guo M. EZH2: Its regulation and roles in immune disturbance of SLE. Front Pharmacol 2022; 13:1002741. [DOI: 10.3389/fphar.2022.1002741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is related to immune homeostasis imbalance. Epigenetic mechanisms have played a significant role in breaking immune tolerance. Enhancer of zeste homolog 2 (EZH2), the specific methylation transferase of lysine at position 27 of histone 3, is currently found to participate in the pathogenesis of SLE through affecting multiple components of the immune system. This review mainly expounds the mechanisms underlying EZH2-mediated disruption of immune homeostasis in SLE patients, hoping to provide new ideas in the pathogenesis of SLE and new targets for future treatment.
Collapse
|
15
|
Xu WD, Huang Q, Huang AF. Emerging role of EZH2 in rheumatic diseases: A comprehensive review. Int J Rheum Dis 2022; 25:1230-1238. [PMID: 35933601 DOI: 10.1111/1756-185x.14416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/03/2022] [Accepted: 07/23/2022] [Indexed: 11/29/2022]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone methylated enzyme. It trimethylates histone 3 lysine 27 (H3K27) to regulate epigenetic processes. Recently, studies showed excessive expression of EZH2 in rheumatic diseases, such as systemic lupus erythematosus, rheumatoid arthritis, osteoarthritis, and systemic sclerosis. Moreover, epigenetic modification of EZH2 regulates differentiation and proliferation of different immune cells. Therefore, in this review, we comprehensively discuss the role of EZH2 in rheumatic diseases. Collection of the evidence may provide a basis for further understanding the role of EZH2 and give potential for targeting these diseases.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, China
| | - Qi Huang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
16
|
Liu Y, Cui DX, Pan Y, Yu SH, Zheng LW, Wan M. Metabolic-epigenetic nexus in regulation of stem cell fate. World J Stem Cells 2022; 14:490-502. [PMID: 36157525 PMCID: PMC9350619 DOI: 10.4252/wjsc.v14.i7.490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
Stem cell fate determination is one of the central questions in stem cell biology, and although its regulation has been studied at genomic and proteomic levels, a variety of biological activities in cells occur at the metabolic level. Metabolomics studies have established the metabolome during stem cell differentiation and have revealed the role of metabolites in stem cell fate determination. While metabolism is considered to play a biological regulatory role as an energy source, recent studies have suggested the nexus between metabolism and epigenetics because several metabolites function as cofactors and substrates in epigenetic mechanisms, including histone modification, DNA methylation, and microRNAs. Additionally, the epigenetic modification is sensitive to the dynamic metabolites and consequently leads to changes in transcription. The nexus between metabolism and epigenetics proposes a novel stem cell-based therapeutic strategy through manipulating metabolites. In the present review, we summarize the possible nexus between metabolic and epigenetic regulation in stem cell fate determination, and discuss the potential preventive and therapeutic strategies via targeting metabolites.
Collapse
Affiliation(s)
- Yi Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Di-Xin Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yue Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Si-Han Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Wei Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mian Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
17
|
He C, Yang Y, Chen Z, Liu S, Lyu T, Zeng L, Wang L, Li Y, Wang M, Chen H, Zhang F. EZH2 Promotes T Follicular Helper Cell Differentiation Through Enhancing STAT3 Phosphorylation in Patients With Primary Sjögren’s Syndrome. Front Immunol 2022; 13:922871. [PMID: 35795677 PMCID: PMC9252457 DOI: 10.3389/fimmu.2022.922871] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesEnhancer of zeste homolog 2 (EZH2) is an epigenetic regulator that plays an essential role in immune system development and autoimmune diseases. This study aimed to characterize the role of EZH2 in the pathogenesis of primary Sjögren’s syndrome (pSS).MethodsWe analyzed EZH2 expression in two transcriptomic datasets of peripheral blood mononuclear cells (PBMCs) from pSS patients and healthy controls. We measured EZH2 expression in CD4+ T cells, CD8+ T cells, and CD19+ B cells from pSS patients and healthy controls and correlated EZH2 expression with clinical parameters. We also examined the activation, proliferation, and T-cell differentiation of CD4+ T cells using the EZH2 inhibitor GSK126, EZH2 siRNA, and EZH2-expressing vector. We further examined the STAT3 signaling pathway after EZH2 inhibition and detected Tfh differentiation in EZH2-overexpressed CD4+ T cells with STAT3 knocked down.ResultsEZH2 was upregulated in GSE164885 and GSE48378. EZH2 expression was higher in pSS CD4+ and CD8+ T cells, and EZH2 expression in circulating pSS CD4+ T cells was positively correlated with IgG, IgA, ESR, RF, and the circulating Tfh population. EZH2 inhibition and silencing EZH2 suppressed activation, proliferation, and Tfh differentiation. Furthermore, overexpressing EZH2 promoted activation, proliferation, and Tfh differentiation in CD4+ T cells. EZH2 inhibition attenuated STAT3 phosphorylation in CD4+ T cells. STAT3 knockdown abrogated EZH2-promoted Tfh differentiation.ConclusionsEZH2 expression was abnormally elevated in pSS CD4+ T cells, which facilitated Tfh differentiation of CD4+ T cells by enhancing STAT3 phosphorylation. EZH2 promotes Tfh differentiation and might be implicated in pSS pathogenesis.
Collapse
Affiliation(s)
- Chengmei He
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yanlei Yang
- Medical Science Research Centre, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhilei Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Suying Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Taibiao Lyu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Mu Wang
- Department of Stomatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
- *Correspondence: Hua Chen, ; Fengchun Zhang,
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
- *Correspondence: Hua Chen, ; Fengchun Zhang,
| |
Collapse
|
18
|
Fu Y, Wang L, Yu B, Xu D, Chu Y. Immunometabolism shapes B cell fate and functions. Immunology 2022; 166:444-457. [PMID: 35569110 DOI: 10.1111/imm.13499] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ying Fu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
- Department of Endocrinology and Metabolism, Shanghai Fifth People's Hospital Fudan University Shanghai China
- Biotherapy Research Center Fudan University Shanghai China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
| | - Damo Xu
- School of Medicine Shenzhen University Shenzhen China
- Third Affiliated Hospital of Shenzhen University Shenzhen Luohu Hospital Group Shenzhen China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences Fudan University Shanghai China
- Biotherapy Research Center Fudan University Shanghai China
| |
Collapse
|
19
|
Calciolari B, Scarpinello G, Tubi LQ, Piazza F, Carrer A. Metabolic control of epigenetic rearrangements in B cell pathophysiology. Open Biol 2022; 12:220038. [PMID: 35580618 PMCID: PMC9113833 DOI: 10.1098/rsob.220038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/26/2022] [Indexed: 01/04/2023] Open
Abstract
Both epigenetic and metabolic reprogramming guide lymphocyte differentiation and can be linked, in that metabolic inputs can be integrated into the epigenome to inform cell fate decisions. This framework has been thoroughly investigated in several pathophysiological contexts, including haematopoietic cell differentiation. In fact, metabolite availability dictates chromatin architecture and lymphocyte specification, a multi-step process where haematopoietic stem cells become terminally differentiated lymphocytes (effector or memory) to mount the adaptive immune response. B and T cell precursors reprogram their cellular metabolism across developmental stages, not only to meet ever-changing energetic demands but to impose chromatin accessibility and regulate the function of master transcription factors. Metabolic control of the epigenome has been extensively investigated in T lymphocytes, but how this impacts type-B life cycle remains poorly appreciated. This assay will review our current understanding of the connection between cell metabolism and epigenetics at crucial steps of B cell maturation and how its dysregulation contributes to malignant transformation.
Collapse
Affiliation(s)
- Beatrice Calciolari
- Department of Biology (DiBio), of the University of Padova, Padova, Italy
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, of the University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Greta Scarpinello
- Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), of the University of Padova, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, of the University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, of the University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Alessandro Carrer
- Department of Biology (DiBio), of the University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
20
|
Abstract
Significance: Epigenetic dysregulation plays an important role in the pathogenesis and development of autoimmune diseases. Oxidative stress is associated with autoimmunity and is also known to alter epigenetic mechanisms. Understanding the interplay between oxidative stress and epigenetics will provide insights into the role of environmental triggers in the development of autoimmunity in genetically susceptible individuals. Recent Advances: Abnormal DNA and histone methylation patterns in genes and pathways involved in interferon and tumor necrosis factor signaling, cellular survival, proliferation, metabolism, organ development, and autoantibody production have been described in autoimmunity. Inhibitors of DNA and histone methyltransferases showed potential therapeutic effects in animal models of autoimmune diseases. Oxidative stress can regulate epigenetic mechanisms via effects on DNA damage repair mechanisms, cellular metabolism and the local redox environment, and redox-sensitive transcription factors and pathways. Critical Issues: Studies looking into oxidative stress and epigenetics in autoimmunity are relatively limited. The number of available longitudinal studies to explore the role of DNA methylation in the development of autoimmune diseases is small. Future Directions: Exploring the relationship between oxidative stress and epigenetics in autoimmunity will provide clues for potential preventative measures and treatment strategies. Inception cohorts with longitudinal follow-up would help to evaluate epigenetic marks as potential biomarkers for disease development, progression, and treatment response in autoimmunity. Antioxid. Redox Signal. 36, 423-440.
Collapse
Affiliation(s)
- Xiaoqing Zheng
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
21
|
Iwata S, Tanaka Y. Association of Viral Infection With the Development and Pathogenesis of Systemic Lupus Erythematosus. Front Med (Lausanne) 2022; 9:849120. [PMID: 35280878 PMCID: PMC8914279 DOI: 10.3389/fmed.2022.849120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that causes multiple organ damage in women of childbearing age and has a relapsing-remitting course. SLE is caused by the interaction between genetic and environmental factors, however, its underlying triggers remain unknown. Among the environmental factors, the involvement of infections as a trigger for SLE, especially those of viral etiology, has been widely reported. Human endogenous retroviruses (HERVs) may put patients at a genetic predisposition to SLE, while the Epstein-Barr virus (EBV) may play a role as an environmental factor that triggers the development of SLE. It has been suggested that EBV-infected B-cells may become resistant to apoptosis, resulting in the activation, proliferation, and antibody production of autoreactive B-cells, which cause tissue damage in SLE. However, the interaction between the virus and immune cells, as well as the impact of the virus on the differentiation and dysfunction of immune cells, remain unclear. In this review, we focus on the relationship between the development and pathogenesis of SLE and viral infections, as well as the mechanism of SLE exacerbation via activation of immune cells, such as B-cells, based on the latest findings.
Collapse
|
22
|
Sumikawa MH, Iwata S, Zhang M, Miyata H, Ueno M, Todoroki Y, Nagayasu A, Kanda R, Sonomoto K, Torimoto K, Lee S, Nakayamada S, Yamamoto K, Okada Y, Tanaka Y. An enhanced mitochondrial function through glutamine metabolism in plasmablast differentiation in systemic lupus erythematosus. Rheumatology (Oxford) 2021; 61:3049-3059. [PMID: 34730825 DOI: 10.1093/rheumatology/keab824] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/23/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To evaluate the dysfunction of B cell metabolism and its involvement in SLE pathology. METHODS We assessed the expression of metabolic markers of B cells in the peripheral blood of healthy controls (HCs) and SLE patients by using flow cytometry. In vitro, peripheral B cells were isolated from HCs and SLE patients to investigate the metabolic regulation mechanisms involved in their differentiation. RESULTS The expression level of DiOc6 (mitochondrial membrane hyperpolarization) was higher in B cells from SLE patients than in HCs, and correlated to the percentage of plasmablasts in CD19+ cells and with SLEDAI, a disease activity score. Stimulation of CD19+ cells with the Toll-like receptor 9 (TLR9) ligand CpG and IFN-α enhanced glycolysis, oxidative phosphorylation (OXPHOS), DiOc6 expression, and plasmablast differentiation in vitro. In the absence of glutamine, both glycolysis and OXPHOS were reduced, and plasmablast differentiation was suppressed, whereas there was no change in the absence of glucose. As glutamine is an important nutrient for protein synthesis, we further investigated the effect of the glutaminase inhibitor BPTES, which inhibits glutamine degradation, on metabolic regulation. BPTES reduced DiOc6 expression, OXPHOS, reactive oxygen species (ROS) production, ATP production, plasmablast differentiation without affecting glycolysis. Metformin inhibited CpG- and IFN-α-induced glutamine uptake, mitochondrial functions and suppressed plasmablast differentiation. CONCLUSIONS Mitochondrial dysfunction in B cells is associated with plasmablast differentiation and disease activity in SLE. Enhanced mitochondrial functions mediated by glutamine metabolism are important for plasmablast differentiation, which may be a potential therapeutic target for SLE.
Collapse
Affiliation(s)
- Maiko Hajime Sumikawa
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Shigeru Iwata
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Mingzeng Zhang
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroko Miyata
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Atsushi Nagayasu
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Keiichi Torimoto
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Seunghyun Lee
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Kazuo Yamamoto
- Biomedical Research Support Center, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Yosuke Okada
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
23
|
Yang M, Yi P, Jiang J, Zhao M, Wu H, Lu Q. Dysregulated translational factors and epigenetic regulations orchestrate in B cells contributing to autoimmune diseases. Int Rev Immunol 2021; 42:1-25. [PMID: 34445929 DOI: 10.1080/08830185.2021.1964498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
B cells play a crucial role in antigen presentation, antibody production and pro-/anti-inflammatory cytokine secretion in adaptive immunity. Several translational factors including transcription factors and cytokines participate in the regulation of B cell development, with the cooperation of epigenetic regulations. Autoimmune diseases are generally characterized with autoreactive B cells and high-level pathogenic autoantibodies. The success of B cell depletion therapy in mouse model and clinical trials has proven the role of B cells in pathogenesis of autoimmune diseases. The failure of B cell tolerance in immune checkpoints results in accumulated autoreactive naïve B (BN) cells with aberrant B cell receptor signaling and dysregulated B cell response, contributing to self-antibody-mediated autoimmune reaction. Dysregulation of translational factors and epigenetic alterations in B cells has been demonstrated to correlate with aberrant B cell compartment in autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, primary Sjögren's syndrome, multiple sclerosis, diabetes mellitus and pemphigus. This review is intended to summarize the interaction of translational factors and epigenetic regulations that are involved with development and differentiation of B cells, and the mechanism of dysregulation in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Ming Yang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ping Yi
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jiao Jiang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.,Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| |
Collapse
|
24
|
Iwata S, Tanaka Y. Therapeutic perspectives on the metabolism of lymphocytes in patients with rheumatoid arthritis and systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:1121-1130. [PMID: 34351835 DOI: 10.1080/1744666x.2021.1964957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The activation of autoreactive T- and B-cells and production of autoantibodies by B cells are involved in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Recently, the concept of 'immunometabolism' has attracted significant attention. Immune cells produce large amounts of energy in the form of ATP and biosynthesize biological components such as nucleic acids and lipids via metabolic reprogramming to activate, differentiate, and exert their functions. AREAS COVERED While the mechanisms underlying the metabolism of CD4+ T cells in SLE have been extensively studied, the metabolic changes underlying B cell activation, differentiation, and function remain unclear. Drugs targeting mTOR and AMPK, such as sirolimus, rapamycin, and metformin, have shown some efficacy and tolerability in clinical trials on patients with SLE, but have not led to breakthroughs. In this review, we summarize the current knowledge on the immunometabolic mechanisms involved in SLE and RA and discuss the potential novel therapeutic drugs. EXPERT OPINION The intensity of activation of different immune cells and their metabolic kinetics vary in different autoimmune diseases; thus, understanding the disease- and cell-specific metabolic mechanisms may help in the development of clinically effective immunometabolism-targeting drugs.
Collapse
Affiliation(s)
- Shigeru Iwata
- The First Department of Internal Medicine, Assistant Professor, University of Occupational and Environmental Health, Japan, School of Medicine, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, Professor and Chairman, Deputy Director, University of Occupational and Environmental Health, Japan, the University Hospital, School of Medicine, Kitakyushu, Japan
| |
Collapse
|
25
|
Tanaka Y, Millson D, Iwata S, Nakayamada S. Safety and efficacy of fostamatinib in rheumatoid arthritis patients with an inadequate response to methotrexate in phase II OSKIRA-ASIA-1 and OSKIRA-ASIA-1X study. Rheumatology (Oxford) 2021; 60:2884-2895. [PMID: 33254235 DOI: 10.1093/rheumatology/keaa732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/26/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE The primary objectives of two phase II studies of fostamatinib were to evaluate efficacy (OSKIRA-Asia-1: NCT01569074) and long-term safety/tolerability (OSKIRA-Asia-1X: NCT01640054) in patients from Asia with active RA despite MTX treatment. METHODS OSKIRA-Asia-1 was a 12-week, multicentre, double-blind, placebo-controlled, parallel-group study. Patients were randomized to receive one of four fostamatinib doses (groups A-D; n = 31, 33, 33, 33) or placebo (group E; n = 33). OSKIRA-Asia-1X was a long-term extension study (100 mg fostamatinib qd) of patients who completed OSKIRA-Asia-1. RA signs and symptoms were measured by ACR response criteria and DAS based on a 28-joint count. Physical function status was assessed with the HAQ-Disability Index. Safety findings were monitored. RESULTS In OSKIRA-Asia-1, fostamatinib revealed numerical improvements in ACR 20% response (ACR20) at week 12 in group A (100 mg bid) and group B (100 mg bid, then 150 mg qd) vs placebo. Statistically significant improvements in ACR20 and ACR50 at week 8 and in ACR70 at week 12, and statistically significant achievement in low disease activity (defined as DAS based on a 28-joint count ≤3.2 based on C-reactive protein) occurred in groups A and B. Improvement in physical function was numerically higher in group A. The most common adverse events were hypertension, diarrhoea and neutropenia. In OSKIRA-Asia-1X, the most common adverse events were nasopharyngitis, hypertension, RA and neutropenia. CONCLUSION Fostamatinib achieved both statistically and clinically significant improvements in RA signs and symptoms. The safety and tolerability of fostamatinib (plus MTX) were consistent with previous studies. TRIAL REGISTRATION OSKIRA-Asia-1 trial registration: https://clinicaltrials.gov, NCT01569074; OSKIRA-Asia-1X trial registration: https://clinicaltrials.gov, NCT01640054.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | - Shigeru Iwata
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
26
|
Wu L, Jiang X, Qi C, Zhang C, Qu B, Shen N. EZH2 Inhibition Interferes With the Activation of Type I Interferon Signaling Pathway and Ameliorates Lupus Nephritis in NZB/NZW F1 Mice. Front Immunol 2021; 12:653989. [PMID: 33868295 PMCID: PMC8044841 DOI: 10.3389/fimmu.2021.653989] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone-lysine N-methyltransferase mediating trimethylation of H3K27, which represses gene expression and is critical to immune regulation. Inhibition of EZH2 is proved to have the potential of treating many diseases. However, whether inhibition of EZH2 affects type I interferon (IFN-I) signaling pathway, the abnormality of which is an important pathogenic mechanism for SLE, is still elusive. Here, we report, unexpectedly, a positive regulatory function of EZH2 in IFN-I signaling pathway, which contributes to the overactivation of IFN-I signaling pathway in SLE. We show that the expression of EZH2 was upregulated and positively correlated with the overexpression of interferon stimulated genes (ISGs) in both peripheral blood mononuclear cells and renal tissues of SLE patients. In vitro inhibition of EZH2 by either siRNAs or chemical inhibitors reduced the phosphorylation of STAT1 and the induction of ISGs stimulated by IFN-I. Additionally, inhibition of EZH2 interfered with the in vivo and ex vivo activation of IFN-I signaling pathway elicited by intravenous injection of adenovirus vector expressing mouse IFN-α5 and exogeneous stimulation with IFN-α, respectively. We evaluated the therapeutic effects of EZH2 inhibitor in NZB/NZW F1 mice which depend on IFN-I signaling pathway for the lupus-like disease development. Administration of EZH2 inhibitor prolonged the survival, reduced the levels of anti-dsDNA autoantibodies, and improved lupus nephritis of the mice. What’s more, EZH2 inhibitor attenuated the expression of ISGs in the kidneys of these mice. In summary, we show that excessive EZH2 contributes to the overactivation of IFN-I signaling pathway in SLE. EZH2 inhibitor has the potential to inhibit IFN-I signaling pathway and alleviate lupus nephritis. Additionally, diverse disease driving pathways exist among systemic lupus erythematosus (SLE) patient, and even in the same patients. Common regulators of different pathogenic pathways can be multivalent therapeutic targets. Together with previous studies showing EZH2 is involved in T-cell and B-cell mediated immune responses, EZH2 could be a potent multivalent therapeutic target for SLE.
Collapse
Affiliation(s)
- Lingling Wu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Jiang
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaojun Qi
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunyan Zhang
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Qu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
27
|
Acevedo N, Alashkar Alhamwe B, Caraballo L, Ding M, Ferrante A, Garn H, Garssen J, Hii CS, Irvine J, Llinás-Caballero K, López JF, Miethe S, Perveen K, Pogge von Strandmann E, Sokolowska M, Potaczek DP, van Esch BCAM. Perinatal and Early-Life Nutrition, Epigenetics, and Allergy. Nutrients 2021; 13:724. [PMID: 33668787 PMCID: PMC7996340 DOI: 10.3390/nu13030724] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Epidemiological studies have shown a dramatic increase in the incidence and the prevalence of allergic diseases over the last several decades. Environmental triggers including risk factors (e.g., pollution), the loss of rural living conditions (e.g., farming conditions), and nutritional status (e.g., maternal, breastfeeding) are considered major contributors to this increase. The influences of these environmental factors are thought to be mediated by epigenetic mechanisms which are heritable, reversible, and biologically relevant biochemical modifications of the chromatin carrying the genetic information without changing the nucleotide sequence of the genome. An important feature characterizing epigenetically-mediated processes is the existence of a time frame where the induced effects are the strongest and therefore most crucial. This period between conception, pregnancy, and the first years of life (e.g., first 1000 days) is considered the optimal time for environmental factors, such as nutrition, to exert their beneficial epigenetic effects. In the current review, we discussed the impact of the exposure to bacteria, viruses, parasites, fungal components, microbiome metabolites, and specific nutritional components (e.g., polyunsaturated fatty acids (PUFA), vitamins, plant- and animal-derived microRNAs, breast milk) on the epigenetic patterns related to allergic manifestations. We gave insight into the epigenetic signature of bioactive milk components and the effects of specific nutrition on neonatal T cell development. Several lines of evidence suggest that atypical metabolic reprogramming induced by extrinsic factors such as allergens, viruses, pollutants, diet, or microbiome might drive cellular metabolic dysfunctions and defective immune responses in allergic disease. Therefore, we described the current knowledge on the relationship between immunometabolism and allergy mediated by epigenetic mechanisms. The knowledge as presented will give insight into epigenetic changes and the potential of maternal and post-natal nutrition on the development of allergic disease.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Bilal Alashkar Alhamwe
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - James Irvine
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kevin Llinás-Caballero
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Juan Felipe López
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
28
|
Bacalao MA, Satterthwaite AB. Recent Advances in Lupus B Cell Biology: PI3K, IFNγ, and Chromatin. Front Immunol 2021; 11:615673. [PMID: 33519824 PMCID: PMC7841329 DOI: 10.3389/fimmu.2020.615673] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/26/2020] [Indexed: 12/18/2022] Open
Abstract
In the autoimmune disease Systemic Lupus Erythematosus (SLE), autoantibodies are formed that promote inflammation and tissue damage. There has been significant interest in understanding the B cell derangements involved in SLE pathogenesis. The past few years have been particularly fruitful in three domains: the role of PI3K signaling in loss of B cell tolerance, the role of IFNγ signaling in the development of autoimmunity, and the characterization of changes in chromatin accessibility in SLE B cells. The PI3K pathway coordinates various downstream signaling molecules involved in B cell development and activation. It is governed by the phosphatases PTEN and SHIP-1. Murine models lacking either of these phosphatases in B cells develop autoimmune disease and exhibit defects in B cell tolerance. Limited studies of human SLE B cells demonstrate reduced expression of PTEN or increased signaling events downstream of PI3K in some patients. IFNγ has long been known to be elevated in both SLE patients and mouse models of lupus. New data suggests that IFNγR expression on B cells is required to develop autoreactive germinal centers (GC) and autoantibodies in murine lupus. Furthermore, IFNγ promotes increased transcription of BCL6, IL-6 and T-bet in B cells, which also promote GC and autoantibody formation. IFNγ also induces epigenetic changes in human B cells. SLE B cells demonstrate significant epigenetic reprogramming, including enhanced chromatin accessibility at transcription factor motifs involved in B cell activation and plasma cell (PC) differentiation as well as alterations in DNA methylation and histone modifications. Histone deacetylase inhibitors limit disease development in murine lupus models, at least in part via their ability to prevent B cell class switching and differentiation into plasma cells. This review will discuss relevant discoveries of the past several years pertaining to these areas of SLE B cell biology.
Collapse
Affiliation(s)
- Maria A. Bacalao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Anne B. Satterthwaite
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
29
|
Zhang B, Vogelzang A, Miyajima M, Sugiura Y, Wu Y, Chamoto K, Nakano R, Hatae R, Menzies RJ, Sonomura K, Hojo N, Ogawa T, Kobayashi W, Tsutsui Y, Yamamoto S, Maruya M, Narushima S, Suzuki K, Sugiya H, Murakami K, Hashimoto M, Ueno H, Kobayashi T, Ito K, Hirano T, Shiroguchi K, Matsuda F, Suematsu M, Honjo T, Fagarasan S. B cell-derived GABA elicits IL-10 + macrophages to limit anti-tumour immunity. Nature 2021; 599:471-476. [PMID: 34732892 PMCID: PMC8599023 DOI: 10.1038/s41586-021-04082-1] [Citation(s) in RCA: 228] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/28/2021] [Indexed: 01/16/2023]
Abstract
Small, soluble metabolites not only are essential intermediates in intracellular biochemical processes, but can also influence neighbouring cells when released into the extracellular milieu1-3. Here we identify the metabolite and neurotransmitter GABA as a candidate signalling molecule synthesized and secreted by activated B cells and plasma cells. We show that B cell-derived GABA promotes monocyte differentiation into anti-inflammatory macrophages that secrete interleukin-10 and inhibit CD8+ T cell killer function. In mice, B cell deficiency or B cell-specific inactivation of the GABA-generating enzyme GAD67 enhances anti-tumour responses. Our study reveals that, in addition to cytokines and membrane proteins, small metabolites derived from B-lineage cells have immunoregulatory functions, which may be pharmaceutical targets allowing fine-tuning of immune responses.
Collapse
Affiliation(s)
- Baihao Zhang
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Alexis Vogelzang
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Michio Miyajima
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Yuki Sugiura
- grid.26091.3c0000 0004 1936 9959Department of Biochemistry and Integrative Biology, Keio University, Tokyo, Japan
| | - Yibo Wu
- grid.7597.c0000000094465255YCI Laboratory for Next-Generation Proteomics, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Kenji Chamoto
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rei Nakano
- grid.260969.20000 0001 2149 8846Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Ryusuke Hatae
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rosemary J. Menzies
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Sonomura
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nozomi Hojo
- grid.508743.dLaboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Taisaku Ogawa
- grid.508743.dLaboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Wakana Kobayashi
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Yumi Tsutsui
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Sachiko Yamamoto
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Mikako Maruya
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Seiko Narushima
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Keiichiro Suzuki
- grid.7597.c0000000094465255Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Hiroshi Sugiya
- grid.260969.20000 0001 2149 8846Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Kosaku Murakami
- grid.258799.80000 0004 0372 2033Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motomu Hashimoto
- grid.258799.80000 0004 0372 2033Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Ueno
- grid.258799.80000 0004 0372 2033Department of Immunology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kobayashi
- grid.258799.80000 0004 0372 2033Department of Urology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuhiro Ito
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan ,grid.258799.80000 0004 0372 2033Department of Urology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Hirano
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Katsuyuki Shiroguchi
- grid.508743.dLaboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Osaka, Japan
| | - Fumihiko Matsuda
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makoto Suematsu
- grid.26091.3c0000 0004 1936 9959Department of Biochemistry and Integrative Biology, Keio University, Tokyo, Japan
| | - Tasuku Honjo
- grid.258799.80000 0004 0372 2033Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan. .,Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
30
|
Lio CWJ, Huang SCC. Circles of Life: linking metabolic and epigenetic cycles to immunity. Immunology 2020; 161:165-174. [PMID: 32418209 DOI: 10.1111/imm.13207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolites are the essential substrates for epigenetic modification enzymes to write or erase the epigenetic blueprint in cells. Hence, the availability of nutrients and activity of metabolic pathways strongly influence the enzymatic function. Recent studies have shed light on the choreography between metabolome and epigenome in the control of immune cell differentiation and function, with a major focus on histone modifications. Yet, despite its importance in gene regulation, DNA methylation and its relationship with metabolism is relatively unclear. In this review, we will describe how the metabolic flux can influence epigenetic networks in innate and adaptive immune cells, with a focus on the DNA methylation cycle and the metabolites S-adenosylmethionine and α-ketoglutarate. Future directions will be discussed for this rapidly emerging field.
Collapse
Affiliation(s)
- Chan-Wang Jerry Lio
- Division of Signaling and Gene Expression, La Jolla Institute, San Diego, CA, USA.,Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Stanley Ching-Cheng Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|