1
|
Lv H, Yu X, Wang P, Luo M, Luo Y, Lu H, Wang K, Xi A, Wen C, Xu Z. Locus coeruleus tyrosine hydroxylase positive neurons mediated the peripheral and central therapeutic effects of transcutaneous auricular vagus nerve stimulation (taVNS) in MRL/lpr mice. Brain Stimul 2024; 17:49-64. [PMID: 38145753 DOI: 10.1016/j.brs.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVE This study aims to investigate the effects of transcutaneous auricular vagus nerve stimulation (taVNS) on the development of systemic lupus erythematosus (SLE) in MRL/lpr mice. METHODS MRL/lpr mice were treated with taVNS for ten weeks. Locus coeruleus (LC) tyrosine hydroxylase positive (TH+) neurons were selectively lesioned by stereotactic injection of 6-hydroxydopamine (6-OHDA) or selectively activated by chemogenetic methods. Sympathetic denervation was conducted by intraperitoneal injection of 6-OHDA. RESULTS TaVNS activated the TH + neurons in LC. TaVNS produced central therapeutic effects by reducing the number of hippocampal microglia, and increasing the number of surviving LC TH+ neurons in MRL/lpr mice. TaVNS also retarded the development of lymphadenectasis and splenomegaly, decreased the proportion of double-negative T (DNT) cells, and alleviated nephritis in MRL/lpr mice. The lesion of LC TH+ neurons eliminated both these central and peripheral therapeutic effects of taVNS, while chemogenetic activation of LC TH+ neurons mimicked most central and peripheral protective effects of taVNS in MRL/lpr mice. Furthermore, taVNS regulated the autonomic nervous system in MRL/lpr mice. CONCLUSION This study provides direct evidence that taVNS can retard the development of peripheral and central symptoms of SLE, which is mediated by the LC TH+ neurons.
Collapse
Affiliation(s)
- Hongjie Lv
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiu Yu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ping Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mengxian Luo
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yijun Luo
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haimei Lu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Keer Wang
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Anran Xi
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chengping Wen
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Zhenghao Xu
- Laboratory of Rheumatology & Institute of TCM Clinical Basic Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Jiang B, Zhang Y, Li Y, Chen Y, Sha S, Zhao L, Li D, Wen J, Lan J, Lou Y, Su H, Zhang C, Zhu J, Tao J. A Tissue-Tended Mycophenolate-Modified Nanoparticle Alleviates Systemic Lupus Erythematosus in MRL/Lpr Mouse Model Mainly by Promoting Local M2-Like Macrophagocytes Polarization. Int J Nanomedicine 2022; 17:3251-3267. [PMID: 35924257 PMCID: PMC9342721 DOI: 10.2147/ijn.s361400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background Mycophenolate mofetil (MMF), for which the bioactive metabolite is mycophenolic acid (MPA), is a frequently used immunosuppressant for systemic lupus erythematosus (SLE). However, its short half-life and poor biodistribution into cells and tissues hinder its clinical efficacy. Our dextran mycophenolate-based nanoparticles (MPA@Dex-MPA NPs) have greatly improved the pharmacokinetics of MMF/MPA. We here tested the therapeutic efficacy of MPA@Dex-MPA NPs against SLE and investigated the underlying mechanism. Methods The tissue and immune cell biodistributions of MPA@Dex-MPA NPs were traced using live fluorescence imaging system and flow cytometry, respectively. Serological proinflammatory mediators and kidney damage were detected to assess the efficacy of MPA@Dex-MPA NPs treatments of MRL/lpr lupus-prone mice. Immune cell changes in the kidney and spleen were further analyzed post-treatment via flow cytometry. Bone marrow-derived macrophages were used to investigate the potential mechanism. Results MPA@Dex-MPA NPs exhibited superior therapeutic efficacy and safety in the MRL/lpr mice using significantly lower administration dosage (one-fifth) and frequency (once/3 days) compared to MMF/MPA used in ordinary practice. The overall prognosis of the mice was improved as they showed lower levels of serological proinflammatory mediators. Moreover, kidney injury was alleviated with reduced pathological signs and decreased urine protein-creatinine ratio. Further investigations of the underlying mechanism revealed a preferential penetration and persistent retention of MPA@Dex-MPA NPs in the spleen and kidney, where they were mostly phagocytosed by macrophages. The macrophages were found to be polarized towards a CD206+ M2-like phenotype, with a downregulation of surface CD80 and CD40, and reduced TNF-α production in the spleen and kidney and in vitro. The expansion of T cells was also significantly inhibited in these two organs. Conclusion Our research improved the efficacy of MPA for MRL/lpr mice through synthesizing MPA@Dex-MPA NPs to enhance its tissue biodistribution and explored the possible mechanism, providing a promising strategy for SLE therapy.
Collapse
Affiliation(s)
- Biling Jiang
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yamin Zhang
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yuce Li
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Yu Chen
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Shanshan Sha
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Liang Zhao
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Danqi Li
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Jingjing Wen
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Jiajia Lan
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Yuchen Lou
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, HUST, Wuhan, People’s Republic of China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, HUST, Wuhan, People’s Republic of China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, HUST, Wuhan, People’s Republic of China
| | - Juan Tao
- Hubei Engineering Research Center of Skin Disease Theranostics and Health, Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, People’s Republic of China
- Correspondence: Juan Tao; Jintao Zhu, Email ;
| |
Collapse
|
3
|
Ning W, Cheng D, Howe PH, Bian C, Kamen DL, Luo Z, Fu X, Ogunrinde E, Yang L, Wang X, Li QZ, Oates J, Zhang W, White D, Wan Z, Gilkeson GS, Jiang W. Staphylococcus aureus peptidoglycan (PGN) induces pathogenic autoantibody production via autoreactive B cell receptor clonal selection, implications in systemic lupus erythematosus. J Autoimmun 2022; 131:102860. [PMID: 35810689 PMCID: PMC9397544 DOI: 10.1016/j.jaut.2022.102860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVES There is an intricate interplay between the microbiome and the immune response impacting development of normal immunity and autoimmunity. However, we do not fully understand how the microbiome affects production of natural-like and pathogenic autoantibodies. Peptidoglycan (PGN) is a component of the bacterial cell wall which is highly antigenic. PGNs from different bacteria can differ in their immune regulatory activities. METHODS C57BL/6 and MRL/lpr mice were intraperitoneally injected with saline or PGN from Staphylococcus aureus or Bacillus subtilis. Spleen anti-double-stranded DNA (dsDNA) IgG + B cells were sorted for B-cell receptor sequencing. Serum autoantibody levels and kidney damage were analyzed. Further, the association between plasma S. aureus translocation and systemic lupus erythematosus (SLE) pathogenesis was assessed in women. RESULTS Administration of B. subtilis PGN induced natural-like anti-dsDNA autoantibodies (e.g., IgM, short lived IgG response, and no tissue damage), whereas S. aureus PGN induced pathogenic anti-dsDNA autoantibodies (e.g., prolonged IgG production, low IgM, autoantibody-mediated kidney damage) in C57BL/6 and/or MRL/lpr mice. However, serum total IgG did not differ. S. aureus PGN induced antibodies with reduced clonality and greater hypermutation of IGHV3-74 in splenic anti-dsDNA IgG + B cells from C57BL/6 mice. Further, S. aureus PGN promoted IgG class switch recombination via toll-like receptor 2. Plasma S. aureus DNA levels were increased in women with SLE versus control women and correlated with levels of lupus-related autoantibodies and renal involvement. CONCLUSIONS S. aureus PGN induces pathogenic autoantibody production, whereas B. subtilis PGN drives production of natural nonpathogenic autoantibodies.
Collapse
Affiliation(s)
- Wangbin Ning
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Da Cheng
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, SC, USA
| | - Chuanxiu Bian
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Diane L Kamen
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA
| | - Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Xiaoyu Fu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Elizabeth Ogunrinde
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Liuqing Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; The Third People's Hospital of Shenzhen, Guangdong, China
| | - Xu Wang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Department of Urology, Capital Medical University Affiliated XuanWu Hospital, Beijing, China
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jim Oates
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Weiru Zhang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - David White
- Department of Otolaryngology, Medical University of South Carolina, Charleston, SC, USA
| | - Zhuang Wan
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA
| | - Gary S Gilkeson
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, 114 Doughty Street, Strom Thurmond Research Building Room 416, Charleston, SC, 29403, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB208D, Charleston, SC, 29425, USA; Ralph H. Johnson VA Medical Center, Charleston, SC, USA; Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
4
|
Gao C, Han Y, Bai L, Wang Y, Xue F. IK: A novel cell mitosis regulator that contributes to carcinogenesis. Cell Biochem Funct 2021; 39:854-859. [PMID: 34250629 DOI: 10.1002/cbf.3660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/12/2022]
Abstract
Carcinogenesis is characterized by abnormal regulation of cell growth and cell death. IK is a novel cell mitosis regulator that may contribute to carcinogenesis. Previous studies showed that the loss of IK expression resulted in cell mitotic arrest and even cell death. Besides, IK can also inhibit the interferon gamma (IFN-γ)-induced expression of human leukocyte antigen (HLA) class II antigen, which is associated with tumour immune microenvironment. To gain insight into the current research progress regarding IK, we conducted a review and searched the limited literature on IK using PubMed or Web of Science. In this review, we discussed the possible biological functions and mechanisms of IK in cancer and its immune microenvironment. Future perspectives of IK were also mentioned to explore its clinical significance.
Collapse
Affiliation(s)
- Chao Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Yanyan Han
- Department of Pathology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Bai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Nankai University Affiliated Hospital (Tianjin Fourth Hospital), Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| |
Collapse
|
5
|
Hiramatsu-Asano S, Sunahori-Watanabe K, Zeggar S, Katsuyama E, Mukai T, Morita Y, Wada J. Deletion of Mir223 Exacerbates Lupus Nephritis by Targeting S1pr1 in Faslpr/lpr Mice. Front Immunol 2021; 11:616141. [PMID: 33574820 PMCID: PMC7871001 DOI: 10.3389/fimmu.2020.616141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/09/2020] [Indexed: 12/29/2022] Open
Abstract
Objective The micro RNAs (miRNAs) and their target mRNAs are differentially expressed in various immune-mediated cells. Here, we investigated the role of Mir223 and sphingosine-1-phosphate receptor 1 (S1pr1) in the pathogenesis of systemic lupus erythematosus. Methods We analyzed miRNA and mRNA profiling data of CD4+ splenic T cells derived from MRL/MpJ-Faslpr /J mice. We performed 3' untranslated region (UTR) luciferase reporter gene assay using human umbilical vein endothelial cells (HUVECs). We generated the B6-Mir223-/-Faslpr/lpr mice and the lupus phenotypes were analyzed. Results In CD4+ splenic T cells, we identified upregulation of miR-223-3p and downregulation of the possible target, S1pr1 by RNA sequencing of MRL/MpJ-Faslpr /J mice. The transfection with miR-223-3p mimic significantly suppressed a luciferase activity in HUVEC treated with a Lentivirus vector containing 3' UTR of S1pr1. The mRNA levels of S1pr1 were significantly decreased after miR-223-3p overexpression. In B6-Mir223-/-Faslpr/lpr mice, the proportion of CD3+ T cells, CD3+CD4-CD8- cells, B cells, plasma cells, and S1PR1+CD4+ T cells in the spleen was significantly increased compared with that in B6-Mir223+/+Faslpr/lpr mice by flow cytometry. B6-Mir223-/-Faslpr/lpr mice demonstrated the elevation of glomerular and renal vascular scores associated with enhanced intraglomerular infiltration of S1PR1+CD4+ T cells. Conclusion Unexpectedly, the deletion of Mir223 exacerbated the lupus phenotypes associated with increased population of S1PR1+CD4+ T in spleen and the enhanced infiltration of S1PR1+CD4+ T cells in inflamed kidney tissues, suggesting compensatory role of Mir223 in the pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Sumie Hiramatsu-Asano
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Katsue Sunahori-Watanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Sonia Zeggar
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Eri Katsuyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoyuki Mukai
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Yoshitaka Morita
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
6
|
Hwang JT, Yu JW, Nam HJ, Song SK, Sung WY, Kim, Y, Cho JH. Suppressive Effects of a Truncated Inhibitor K562 Protein-Derived Peptide on Two Proinflammatory Cytokines, IL-17 and TNF-α. J Microbiol Biotechnol 2020; 30:1810-1818. [PMID: 32958733 PMCID: PMC9728226 DOI: 10.4014/jmb.2004.04062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Inhibitor K562 (IK) protein was first isolated from the culture medium of K562 cells, a leukemia cell line, and is an inhibitory regulator of interferon-γ-induced major histocompatibility complex class II expression. Recently, exogenous truncated IK (tIK) protein showed potential as a therapeutic agent for inflammation-related diseases. In this study, we designed a novel putative anti-inflammatory peptide derived from tIK protein based on homology modeling of the human interleukin-10 (hIL-10) structure, and investigated whether the peptide exerted inhibitory effects against proinflammatory cytokines such as IL-17 and tumor necrosis factor-α (TNF-α). The peptide contains key residues involved in binding hIL-10 to the IL-10 receptor, and exerted strong inhibitory effects on IL- 17 (43.8%) and TNF-α (50.7%). In addition, we used circular dichroism spectroscopy to confirm that the peptide is usually present in a random coil configuration in aqueous solution. In terms of toxicity, the peptide was found to be biologically safe. The mechanisms by which the short peptide derived from human tIK protein exerts inhibitory effects against IL-17 and TNF-α should be explored further. We also evaluated the feasibility of using this novel peptide in skincare products.
Collapse
Affiliation(s)
- Jong Tae Hwang
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Ji Won Yu
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Department of Biology, Kongju National University, Kongju 3588, Republic of Korea
| | - Hee Jin Nam
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Sun Kwang Song
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Woo Yong Sung
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea
| | - Yongae Kim,
- Department of Chemistry, Hankuk University of Foreign Studies, Yongin 1705, Republic of Korea
| | - Jang-Hee Cho
- Biomaterial Research Center, Cellinbio, Suwon 668, Republic of Korea,Corresponding author Phone: +82-31-695-7959 Fax: +82-31-695-7986 E-mail:
| |
Collapse
|
7
|
Ruacho G, Kvarnström M, Zickert A, Oke V, Rönnelid J, Eketjäll S, Elvin K, Gunnarsson I, Svenungsson E. Sjögren Syndrome in Systemic Lupus Erythematosus: A Subset Characterized by a Systemic Inflammatory State. J Rheumatol 2019; 47:865-875. [PMID: 31523050 DOI: 10.3899/jrheum.190250] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE An often-neglected subset of patients with systemic lupus erythematosus (SLE) is those with secondary Sjögren syndrome (SLE-sSS). Further, primary SS overlaps and can be difficult to delineate from SLE. To shed light on the SLE-sSS subset, we investigated a large and well-characterized SLE cohort, comparing patients with SLE-sSS and SLE patients without SS (SLE-nonsSS) and controls. METHODS We included 504 consecutive patients with SLE, fulfilling the 1982 revised American College of Rheumatology criteria, and 319 controls from the general population, matched for age and sex to the first 319 patients. SLE-sSS was defined according to the American-European Consensus Criteria (AECC). A thorough clinical examination, including subjective and objective quantifications of sicca symptoms, was performed in all participants. Autoantibodies and 20 selected cytokines were measured by luminex and multiplex analysis, respectively. RESULTS SLE-sSS, as defined by AECC, occurred in 23% of the patients with SLE. In comparison to SLE-nonsSS, the SLE-sSS group was older and more frequently female. Leukopenia and peripheral neuropathy were more frequent and nephritis less frequent. Circulating levels of 6/20 investigated proinflammatory cytokines [tumor necrosis factor-α, interleukin (IL) 6, monocyte chemoattractant protein 4, macrophage inflammatory protein 1β, IL-12/IL-23p40, and interferon γ-induced protein 10], total IgG, anti-SSA/Ro52, anti-SSA/Ro60, anti-SSB/La antibodies, and rheumatoid factor (IgM and IgA) were higher in the SLE-sSS group (p < 0.05 for all comparisons). CONCLUSION The frequency of SLE-sSS increased with age and affected roughly one-quarter of all patients with SLE. Despite less internal organ involvement, a systemic inflammatory state with high levels of proinflammatory cytokines is present in the SLE-sSS subgroup. This is a novel observation that may affect future understanding and treatment of the SLE-sSS subset.
Collapse
Affiliation(s)
- Guillermo Ruacho
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Marika Kvarnström
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Agneta Zickert
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Vilija Oke
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Johan Rönnelid
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Susanna Eketjäll
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Kerstin Elvin
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Iva Gunnarsson
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden.,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital
| | - Elisabet Svenungsson
- From the Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, Stockholm; Karolinska University Hospital, Stockholm; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI-AZ ICMC), Stockholm; Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm; Center for Clinical Research, Uppsala University, Sörmland; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala; Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Huddinge, Sweden. .,G. Ruacho, DMD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Center for Clinical Research, Uppsala University; M. Kvarnström, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; A. Zickert, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; V. Oke, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; J. Rönnelid, MD, PhD, Department of Immunology, Genetics and Pathology, Uppsala University; S. Eketjäll, PhD, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, and KI-AZ ICMC; K. Elvin, MD, PhD, Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital; I. Gunnarsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital; E. Svenungsson, MD, PhD, Department of Medicine Solna, Division of Rheumatology, Karolinska Institutet, and Karolinska University Hospital.
| |
Collapse
|
8
|
Li X, Wang M, Hong H, Luo C, Liu Z, Yang R. Sophocarpine attenuates murine lupus nephritis via inhibiting NLRP3 inflammasome and NF-κB activation. Immunol Res 2018; 66:521-527. [DOI: 10.1007/s12026-018-9012-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
9
|
Choi S, Park H, Jung S, Kim EK, Cho ML, Min JK, Moon SJ, Lee SM, Cho JH, Lee DH, Nam JH. Therapeutic Effect of Exogenous Truncated IK Protein in Inflammatory Arthritis. Int J Mol Sci 2017; 18:E1976. [PMID: 28906466 PMCID: PMC5618625 DOI: 10.3390/ijms18091976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/18/2022] Open
Abstract
Inhibitor K562 (IK) protein was first isolated from the culture medium of K562, a leukemia cell line. It is known to be an inhibitory regulator of interferon-γ-induced major histocompatibility complex class (MHC) II expression. Previously, we found that transgenic (Tg) mice constitutively expressing truncated IK (tIK) showed reduced numbers of pathogenic Th1 and Th17 cells, which are known to be involved in the development of rheumatoid arthritis (RA). Here, we investigated whether exogenous tIK protein has a therapeutic effect in arthritis in disease models and analyzed its mechanism. Exogenous tIK protein was produced in an insect expression system and applied to the collagen antibody-induced arthritis (CAIA) mouse disease model. Injection of tIK protein alleviated the symptoms of arthritis in the CAIA model and reduced Th1 and Th17 cell populations. In addition, treatment of cultured T cells with tIK protein induced expression of A20, a negative regulator of nuclear factor-κB (NFκB)-induced inflammation, and reduced expression of several transcription factors related to T cell activation. We conclude that exogenous tIK protein has the potential to act as a new therapeutic agent for RA patients, because it has a different mode of action to biopharmaceutical agents, such as tumor necrosis factor antagonists, that are currently used to treat RA.
Collapse
Affiliation(s)
- Seulgi Choi
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - HyeLim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - SeoYeon Jung
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Sang-Myeong Lee
- Department of Biotechnology, Chonbuk National University, Iksan 54596, Korea.
| | - Jang-Hee Cho
- Biomaterials Research Center, Cellinbio, Suwon 16680, Korea.
| | - Dong-Hee Lee
- Biomaterials Research Center, Cellinbio, Suwon 16680, Korea.
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| |
Collapse
|
10
|
Park HL, Lee SM, Min JK, Moon SJ, Kim I, Kang KW, Park S, Choi S, Jung HN, Lee DH, Nam JH. IK acts as an immunoregulator of inflammatory arthritis by suppressing T H17 cell differentiation and macrophage activation. Sci Rep 2017; 7:40280. [PMID: 28071693 PMCID: PMC5223115 DOI: 10.1038/srep40280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 12/02/2016] [Indexed: 12/15/2022] Open
Abstract
Pathogenic T helper cells (TH) and macrophages have been implicated in the development of rheumatoid arthritis (RA), which can lead to severe synovial inflammation and bone destruction. A range of therapies have been widely used for RA, including specific monoclonal antibodies and chemical inhibitors against inflammatory cytokines produced by these cells. However, these have not been sufficient to meet the medical need. Here, we show that in transgenic mice expressing truncated IK (tIK) cytokine, inflammatory arthritis symptoms were ameliorated as the result of suppression of the differentiation of TH1 and TH17 cells and of macrophage activation. During inflammatory responses, tIK cytokine systemically regulated macrophage functions and TH17 cell differentiation through inactivation of the MAPK and NF-κB pathways. Interestingly, the level of tIK cytokine was higher in synovial fluid of RA patients compared with that in osteoarthritis (OA) patients. Our observations suggest that tIK cytokine can counterbalance the induction of inflammatory cells related to RA and thus could be a new therapeutic agent for the treatment of RA.
Collapse
Affiliation(s)
- Hye-Lim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 420-743, Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan, 570-752, Korea
| | - Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul,137-040, Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul,137-040, Korea
| | - Inki Kim
- Department of Medicine, College of Medicine, University of Ulsan, Seoul 138-736, Korea
| | - Kyung-Won Kang
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Chonbuk National University, Iksan, 570-752, Korea
| | - Sooho Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 420-743, Korea
| | - SeulGi Choi
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 420-743, Korea
| | - Ha-Na Jung
- Biomaterials Research Center, Cellinbio, Suwon, 443-734, Korea
| | - Dong-Hee Lee
- Biomaterials Research Center, Cellinbio, Suwon, 443-734, Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 420-743, Korea
| |
Collapse
|
11
|
Lu A, Li H, Niu J, Wu S, Xue G, Yao X, Guo Q, Wan N, Abliz P, Yang G, An L, Meng G. Hyperactivation of the NLRP3 Inflammasome in Myeloid Cells Leads to Severe Organ Damage in Experimental Lupus. THE JOURNAL OF IMMUNOLOGY 2016; 198:1119-1129. [PMID: 28039299 DOI: 10.4049/jimmunol.1600659] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune syndrome associated with severe organ damage resulting from the activation of immune cells. Recently, a role for caspase-1 in murine lupus was described, indicating an involvement of inflammasomes in the development of SLE. Among multiple inflammasomes identified, the NLRP3 inflammasome was connected to diverse diseases, including autoimmune encephalomyelitis. However, the function of NLRP3 in SLE development remains elusive. In this study, we explored the role of NLRP3 in the development of SLE using the pristane-induced experimental lupus model. It was discovered that more severe lupus-like syndrome developed in Nlrp3-R258W mice carrying the gain-of-function mutation. Nlrp3-R258W mutant mice exhibited significantly higher mortality upon pristane challenge. Moreover, prominent hypercellularity and interstitial nephritis were evident in the glomeruli of Nlrp3-R258W mice. In addition, hyperactivation of the NLRP3 inflammasome in this mouse line resulted in proteinuria and mesangial destruction. Importantly, all of these phenotypes were largely attributed to the Nlrp3-R258W mutation expressed in myeloid cells, because Cre recombinase-mediated depletion of this mutant from such cells rescued mice from experimental lupus. Taken together, our study demonstrates a critical role for NLRP3 in the development of SLE and suggests that modulating the inflammasome signal may help to control the inflammatory damage in autoimmune diseases, including lupus.
Collapse
Affiliation(s)
- Ailing Lu
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Li
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.,College of Life Science, Shandong Normal University, Jinan 250014, China
| | - Junling Niu
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuxian Wu
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guang Xue
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaomin Yao
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiuhong Guo
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nianhong Wan
- Department of Laboratory Medicine, Central Hospital of Zhabei District, Shanghai 200070, China; and
| | - Paride Abliz
- Department of Dermatology, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Guiwen Yang
- College of Life Science, Shandong Normal University, Jinan 250014, China
| | - Liguo An
- College of Life Science, Shandong Normal University, Jinan 250014, China
| | - Guangxun Meng
- Chinese Academy of Sciences Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China;
| |
Collapse
|
12
|
Lee S, Jeong AL, Park JS, Han S, Jang CY, Kim KI, Kim Y, Park JH, Lim JS, Lee MS, Yang Y. IK-guided PP2A suppresses Aurora B activity in the interphase of tumor cells. Cell Mol Life Sci 2016; 73:3375-86. [PMID: 26906715 PMCID: PMC11108362 DOI: 10.1007/s00018-016-2162-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/13/2016] [Accepted: 02/09/2016] [Indexed: 01/02/2023]
Abstract
Aurora B activation is triggered at the mitotic entry and required for proper microtubule-kinetochore attachment at mitotic phase. Therefore, Aurora B should be in inactive form in interphase to prevent aberrant cell cycle progression. However, it is unclear how the inactivation of Aurora B is sustained during interphase. In this study, we find that IK depletion-induced mitotic arrest leads to G2 arrest by Aurora B inhibition, indicating that IK depletion enhances Aurora B activation before mitotic entry. IK binds to Aurora B, and colocalizes on the nuclear foci during interphase. Our data further show that IK inhibits Aurora B activation through recruiting PP2A into IK and Aurora B complex. It is thus believed that IK, as a scaffold protein, guides PP2A into Aurora B to suppress its activity in interphase until mitotic entry.
Collapse
Affiliation(s)
- Sunyi Lee
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Ae Lee Jeong
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Jeong Su Park
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Sora Han
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Chang-Young Jang
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Keun Il Kim
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Yonghwan Kim
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Jong Hoon Park
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Jong-Seok Lim
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Myung Sok Lee
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Young Yang
- Division of Biological Sciences, Department of Life Systems, Research Center for Women's Disease, Sookmyung Women's University, Seoul, 140-742, Republic of Korea.
| |
Collapse
|
13
|
Li X, Liu F, Zhang X, Shi G, Ren J, Ji J, Ding L, Fan H, Dou H, Hou Y. Notch-Hes-1 axis controls TLR7-mediated autophagic death of macrophage via induction of P62 in mice with lupus. Cell Death Dis 2016; 7:e2341. [PMID: 27537524 PMCID: PMC5108329 DOI: 10.1038/cddis.2016.244] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 01/21/2023]
Abstract
The increased death of macrophages has been considered as a pathogenic factor for systemic lupus erythematosus (SLE), and dysfunction of autophagy may contribute to improper cell death. However, the effect of autophagy on macrophage during the pathogenesis of SLE is still unclear. Here we found that the death rate and autophagy level of macrophages significantly increased in MRL/lpr lupus-prone mice. Activation of toll-like receptor 7 (TLR7) triggered macrophage death in an autophagy-dependent but caspase-independent way in vitro. Moreover, P62/SQSTM1 is thought to have an essential role in selective autophagy. We also demonstrated that P62/SQSTM1 was required for TLR7-induced autophagy, and knockdown of P62 suppressed R848-induced cell death and LC3II protein accumulation. As an important mediator for cell-cell communication, Notch signaling is responsible for cell-fate decisions. Our results showed that activation of TLR7 also upregulated the expression of Notch1, especially its downstream target gene Hairy and enhancer of split 1 (Hes-1) in macrophages. Of note, we found that Hes-1, as a transcriptional factor, controlled TLR7-induced autophagy by regulating P62 expression. Furthermore, to confirm the above results in vivo, TLR7 agonist imiquimod (IMQ)-induced lupus mouse model was prepared. Splenic macrophages from IMQ-treated mice exhibited increased autophagy and cell death as well as enhanced expressions of Notch1 and Hes-1. Our results indicate that Notch1-Hes-1 signaling controls TLR7-induced autophagic death of macrophage via regulation of P62 in mice with lupus.
Collapse
Affiliation(s)
- Xiaojing Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Fei Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Xuefang Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Guoping Shi
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Jianjian Ji
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Liang Ding
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Hongye Fan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| |
Collapse
|
14
|
Wu X, Guo J, Ding R, Lv B, Bi L. CXCL13 blockade attenuates lupus nephritis of MRL/lpr mice. Acta Histochem 2015; 117:732-7. [PMID: 26456520 DOI: 10.1016/j.acthis.2015.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/01/2015] [Accepted: 09/09/2015] [Indexed: 12/29/2022]
Abstract
The chemokine CXC ligand 13 protein (CXCL13) is reported to closely related to the disease activity and severity of systemic lupus erythematosus (SLE), moreover, the level of CXCL13 was markedly raised in kidney tissues of lupus nephritis (LN) patients. The aim of the present study was to explore whether the blockade of CXCL13 has therapeutic effects on murine LN. MRL/lpr mice received 50μg anti-CXCL13 neutralizing antibody or isotype IgG by intraperitoneal injection everyday for six weeks, and renal damage of each group was determined. Our results showed that the blockade of CXCL13 significantly reduced urine protein, serum creatinine, and dramatically attenuated renal pathology injury. Treatment with anti-CXCL13Ab also reduced serum anti-dsDNA level, renal immune complex deposition as well as inflammatory cytokines secretion. Meanwhile, Th17/Treg ratio in spleens of MRL/lpr mice was significantly decreased by the blocking of CXCL13. These findings suggested that CXCL13 may be a promising target for the therapy of LN.
Collapse
Affiliation(s)
- Xiaobei Wu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China; The Affiliated Hospital of Northeast Normal University, Changchun 130024, People's Republic of China
| | - Jialong Guo
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Rui Ding
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Bin Lv
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| |
Collapse
|
15
|
Zhao J, Wang H, Dai C, Wang H, Zhang H, Huang Y, Wang S, Gaskin F, Yang N, Fu SM. P2X7 blockade attenuates murine lupus nephritis by inhibiting activation of the NLRP3/ASC/caspase 1 pathway. ACTA ACUST UNITED AC 2014; 65:3176-85. [PMID: 24022661 DOI: 10.1002/art.38174] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 08/22/2013] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The NLRP3 inflammasome plays key roles in inflammation and autoimmunity, and purinergic receptor P2X7 has been proposed to be upstream of NLRP3 activation. The aim of the present study, using murine models, was to investigate whether the P2X7 /NLRP3 inflammasome pathway contributes to the pathogenesis of lupus nephritis (LN). METHODS MRL/lpr mice were treated with the selective P2X7 antagonist brilliant blue G (BBG) for 8 weeks. Following treatment, the severity of renal lesions, production of anti-double-stranded DNA (anti-dsDNA) antibodies, rate of survival, activation of the NLRP3/ASC/caspase 1 inflammasome pathway, and ratio of Th17 cells to Treg cells were evaluated. P2X7 -targeted small interfering RNA (siRNA) was also used for in vivo intervention. Similar evaluations were carried out in NZM2328 mice, a model of LN in which the disease was accelerated by administration of adenovirus-expressing interferon-α (AdIFNα). RESULTS Significant up-regulation of P2X7 /NLRP3 inflammasome signaling molecules was detected in the kidneys of MLR/lpr mice as compared with normal control mice. Blockade of P2X7 activation by BBG suppressed NLRP3/ASC/caspase 1 assembly and the subsequent release of interleukin-1β (IL-1β), resulting in a significant reduction in the severity of nephritis and circulating anti-dsDNA antibodies. The lifespan of the treated mice was significantly prolonged. BBG treatment reduced the serum levels of IL-1β and IL-17 and the Th17:Treg cell ratio. Similar results were obtained by specific siRNA silencing of P2X7 in vivo. The effectiveness of BBG treatment in modulating LN was confirmed in NZM2328 mice with AdIFNα-accelerated disease. CONCLUSION Activation of the P2X7 signaling pathway accelerates murine LN by activating the NLRP3/ASC/caspase 1 inflammasome, resulting in increased IL-1β production and enhanced Th17 cell polarization. Thus, targeting of the P2X7 /NLRP3 pathway should be considered as a novel therapeutic strategy in patients with lupus.
Collapse
Affiliation(s)
- Jijun Zhao
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Park HL, Kim YJ, Na HN, Park MY, Kim JY, Yun CW, Nam JH. IK induced by coxsackievirus B3 infection transiently downregulates expression of MHC class II through increasing cAMP. Viral Immunol 2013; 26:13-24. [PMID: 23409929 DOI: 10.1089/vim.2012.0054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Major histocompatibility complex (MHC) class II expression is critical for the presentation of antigens in the immune response to viral infection. Consequently, some viruses regulate the MHC class II-mediated presentation of viral antigens as a mechanism of immune escape. In this study, we found that Coxsackievirus B3 (CVB3) infection transiently increased IK expression, which reduced the expression of MHC class II (I-A/I-E) on splenic B cells. Interestingly, CVB3-induced IK elevated cAMP, a downstream molecule of the G protein-coupled receptors, which inhibited MHC class II presentation on B cells. Transgenic mice expressing truncated IK showed lower expression of MHC class II on B cells than did wild-type mice after CVB3 infection. Taken together, these results imply that IK plays a role in downregulating MHC class II expression on B cells during CVB3 infection through the induction of cAMP.
Collapse
Affiliation(s)
- Hye-Lim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
17
|
He X, Fuller C, Song Y, Meng Q, Zhang B, Yang X, Li H. Sherlock: detecting gene-disease associations by matching patterns of expression QTL and GWAS. Am J Hum Genet 2013; 92:667-80. [PMID: 23643380 PMCID: PMC3644637 DOI: 10.1016/j.ajhg.2013.03.022] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 03/07/2013] [Accepted: 03/25/2013] [Indexed: 12/26/2022] Open
Abstract
Genetic mapping of complex diseases to date depends on variations inside or close to the genes that perturb their activities. A strong body of evidence suggests that changes in gene expression play a key role in complex diseases and that numerous loci perturb gene expression in trans. The information in trans variants, however, has largely been ignored in the current analysis paradigm. Here we present a statistical framework for genetic mapping by utilizing collective information in both cis and trans variants. We reason that for a disease-associated gene, any genetic variation that perturbs its expression is also likely to influence the disease risk. Thus, the expression quantitative trait loci (eQTL) of the gene, which constitute a unique "genetic signature," should overlap significantly with the set of loci associated with the disease. We translate this idea into a computational algorithm (named Sherlock) to search for gene-disease associations from GWASs, taking advantage of independent eQTL data. Application of this strategy to Crohn disease and type 2 diabetes predicts a number of genes with possible disease roles, including several predictions supported by solid experimental evidence. Importantly, predicted genes are often implicated by multiple trans eQTL with moderate associations. These genes are far from any GWAS association signals and thus cannot be identified from the GWAS alone. Our approach allows analysis of association data from a new perspective and is applicable to any complex phenotype. It is readily generalizable to molecular traits other than gene expression, such as metabolites, noncoding RNAs, and epigenetic modifications.
Collapse
Affiliation(s)
- Xin He
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94143, USA
- Lane Center of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Chris K. Fuller
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Yi Song
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Qingying Meng
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
18
|
|
19
|
Shao R, Liu X, Ding Y, Chen X, Gao R, He J, Wang Y. Characterization of IK cytokine expression in mouse endometrium during early pregnancy and its significance on implantation. Int J Mol Med 2012; 30:615-21. [PMID: 22692555 DOI: 10.3892/ijmm.2012.1019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 04/17/2012] [Indexed: 11/05/2022] Open
Abstract
The expression of IK cytokine was investigated in the mouse endometrium during early pregnancy (D1-D7 of pregnancy) and pseudopregnancy using real-time PCR, western blotting and immunohistochemical analysis, and the effects of IK cytokine on embryo implantation were observed by injection with antisense IK cytokine oligodeoxynucleotides in the uterine horn. Our data showed that the expression of IK cytokine mRNA increased gradually from D1 to D4 of pregnancy and reached a peak level at D4 of pregnancy (P<0.05). Western blotting and immunohistochemical analysis revealed that the expression of IK cytokine protein increased gradually from D1 to D5 of pregnancy and reached a peak level at D5 of pregnancy (P<0.05). The expression of IK cytokine in the pseudopregnant uterus was significantly lower compared to that in the normal pregnant uterus and the level of the protein never showed a high peak during the whole pseudopregnancy. The expression of IK cytokine at the implantation site was much stronger than that in the peri-implantation site on Day 5 of pregnancy. After 24 and 48 h of injection with antisense IK cytokine oligodexynucleotides in the uterine horn on D3 of pregnancy (i.e. implantation window), the expression of IK cytokine in the uterus was remarkably inhibited, while the expression of major histocompatibility complex II (MHC II) increased and the number of implanted embryos significantly decreased in the site of uterine horns receiving antisense IK cytokine (P<0.05). These results suggested that IK cytokine may play a crucial role in implantation.
Collapse
Affiliation(s)
- Ruyue Shao
- Laboratory of Reproductive Biology, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | | | | | | | | | | | | |
Collapse
|
20
|
Zhang L, Yang N, Wang S, Huang B, Li F, Tan H, Liang Y, Chen M, Li Y, Yu X. Adenosine 2A receptor is protective against renal injury in MRL/lpr mice. Lupus 2010; 20:667-77. [PMID: 21183557 DOI: 10.1177/0961203310393262] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Adenosine is considered as a potent endogenous anti-inflammatory and immunosuppressive molecule. We examined the roles of A2A-adenosine receptor (A(2A)R) in the progression of lupus nephritis. METHODS MRL/lpr mice were given a selective A(2A)R agonist, CGS21680 (0.4 mg/kg per day, i.p.) while control mice received saline only. After 8 weeks of treatment, mice were sacrificed for assessment of functional and histological parameters as well as inflammatory infiltration in the kidneys. MCP-1, IFN-γ, MHC-II and A(2A)R mRNA expression was evaluated by RT-PCR. Expression of A(2A)R and nuclear NFκB p65 protein was determined by Western blot analysis. Levels of anti-dsDNA antibody and IFN-γ were measured by ELISA. RESULTS CGS21680 treatment resulted in significant decrease in proteinuria, blood urea and creatinine as well as improvement in renal histology. Renal macrophage and T-cell infiltration were significantly attenuated in association with suppressed expression of MCP-1, IFN-γ and MHC-II. CGS21680 treatment reduced the level of serum anti-dsDNA and renal immune complex deposition. CGS21680 inhibited the activation of NFκB and suppressed the expression of IFN-γ, MCP-1 and MHC-II in MRL/lpr splenocytes. CONCLUSIONS A(2A)R activation suppressed inflammation in the kidneys of MRL/lpr mice and can be considered as a novel therapeutic approach for human lupus nephritis.
Collapse
Affiliation(s)
- L Zhang
- Department of Nephrology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gilhar A. Collapse of Immune Privilege in Alopecia Areata: Coincidental or Substantial? J Invest Dermatol 2010; 130:2535-7. [DOI: 10.1038/jid.2010.260] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Wang S, Yang N, Zhang L, Huang B, Tan H, Liang Y, Li Y, Yu X. Jak/STAT signaling is involved in the inflammatory infiltration of the kidneys in MRL/lpr mice. Lupus 2010; 19:1171-80. [PMID: 20501525 DOI: 10.1177/0961203310367660] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cytokines are known to play an important role in the pathogenesis of lupus nephritis (LN) and the Jak/STAT (Janus kinase-signal transducer and activator of transcription factor) pathway is important in mediating signal transduction of cytokines. This study examined the pathogenic role of Jak/STAT signaling in LN. MRL/lpr mice were either treated with a selective Jak2 inhibitor tyrphostin AG490 or with vehicle alone from 12 weeks of age until being sacrificed at week 20. AG490 significantly inhibited the phosphorylation of Jak2 and STAT1 (p < 0.05). Compared with the vehicle-treated mice, AG490 treatment significantly reduced proteinuria, improved renal function and suppressed histological lesions of the kidneys and salivary glands (p < 0.05). AG490 treatment significantly inhibited the renal expression of monocyte chemotactic protein (MCP)-1, interferon (IFN)-gamma and class II MHC, which was accompanied by reduced renal infiltration of T cells and macrophages (p < 0.05). In addition, AG490 treatment resulted in a decrease in serum anti-double-stranded DNA (anti-dsDNA) antibody and attenuated the deposition of IgG and C3 in the kidneys (p < 0.05). This study demonstrated that Jak/STAT pathway is implicated in the progression of renal inflammation in MRL/lpr mice and targeting this pathway may provide a potential therapeutic approach for LN.
Collapse
Affiliation(s)
- S Wang
- Department of Nephrology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Radaelli E, Del Piero F, Aresu L, Sciarrone F, Vicari N, Mattiello S, Tagliabue S, Fabbi M, Scanziani E. Expression of Major Histocompatibility Complex Class II Antigens in Porcine Leptospiral Nephritis. Vet Pathol 2009; 46:800-9. [DOI: 10.1354/vp.08-vp-0078-r-fl] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Class II major histocompatibility complex (MHCII) is required for the presentation of antigens to CD4 helper T cells. During nephritis, not only primary antigen presenting cells such as histiocytes and lymphocytes, but also cytokine-stimulated tubular epithelial cells express MHCII. Leptospirosis in fattening pigs is characterized by several degrees of nephritis, from absence of lesions to severe multifocal tubulo-interstitial inflammation. Renal tissue from 20 8-month-old pigs with spontaneous nephritis and 6 control pigs without renal lesions were investigated for leptospirosis by indirect immunohistochemistry (IHC) and polymerase chain reaction (PCR). IHC for MHCII also was performed on renal samples. Serum samples were tested for different serovars of Leptospira interrogans. Control pigs were free of interstitial nephritis and negative for leptospirosis by all tests. In pigs with nephritis, serology was positive for serovar Pomona in 19/20 pigs. In 16 of these 19 pigs, leptospiral renal infection was confirmed by PCR and/or indirect IHC. Nephritic lesions were classified histologically into perivascular lymphocytic (4 pigs), lymphofollicular (6 pigs), lymphohistiocytic (8 pigs), and neutrophilic (2 pigs) pattern. MHCII expression by histiocytes and lymphocytes was observed in all lesions. Prominent MHCII expression in regenerating tubular epithelium was observed in lymphofollicular and lymphohistiocytic nephritis. No tubular colocalization between leptospiral and MHCII antigen was observed. Results suggest that during leptospiral nephritis, MHCII contributes to the intensity of the inflammatory response. Furthermore de novo MHCII expression in regenerating tubules may play a role in the defence mechanism against leptospiral tubular colonization.
Collapse
Affiliation(s)
- E. Radaelli
- Department of Veterinary Pathology, Hygiene and Public Health, Section of Veterinary and Avian Pathology, Faculty of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy
| | - F. Del Piero
- School of Veterinary Medicine, Department of Pathobiology and Department of Clinical Studies, New Bolton Center, University of Pennsylvania, PA
| | - L. Aresu
- Department of Public Health, Veterinary Comparative Pathology and Hygiene, University of Padova, Legnaro, Padova, Italy
| | | | - N. Vicari
- Experimental Zooprophylactic Institute of Lombardia and Emilia Romagna, Diagnostic Section of Pavia, Pavia, Italy
| | - S. Mattiello
- Department of Animal Science, Faculty of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy
| | - S. Tagliabue
- Experimental Zooprophylactic Institute of Lombardia and Emilia Romagna, Diagnostic Section of Pavia, Pavia, Italy
| | - M. Fabbi
- Experimental Zooprophylactic Institute of Lombardia and Emilia Romagna, Diagnostic Section of Pavia, Pavia, Italy
| | - E. Scanziani
- Department of Veterinary Pathology, Hygiene and Public Health, Section of Veterinary and Avian Pathology, Faculty of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
24
|
Okamoto A, Fujio K, van Rooijen N, Tsuno NH, Takahashi K, Tsurui H, Hirose S, Elkon KB, Yamamoto K. Splenic phagocytes promote responses to nucleosomes in (NZB x NZW) F1 mice. THE JOURNAL OF IMMUNOLOGY 2008; 181:5264-71. [PMID: 18832681 DOI: 10.4049/jimmunol.181.8.5264] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Autoantigen presentation to T cells is crucial for the development of autoimmune disease. However, the mechanisms of autoantigen presentation are poorly understood. In this study, we show that splenic phagocytes play an important role in autoantigen presentation in murine lupus. Nucleosomes are major autoantigens in systemic lupus erythematosus. We found that nucleosome-specific T cells were stimulated dominantly in the spleen, compared with lymph nodes, lung, and thymus. Among splenic APCs, F4/80(+) macrophages and CD11b(+)CD11c(+) dendritic cells were strong stimulators for nucleosome-specific T cells. When splenic phagocytes were depleted in (NZB x NZW) F(1) (NZB/W F(1)) mice, nucleosome presentation in the spleen was dramatically suppressed. Moreover, depletion of splenic phagocytes significantly suppressed anti-nucleosome Ab and anti-dsDNA Ab production. Proteinuria progression was delayed and survival was prolonged in phagocyte-depleted mice. The numbers of autoantibody- secreting cells were decreased in the spleen from phagocyte-depleted mice. Multiple injections of splenic F4/80(+) macrophages, not those of splenic CD11c(+) dendritic cells, induced autoantibody production and proteinuria progression in NZB/W F(1) mice. These results indicate that autoantigen presentation by splenic phagocytes including macrophages significantly contributes to autoantibody production and disease progression in lupus-prone mice.
Collapse
Affiliation(s)
- Akiko Okamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|