1
|
Zhou Y, Adeluwa T, Zhu L, Salazar-Magaña S, Sumner S, Kim H, Gona S, Nyasimi F, Kulkarni R, Powell JE, Madduri R, Liu B, Chen M, Im HK. scPrediXcan integrates deep learning methods and single-cell data into a cell-type-specific transcriptome-wide association study framework. CELL GENOMICS 2025; 5:100875. [PMID: 40373737 DOI: 10.1016/j.xgen.2025.100875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/31/2024] [Accepted: 04/14/2025] [Indexed: 05/17/2025]
Abstract
Transcriptome-wide association studies (TWASs) help identify disease-causing genes but often fail to pinpoint disease mechanisms at the cellular level because of the limited sample sizes and sparsity of cell-type-specific expression data. Here, we propose scPrediXcan, which integrates state-of-the-art deep learning approaches that predict epigenetic features from DNA sequences with the canonical TWAS framework. Our prediction approach, ctPred, predicts cell-type-specific expression with high accuracy and captures complex gene-regulatory grammar that linear models overlook. Applied to type 2 diabetes (T2D) and systemic lupus erythematosus (SLE), scPrediXcan outperformed the canonical TWAS framework by identifying more candidate causal genes, explaining more genome-wide association study (GWAS) loci and providing insights into the cellular specificity of TWAS hits. Overall, our results demonstrate that scPrediXcan represents a significant advance, promising to deepen our understanding of the cellular mechanisms underlying complex diseases.
Collapse
Affiliation(s)
- Yichao Zhou
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Temidayo Adeluwa
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Lisha Zhu
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sofia Salazar-Magaña
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sarah Sumner
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Hyunki Kim
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Saideep Gona
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Festus Nyasimi
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Rohit Kulkarni
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph E Powell
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW 2052, Australia; Translational Genomics, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Ravi Madduri
- Data Science and Learning Division, Argonne National Laboratory, Chicago, IL 60439, USA
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 119077, Singapore
| | - Mengjie Chen
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Qin X, Zhang M, Liang J, Xu S, Fu X, Liu Z, Tian T, Song J, Lin Y. Nanoparticles encapsulating antigenic peptides induce tolerogenic dendritic cells in situ for treating systemic lupus erythematosus. J Control Release 2025; 380:943-956. [PMID: 39983922 DOI: 10.1016/j.jconrel.2025.02.049] [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: 01/15/2025] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Using Tetrahedral framework nucleic acids, we combined antigenic peptides to create the "DART" vaccine: DNA framework-Antigenic peptide-RNA modification-Targeting aptamer coupling. Generating antigen-specific tolerogenic dendritic cells (tolDCs), for systemic lupus erythematosus (SLE) is a potential therapeutic strategy for addressing compromised autoimmune tolerance. However, simple antigenic peptides degrade easily, lack specificity for delivery to dendritic cells (DCs), and cannot transform DCs to tolDCs. Therefore, this study aims to employ DART to generate tolDCs and compare DART-treated DCs to tolDCs. DART improved peptide stability, specifically targeted DCs, induced tolDCs in situ, and showed promising outcomes in mitigating SLE symptoms in the MRL/lpr mouse model. DART effectively normalized the plasma cytokine levels, glomerulonephritis, and joint lesions in MRL/lpr mice. These findings highlight the potential of the DART vaccine to induce transformation of DCs to tolDCs and address SLE symptoms, suggesting novel therapeutic utility. These findings may advance vaccine design for various autoimmune diseases.
Collapse
Affiliation(s)
- Xin Qin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Mei Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiale Liang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Xu
- The Affiliated Hainan Hospital of Hainan Medical University, Haikou 570101, China
| | - Xiao Fu
- The Affiliated Hainan Hospital of Hainan Medical University, Haikou 570101, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China.
| |
Collapse
|
3
|
Talwar S, Harker JA, Openshaw PJM, Thwaites RS. Autoimmunity in long COVID. J Allergy Clin Immunol 2025; 155:1082-1094. [PMID: 39956285 DOI: 10.1016/j.jaci.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Long COVID (also termed postacute sequelae of SARS-CoV-2, or PASC) affects up to 10% of people recovering from infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnosis is hampered by diffuse symptomatology, lack of biomarkers, incomplete understanding of pathogenesis, and lack of validated treatments. In terms of pathogenesis, hypothesized causes include virus persistence, the legacy of endotheliitis and thrombosis, low-grade tissue-based inflammation and/or scarring, perturbation of the host virome/microbiome, or triggering of autoimmunity. Several studies show preexisting and/or de novo production of autoantibodies after infection with SARS-CoV-2, but the persistence of these antibodies and their role in causing long COVID is debated. Here, we review the mechanisms through which autoimmune responses can arise during and after viral infection, focusing on the evidence for B-cell dysregulation and autoantibody production in acute and long COVID.
Collapse
Affiliation(s)
- Shubha Talwar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - James A Harker
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J M Openshaw
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
4
|
Zhou Y, Adeluwa T, Zhu L, Salazar-Magaña S, Sumner S, Kim H, Gona S, Nyasimi F, Kulkarni R, Powell J, Madduri R, Liu B, Chen M, Im HK. scPrediXcan integrates advances in deep learning and single-cell data into a powerful cell-type-specific transcriptome-wide association study framework. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.11.623049. [PMID: 39605417 PMCID: PMC11601274 DOI: 10.1101/2024.11.11.623049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Transcriptome-wide association studies (TWAS) help identify disease causing genes, but often fail to pinpoint disease mechanisms at the cellular level because of the limited sample sizes and sparsity of cell-type-specific expression data. Here we propose scPrediXcan which integrates state-of-the-art deep learning approaches that predict epigenetic features from DNA sequences with the canonical TWAS framework. Our prediction approach, ctPred, predicts cell-type-specific expression with high accuracy and captures complex gene regulatory grammar that linear models overlook. Applied to type 2 diabetes and systemic lupus erythematosus, scPrediXcan outperformed the canonical TWAS framework by identifying more candidate causal genes, explaining more genome-wide association studies (GWAS) loci, and providing insights into the cellular specificity of TWAS hits. Overall, our results demonstrate that scPrediXcan represents a significant advance, promising to deepen our understanding of the cellular mechanisms underlying complex diseases.
Collapse
Affiliation(s)
- Yichao Zhou
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Temidayo Adeluwa
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Lisha Zhu
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Sofia Salazar-Magaña
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Sarah Sumner
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Hyunki Kim
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Saideep Gona
- Committee of Genetic, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Festus Nyasimi
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| | - Rohit Kulkarni
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joseph Powell
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, Australia
| | - Ravi Madduri
- Data Science and Learning Division, Argonne National Laboratory, Chicago, Illinois, United States of America
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore
| | - Mengjie Chen
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Hae Kyung Im
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
5
|
Lauver MD, Katz ZE, Markus H, Derosia NM, Jin G, Ayers KN, Butic AB, Bushey K, Abendroth CS, Liu DJ, Lukacher AE. The CXCR6-CXCL16 axis mediates T cell control of polyomavirus infection in the kidney. PLoS Pathog 2025; 21:e1012969. [PMID: 40043065 PMCID: PMC11922244 DOI: 10.1371/journal.ppat.1012969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/19/2025] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
BK polyomavirus (PyV) establishes lifelong asymptomatic infections in the reno-urinary system of most humans. BKPyV-associated nephropathy is the leading infectious cause of kidney allograft loss. Using mouse PyV, a natural murine pathogen that also persists in the kidney, we define a dominant chemokine receptor-chemokine axis that directs T cell infiltration of the kidney. We found that CXCR6 was required for CD4+ and CD8+ T cells to be recruited to and retained in the kidney, respectively. Absence of CXCR6 impaired virus control in the kidney. The soluble form of CXCL16 was increased in kidneys of infected mice and in vivo CXCL16 neutralization reduced numbers of virus-specific CD8+ T cells infiltrating the kidney. In vivo administration of IL-12 upregulated CXCR6 expression on virus-specific CD8+ T cells, improved T cell recruitment to the infected kidney, and reduced virus levels. Notably, T cells in kidney biopsies from PyV-associated nephropathy patients express CXCR6 and transcriptional analysis shows significant upregulation of CXCR6 and CXCL16. These findings demonstrate the importance of the CXCR6-CXCL16 axis in regulating T cell responses in the kidney to PyV infection.
Collapse
MESH Headings
- Animals
- Chemokine CXCL16
- Receptors, CXCR6
- Mice
- Polyomavirus Infections/immunology
- Polyomavirus Infections/metabolism
- Polyomavirus Infections/pathology
- Polyomavirus Infections/virology
- CD8-Positive T-Lymphocytes/immunology
- Humans
- Kidney/immunology
- Kidney/virology
- Kidney/pathology
- Mice, Inbred C57BL
- Chemokines, CXC/immunology
- Chemokines, CXC/metabolism
- Chemokines, CXC/genetics
- Receptors, Scavenger/immunology
- Receptors, Scavenger/metabolism
- Receptors, Scavenger/genetics
- CD4-Positive T-Lymphocytes/immunology
- Tumor Virus Infections/immunology
- Mice, Knockout
- BK Virus/immunology
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Receptors, Virus/genetics
- Receptors, Chemokine/immunology
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/genetics
- Kidney Diseases/immunology
- Kidney Diseases/virology
Collapse
Affiliation(s)
- Matthew D. Lauver
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Zoe E. Katz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Havell Markus
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Nicole M. Derosia
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Ge Jin
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Katelyn N. Ayers
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Arrienne B. Butic
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Kaitlyn Bushey
- Bio X Cell, Inc., Lebanon, New Hampshire, United States of America
| | - Catherine S. Abendroth
- Department of Pathology, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Dajiang J. Liu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Aron E. Lukacher
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
6
|
Shi Y, Qin Y, Li Y, Jiang P, Wei K, Zhao J, Shan Y, Zheng Y, Zhao F, Zhou M, Li L, Shen Y, Lv X, Zheng Y, Guo S, Ding Q, Chang C, He D. Comparative Analysis of CXCR5 Circulating DNA Methylation Levels in Autoimmune Rheumatic Diseases. Immun Inflamm Dis 2025; 13:e70128. [PMID: 39835879 PMCID: PMC11748209 DOI: 10.1002/iid3.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/08/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE To assess CXC chemokine receptor 5 (CXCR5) circulating DNA methylation differences in autoimmune rheumatic diseases and their relation with clinical features. METHODS Targeted methylation sequencing was performed using peripheral blood from 164 rheumatoid arthritis (RA), 30 systemic lupus erythematosus (SLE), 30 ankylosing spondylitis (AS), 30 psoriatic arthritis (PsA), 24 Sjögren's syndrome (SS) patients, and 30 healthy controls (HC). RESULTS Significant differences in CXCR5 cg19599951 methylation were found between RA and HC, as well as AS and SLE. RA patients exhibited higher methylation than HC and AS (p < 0.01) but lower than SLE (p < 0.05). SLE patients showed higher methylation compared to HC, AS, and PsA (p < 0.001, 0.01, and 0.05, respectively). No significant differences were found in patients with SS compared to other autoimmune diseases and HC. Methylation at cg19599951_103 (r = 0.17, p < 0.05) and cg19599951_209 (r = 0.22, p < 0.01), along with the CC haplotype (r = 0.21, p < 0.01), showed significant positive correlations with erythrocyte sedimentation rate (ESR), while the CT (r = -0.27, p < 0.001) and TT haplotypes (r = -0.19, p < 0.05) were negatively correlated. For C-reactive protein (CRP), methylation at cg19599951_103 (r = 0.29, p < 0.001) and cg19599951_209 (r = 0.33, p < 0.0001), and the CC haplotype (r = 0.34, p < 0.0001) was positively correlated, whereas the CT (r = -0.36, p < 0.0001) and TT (r = -0.30, p < 0.0001) haplotypes were negatively correlated. Significant negative correlations were observed between the CT haplotype and rheumatoid factor (r = -0.25, p < 0.01), and anti-citrullinated protein antibody (r = -0.20, p < 0.05). No significant correlations were found in patients with SLE, AS, and SS. Receiver operating characteristic analysis showed CXCR5 methylation could classify patients with RA versus those with AS (AUC: 0.624-0.967). CONCLUSION Differential circulating CXCR5 methylation levels were observed in autoimmune rheumatic diseases, which correlated with inflammatory mediators in RA and may serve as potential biomarkers for RA diagnosis.
Collapse
Affiliation(s)
- Yiming Shi
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Yingying Qin
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Yunshen Li
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Ping Jiang
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Kai Wei
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Jianan Zhao
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Yu Shan
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Yixin Zheng
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Fuyu Zhao
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Mi Zhou
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Li Li
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Yu Shen
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Xinliang Lv
- Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous RegionInner Mongolia Autonomous RegionHohhotChina
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical SciencesShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shicheng Guo
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qin Ding
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Cen Chang
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| | - Dongyi He
- Department of RheumatologyGuanghua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
- Guanghua Clinical Medical CollegeShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Arthritis Research in Integrative MedicineShanghai Academy of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
7
|
Gao R, Yang H, Wang Y. SETD3 functions beyond histidine methylation. Life Sci 2024; 357:123064. [PMID: 39299385 DOI: 10.1016/j.lfs.2024.123064] [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: 08/07/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
SETD3 is a member of SET domain-containing proteins. It has been discovered as the first metazoan protein (actin) histidine methyltransferase. In addition to this well-characterized molecular function of SETD3, it has been clearly shown to be involved in multiple biological processes, such as cell differentiation, tumorigenesis and viral infection. Here, we summarize the current knowledge on the roles of SETD3 beyond its histidine methyltransferase activity, and outline its cellular and molecular modes of action, as well as the upstream regulation on SETD3, therefore providing insights for the molecular basis of how SETD3 fine regulates multiple physiological and pathological processes.
Collapse
Affiliation(s)
- Rui Gao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen 361000, China.
| | - Hao Yang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen 361000, China
| | - Yan Wang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen 361000, China
| |
Collapse
|
8
|
Moysidou E, Christodoulou M, Lioulios G, Stai S, Karamitsos T, Dimitroulas T, Fylaktou A, Stangou M. Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis. Int J Mol Sci 2024; 25:10905. [PMID: 39456692 PMCID: PMC11508046 DOI: 10.3390/ijms252010905] [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: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by considerable changes in peripheral lymphocyte structure and function, that plays a critical role in commencing and reviving the inflammatory and immune signaling pathways. In healthy individuals, B lymphocytes have a major role in guiding and directing defense mechanisms against pathogens. Certain changes in B lymphocyte phenotype, including alterations in surface and endosomal receptors, occur in the presence of SLE and lead to dysregulation of peripheral B lymphocyte subpopulations. Functional changes are characterized by loss of self-tolerance, intra- and extrafollicular activation, and increased cytokine and autoantibody production. T lymphocytes seem to have a supporting, rather than a leading, role in the disease pathogenesis. Substantial aberrations in peripheral T lymphocyte subsets are evident, and include a reduction of cytotoxic, regulatory, and advanced differentiated subtypes, together with an increase of activated and autoreactive forms and abnormalities in follicular T cells. Up-regulated subpopulations, such as central and effector memory T cells, produce pre-inflammatory cytokines, activate B lymphocytes, and stimulate cell signaling pathways. This review explores the pivotal roles of B and T lymphocytes in the pathogenesis of SLE and Lupus Nephritis, emphasizing the multifaceted mechanisms and interactions and their phenotypic and functional dysregulations.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Theodoros Karamitsos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Cardiology, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros Dimitroulas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 4th Department of Medicine, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
9
|
Wiley MM, Khatri B, Joachims ML, Tessneer KL, Stolarczyk AM, Rasmussen A, Anaya JM, Aqrawi LA, Bae SC, Baecklund E, Björk A, Brun JG, Bucher SM, Dand N, Eloranta ML, Engelke F, Forsblad-d’Elia H, Fugmann C, Glenn SB, Gong C, Gottenberg JE, Hammenfors D, Imgenberg-Kreuz J, Jensen JL, Johnsen SJA, Jonsson MV, Kelly JA, Khanam S, Kim K, Kvarnström M, Mandl T, Martín J, Morris DL, Nocturne G, Norheim KB, Olsson P, Palm Ø, Pers JO, Rhodus NL, Sjöwall C, Skarstein K, Taylor KE, Tombleson P, Thorlacius GE, Venuturupalli S, Vital EM, Wallace DJ, Grundahl KM, Radfar L, Brennan MT, James JA, Scofield RH, Gaffney PM, Criswell LA, Jonsson R, Appel S, Eriksson P, Bowman SJ, Omdal R, Rönnblom L, Warner BM, Rischmueller M, Witte T, Farris AD, Mariette X, Shiboski CH, Sjögren’s International Collaborative Clinical Alliance (SICCA), Wahren-Herlenius M, Alarcón-Riquelme ME, PRECISESADS Clinical Consortium, Ng WF, UK Primary Sjögren’s Syndrome Registry, Sivils KL, Guthridge JM, Adrianto I, Vyse TJ, Tsao BP, Nordmark G, Lessard CJ. Variants in the DDX6-CXCR5 autoimmune disease risk locus influence the regulatory network in immune cells and salivary gland. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561076. [PMID: 39071447 PMCID: PMC11275775 DOI: 10.1101/2023.10.05.561076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Fine mapping and bioinformatic analysis of the DDX6-CXCR5 genetic risk association in Sjögren's Disease (SjD) and Systemic Lupus Erythematosus (SLE) identified five common SNPs with functional evidence in immune cell types: rs4938573, rs57494551, rs4938572, rs4936443, rs7117261. Functional interrogation of nuclear protein binding affinity, enhancer/promoter regulatory activity, and chromatin-chromatin interactions in immune, salivary gland epithelial, and kidney epithelial cells revealed cell type-specific allelic effects for all five SNPs that expanded regulation beyond effects on DDX6 and CXCR5 expression. Mapping the local chromatin regulatory network revealed several additional genes of interest, including lnc-PHLDB1-1. Collectively, functional characterization implicated the risk alleles of these SNPs as modulators of promoter and/or enhancer activities that regulate cell type-specific expression of DDX6, CXCR5, and lnc-PHLDB1-1, among others. Further, these findings emphasize the importance of exploring the functional significance of SNPs in the context of complex chromatin architecture in disease-relevant cell types and tissues.
Collapse
Affiliation(s)
- Mandi M. Wiley
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Bhuwan Khatri
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Michelle L. Joachims
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
| | - Kandice L. Tessneer
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Anna M. Stolarczyk
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Astrid Rasmussen
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | | | - Lara A. Aqrawi
- Department of Health Sciences, Kristiania University College, Oslo, Norway
- University of Oslo, Norway
| | | | | | | | - Johan G. Brun
- University of Bergen, Bergen, Norway
- Haukeland University Hospital, Bergen, Norway
| | | | - Nick Dand
- King’s College London, London, United Kingdom
| | | | | | | | | | - Stuart B. Glenn
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Chen Gong
- King’s College London, London, United Kingdom
| | | | | | | | | | | | | | - Jennifer A. Kelly
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Sharmily Khanam
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
| | | | | | | | - Javier Martín
- Instituto de Biomedicina y Parasitología López-Neyra, Granada, Spain
| | | | - Gaetane Nocturne
- Université Paris-Saclay, Paris, France
- Assistance Publique – Hôpitaux de Paris, Hôpital Bicêtre, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kiely M. Grundahl
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
| | - Lida Radfar
- University of Oklahoma College of Dentistry, Oklahoma City, Oklahoma, USA
| | | | - Judith A. James
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - R. Hal Scofield
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- US Department of Veteran Affairs Medical Center, Oklahoma City, Oklahoma, USA
| | - Patrick M. Gaffney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lindsey A. Criswell
- University of California San Francisco, San Francisco, California, USA
- National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | | | | | | | - Simon J. Bowman
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Roald Omdal
- University of Bergen, Bergen, Norway
- Stavanger University Hospital, Stavanger, Norway
| | | | - Blake M. Warner
- National Institute of Dental and Craniofacial Research, Bethesda, Maryland, USA
| | | | | | - A. Darise Farris
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xavier Mariette
- Université Paris-Saclay, Paris, France
- Assistance Publique – Hôpitaux de Paris, Hôpital Bicêtre, Paris, France
| | | | | | | | - Marta E. Alarcón-Riquelme
- Karolinska Institutet, Solna, Sweden
- Genyo, Center for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Spain
| | | | - Wan-Fai Ng
- NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | | | - Kathy L. Sivils
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
| | - Joel M. Guthridge
- Arthritis and Clinical Immunology Research Program, OMRF, Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Indra Adrianto
- Center for Bioinformatics, Department of Public Health Sciences, Henry Ford Health, Detroit, Michigan, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | | | - Betty P. Tsao
- Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Christopher J. Lessard
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
10
|
Sato S, Zhang XK, Matsuoka N, Sumichika Y, Saito K, Yoshida S, Matsumoto H, Temmoku J, Fujita Y, Asano T, Migita K. Transcription factor Fli-1 impacts the expression of CXCL13 and regulates immune cell infiltration into the kidney in MRL/lpr mouse. Lupus Sci Med 2023; 10:10/1/e000870. [PMID: 37094946 PMCID: PMC10152041 DOI: 10.1136/lupus-2022-000870] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE Friend leukaemia virus integration 1 (Fli-1) regulates chemokine/cytokine expression and thus plays an important role in the development of lupus nephritis. Chemokine CXC ligand 13 (CXCL13) is a chemokine that promotes the formation of ectopic lymphoid structures and has been reported to be associated with the pathogenesis of lupus nephritis. The relationship between Fli-1 and CXCL13 is unknown. This study aims to elucidate whether Fli-1 impacts CXCL13 expression and contributes to the progression of lupus-like nephritis in adult MRL/lpr mouse. METHODS Serum CXCL13 levels were measured in adult wild-type (WT) MRL/lpr mice and Fli-1 heterozygote knockout (Fli-1+/-) MRL/lpr mice (4 months old or older) using ELISA. Renal mRNA expression (CXCL13 and related molecules) was measured using real-time PCR method. Kidneys were removed, stained and evaluated using a pathology scoring system. The grade of CXCL13 or CXC-chemokine receptor type 5 (CXCR5)-positive immune cell infiltration into the kidney was evaluated using immunostaining with anti-CXCL13 or anti-CXCR5 antibodies. We also used immunofluorescence staining with CXCL13- and CD11b-specific antibodies to detect the infiltration of CXCL13/CD11b double-positive immune cells. RESULTS Serum CXCL13 levels in Fli-1+/- MRL/lpr mice were significantly lower than that in WT MRL/lpr mice (545.5 and 960.5 pg/mL, p=0.02). Renal expression of CXCL13 mRNA and SRY-related HMG box4 (Sox4) (an important factor for B-cell development) levels were significantly lower in Fli-1+/- MRL/lpr mice. Renal histology scores in WT MRL/lpr mice revealed significantly increased glomerular inflammation. Despite similar interstitial immune cell infiltration into the kidney, the number of CXCL13- and CXCR5-positive cells was significantly lower in Fli-1+/- MRL/lpr mice than in WT mice. Furthermore, immunofluorescence staining revealed that Fli-1+/-MRL/lpr mice had significantly fewer CXCL13/CD11b double-positive immune cells. CONCLUSION Fli-1 regulates renal Sox4 mRNA expression and infiltration of CXCR5-positive cells as well as CXCL13/CD11b double-positive immune cells into the kidney, which affects CXCL13 expression and lupus-like nephritis.
Collapse
Affiliation(s)
- Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Xian K Zhang
- Department of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
11
|
Newman-Rivera AM, Kurzhagen JT, Rabb H. TCRαβ+ CD4-/CD8- "double negative" T cells in health and disease-implications for the kidney. Kidney Int 2022; 102:25-37. [PMID: 35413379 PMCID: PMC9233047 DOI: 10.1016/j.kint.2022.02.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 12/22/2022]
Abstract
Double negative (DN) T cells, one of the least studied T lymphocyte subgroups, express T cell receptor αβ but lack CD4 and CD8 coreceptors. DN T cells are found in multiple organs including kidney, lung, heart, gastrointestinal tract, liver, genital tract, and central nervous system. DN T cells suppress inflammatory responses in different disease models including experimental acute kidney injury, and significant evidence supports an important role in the pathogenesis of systemic lupus erythematosus. However, little is known about these cells in other kidney diseases. Therefore, it is important to better understand different functions of DN T cells and their signaling pathways as promising therapeutic targets, particularly with the increasing application of T cell-directed therapy in humans. In this review, we aim to summarize studies performed on DN T cells in normal and diseased organs in the setting of different disease models with a focus on kidney.
Collapse
Affiliation(s)
| | | | - Hamid Rabb
- Nephrology Division, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
12
|
Chen X, Wang D, Zhu X. Application of double-negative T cells in haematological malignancies: recent progress and future directions. Biomark Res 2022; 10:11. [PMID: 35287737 PMCID: PMC8919567 DOI: 10.1186/s40364-022-00360-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/26/2022] [Indexed: 12/16/2022] Open
Abstract
Haematologic malignancies account for a large proportion of cancers worldwide. The high occurrence and mortality of haematologic malignancies create a heavy social burden. Allogeneic haematopoietic stem cell transplantation is widely used in the treatment of haematologic malignancies. However, graft-versus-host disease and relapse after allogeneic haematopoietic stem cell transplantation are inevitable. An emerging treatment method, adoptive cellular therapy, has been effectively used in the treatment of haematologic malignancies. T cells, natural killer (NK) cells and tumour-infiltrating lymphocytes (TILs) all have great potential in therapeutic applications, and chimeric antigen receptor T (CAR-T) cell therapy especially has potential, but cytokine release syndrome and off-target effects are common. Efficient anticancer measures are urgently needed. In recent years, double-negative T cells (CD3+CD4-CD8-) have been found to have great potential in preventing allograft/xenograft rejection and inhibiting graft-versus-host disease. They also have substantial ability to kill various cell lines derived from haematologic malignancies in an MHC-unrestricted manner. In addition, healthy donor expanded double-negative T cells retain their antitumour abilities and ability to inhibit graft-versus-host disease after cryopreservation under good manufacturing practice (GMP) conditions, indicating that double-negative T cells may be able to be used as an off-the-shelf product. In this review, we shed light on the potential therapeutic ability of double-negative T cells in treating haematologic malignancies. We hope to exploit these cells as a novel therapy for haematologic malignancies.
Collapse
Affiliation(s)
- Xingchi Chen
- Department of hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.,Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.,Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, 230001, Anhui, China
| | - Dongyao Wang
- Department of hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.,Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.,Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, 230001, Anhui, China
| | - Xiaoyu Zhu
- Department of hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China. .,Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China. .,Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, 230001, Anhui, China.
| |
Collapse
|
13
|
Pan Z, Zhu T, Liu Y, Zhang N. Role of the CXCL13/CXCR5 Axis in Autoimmune Diseases. Front Immunol 2022; 13:850998. [PMID: 35309354 PMCID: PMC8931035 DOI: 10.3389/fimmu.2022.850998] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
CXCL13 is a B-cell chemokine produced mainly by mesenchymal lymphoid tissue organizer cells, follicular dendritic cells, and human T follicular helper cells. By binding to its receptor, CXCR5, CXCL13 plays an important role in lymphoid neogenesis, lymphoid organization, and immune responses. Recent studies have found that CXCL13 and its receptor CXCR5 are implicated in the pathogenesis of several autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, primary Sjögren’s syndrome, myasthenia gravis, and inflammatory bowel disease. In this review, we discuss the biological features of CXCL13 and CXCR5 and the recent findings on the pathogenic roles of the CXCL13/CXCR5 axis in autoimmune diseases. Furthermore, we discuss the potential role of CXCL13 as a disease biomarker and therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Zijian Pan
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tong Zhu
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjun Liu
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Nannan Zhang
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, and State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
- *Correspondence: Nannan Zhang,
| |
Collapse
|
14
|
Bekele Feyissa Y, Chiodi F, Sui Y, Berzofsky JA. The Role of CXCL13 in Antibody Responses to HIV-1 Infection and Vaccination. Front Immunol 2021; 12:638872. [PMID: 33732259 PMCID: PMC7959754 DOI: 10.3389/fimmu.2021.638872] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
CXCL13 signals through the G protein-coupled chemokine receptor CXCR5 to drive development of secondary lymphoid tissue as well as B cell and Tfh cell trafficking to germinal centers (GC), which leads to the differentiation of B cells to plasma cells and memory B cells. CXCL13 has been proposed as a general plasma biomarker for GC activities. In HIV-1 infected individuals, plasma CXCL13 levels have been associated with the rate of disease progression to AIDS. Moreover, CXCL13 production has been reported to be increased in HIV-1-infected lymph nodes, which may drive increased downregulation of CXCR5. In this review, we address the role of CXCL13 in HIV-1 infected individuals with regard to GC formation, generation of broadly neutralizing antibodies after infection and vaccination, and AIDS-related B cell lymphoma.
Collapse
Affiliation(s)
- Yonas Bekele Feyissa
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Solna, Sweden
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
15
|
Bhamidipati K, Silberstein JL, Chaichian Y, Baker MC, Lanz TV, Zia A, Rasheed YS, Cochran JR, Robinson WH. CD52 Is Elevated on B cells of SLE Patients and Regulates B Cell Function. Front Immunol 2021; 11:626820. [PMID: 33658999 PMCID: PMC7917337 DOI: 10.3389/fimmu.2020.626820] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by B cell dysregulation and breaks in tolerance that lead to the production of pathogenic autoantibodies. We performed single-cell RNA sequencing of B cells from healthy donors and individuals with SLE which revealed upregulated CD52 expression in SLE patients. We further demonstrate that SLE patients exhibit significantly increased levels of B cell surface CD52 expression and plasma soluble CD52, and levels of soluble CD52 positively correlate with measures of lupus disease activity. Using CD52-deficient JeKo-1 cells, we show that cells lacking surface CD52 expression are hyperresponsive to B cell receptor (BCR) signaling, suggesting an inhibitory role for the surface-bound protein. In healthy donor B cells, antigen-specific BCR-activation initiated CD52 cleavage in a phospholipase C dependent manner, significantly reducing cell surface levels. Experiments with recombinant CD52-Fc showed that soluble CD52 inhibits BCR signaling in a manner partially-dependent on Siglec-10. Moreover, incubation of unstimulated B cells with CD52-Fc resulted in the reduction of surface immunoglobulin and CXCR5. Prolonged incubation of B cells with CD52 resulted in the expansion of IgD+IgMlo anergic B cells. In summary, our findings suggest that CD52 functions as a homeostatic protein on B cells, by inhibiting responses to BCR signaling. Further, our data demonstrate that CD52 is cleaved from the B cell surface upon antigen engagement, and can suppress B cell function in an autocrine and paracrine manner. We propose that increased expression of CD52 by B cells in SLE represents a homeostatic mechanism to suppress B cell hyperactivity.
Collapse
Affiliation(s)
- Kartik Bhamidipati
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - John L. Silberstein
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Yashaar Chaichian
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Matthew C. Baker
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Tobias V. Lanz
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Amin Zia
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Yusuf S. Rasheed
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Jennifer R. Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - William H. Robinson
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
16
|
Yang B, Zhao M, Wu H, Lu Q. A Comprehensive Review of Biological Agents for Lupus: Beyond Single Target. Front Immunol 2020; 11:539797. [PMID: 33123125 PMCID: PMC7573553 DOI: 10.3389/fimmu.2020.539797] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that involves multiple immune cells. Due to its complex pathogenesis, the effectiveness of traditional treatment methods is limited. Many patients have developed resistance to conventional treatment or are not sensitive to steroid and immunosuppressant therapy, and so emerging therapeutic antibodies have become an alternative and have been shown to work well in many patients with moderate and severe SLE. This review summarizes the biological agents that are in the preclinical and clinical trial study of SLE. In addition to the various monoclonal antibodies that have been studied for a long time, such as belimumab and rituximab, we focused on another treatment for SLE, bispecific antibodies (BsAbs) such as tibulizumab, which simultaneously targets multiple pathogenic cytokines or pathways. Although the application of BsAbs in cancer has been intensively studied, their application in autoimmune diseases is still in the infant stage. This unique combined mechanism of action may provide a novel therapeutic strategy for SLE.
Collapse
Affiliation(s)
- Bingyi Yang
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming Zhao
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, The Second Xiangya Hospital of Central South University; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
17
|
Tocut M, Shoenfeld Y, Zandman-Goddard G. Systemic lupus erythematosus: an expert insight into emerging therapy agents in preclinical and early clinical development. Expert Opin Investig Drugs 2020; 29:1151-1162. [PMID: 32755494 DOI: 10.1080/13543784.2020.1807004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic disease that is potentially fatal. There is no cure for SLE and the medications used are associated with toxic side effects. In the era of revolutionary emerging novel biologic agents, the design and investigation of targeted therapy for these patients is necessary. Novel therapies under investigation in phase II-III clinical trials showed promising results. Therapies can target various pathways involved in SLE including cytokines, signal transduction inhibitors, B-cell depletion and interference with co-stimulation. Of interest is the proof of concept of sequential therapy. AREAS COVERED We performed an extensive literature search via PubMed, Medline, Elsevier Science and Springer Link databases between the years 2014-2020 using the following terms: SLE, novel treatments. We have reviewed 232 articles and selected those articles that (i) focus on phase II-III emerging therapies and (ii) offer new findings from existing therapies, which reveal breakthrough concepts in SLE treatment. EXPERT OPINION It is still difficult to crack the puzzle of a successful SLE treatment approach. New strategies with potential may encompass the targeting of more than one protein. Another way forward is to identify each SLE patient and personalize therapy by clinical manifestations, disease activity, serology and activated protein.
Collapse
Affiliation(s)
- Milena Tocut
- Department of Internal Medicine C, Wolfson Medical Center , Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel
| | - Yehuda Shoenfeld
- Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel.,Center for Autoimmune Diseases, Sheba Medical Center , Ramat Gan, Israel.,I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
| | - Gisele Zandman-Goddard
- Department of Internal Medicine C, Wolfson Medical Center , Holon, Israel.,Sackler Faculty of Medicine, Tel-Aviv University , Tel Aviv, Israel
| |
Collapse
|
18
|
Ganugula R, Arora M, Zou D, Agarwal SK, Mohan C, Kumar MNVR. A highly potent lymphatic system-targeting nanoparticle cyclosporine prevents glomerulonephritis in mouse model of lupus. SCIENCE ADVANCES 2020; 6:eabb3900. [PMID: 32582860 PMCID: PMC7292630 DOI: 10.1126/sciadv.abb3900] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Cyclosporine A (CsA) is a powerful immunosuppressant, but it is an ineffective stand-alone treatment for systemic lupus erythematosus (SLE) due to poor target tissue distribution and renal toxicity. We hypothesized that CD71 (transferrin receptor 1)-directed delivery of CsA to the lymphatic system would improve SLE outcomes in a murine model. We synthesized biodegradable, ligand-conjugated nanoparticles [P2Ns-gambogic acid (GA)] targeting CD71. GA conjugation substantially increased nanoparticle association with CD3+ or CD20+ lymphocytes and with intestinal lymphoid tissues. In orally dosed MRL-lpr mice, P2Ns-GA-encapsulated CsA increased lymphatic drug delivery 4- to 18-fold over the ligand-free formulation and a commercial CsA capsule, respectively. Improved lymphatic bioavailability of CsA was paralleled by normalization of anti-double-stranded DNA immunoglobulin G titer, plasma cytokines, and glomerulonephritis. Thus, this study demonstrates the translational potential of nanoparticles that enhance the targeting of lymphatic tissues, transforming CsA into a potent single therapeutic for SLE.
Collapse
Affiliation(s)
- Raghu Ganugula
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Meenakshi Arora
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Dianxiong Zou
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Sandeep K. Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Biology of Inflammation Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | | |
Collapse
|
19
|
Liao J, Luo S, Yang M, Lu Q. Overexpression of CXCR5 in CD4+ T cells of SLE patients caused by excessive SETD3. Clin Immunol 2020; 214:108406. [DOI: 10.1016/j.clim.2020.108406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/15/2022]
|
20
|
Yu J, Zeng T, Wu Y, Tian Y, Tan L, Duan X, Wu Q, Li H, Yu L. Neutrophil-to-C3 ratio and neutrophil-to-lymphocyte ratio were associated with disease activity in patients with systemic lupus erythematosus. J Clin Lab Anal 2018; 33:e22633. [PMID: 30129188 DOI: 10.1002/jcla.22633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus is prone to recurrent attacks, and its treatment is related to disease activities. It is important to accurately assess the patient's disease activity. So, the purpose of this study was to investigate the relation between neutrophil-to-C3 ratio (NC3 R), neutrophil-to-lymphocyte ratio (NLR), and disease activity in patients with Systemic lupus erythematosus (SLE). METHODS This was a retrospective study. One hundred and ninety-four patients with SLE and 71 healthy controls were included in this study. We divided the patients into two groups according to the SLE disease activity (SLEDAI). Group 1 included patients with a score of >9 (patients with severe disease activity), and Group 2 included patients with a score of 9 and lower (patients with mild disease activity). Correlations between NC3 R, NLR, and disease activity were analyzed. RESULTS NC3 R and NLR in patients with SLE were obviously higher compared to healthy controls (P < 0.05). There was an obviously significant difference in NC3 R and NLR between Group 1 and Group 2 (P < 0.05). SLEDAI scores were positively correlated with NC3 R (r = 0.353, P < 0.01) and NLR (r = 0.237, P = 0.01). Receiver operating characteristic (ROC) curve analysis showed that the cutoff value of NC3 R to identify SLE with high disease activity was 5.935, with sensitivity and specificity being 75.9% and 67.0%, while that of NLR was 2.293, with sensitivity being 68.9% and specificity being 82.8%. CONCLUSION NC3 R and NLR are two useful inflammatory markers for evaluating disease activity in patients with SLE.
Collapse
Affiliation(s)
- Jianlin Yu
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Tingting Zeng
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yang Wu
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yongjian Tian
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Liming Tan
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xinwang Duan
- Department of Rheumatology of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Qiong Wu
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Hua Li
- Department of Clinical Laboratory of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Le Yu
- Department of Rheumatology of the Second Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
21
|
CD11c-Specific Deletion Reveals CREB as a Critical Regulator of DC Function during the Germinal Center Response. J Immunol Res 2018; 2018:8947230. [PMID: 29854847 PMCID: PMC5964551 DOI: 10.1155/2018/8947230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/24/2018] [Accepted: 03/11/2018] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells (DCs) are crucial for the balance between immune response and tolerance, but the molecular mechanism regulating development, differentiation, and homeostasis are poorly understood. The transcriptional activator CREB is involved in regulating different cells of the innate and adaptive immune system and is a transcriptional regulator of development, survival, activation, or proliferation in macrophages, dendritic cells, B cells, and T cells. To directly examine the role of CREB in the regulation of DCs, the CREB gene was targeted for deletion with a CD11c-cre transgene. The deletion of CREB in CD11c+ cells did not involve any developmental or systemic defects within DC populations. However, CREB deficiency in CD11c+ cells reduced germinal center (GC) B cells in steady state, and immunization with NP-CGG resulted in a reduced formation of GCs, paralleled by the reduced production of IgGs in sera of immunized mice. In conclusion, we demonstrate that CREB expression in CD11c+ cells enhances germinal center responses, most likely by altering DC function, which might have implications for autoimmune diseases that are associated with dysregulated GC responses.
Collapse
|
22
|
Stabilized β-Catenin Ameliorates ALPS-Like Symptoms of B6/ lpr Mice. J Immunol Res 2017; 2017:3469108. [PMID: 29250557 PMCID: PMC5700472 DOI: 10.1155/2017/3469108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/15/2017] [Indexed: 01/05/2023] Open
Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is an incurable disease mainly caused by the defect of Fas-mediated apoptosis and characterized by nonmalignant autoimmune lymphoproliferation. Stabilized β-catenin could not only potentiate Fas-mediated T cell apoptosis via upregulating the expression of Fas on activated T cells, but also potentiate T cell apoptosis via intrinsic apoptotic pathway. In the present study, we introduced β-catTg into lpr/lpr mice and aimed to explore the potential role of stabilized β-catenin (β-catTg) in the development of ALPS-like phenotypes of lpr/lpr mice. We found that the total splenocyte cells and some compositions were slightly downregulated in β-catTglpr/lpr mice, especially the CD4 and CD8 TEM cells were significantly reduced. Meanwhile, stabilized β-catenin obviously decreased the numbers of spleen TCRβ+CD4−CD8− T (DNT) cells, and the levels of some serum proinflammatory factors also were lowered in β-catTglpr/lpr mice. Beyond that, stabilized β-catenin slightly lowered the levels of the serum autoantibodies and the scores of kidney histopathology of β-catTglpr/lpr mice compared with lpr/lpr mice. Our study suggested that stabilized β-catenin ameliorated some ALPS-like symptoms of lpr/lpr mice by potentiating Fas-independent signal-mediated T cell apoptosis, which might uncover a potential novel therapeutic direction for ALPS.
Collapse
|
23
|
Fang C, Luo T, Lin L. The correlational research among serum CXCL13 levels, circulating plasmablasts and memory B cells in patients with systemic lupus erythematosus: A STROBE-compliant article. Medicine (Baltimore) 2017; 96:e8675. [PMID: 29310341 PMCID: PMC5728742 DOI: 10.1097/md.0000000000008675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We investigated whether serum CXC ligand 13 protein (CXCL13) levels correlate with the circulating plasmablasts and memory B-cells alteration in systemic lupus erythematosus (SLE) patients. The diagnostic use of CXCL13 concentrations in active lupus was also analyzed.A total of 36 SLE patients and 18 healthy controls were included. Serum CXCL13 levels were examined by enzyme-linked immunosorbent assay. The frequency and absolute count of circulating plasmablasts and memory B cells were analyzed by flow cytometry. Receiver operating characteristic curves (ROC curves) were generated to analyze the utility of serum CXCL13 level and plasmablasts frequency as tools for the recognition of active SLE.Elevation of serum CXCL13 levels, higher plasmablasts frequency, and reduction of memory B-cells count were observed in SLE patients, compared with healthy controls. Interestingly, correlational analyses showed not only significantly positive association between CXCL13 levels and SLE Disease Activity Index (SLEDAI) or plasmablasts frequency, but an inverse correlation between CXCL13 concentration and memory B-cell count. ROC curves showed that serum CXCL13 level and plasmablasts frequency were practical in identifying active disease from overall SLE patients, with considerable accuracy.Serum CXCL13 levels correlate with the alteration of plasmablasts and memory B cells in SLE. CXCL13 may be used as a practical tool in judgment of active SLE.
Collapse
Affiliation(s)
| | - Tingting Luo
- Ultrasonic Cardiogram Department, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | | |
Collapse
|
24
|
Lupus érythémateux systémique et lymphopénie : aspects cliniques et physiopathologiques. Rev Med Interne 2017; 38:603-613. [DOI: 10.1016/j.revmed.2017.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/20/2022]
|
25
|
Davis LS, Reimold AM. Research and therapeutics-traditional and emerging therapies in systemic lupus erythematosus. Rheumatology (Oxford) 2017; 56:i100-i113. [PMID: 28375452 DOI: 10.1093/rheumatology/kew417] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Indexed: 12/21/2022] Open
Abstract
This review summarizes traditional and emerging therapies for SLE. Evidence suggests that the heterogeneity of SLE is a crucial aspect contributing to the failure of large clinical trials for new targeted therapies. A clearer understanding of the mechanisms driving disease pathogenesis combined with recent advances in medical science are predicted to enable accelerated progress towards improved SLE diagnosis and personalized approaches to treatment.
Collapse
Affiliation(s)
- Laurie S Davis
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center
| | - Andreas M Reimold
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center.,Dallas VA Medical Center, Dallas, TX, USA
| |
Collapse
|
26
|
Xu X, Yu B, Cai W, Huang Z. TCF1 deficiency ameliorates autoimmune lymphoproliferative syndrome (ALPS)-like phenotypes oflpr/lprmice. Scand J Immunol 2017; 85:406-416. [DOI: 10.1111/sji.12546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Affiliation(s)
- X. Xu
- Institute of Human Virology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
- Key Laboratory of Tropical Diseases Control; Ministry of Education in China; Sun Yat-sen University; Guangzhou China
| | - B. Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province; Third Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - W. Cai
- Department of Biochemistry; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
| | - Z. Huang
- Institute of Human Virology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
- Key Laboratory of Tropical Diseases Control; Ministry of Education in China; Sun Yat-sen University; Guangzhou China
- Department of Biochemistry; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou China
| |
Collapse
|
27
|
Liao X, Pirapakaran T, Luo XM. Chemokines and Chemokine Receptors in the Development of Lupus Nephritis. Mediators Inflamm 2016; 2016:6012715. [PMID: 27403037 PMCID: PMC4923605 DOI: 10.1155/2016/6012715] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/11/2016] [Accepted: 05/17/2016] [Indexed: 11/18/2022] Open
Abstract
Lupus nephritis (LN) is a major cause of morbidity and mortality in the patients with systemic lupus erythematosus (SLE), an autoimmune disease with damage to multiple organs. Leukocyte recruitment into the inflamed kidney is a critical step to promote LN progression, and the chemokine/chemokine receptor system is necessary for leukocyte recruitment. In this review, we summarize recent studies on the roles of chemokines and chemokine receptors in the development of LN and discuss the potential and hurdles of developing novel, chemokine-based drugs to treat LN.
Collapse
Affiliation(s)
- Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Tharshikha Pirapakaran
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|