1
|
A B cell receptor variant confers evolutionary protection against pathogens. Nat Immunol 2024; 25:1791-1792. [PMID: 39289559 DOI: 10.1038/s41590-024-01953-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
|
2
|
Sun W, Yang T, Sun F, Liu P, Gao J, Lan X, Xu W, Pang Y, Li T, Li C, Liang Q, Chen H, Liu X, Tan W, Zhu H, Wang F, Cheng F, Zhai W, Kim HN, Zhang J, Zhang L, Lu L, Xi Q, Deng G, Huang Y, Jin X, Chen X, Liu W. An IGHG1 variant exhibits polarized prevalence and confers enhanced IgG1 antibody responses against life-threatening organisms. Nat Immunol 2024; 25:1809-1819. [PMID: 39261722 DOI: 10.1038/s41590-024-01944-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024]
Abstract
Evolutionary pressures sculpt population genetics, whereas immune adaptation fortifies humans against life-threatening organisms. How the evolution of selective genetic variation in adaptive immune receptors orchestrates the adaptation of human populations to contextual perturbations remains elusive. Here, we show that the G396R coding variant within the human immunoglobulin G1 (IgG1) heavy chain presents a concentrated prevalence in Southeast Asian populations. We uncovered a 190-kb genomic linkage disequilibrium block peaked in close proximity to this variant, suggestive of potential Darwinian selection. This variant confers heightened immune resilience against various pathogens and viper toxins in mice. Mechanistic studies involving severe acute respiratory syndrome coronavirus 2 infection and vaccinated individuals reveal that this variant enhances pathogen-specific IgG1+ memory B cell activation and antibody production. This G396R variant may have arisen on a Neanderthal haplotype background. These findings underscore the importance of an IGHG1 variant in reinforcing IgG1 antibody responses against life-threatening organisms, unraveling the intricate interplay between human evolution and immune adaptation.
Collapse
Affiliation(s)
- Wenbo Sun
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Tingyu Yang
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Fengming Sun
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Chongqing Key Laboratory for Research of Infectious Diseases, Chongqing, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | | | - Ji Gao
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xianmei Lan
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, Fudan University, Shanghai, China
| | - Yuhong Pang
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Tong Li
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Cuifeng Li
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Qingtai Liang
- NexVac Research Center, Center for Infectious Disease Research, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haoze Chen
- NexVac Research Center, Center for Infectious Disease Research, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohang Liu
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Wenting Tan
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | | | - Fang Wang
- The Third People's Hospital of Shenzhen, National Clinical Research Center for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Fanjun Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiwei Zhai
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Han-Na Kim
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Biomedical Statistics Center Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Jingren Zhang
- NexVac Research Center, Center for Infectious Disease Research, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linqi Zhang
- NexVac Research Center, Center for Infectious Disease Research, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, Fudan University, Shanghai, China.
| | - Qiaoran Xi
- Key Laboratory of Protein Sciences (Ministry of Education), State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Guohong Deng
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing Key Laboratory for Research of Infectious Diseases, Chongqing, China.
| | - Yanyi Huang
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China.
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China.
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, China.
- School of Medicine, South China University of Technology, Guangzhou, China.
| | - Xiangjun Chen
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, China.
| | - Wanli Liu
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
3
|
Ma X, Lu Y, Xu S. Adaptive Evolution of Two Distinct Adaptive Haplotypes of Neanderthal Origin at the Immunoglobulin Heavy-chain Locus in East Asian and European Populations. Mol Biol Evol 2024; 41:msae147. [PMID: 39011558 DOI: 10.1093/molbev/msae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
Immunoglobulins (Igs) have a crucial role in humoral immunity. Two recent studies have reported a high-frequency Neanderthal-introgressed haplotype throughout Eurasia and a high-frequency Neanderthal-introgressed haplotype specific to southern East Asia at the immunoglobulin heavy-chain (IGH) gene locus on chromosome 14q32.33. Surprisingly, we found the previously reported high-frequency Neanderthal-introgressed haplotype does not exist throughout Eurasia. Instead, our study identified two distinct high-frequency haplotypes of putative Neanderthal origin in East Asia and Europe, although they shared introgressed alleles. Notably, the alleles of putative Neanderthal origin reduced the expression of IGHG1 and increased the expression of IGHG2 and IGHG3 in various tissues. These putatively introgressed alleles also affected the production of IgG1 upon antigen stimulation and increased the risk of systemic lupus erythematosus. Additionally, the greatest genetic differentiation across the whole genome between southern and northern East Asians was observed for the East Asian haplotype of putative Neanderthal origin. The frequency decreased from southern to northern East Asia and correlated positively with the genome-wide proportion of southern East Asian ancestry, indicating that this putative positive selection likely occurred in the common ancestor of southern East Asian populations before the admixture with northern East Asian populations.
Collapse
Affiliation(s)
- Xixian Ma
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Yan Lu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Sun W, Zhu C, Li Y, Wu X, Shi X, Liu W. B cell activation and autoantibody production in autoimmune diseases. Best Pract Res Clin Rheumatol 2024; 38:101936. [PMID: 38326197 DOI: 10.1016/j.berh.2024.101936] [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: 01/03/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
B cells are central players in the immune system, responsible for producing antibodies and modulating immune responses. This review explores the intricate relationship between aberrant B cell activation and the development of autoimmune diseases, emphasizing the essential role of B cells in these conditions. We also summarize B cell receptor signaling and Toll-like receptor signaling in B cell activation, as well as their association with autoimmune diseases, shedding light on the molecular mechanisms behind these associations. Additionally, we explore the clinical observations involving B cell activation and their significance in autoimmune disease management. Various clinical studies related to B cell-targeted therapies are also discussed, offering insights into potential avenues for improving treatment strategies. Overall, this review serves as a resource for researchers and clinicians in the field of immunology and autoimmune diseases, providing a general view of B cell signaling and its role in autoimmunity.
Collapse
Affiliation(s)
- Wenbo Sun
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, No. 1, Qinghua Yuan, New Biology Bldg, Haidian District, Beijing, 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China; The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei, 230032, China.
| | - Can Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, No. 1, Qinghua Yuan, New Biology Bldg, Haidian District, Beijing, 100084, China.
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, No. 1, Qinghua Yuan, New Biology Bldg, Haidian District, Beijing, 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Xinfeng Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 636, Guanlin Road, 471000, Luoyang, China.
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 636, Guanlin Road, 471000, Luoyang, China.
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, No. 1, Qinghua Yuan, New Biology Bldg, Haidian District, Beijing, 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China; The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Purcell RA, Aurelia LC, Esterbauer R, Allen LF, Bond KA, Williamson DA, Trevillyan JM, Trubiano JA, Juno JJ, Wheatley AK, Davenport MP, Nguyen THO, Kedzierska K, Kent SJ, Selva KJ, Chung AW. Immunoglobulin G genetic variation can confound assessment of antibody levels via altered binding to detection reagents. Clin Transl Immunology 2024; 13:e1494. [PMID: 38433763 PMCID: PMC10902689 DOI: 10.1002/cti2.1494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
Objectives Amino acid variations across more than 30 immunoglobulin (Ig) allotypes may introduce structural changes that influence recognition by anti-Ig detection reagents, consequently confounding interpretation of antibody responses, particularly in genetically diverse cohorts. Here, we assessed a panel of commercial monoclonal anti-IgG1 clones for capacity to universally recognise two dominant IgG1 haplotypes (G1m-1,3 and G1m1,17). Methods Four commercial monoclonal anti-human IgG1 clones were assessed via ELISAs and multiplex bead-based assays for their ability to bind G1m-1,3 and G1m1,17 IgG1 variants. Detection antibodies were validated against monoclonal IgG1 allotype standards and tested for capacity to recognise antigen-specific plasma IgG1 from G1m-1,3 and G1m1,17 homozygous and heterozygous SARS-CoV-2 BNT162b2 vaccinated (n = 28) and COVID-19 convalescent (n = 44) individuals. An Fc-specific pan-IgG detection antibody corroborated differences between hinge- and Fc-specific anti-IgG1 responses. Results Hinge-specific anti-IgG1 clone 4E3 preferentially bound G1m1,17 compared to G1m-1,3 IgG1. Consequently, SARS-CoV-2 Spike-specific IgG1 levels detected in G1m1,17/G1m1,17 BNT162b2 vaccinees appeared 9- to 17-fold higher than in G1m-1,3/G1m-1,3 vaccinees. Fc-specific IgG1 and pan-IgG detection antibodies equivalently bound G1m-1,3 and G1m1,17 IgG1 variants, and detected comparable Spike-specific IgG1 levels between haplotypes. IgG1 responses against other human coronaviruses and influenza were similarly poorly detected by 4E3 anti-IgG1 in G1m-1,3/G1m-1,3 subjects. Conclusion Anti-IgG1 clone 4E3 confounds assessment of antibody responses in clinical cohorts owing to bias towards detection of G1m1,17 IgG1 variants. Validation of anti-Ig clones should include evaluation of binding to relevant antibody variants, particularly as the role of immunogenetics upon humoral immunity is increasingly explored in diverse populations.
Collapse
Affiliation(s)
- Ruth A Purcell
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - L Carissa Aurelia
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Robyn Esterbauer
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Lilith F Allen
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Katherine A Bond
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Victorian Infectious Diseases Reference Laboratory (VIDRL)The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory (VIDRL)The Peter Doherty Institute for Infection and ImmunityMelbourneVICAustralia
- Walter and Eliza Hall Institute of Medical ResearchParkvilleVICAustralia
- Department of Infectious DiseasesThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Janine M Trevillyan
- Department of Infectious DiseasesThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Centre for Antibiotic Allergy and Research, Department of Infectious DiseasesAustin HealthHeidelbergVICAustralia
| | - Jason A Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious DiseasesAustin HealthHeidelbergVICAustralia
- Department of MedicineUniversity of MelbourneParkvilleVICAustralia
- Department of Infectious DiseasesPeter MacCallum Cancer CentreMelbourneVICAustralia
- National Centre for Infections in CancerPeter MacCallum Cancer CentreMelbourneVICAustralia
| | - Jennifer J Juno
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | | | - Thi HO Nguyen
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Katherine Kedzierska
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Kevin John Selva
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| | - Amy W Chung
- Department of Microbiology and ImmunologyThe Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneVICAustralia
| |
Collapse
|
6
|
Zhou X, Wang X, Xu J, Tang Q, Bergquist R, Shi L, Qin Z. High-throughput autoantibody profiling of different stages of Schistosomiasis japonica. Autoimmunity 2023; 56:2250102. [PMID: 37599561 DOI: 10.1080/08916934.2023.2250102] [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: 04/19/2023] [Revised: 06/13/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
Infection by the Schistosoma japonicum can result in acute, chronic and late-stage manifestations. The latter often presents with severe organ failures and premature death. Importantly, infection can also produce autoimmune phenomena reflected by the development of autoantibodies. We wished to explore and profile the presence of autoantibodies in sera of patients with different stages of S. japonicum infection with the added aim of providing a reference assisting diagnosis. Blood samples from 55 patients with chronic and 20 patients with late-stage schistosomiasis japonica together, with a control group of 50 healthy people were randomly investigated against a microarray of 121 different autoantigens. In addition, the frequency of antibodies against Schistosoma egg antigen (SEA) was examined. In the sera from patients with chronic schistosomiasis japonica, 14 different highly expressed autoantibodies were detected, while patients with late-stage schistosomiasis were found to express as many as 43 autoantibody specificities together with a significantly higher frequency of antibodies against SEA compared to the control group. The findings presented suggest that autoantibody-based classification of schistosomiasis japonica represents a promising approach for the elucidation of subtypes of the disease. This approach may reflect differential disease mechanisms, which could ultimately lead to better treatment.
Collapse
Affiliation(s)
- Xiaorong Zhou
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, China
| | - Xi Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| | - Jing Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| | - Qi Tang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| | - Robert Bergquist
- UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases (TDR), Ingerod, Brastad, Sweden
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Zhiqiang Qin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| |
Collapse
|
7
|
Luo H, Zhang P, Zhang W, Zheng Y, Hao D, Shi Y, Niu Y, Song T, Li Y, Zhao S, Chen H, Xu T, He S. Recent positive selection signatures reveal phenotypic evolution in the Han Chinese population. Sci Bull (Beijing) 2023; 68:2391-2404. [PMID: 37661541 DOI: 10.1016/j.scib.2023.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/08/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023]
Abstract
Characterizing natural selection signatures and relationships with phenotype spectra is important for understanding human evolution and both biological and pathological mechanisms. Here, we identified 24 genetic loci under recent selection by analyzing rare singletons in 3946 high-depth whole-genome sequencing data of Han Chinese. The loci include immune-related gene regions (MHC cluster, IGH cluster, STING1, and PSG), alcohol metabolism-related gene regions (ADH1B, ALDH2, and ALDH3B2), and the olfactory perception gene OR4C16, in which the MHC cluster, ADH1B, and ALDH2 were also identified by TOPMed and WestLake Biobank. Among the signals, the IGH cluster is particularly interesting, in which the favored allele of variant 14_105737776_C_T (rs117518546, IgG1-G396R) promotes immune response, but also increases the risk of an autoimmune disease systemic lupus erythematosus (SLE). It is also surprising that our newly discovered ALDH3B2 evolved in the opposite direction to ALDH2 for alcohol metabolism. Besides monogenic traits, we found that multiple complex traits experienced polygenic adaptation. Particularly, multi-methods consistently revealed that lower blood pressure was favored in natural selection. Finally, we built a database named RePoS (recent positive selection, http://bigdata.ibp.ac.cn/RePoS/) to integrate and display multi-population selection signals. Our study extended our understanding of natural evolution and phenotype adaptation in Han Chinese as well as other populations.
Collapse
Affiliation(s)
- Huaxia Luo
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Peng Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wanyu Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Zheng
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Hao
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yirong Shi
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiwei Niu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingrui Song
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanyan Li
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shilei Zhao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China
| | - Hua Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, China.
| | - Shunmin He
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
8
|
Jia YJ, Chen HL. Risk factors of progression from discoid lupus to severe systemic lupus erythematosus: More evidence will be needed. J Am Acad Dermatol 2023; 89:e163-e164. [PMID: 37269925 DOI: 10.1016/j.jaad.2023.04.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 06/05/2023]
Affiliation(s)
- Yi-Jie Jia
- Medical School of Nantong University, Nantong, Jiangsu, PR China
| | - Hong-Lin Chen
- School of Public Health, Nantong University, Nantong, Jiangsu, PR China.
| |
Collapse
|
9
|
Zhang Y, Niazi B, Auda A, Chacko AA, Jarri A, Mohamed A, Ali S, Zhu H, Sirajuddin S. A Novel Presentation of Autoimmune Hepatitis with IgG1 Elevation. Case Rep Gastroenterol 2023; 17:281-286. [PMID: 37928974 PMCID: PMC10624941 DOI: 10.1159/000530517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/27/2023] [Indexed: 11/07/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a common and debilitating pathology that has acute, subacute, and chronic presentation, requiring prompt diagnosis and early intervention. Several serologic markers are found to be associated with the pathogenesis and progression of autoimmune hepatitis, most notably antinuclear antibodies and anti-smooth muscle antibodies [Front Immunol. 2018;9:609]. In addition, AIH is also characterized by the elevation of gamma globulin levels, mainly immunoglobulin G (IgG) [World J Gastroenterol. 2015;21(1):60-83]. Although the literature has well established the presence of increased IgG levels in AIH, few studies have evaluated the subtypes of IgG and their differential levels associated with AIH. Here, we present a rare case of AIH that lacks the common serologic markers but instead reveals an elevation in IgG1 level. Our patient was subsequently placed on corticosteroids, and her symptoms quickly resolved. We intend to introduce this case to the medical community in the hope of aiding in the proper diagnosis and timely intervention of subsequent cases with similar presentations.
Collapse
Affiliation(s)
- Yujiao Zhang
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Bilal Niazi
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Auda Auda
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Angel Ann Chacko
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Amer Jarri
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Abdifatah Mohamed
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Saad Ali
- Department of Gastroenterology, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Hongfa Zhu
- Department of Pathology, HMH-Palisades Medical Center, North Bergen, NJ, USA
| | - Syed Sirajuddin
- Department of Internal Medicine, HMH-Palisades Medical Center, North Bergen, NJ, USA
| |
Collapse
|
10
|
Xu L, Zhao J, Sun Q, Xu X, Wang L, Liu T, Wu Y, Zhu J, Geng L, Deng Y, Awgulewitsch A, Kamen DL, Oates JC, Raj P, Wakeland EK, Scofield RH, Guthridge JM, James JA, Hahn BH, McCurdy DK, Wang F, Zhang M, Tan W, Gilkeson GS, Tsao BP. Loss-of-function variants in SAT1 cause X-linked childhood-onset systemic lupus erythematosus. Ann Rheum Dis 2022; 81:1712-1721. [PMID: 35977808 PMCID: PMC10394691 DOI: 10.1136/ard-2022-222795] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/28/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Families that contain multiple siblings affected with childhood onset of systemic lupus erythematosus (SLE) likely have strong genetic predispositions. We performed whole exome sequencing (WES) to identify familial rare risk variants and to assess their effects in lupus. METHODS Sanger sequencing validated the two ultra-rare, predicted pathogenic risk variants discovered by WES and identified additional variants in 562 additional patients with SLE. Effects of a splice site variant and a frameshift variant were assessed using a Minigene assay and CRISPR/Cas9-mediated knock-in (KI) mice, respectively. RESULTS The two familial ultra-rare, predicted loss-of-function (LOF) SAT1 variants exhibited X-linked recessive Mendelian inheritance in two unrelated African-American families. Each LOF variant was transmitted from the heterozygous unaffected mother to her two sons with childhood-onset SLE. The p.Asp40Tyr variant affected a splice donor site causing deleterious transcripts. The young hemizygous male and homozygous female Sat1 p.Glu92Leufs*6 KI mice spontaneously developed splenomegaly, enlarged glomeruli with leucocyte infiltration, proteinuria and elevated expression of type I interferon-inducible genes. SAT1 is highly expressed in neutrophils and encodes spermidine/spermine-N1-acetyltransferase 1 (SSAT1), a rate-limiting enzyme in polyamine catabolism. Young male KI mice exhibited neutrophil defects and decreased proportions of Foxp3 +CD4+ T-cell subsets. Circulating neutrophil counts and proportions of Foxp3 +CD4+ T cells correlated with decreased plasma levels of spermine in treatment-naive, incipient SLE patients. CONCLUSIONS We identified two novel SAT1 LOF variants, showed the ability of the frameshift variant to confer murine lupus, highlighted the pathogenic role of dysregulated polyamine catabolism and identified SAT1 LOF variants as new monogenic causes for SLE.
Collapse
Affiliation(s)
- Lingxiao Xu
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jian Zhao
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Qing Sun
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xue Xu
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lei Wang
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ting Liu
- Department of Rheumatology and Immunology, Wuxi People's Hospital, Wuxi, Jiangsu, People's Republic of China
| | - Yunjuan Wu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jingfeng Zhu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Linyu Geng
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yun Deng
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexander Awgulewitsch
- Department of Regenerative Medicine and Cell Biology, Transgenic and Gene Function Core, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Diane L Kamen
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jim C Oates
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - R Hal Scofield
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- US Department of Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Joel M Guthridge
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Judith A James
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Bevra H Hahn
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Deborah K McCurdy
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA
| | - Fang Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Miaojia Zhang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Gary S Gilkeson
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Betty P Tsao
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
11
|
Contribution of rare mutational outcomes to broadly neutralizing antibodies. Acta Biochim Biophys Sin (Shanghai) 2022; 54:820-827. [PMID: 35713319 PMCID: PMC9828561 DOI: 10.3724/abbs.2022065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Antibodies are important immune molecules that are elicited by B cells to protect our bodies during viral infections or vaccinations. In humans, the antibody repertoire is diversified by programmed DNA lesion processes to ensure specific and high affinity binding to various antigens. Broadly neutralizing antibodies (bnAbs) are antibodies that have strong neutralizing activities against different variants of a virus. bnAbs such as anti-HIV bnAbs often have special characteristics including insertions and deletions, long complementarity determining region 3 (CDR3), and high frequencies of mutations, often at improbable sites of the variable regions. These unique features are rare mutational outcomes that are acquired during antibody diversification processes. In this review, we will discuss possible mechanisms that generate these rare antibody mutational outcomes. The understanding of the mechanisms that generate these rare mutational outcomes during antibody diversification will have implications in vaccine design strategies to elicit bnAbs.
Collapse
|
12
|
Abstract
Tumor-infiltrating B cells exert antitumor effects by producing antibodies against tumor-associated antigens. Conversely, B cells may promote tumors through the production of factors that dampen antitumor immunity. In this issue of the JCI, Bing Yang, Zhen Zhang, et al. investigated the roles of B cell receptor (BCR) signaling in antitumor immunity, focusing on the role of an Asia-specific variant of human immunoglobulin G1 (IgG1) containing a Gly396 to Arg396 substitution (hIgG1-G396R) in colorectal cancer (CRC). Epidemiological analysis revealed an association between hIgG1-G396R and progression-free survival in CRC. Human samples and mouse models of CRC showed plasma cells, as opposed to B cells, infiltrating the tumor microenvironment. Notably, patients with the hIgG1-G396R variant had increased CD8+ T cells, dendritic cells, and tertiary lymphoid structure density. These findings indicate that the hIgG1-G396R variant represses tumorigenesis by enhancing B cell responses, and suggest that modulating BCR signaling could improve the efficacy of immunotherapy in cancer.
Collapse
|
13
|
Yang B, Zhang Z, Chen X, Wang XY, Qin S, Du L, Yang C, Zhu L, Sun W, Zhu Y, Zheng Q, Zhao S, Wang Q, Zhao L, Lin Y, Huang J, Wu F, Lu L, Wang F, Zheng W, Zhou XH, Zhao X, Wang Z, Sun X, Ye Y, Wang S, Li Z, Qi H, Zhang Z, Kuang DM, Zhang L, Shen Z, Liu W. An Asian-specific variant in human IgG1 represses colorectal tumorigenesis by shaping the tumor microenvironment. J Clin Invest 2022; 132:153454. [PMID: 35133976 PMCID: PMC8920342 DOI: 10.1172/jci153454] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Emerging studies have focused on ways to treat cancers by modulating T cell activation. However, whether B cell receptor signaling in the tumor microenvironment (TME) can be harnessed for immunotherapy is unclear. Here, we report that an Asia-specific variant of human IgG1 containing a Gly396 to Arg396 substitution (hIgG1-G396R) conferred improved survival of patients with colorectal cancer (CRC). Mice with knockin of the murine functional homolog mIgG2c-G400R recapitulated the alleviated tumorigenesis and progression in murine colon carcinoma models. Immune profiling of the TME revealed broad mobilizations of IgG1+ plasma cells, CD8+ T cells, CD103+ DCs, and active tertiary lymphoid structure formation, suggesting an effective antitumor microenvironment in hIgG1-G396R CRC patients. Mechanistically, this variant potentiated tumor-associated antigen–specific (TAA-specific) plasma cell differentiation and thus antibody production. These elevated TAA-specific IgG2c antibodies in turn efficiently boosted the antibody-dependent tumor cell phagocytosis and TAA presentation to effector CD8+ T cells. Notably, adoptive transfer of TAA-specific class-switched memory B cells harboring this variant exhibited therapeutic efficacy in murine tumor models, indicating their clinical potential. All these results prompted a prospective investigation of hIgG1-G396R in patients with CRC as a biomarker for clinical prognosis and demonstrated that manipulating the functionality of IgG1+ memory B cells in tumors could improve immunotherapy outcomes.
Collapse
Affiliation(s)
- Bing Yang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhen Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Xiangjun Chen
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xu-Yan Wang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shishang Qin
- BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Liaoqi Du
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Changjiang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Liyu Zhu
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Wenbo Sun
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yongjie Zhu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Qinwen Zheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shidong Zhao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Quan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Long Zhao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Yilin Lin
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Jinghe Huang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Fan Wu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lu Lu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Fei Wang
- Center for Natural Products Research, Chinese Academy of Sciences,, Chengdu, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Xiao-Hua Zhou
- School of Public Health, Peking University, Beijing, China
| | - Xiaozhen Zhao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Ziye Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, People's Hospital Peking University, Beijing, China
| | - Hai Qi
- Department of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Zemin Zhang
- BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Dong-Ming Kuang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhang
- BIOPIC and School of Life Sciences, Peking University, Beijing, China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Wanli Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
14
|
Nimesh S, Ahmad MI, Dhama S, Kumar P, Akram M, Hasaroeih NEN. Systemic Lupus Erythematosus Disease: An Overview of the Clinical Approach to Pathogenesis, Diagnosis, and Treatment. BORNEO JOURNAL OF PHARMACY 2021. [DOI: 10.33084/bjop.v4i2.1950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The systemic lupus erythematosus (SLE), commonly known as Lupus, is a rare and complex multisystem autoimmune disease where one’s immune system is overactive, and the body attacks its organ systems. SLE is a historically old disease described already in antiquity; it is an example of a chronic disease with physical, psychological, financial, and social implications for individuals diagnosed. It has inspired medical and basic biological scientists that focus on molecular biology, basic immunology, immunopathology, clinical science, genetics, and epidemiology. The syndrome is real in its existence-although hidden behind obstacles, cumbersome for patients and clinicians, and rebellious for scientists. There is currently no cure for SLE. The goal of treatment is to ease symptoms. This article will review information on the general approach to SLE therapy, focusing on currently approved therapies and novel approaches that might be used in the future.
Collapse
|
15
|
Zhao X, Zhang J, Liang Y, Li J, Ding S, Wang Y, Chen Y, Liu J. Advances in Drug Therapy for Systemic Lupus Erythematosus. Curr Med Chem 2021; 28:1251-1268. [PMID: 32586244 DOI: 10.2174/0929867327666200625150408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/27/2020] [Accepted: 06/04/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by a local or systemic inflammatory response. At present, the increasing research results show that the pathogenesis of the disease is complex, and the methods of clinical treatment also show diversity. This review analyzes and summarizes the existing mechanism research and drug treatment methods in order to provide a reference value for further drug research and development. METHOD We carried out a thorough literature search using databases. According to the main purpose of the article, irrelevant articles were excluded after further examination and directly relevant articles were included. Finally, the information related to the article was summarized. RESULT In this article, seventy-four articles are included. According to related articles, there are mainly four kinds of drugs, namely antimalarial drugs, glucocorticoids, immunosuppressive agents and biological agents. About fifty-five articles summarized the drugs for the treatment of systemic lupus erythematosus. The rest of the articles were related to the research progress of the mechanism of systemic lupus erythematosus. CONCLUSION This article describes the pathogenesis of systemic lupus erythematosus, and summarizes the traditional and new therapeutic drugs, which is not only beneficial to the treatment of lupus erythematosus patients, but also plays a vital reference significance for the future development of new systemic lupus erythematosus drugs.
Collapse
Affiliation(s)
- Xinghua Zhao
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Jiaojiao Zhang
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Yutong Liang
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Jie Li
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Shi Ding
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Yang Wang
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Ye Chen
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| | - Ju Liu
- Department of Medicinal, College of Pharmacy, Liaoning University, Shenyang, China
| |
Collapse
|
16
|
Zhong X, Chen C, Sun X, Wang J, Li R, Chang Y, Fan P, Wang Y, Wu Y, Peng L, Lu Z, Qiu W. Whole-exome sequencing reveals the major genetic factors contributing to neuromyelitis optica spectrum disorder in Chinese patients with aquaporin 4-IgG seropositivity. Eur J Neurol 2021; 28:2294-2304. [PMID: 33559384 DOI: 10.1111/ene.14771] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/15/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune disease. Although genetic factors are involved in its pathogenesis, limited evidence is available in this area. The aim of the present study was to identify the major genetic factors contributing to NMOSD in Chinese patients with aquaporin 4 (AQP4)-IgG seropositivity. METHODS Whole-exome sequencing (WES) was performed on 228 Chinese NMOSD patients seropositive for AQP4-IgG and 1400 healthy controls in Guangzhou, South China. Human leukocyte antigen (HLA) sequencing was also utilized. Genotype model and haplotype, gene burden, and enrichment analyses were conducted. RESULTS A significant region of the HLA composition is on chromosome 6, and great variation was observed in DQB1, DQA2 and DQA1. HLA sequencing confirmed that the most significant allele was HLA-DQB1*05:02 (p < 0.01, odds ratio [OR] 3.73). The genotype model analysis revealed that HLA-DQB1*05:02 was significantly associated with NMOSD in the additive effect model and dominant effect model (p < 0.05). The proportion of haplotype "HLA-DQB1*05:02-DRB1*15:01" was significantly greater in the NMOSD patients than the controls, at 8.42% and 1.23%, respectively (p < 0.001, OR 7.39). The gene burden analysis demonstrated that loss-of-function mutations in NOP16 were more common in the NMOSD patients (11.84%) than the controls (5.71%; p < 0.001, OR 2.22). The IgG1-G390R variant was significantly more common in NMOSD, and the rate of the T allele was 0.605 in patients and 0.345 in the controls (p < 0.01, OR 2.92). The enrichment analysis indicated that most of the genetic factors were mainly correlated with nervous and immune processes. CONCLUSIONS Human leukocyte antigen is highly correlated with NMOSD. NOP16 and IgG1-G390R play important roles in disease susceptibility.
Collapse
Affiliation(s)
- Xiaonan Zhong
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaobo Sun
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingqi Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rui Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanyu Chang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ping Fan
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunting Wu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lisheng Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Wang YF, Zhang Y, Lin Z, Zhang H, Wang TY, Cao Y, Morris DL, Sheng Y, Yin X, Zhong SL, Gu X, Lei Y, He J, Wu Q, Shen JJ, Yang J, Lam TH, Lin JH, Mai ZM, Guo M, Tang Y, Chen Y, Song Q, Ban B, Mok CC, Cui Y, Lu L, Shen N, Sham PC, Lau CS, Smith DK, Vyse TJ, Zhang X, Lau YL, Yang W. Identification of 38 novel loci for systemic lupus erythematosus and genetic heterogeneity between ancestral groups. Nat Commun 2021; 12:772. [PMID: 33536424 PMCID: PMC7858632 DOI: 10.1038/s41467-021-21049-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Systemic lupus erythematosus (SLE), a worldwide autoimmune disease with high heritability, shows differences in prevalence, severity and age of onset among different ancestral groups. Previous genetic studies have focused more on European populations, which appear to be the least affected. Consequently, the genetic variations that underlie the commonalities, differences and treatment options in SLE among ancestral groups have not been well elucidated. To address this, we undertake a genome-wide association study, increasing the sample size of Chinese populations to the level of existing European studies. Thirty-eight novel SLE-associated loci and incomplete sharing of genetic architecture are identified. In addition to the human leukocyte antigen (HLA) region, nine disease loci show clear ancestral differences and implicate antibody production as a potential mechanism for differences in disease manifestation. Polygenic risk scores perform significantly better when trained on ancestry-matched data sets. These analyses help to reveal the genetic basis for disparities in SLE among ancestral groups.
Collapse
Affiliation(s)
- Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhiming Lin
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huoru Zhang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Ting-You Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
- The Hormel Institute, University of Minnesota, Austin, USA
| | - Yujie Cao
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - David L Morris
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Yujun Sheng
- Department of Dermatology, No.1 Hospital, Anhui Medical University, Hefei, China
| | - Xianyong Yin
- Department of Dermatology, No.1 Hospital, Anhui Medical University, Hefei, China
| | - Shi-Long Zhong
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Yao Lei
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qi Wu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiangshan Jane Shen
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Jing Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Tai-Hing Lam
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Jia-Huang Lin
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Zhi-Ming Mai
- School of Public Health, The University of Hong Kong, Hong Kong, China
- Radiation Epidemiology Branch, Division of Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Mengbiao Guo
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhui Chen
- Department of Pediatrics, Union Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Qin Song
- Department of Rheumatology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Bo Ban
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Chaoyang, China
| | - Liangjing Lu
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | - Chak Sing Lau
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - David K Smith
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Timothy J Vyse
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | - Xuejun Zhang
- Department of Dermatology, No.1 Hospital, Anhui Medical University, Hefei, China
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China.
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, China.
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Warrender AK, Kelton W. Beyond Allotypes: The Influence of Allelic Diversity in Antibody Constant Domains. Front Immunol 2020; 11:2016. [PMID: 32973808 PMCID: PMC7461860 DOI: 10.3389/fimmu.2020.02016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/24/2020] [Indexed: 01/25/2023] Open
Abstract
Polymorphic diversity in antibody constant domains has long been defined by allotypic motifs that cross react with the sera of other individuals. Improvements in sequencing technologies have led to the discovery of a large number of new allelic sequences that underlie this diversity. Many of the point mutations lie outside traditional allotypic motifs suggesting they do not elicit immunogenic responses. As antibodies play an important role in immune defense and biotechnology, understanding how this newly resolved diversity influences the function of antibodies is important. This review investigates the current known diversity of antibody alleles at a protein level for each antibody isotype as well as the kappa and lambda light chains. We focus on evidence emerging for how these mutations perturb antibody interactions with antigens and Fc receptors that are critical for function, as well as the influence this might have on the use of antibodies as therapeutics and reagents.
Collapse
Affiliation(s)
| | - William Kelton
- Te Huataki Waiora School of Health, The University of Waikato, Hamilton, New Zealand
| |
Collapse
|
19
|
Tsokos GC. Autoimmunity and organ damage in systemic lupus erythematosus. Nat Immunol 2020; 21:605-614. [PMID: 32367037 PMCID: PMC8135909 DOI: 10.1038/s41590-020-0677-6] [Citation(s) in RCA: 280] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/31/2020] [Indexed: 01/07/2023]
Abstract
Impressive progress has been made over the last several years toward understanding how almost every aspect of the immune system contributes to the expression of systemic autoimmunity. In parallel, studies have shed light on the mechanisms that contribute to organ inflammation and damage. New approaches that address the complicated interaction between genetic variants, epigenetic processes, sex and the environment promise to enlighten the multitude of pathways that lead to what is clinically defined as systemic lupus erythematosus. It is expected that each patient owns a unique 'interactome', which will dictate specific treatment.
Collapse
Affiliation(s)
- George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
20
|
Li Y, Jia R, Liu Y, Tang S, Ma X, Shi L, Zhao J, Hu F, Li Z. Antibodies against carbamylated vimentin exist in systemic lupus erythematosus and correlate with disease activity. Lupus 2020; 29:239-247. [PMID: 31930936 DOI: 10.1177/0961203319897127] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Antibodies against carbamylated protein (anti-CarP) were found to be a promising marker to evaluate joint damage and disease activity in patients with rheumatoid arthritis (RA). However, whether anti-CarP antibodies were present in systemic lupus erythematosus (SLE) remained ambiguous. We have therefore undertaken this study to assess the levels of serum anti-CarP antibodies and to evaluate their clinical value in SLE. METHODS Serum levels of antibodies against carbamylated-vimentin (anti-Carp) were measured by enzyme immunosorbent assay in 100 patients with SLE, 76 with RA, 17 with primary Sjögren syndrome (pSS), and 68 healthy controls. Data analyses between anti-Carp antibodies and other laboratory measures were performed using SPSS 24 software for Windows. RESULTS The levels of serum anti-CarP antibodies in patients with SLE were significantly higher than those in healthy controls. In addition, anti-CarP antibodies were present in SLE patients lacking the disease-specific antibodies, including anti-Smith-negative patients (24.4%, 21/86), anti-dsDNA-negative patients (29.3%, 12/41), anti-nucleosome-negative patients (21.4%, 9/42), and antiribosomal P protein antibody-negative patients (23.7%, 18/76). There were significant differences between the anti-CarP-positive and anti-CarP-negative SLE patients in clinical and laboratory features, such as age, erythrocyte sedimentation rate (ESR), C-reactive protein, rheumatoid factor, third-generation cyclic citrullinated peptide (CCP3), anticardiolipin, D-dipolymer, complement 3, immunoglobulin G (IgG), red blood cell count (RBC) and hemoglobin. After adjusting for age and disease duration, the high levels of anti-CarP antibodies were still correlated with low RBC, hemoglobin and high ESR, IgG and CCP3. Active SLE patients demonstrated higher anti-CarP IgG than inactive patients. Moreover, the levels of anti-CarP were significantly higher in SLE patients with arthralgia and/or arthritis than in those without joint involvement. CONCLUSIONS Anti-CarP antibodies were present in SLE patients and associated with the disease severity. These might provide a potential supplement to other specific autoantibodies for diagnosis of SLE and serve as a promising marker for measuring joint damage in the disease.
Collapse
Affiliation(s)
- Y Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - R Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Y Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - S Tang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - X Ma
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,Department of Rheumatology and Immunology, Handan First Hospital, Hebei Province, China
| | - L Shi
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,Department of Rheumatology and Immunology, Peking University International Hospital, Beijing, China
| | - J Zhao
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - F Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Z Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
21
|
Feng Y, Wang Y, Zhang S, Haneef K, Liu W. Structural and immunogenomic insights into B-cell receptor activation. J Genet Genomics 2020; 47:27-35. [PMID: 32111437 DOI: 10.1016/j.jgg.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023]
Abstract
B cells express B-cell receptors (BCRs) which recognize antigen to trigger signaling cascades for B-cell activation and subsequent antibody production. BCR activation has a crucial influence on B-cell fate. How BCR is activated upon encountering antigen remains to be solved, although tremendous progresses have been achieved in the past few years. Here, we summarize the models that have been proposed to explain BCR activation, including the cross-linking model, the conformation-induced oligomerization model, the dissociation activation model, and the conformational change model. Especially, we elucidate the partially resolved structures of antibodies and/or BCRs by far and discusse how these current structural and further immunogenomic messages and more importantly the future studies may shed light on the explanation of BCR activation and the relevant diseases in the case of dysregulation.
Collapse
Affiliation(s)
- Yangyang Feng
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Shaocun Zhang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Kabeer Haneef
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
22
|
Abstract
Autoimmune diseases, such as rheumatoid arthritis, systematic lupus erythematosus and Sjögren's syndrome, are a group of diseases characterized by the activation of immune cells and excessive production of autoantibodies. Although the pathogenesis of these diseases is still not completely understood, studies have shown that multiple factors including genetics, environment and immune responses play important roles in the development and progression of the diseases. In China, there are great achievements in the mechanisms of autoimmune diseases during the last decades. These studies provide new insight to understand the diseases and also shed light on the development of novel therapy.
Collapse
Affiliation(s)
- Ru Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Xing Sun
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xu Liu
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yue Yang
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Zhanguo Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
23
|
Dong G, Yang Y, Li X, Yao X, Zhu Y, Zhang H, Wang H, Ma Q, Zhang J, Shi H, Ning Z, Yan F, Zhai W, Dai J, Li Z, Li C, Ming J, Xue Q, Meng X, Si C, Xiong H. Granulocytic myeloid-derived suppressor cells contribute to IFN-I signaling activation of B cells and disease progression through the lncRNA NEAT1-BAFF axis in systemic lupus erythematosus. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165554. [PMID: 31513833 DOI: 10.1016/j.bbadis.2019.165554] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/16/2019] [Accepted: 09/07/2019] [Indexed: 12/20/2022]
Abstract
Activation of interferon (IFN)-I signaling in B cells contributes to the pathogenesis of systemic lupus erythematosus (SLE). Recent studies have shown that myeloid-derived suppressor cells (MDSCs) significantly expand in SLE patients and lupus-prone MRL/lpr mice and contribute to the pathogenesis of SLE. However, the role of SLE-derived MDSCs in regulating IFN-I signaling activation of B cells remains unknown. Here, we demonstrate that expansions of MDSCs, including granulocyte (G)-MDSCs and monocytic (M)-MDSCs, during the progression of SLE were correlated with the IFN-I signature of B cells. Interestingly, G-MDSCs from MRL/lpr mice, but not M-MDSCs, could significantly promote IFN-I signaling activation of B cells and contribute to the pathogenesis of SLE. Mechanistically, we identified that the long non-coding RNA NEAT1 was over-expressed in G-MDSCs from MRL/lpr mice and could induce the promotion of G-MDSCs on IFN-I signaling activation of B cells through B cell-activating factor (BAFF) secretion. Importantly, NEAT1 deficiency significantly attenuated the lupus symptoms in pristane-induced lupus mice. In addition, there was a positive correlation between NEAT1 and BAFF with the IFN signature in SLE patients. In conclusion, G-MDSCs may contribute to the IFN signature in SLE B cells through the NEAT1-BAFF axis, highlighting G-MDSCs as a potential therapeutic target to treat SLE.
Collapse
Affiliation(s)
- Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yonghong Yang
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong 272067, China
| | - Xuehui Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Xiaoying Yao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yuzhen Zhu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Haiyan Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Qun Ma
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Hui Shi
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Weiwei Zhai
- Department of Clinical Laboratory, Jining NO.1 People's Hospital, Jining, Shandong 272067, China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhihua Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Jiankuo Ming
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Qingjie Xue
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Xiangzhi Meng
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Chuanping Si
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China.
| | - Huabao Xiong
- Department of Medicine, Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA..
| |
Collapse
|
24
|
Wang C, Ming B, Wu X, Wu T, Cai S, Hu P, Tang J, Tan Z, Liu C, Zhong J, Zheng F, Dong L. Sphingomyelin synthase 1 enhances BCR signaling to promote lupus-like autoimmune response. EBioMedicine 2019; 45:578-587. [PMID: 31262710 PMCID: PMC6642282 DOI: 10.1016/j.ebiom.2019.06.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Sphingomyelin synthase 1 (SMS1) has been reported to participate in hepatitis and atherosclerosis. However, its role in autoimmune response is not clear. This study investigates the possible involvement of SMS1 in B-cell activation and lupus-like autoimmunity. METHODS SMS1 knockout lupus-like animal model and SLE patient samples were utilized. B-cell activation and associated signal transduction were detected by flow cytometry, confocal analysis and western blotting. The SMS1 expression in B cells was measured by real-time qPCR. FINDINGS SMS1 deficiency suppressed B-cell activation in culture, which was restored by exogenous SM supplementation. The BCR-mediated early signal transduction including the colocalization of BCR with F-actin or pY/pBtk, and the phosphorylation of intracellular Fyn and Syk were impaired in SMS1 knockout B cells. Furthermore, SMS1 knockout mice showed reduced production and deposition of autoantibodies, accompanied by less severe kidney pathological changes after pristane induction. SMS1 deficiency also displayed lower autoantibody titers and 24 h urine protein excretion in bm12-induced lupus, which were associated with reduced B-cell activation. Adoptively transferred wide-type B cells partially recovered B-cell activation and autoantibody production in SMS1 deficient bm12-induced lupus mice. Moreover, the SMS1 mRNA level in B cells of SLE patients was increased and positively correlated with the serum anti-dsDNA level, IgG and globulin titers. INTERPRETATION These data suggest that SMS1 is involved in lupus-like autoimmunity via regulating BCR signal transduction and B cell activation. (Word count for the abstract: 230).
Collapse
Affiliation(s)
- Chenqiong Wang
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Bingxia Ming
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Xuefen Wu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Tong Wu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Shaozhe Cai
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Peng Hu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Jungen Tang
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Jixin Zhong
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH, United States
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China.
| |
Collapse
|
25
|
Refae S, Gal J, Brest P, Milano G. Germinal immunogenetics as a predictive factor for immunotherapy. Crit Rev Oncol Hematol 2019; 141:146-152. [PMID: 31301542 DOI: 10.1016/j.critrevonc.2019.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 01/17/2023] Open
Abstract
Clinical response to checkpoint inhibitors-based (CPIs) therapies can vary among tumor types and between patients. This led to a significant amount of pre-clinical and clinical research into biomarker identification. Biomarkers have been found to cover both the tumor itself and the tumor microenvironment. Entering host-related parameters into the equation should provide a valuable strategy for identifying not only factors predictive of treatment efficacy but also of treatment-related toxicity. It is clear that germline variants can offer efficient and easily-assessable indicators (blood DNA) to enlarge the spectrum of predictive markers for CPI-based treatment. A major issue concerns the real functional significance of the reported single-nucleotide polymorphisms (SNPs) linked to CPI-treatment outcome. Powered calculations should lead to an optimal trade-off between sample size and allele frequency. New molecular technologies and new analytical methods should provide opportunities to bridge the knowledge gap between SNP-CPI treatment associations and the functional impact of these SNPs.
Collapse
Affiliation(s)
- Sadal Refae
- Centre Antoine Lacassagne, Medical Oncology Department, University Côte d'Azur, Nice, F-06189, France
| | - Jocelyn Gal
- Centre Antoine Lacassagne, Epidemiology and Biostatistics Department, University Côte d'Azur, Nice, F-06189, France
| | - Patrick Brest
- Centre Antoine Lacassagne, Cnrs, Inserm, Ircan, FHU-Oncoage, University Côte d'Azur, Nice, F-06189, France
| | - Gerard Milano
- Centre Antoine Lacassagne, Oncopharmacology Unit, University Côte d'Azur, Nice, F-06189, France.
| |
Collapse
|
26
|
Cyster JG, Allen CDC. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell 2019; 177:524-540. [PMID: 31002794 PMCID: PMC6538279 DOI: 10.1016/j.cell.2019.03.016] [Citation(s) in RCA: 528] [Impact Index Per Article: 105.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 12/21/2022]
Abstract
B cells and the antibodies they produce have a deeply penetrating influence on human physiology. Here, we review current understanding of how B cell responses are initiated; the different paths to generate short- and long-lived plasma cells, germinal center cells, and memory cells; and how each path impacts antibody diversity, selectivity, and affinity. We discuss how basic research is informing efforts to generate vaccines that induce broadly neutralizing antibodies against viral pathogens, revealing the special features associated with allergen-reactive IgE responses and uncovering the antibody-independent mechanisms by which B cells contribute to health and disease.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Christopher D C Allen
- Cardiovascular Research Institute, Department of Anatomy, and Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
27
|
Tsokos GC. A new checkpoint in lupus. J Allergy Clin Immunol 2019; 143:1351-1352. [PMID: 30639348 DOI: 10.1016/j.jaci.2018.12.996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/18/2018] [Accepted: 12/26/2018] [Indexed: 11/19/2022]
Affiliation(s)
- George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass.
| |
Collapse
|
28
|
IgG1 variant promotes autoimmunity. Nat Rev Rheumatol 2018; 14:683. [PMID: 30425328 DOI: 10.1038/s41584-018-0127-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|