Peripheral immune landscape and natural killer-like B cells in human Vogt-Koyanagi-Harada disease

He Li , Lei Zhu , Xiuxing Liu , Lihui Xie , Rong Wang , Zhaohuai Li , Zhaohao Huang , Shizhao Yang , Binyao Chen , Jinguo Ye , Yingfeng Zheng , Wenru Su

Life Medicine ›› 2022, Vol. 1 ›› Issue (3) : 387 -400.

PDF (3207KB)
Life Medicine ›› 2022, Vol. 1 ›› Issue (3) : 387 -400. DOI: 10.1093/lifemedi/lnac047
Article
Article

Peripheral immune landscape and natural killer-like B cells in human Vogt-Koyanagi-Harada disease

Author information +
History +
PDF (3207KB)

Abstract

Vogt-Koyanagi-Harada (VKH) disease is a systemic autoimmune disorder threatening the eyesight. The pathogenic mechanisms and biomarkers reflecting disease severity and predicting treatment response require further exploration. Here, we performed a single-cell analysis of peripheral blood mononuclear cells (PBMC) obtained from eight patients with VKH disease and eight healthy controls to comprehensively delineate the changes in VKH disease. We showed a mixture of inflammation, effector, and exhausted states for PBMCs in VKH disease. Notably, our study implicated a newly identified B cell subset, natural killer-like B cells (K-BC) characterized by expressing CD19 and CD56, was correlated with VKH disease. K-BCs expanded in VKH disease, fell back after effective treatment, and promoted the differentiation of pathogenic T cells. Overall, we mapped the peripheral immune cell atlas in VKH disease and indicated the pathogenic role and potential value in predicting treatment response of K-BCs.

Keywords

Vogt-Koyanagi-Harada disease / single-cell RNA sequencing / natural killer-like B cells / autoimmunity

Cite this article

Download citation ▾
He Li, Lei Zhu, Xiuxing Liu, Lihui Xie, Rong Wang, Zhaohuai Li, Zhaohao Huang, Shizhao Yang, Binyao Chen, Jinguo Ye, Yingfeng Zheng, Wenru Su. Peripheral immune landscape and natural killer-like B cells in human Vogt-Koyanagi-Harada disease. Life Medicine, 2022, 1(3): 387-400 DOI:10.1093/lifemedi/lnac047

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Du L, Kijlstra A, Yang P. Vogt-Koyanagi-Harada disease: novel insights into pathophysiology, diagnosis and treatment. Prog Retin Eye Res 2016;52:84–111.

[2]

Read RW, Holland GN, Rao NA, et al. Revised diagnostic criteria for Vogt-Koyanagi-Harada disease: report of an international committee on nomenclature. Am J Ophthalmol 2001;131:647–52.

[3]

Hou S, Li N, Liao X, et al. Uveitis genetics. Exp Eye Res 2020;190:107853.

[4]

Rao NA, Gupta A, Dustin L, et al. Frequency of distinguishing clinical features in Vogt-Koyanagi-Harada disease. Ophthalmology 2010;117:591–9, 599.e1.

[5]

Yang P, Zhong Y, Du L, et al. Development and evaluation of diagnostic criteria for Vogt-Koyanagi-Harada disease. JAMA Ophthalmol 2018;136:1025–31.

[6]

Abu El-Asrar AM, Van Damme J, Struyf S, et al. New perspectives on the immunopathogenesis and treatment of uveitis associated with Vogt-Koyanagi-Harada disease. Front Med 2021;8:705796.

[7]

Lin P, Suhler EB, Rosenbaum JT. The future of uveitis treatment. Ophthalmology 2014;121:365–76.

[8]

Uchiyama E, Papaliodis GN, Lobo AM, et al. Side-effects of anti-inflammatory therapy in uveitis. Semin Ophthalmol 2014;29:456–67.

[9]

Imrie FR, Dick AD. Biologics in the treatment of uveitis. Curr Opin Ophthalmol 2007;18:481–6.

[10]

Damico FM, Cunha-Neto E, Goldberg AC, et al. T-cell recognition and cytokine profile induced by melanocyte epitopes in patients with HLADRB1* 0405-positive and -negative Vogt-Koyanagi-Harada uveitis. Invest Ophthalmol Vis Sci 2005;46:2465–71.

[11]

Sugita S, Takase H, Kawaguchi T, et al. Cross-reaction between tyrosinase peptides and cytomegalovirus antigen by T cells from patients with Vogt-Koyanagi-Harada disease. Int Ophthalmol 2007;27:87–95.

[12]

Hammer H. Cellular hypersensitivity to uveal pigment confirmed by leucocyte migration tests in sympathetic ophthalmitis and the Vogt-Koyanagi-Harada syndrome. Br J Ophthalmol 1974;58:773773776–776.

[13]

Zhong Z, Su G, Kijlstra A, et al. Activation of the interleukin-23/interleukin-17 signalling pathway in autoinflammatory and autoimmune uveitis. Prog Retin Eye Res 2021;80:100866.

[14]

Wu S, Ma R, Zhong Y, et al. Deficiency of IL-27 signaling exacerbates experimental autoimmune uveitis with elevated uveitogenic Th1 and Th17 responses. Int J Mol Sci 2021;22:7517.

[15]

Tsuruta D, Hamada T, Teramae H, et al. Inflammatory vitiligo in Vogt-Koyanagi-Harada disease. J Am Acad Dermatol 2001;44:129–31.

[16]

Chi W, Yang P, Li B, et al. IL-23 promotes CD4+ T cells to produce IL-17 in Vogt-Koyanagi-Harada disease. J Allergy Clin Immunol 2007;119:1218–24.

[17]

Ohta K, Yoshimura N. Bcl-2 expression by CD4 T lymphocytes in Vogt-Koyanagi-Harada disease. Ocul Immunol Inflamm 2002;10:93–103.

[18]

Caspi RR, Silver PB, Luger D, et al. Mouse models of experimental autoimmune uveitis. Ophthalmic Res 2008;40:169–74.

[19]

Hu Y, Hu Y, Xiao Y, et al. Genetic landscape and autoimmunity of monocytes in developing Vogt-Koyanagi-Harada disease. Proc Natl Acad Sci USA 2020;117:25712–21.

[20]

Chan CC, Palestine AG, Nussenblatt RB, et al. Anti-retinal auto-antibodies in Vogt-Koyanagi-Harada syndrome, Behcet’s disease, and sympathetic ophthalmia. Ophthalmology 1985;92:1025–8.

[21]

El-Asrar AMA, Berghmans N, Al-Obeidan SA, et al. Differential CXC and CX3C chemokine expression profiles in aqueous humor of patients with specific endogenous uveitic entities. Invest Ophthalmol Vis Sci 2018;59:2222–8.

[22]

Abu El-Asrar AM, Berghmans N, Al-Obeidan SA, et al. Local cytokine expression profiling in patients with specific autoimmune uveitic entities. Ocul Immunol Inflamm 2020;28:453462.

[23]

Stuart T, Satija R. Integrative single-cell analysis. Nat Rev Genet 2019;20:257–72.

[24]

Yamada S, Nomura S. Review of single-cell RNA sequencing in the heart. Int J Mol Sci 2020;21:8345.

[25]

Zhang Y, Wang D, Peng M, et al. Single-cell RNA sequencing in cancer research. J Exp Clin Cancer Res 2021;40:81.

[26]

Han J, DePinho RA, Maitra A. Single-cell RNA sequencing in pancreatic cancer. Nat Rev Gastroenterol Hepatol 2021;18:451–2.

[27]

Dutertre CA, Becht E, Irac SE, et al. Single-cell analysis of human mononuclear phagocytes reveals subset-defining markers and identifies circulating inflammatory dendritic cells. Immunity 2019;51:573–589.e578.

[28]

Harasymowicz NS, Rashidi N, Savadipour A, et al. Single-cell RNA sequencing reveals the induction of novel myeloid and myeloid-associated cell populations in visceral fat with long-term obesity. FASEB J 2021;35:e21417.

[29]

Xie X, Shi Q, Wu P, et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol 2020;21:1119–33.

[30]

Wilk AJ, Rustagi A, Zhao NQ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med 2020;26:1070–6.

[31]

Zhang W, Yang B, Weng L, et al. Single cell sequencing reveals cell populations that predict primary resistance to imatinib in chronic myeloid leukemia. Aging 2020;12:25337–55.

[32]

Blair TA, Michelson AD, Frelinger AL, et al. Mass cytometry reveals distinct platelet subtypes in healthy subjects and novel alterations in surface glycoproteins in Glanzmann thrombasthenia. Sci Rep 2018;8:10300.

[33]

Wang S, Xia P, Chen Y, et al. Natural killer-like B cells prime innate lymphocytes against microbial infection. Immunity 2016;45:131–44.

[34]

Wang S, Xia P, Fan Z. Natural-killer-like B Cells function as a separate subset of innate B cells. Immunity 2017;47:201–2.

[35]

Zhang Y, Kuang W, Li D, et al. Natural killer-like B cells secreting interleukin-18 induces a proinflammatory response in periodontitis. Front Immunol 2021;12:641562.

[36]

Liu S, Yang L, Jia S, et al. Interleukin-35 suppresses the activity of natural killer-like B cells in patients with hepatocellular carcinoma. Int Immunopharmacol 2021;100:108161.

[37]

Ahmed R, Omidian Z, Giwa A, et al. A public BCR present in a unique dual-receptor-expressing lymphocyte from Type 1 diabetes patients encodes a potent T cell autoantigen. Cell 2019;177:1583–1599.e16.

[38]

Yang J, Sundrud MS, Skepner J, et al. Targeting Th17 cells in autoimmune diseases. Trends Pharmacol Sci 2014;35:493–500.

[39]

Nicholson LB, Kuchroo VK. Manipulation of the Th1/Th2 balance in autoimmune disease. Curr Opin Immunol 1996;8:837–42.

[40]

Zhou L, Ivanov II, Spolski R, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 2007;8:967–74.

[41]

McGinley AM, Sutton CE, Edwards SC, et al. Interleukin-17A serves a priming role in autoimmunity by recruiting IL-1β-producing myeloid cells that promote pathogenic T cells. Immunity 2020;52:342–356.e6.

[42]

Dinarello CA. IL-18: a TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol 1999;103:11–24.

[43]

Shevach EM, Chang JT, Segal BM. The critical role of IL-12 and the IL-12R beta 2 subunit in the generation of pathogenic autoreactive Th1 cells. Springer Semin Immunopathol 1999;21:249–62.

[44]

Abbas AK, Trotta E, Simeonov DR, et al. Revisiting IL-2: biology and therapeutic prospects. Sci Immunol 2018;3:eaat1482.

[45]

Picelli S. Single-cell RNA-sequencing: the future of genome biology is now. RNA Biol 2017;14:637–50.

[46]

Silpa-Archa S, Silpa-Archa N, Preble JM, et al. Vogt-Koyanagi-Harada syndrome: perspectives for immunogenetics, multimodal imaging, and therapeutic options. Autoimmun Rev 2016;15:809–19.

[47]

Greco A, Fusconi M, Gallo A, et al. Vogt-Koyanagi-Harada syndrome. Autoimmun Rev 2013;12:1033–8.

[48]

Nicholson LB, Raveney BJ, Munder M. Monocyte dependent regulation of autoimmune inflammation. Curr Mol Med 2009;9:23–9.

[49]

Flaherty S, Reynolds JM. Mouse naïve CD4+ T cell isolation and in vitro differentiation into T cell subsets. J Vis Exp JoVE 2015;(98):52739.

[50]

Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015;15:486–99.

[51]

Musette P, Bouaziz JD. B cell modulation strategies in autoimmune diseases: new concepts. Front Immunol 2018;9:622.

[52]

Myhr KM, Torkildsen O, Lossius A, et al. B cell depletion in the treatment of multiple sclerosis. Expert Opin Biol Ther 2019;19:261–71.

[53]

Guo L, Kapur R, Aslam R, et al. CD20+ B-cell depletion therapy suppresses murine CD8+ T-cell-mediated immune thrombocytopenia. Blood 2016;127:735–8.

[54]

Jagessar SA, Heijmans N, Bauer J, et al. B-cell depletion abrogates T cell-mediated demyelination in an antibody-nondependent common marmoset experimental autoimmune encephalomyelitis model. J Neuropathol Exp Neurol 2012;71:716–28.

[55]

Zheng Y, Liu X, Le W, et al. A human circulating immune cell landscape in aging and COVID-19. Protein Cell 2020;11:740–70.

[56]

Bing SJ, Shemesh I, Chong WP, et al. AS101 ameliorates experimental autoimmune uveitis by regulating Th1 and Th17 responses and inducing Treg cells. J Autoimmun 2019;100:52–61.

[57]

Yasuda K, Takeuchi Y, Hirota K. The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol 2019;41:283–97.

[58]

Zhou Y, Zhou B, Pache L, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun 2019;10:1523.

[59]

Chen TJ, Kotecha N. Cytobank: providing an analytics platform for community cytometry data analysis and collaboration. High-dimensional single cell analysis: Mass cytometry, multi-parametric flow cytometry and bioinformatic techniques 2014;127–157.

RIGHTS & PERMISSIONS

The Author(s) 2022. Published by Oxford University Press on behalf of Higher Education Press.

AI Summary AI Mindmap
PDF (3207KB)

Supplementary files

lnac047_suppl_supplementary_material

lnac047_suppl_supplementary_table_s1

804

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/