LYMPHOCYTES IN PATIENTS WITH CHRONIC ACTIVE EPSTEIN-BARR VIRUS INFECTION EXHIBITED ELEVATED PD-1/PD-L1 EXPRESSION AND A PREVAILING TH2 IMMUNE RESPONSE. EPSTEIN-BARR VIRUS INFECTION EXHIBITED ELEVATED PD-1/PD-L1

Main Article Content

kang sun

Keywords

chronic active Epstein-Barr virus, lymphocyte subsets, programmed cell death protein 1, programmed death-ligand 1, helper T cells 1, helper T cells 2

Abstract

Background And Objectives: Chronic active Epstein-Barr virus infection (CAEBV) is a lymphoproliferative disorder characterized by the increased numbers of EBV-infected T/natural killer (NK) cells and ongoing symptoms resembling infectious mononucleosis. Nonetheless, the exact correlation between peripheral lymphocyte subsets and the development of CAEBV remains uncertain. Methods: Consequently, we examined the levels of programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) expression, the status of EBV infection, and the frequencies of peripheral lymphocyte subpopulations in 24 patients with CAEBV and 15 EBV-seronegative donors using flow cytometry.


Results: Patients with CAEBV showed a notable rise in the expression levels of PD-1 and PD-L1 in peripheral T and NK cells compared to healthy donors (P < 0.05). The induction of PD-L1 expression was attributed to EBV infection. Moreover, most of the T cells infected with EBV displayed a memory phenotype characterized by the presence of CD45RO+. Additionally, patients with CAEBV showed markedly decreased frequency of helper T cells 1 (Th1) and naïve T (Tn) cells, and significantly elevated frequency of Th2 and effector-memory T (Tem) cells.


Conclusions: To summarize, EBV might evade the immune system by inducing PD-1/PD-L1 expression in peripheral T and NK cells, and promoting Th2 immune response dominance. Additionally, it may establish long-term persistence by infecting memory T cells in CAEBV.

Downloads

Download data is not yet available.


Abstract 1372
PDF Downloads 105
HTML Downloads 5

References

1. Okuno Y, Murata T, Sato Y, et al. Defective Epstein–Barr virus in chronic active infection and haematological malignancy. Nat Microbiol. 2019; 4(3): 404-413.
https://www.nature.com/articles/s41564-018-0334-0
PMid: 30664667
2. Chakravorty S, Afzali B, Kazemian M. EBV-associated diseases: Current therapeutics and emerging technologies. Front Immunol. 2022; 13:1059133.
https://www.frontiersin.org/articles/10.3389/fimmu.2022.1059133/full
PMid:36389670
3. Cui X, Snapper CM. Epstein Barr Virus: Development of Vaccines and Immune Cell Therapy for EBV-Associated Diseases. Front Immunol. 2021; 12:734471.
https://www.frontiersin.org/articles/10.3389/fimmu.2021.734471/full
PMid: 34691042
4. Leticia QM, Steven HS, Thomas T, et al. New concepts in EBV-associated B, T, and NK cell lymphoproliferative disorders. Virchows Arch. 2023; 482(1): 227-244.
https://link.springer.com/article/10.1007/s00428-022-03414-4
PMid: 36216980
5. Elias C, Elaine SJ, James RC, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood. 2022;140(11):1229-1253.
https://ashpublications.org/blood/article/140/11/1229/485458/The-International-Consensus-Classification-of
PMid: 35653592
6. Yonese I, Sakashita C, Imadome KI, et al. Nationwide survey of systemic chronic active EBV infection in Japan in accordance with the new WHO classification. Blood Adv. 2020; 4(13): 2918-2926.
https://ashpublications.org/bloodadvances/article/4/13/2918/461171/Nationwide-survey-of-systemic-chronic-active-EBV
PMid: 32598475
7. Austen JJW, Charlotte JH, Claire B. Severe Epstein-Barr virus infection in primary immunodeficiency and the normal host. Br J Haematol. 2016;175(4): 559-576.
https://onlinelibrary.wiley.com/doi/full/10.1111/bjh.14339?sid=nlm%3Apubmed
PMid: 27748521
8. Jiancheng L, Xiaokang C, Haiming W, et al. Peripheral blood lymphocyte counts in patients with infectious mononucleosis or chronic active Epstein-Barr virus infection and prognostic risk factors of chronic active Epstein-Barr virus infection. Am J Transl Res. 2021; 13 (11): 12797-12806.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8661241/
PMid: 34956494
9. Ming Y, Dechao J, Hanxiao X, et al. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer. 2018;17(1):129-143.
https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-018-0864-3
PMid: 30139382
10. Xi-Wen B, Hua W, Wen-Wen Z, et al. PD-L1 is upregulated by EBV-driven LMP1 through NF-κB pathway and correlates with poor prognosis in natural killer/T-cell lymphoma. J Hematol Oncol. 2016; 9(1):109-121.
https://jhoonline.biomedcentral.com/articles/10.1186/s13045-016-0341-7
PMid: 27737703
11. Jie W, Junshang G, Yian W, et al. EBV miRNAs BART11 and BART17-3p promote immune escape through the enhancer-mediated transcription of PD-L1. Nat Commun. 2022;13(1):866-885.
https://www.nature.com/articles/s41467-022-28479-2
PMid: 35165282
12. Junshang G, Jie W, Fang X, et al. Epstein-Barr Virus-Encoded Circular RNA CircBART2.2 Promotes Immune Escape of Nasopharyngeal Carcinoma by Regulating PD-L1. Cancer Res. 2021; 81(19):5074-5088.
https://aacrjournals.org/cancerres/article/81/19/5074/670309/Epstein-Barr-Virus-Encoded-Circular-RNA-CircBART2
PMid: 34321242
13. Yue S, Jingshi W, Yini W, et al. PD-1 blockade and lenalidomide combination therapy for chronic active Epstein-Barr virus infection. Clin Microbiol Infect. 2023; 29(6): e7-e13
https://www.clinicalmicrobiologyandinfection.com/article/S1198-743X(23)00039-3/fulltext
PMid: 36702399
14. Sebastian FP, Oscar S, Yasodha N. Defining the elusive boundaries of chronic active Epstein-Barr virus infection. Haematologica. 2018; 103(6): 924-927.
https://haematologica.org/article/view/8487
PMid: 29866887
15. Benjamin F, David B, Julie B, et al. Rapid identification and characterization of infected cells in blood during chronic active Epstein-Barr virus infection. J Exp Med. 2020; 217(11): e20192262.
https://rupress.org/jem/article/217/11/e20192262/152032/Rapid-identification-and-characterization-of
PMid: 32812031
16. Ann WNA, Carey LS, Alyssa L, et al. Naïve CD4+ T Cell Lymphopenia and Apoptosis in Chronic Hepatitis C Virus Infection Is Driven by the CD31+ Subset and Is Partially Normalized in Direct-Acting Antiviral Treated Persons. Front Immunol. 2021; 12: 641230.
https://www.frontiersin.org/articles/10.3389/fimmu.2021.641230/full
PMid: 33912168
17. Ann WNA, Carey LS, Lenche K, et al. Variable Normalization of Naïve CD4+ Lymphopenia and Markers of Monocyte and T Cell Activation over the Course of Direct-Acting Anti-Viral Treatment of Chronic Hepatitis C Virus Infection. Viruses. 2021; 14(1): 50-60.
https://www.mdpi.com/1999-4915/14/1/50
PMid: 35062255
18. Poonam S, Bhagyashri T, Prachi A, et al. Lymphopenia with Altered T Cell Subsets in Hospitalized COVID-19 Patients in Pune, India. Viral Immunol. 2023; 36(3): 163-175.
https://www.liebertpub.com/doi/10.1089/vim.2022.0123?url_ver=Z39.882-003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub++0pubmed
PMid: 36897333
19. Quirin N, Marc S, Florian W, et al. Pro- and Anti-Inflammatory Responses in Severe COVID-19-Induced Acute Respiratory Distress Syndrome—An Observational Pilot Study. Front Immunol. 2020; 11: 581338.
https://www.frontiersin.org/articles/10.3389/fimmu.2020.581338/full
PMid: 33123167
20. Zhanfeng L, Xue D, Zhaoqi Z, et al. Age‐related thymic involution: Mechanisms and functional impact. Aging Cell. 2022; 21(8): e13671.
https://onlinelibrary.wiley.com/doi/10.1111/acel.13671
PMid: 35822239
21. Hui S, Chen Y, Ya-Nan G, et al. Recirculating Th2 cells induce severe thymic dysfunction via IL-4/STAT6 signaling pathway. Biochem Biophys Res Commun. 2018; 501(1): 320-327.
https://www.sciencedirect.com/science/article/pii/S0006291X18310799
PMid: 29738764
22. Francesco A, Chiara R, Sergio R. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol. 2015;135(3): 626-35.
https://www.jacionline.org/article/S0091-6749(14)01585-1/fulltext
PMid: 25528359
23. Weihang L, Jindong H, Weifang X, et al. Distinct spatial and temporal roles for Th1, Th2, and Th17 cells in asthma. Front Immunol. 2022; 13: 974066.
https://www.frontiersin.org/articles/10.3389/fimmu.2022.974066/full
PMid: 36032162
24. Charles AD. The IL-1 family of cytokines and receptors in rheumatic diseases. Nat Rev Rheumatol. 2019;15(10): 612-632.
https://www.nature.com/articles/s41584-019-0277-8
PMid: 31515542
25. Punniyakoti VT, Srinivasan R, Poojitha M, et al. Modulation of IL-33/ST2 signaling as a potential new therapeutic target for cardiovascular diseases. Cytokine Growth Factor Rev. 2023; 71-72: 94-104.
https://www.sciencedirect.com/science/article/pii/S1359610123000266?via%3Dihub
PMid: 37422366
26. Jochen S, Alexander O, Elizabeth O, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005; 23(5): 479-90.
https://www.cell.com/immunity/fulltext/S1074-7613(05)00311-0?_returnURL=https%3A%2F%2Flinkinghub.elsevier.com%2Fretrieve%2Fpii%2FS1074761305003110%3Fshowall%3Dtrue
PMid: 16286016
27. Jaewoo H, Soohyun K, P CL. Interleukin-33 and ST2 Signaling in Tumor Microenvironment. J Interferon Cytokine Res. 2019; 39(1): 61-71.
https://www.liebertpub.com/doi/10.1089/jir.2018.0044?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub++0pubmed
PMid: 30256696
28. Corinne C, Jean-PG. Interleukin-33 (IL-33): A critical review of its biology and the mechanisms involved in its release as a potent extracellular cytokine. Cytokine. 2022; 156: 155891-155906.
https://www.sciencedirect.com/science/article/pii/S1043466622001004?via%3Dihub
PMid: 35640416
29. Wenyi J, Jingyao L, Ying Y, et al. IL‐33/ST2 as a potential target for tumor immunotherapy. Eur J Immunol. 2021; 51(8): 1943-1955.
https://onlinelibrary.wiley.com/doi/10.1002/eji.202149175
PMid: 34131922
30. Siyan C, Luxi W, Lirong P, et al. Hepatitis B virus X protein (HBx) promotes ST2 expression by GATA2 in liver cells. Mol Immunol. 2020; 123: 32-39.
https://www.sciencedirect.com/science/article/pii/S016158901930848X?via%3Dihub
PMid: 32413787
31. Tiago LF, Andreas HG, Christian L, et al. Macrophage: A Potential Target on Cartilage Regeneration. Front Immunol. 2020; 11(11):111-120.
https://www.frontiersin.org/articles/10.3389/fimmu.2020.00111/full
PMid: 32117263
32. Anna B, Slaven C, Valentina B, et al. IL-1 receptor blockade skews inflammation towards Th2 in a mouse model of systemic sclerosis. Eur Respir J. 2019; 54(3): 1900154-1900165.
https://erj.ersjournals.com/content/54/3/1900154.long
PMid: 31320452
33. Carl RH, Bojana SM, Crissy F, et al. The role of Interleukin 1 receptor antagonist in mesenchymal stem cell‐based tissue repair and regeneration. Biofactors. 2020; 46(2): 263-275.
https://iubmb.onlinelibrary.wiley.com/doi/10.1002/biof.1587
PMid: 31755595