Differential alterations in peripheral lymphocyte subsets in COVID-19 patients: upregulation of double-positive and double-negative T cells

Differential alterations in peripheral lymphocyte subsets in COVID-19 patients: upregulation of double-positive and double-negative T cells

Authors

  • Asmaa M. Zahran Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut
  • Zeinab Albadry M. Zahran Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut
  • Yasmeen H. Mady Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut https://orcid.org/0000-0002-7746-7894
  • Essam Eldeen M.O. Mahran Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut
  • Alaa Rashad Department Of Chest Diseases and Tuberculosis, Qena Faculty of Medicine, South Vally-University, Qena https://orcid.org/0000-0003-2727-8187
  • Ahmed Makboul Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut https://orcid.org/0000-0002-3574-0362
  • Khalid A. Nasif Department of Biochemistry, Faculty of Medicine, Minia University, Minia
  • Aida A. Abdelmaksoud Department of E.N.T., Qena Faculty of Medicine, South Valley University, Qena https://orcid.org/0000-0002-5347-0693
  • Omnia El-Badawy Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut

Keywords:

COVID-19, SARS-CoV-2, lymphocyte subsets, double-positive T cells, double-negative T cells

Abstract

Background: Viral infections cause alteration in the total number of lymphocytes and their subset distribution. We aimed to study peripheral blood lymphocyte subsets in COVID-19 patients and to correlate these subsets with clinical and laboratory data, which may help in clarifying the pathogenesis to develop novel diagnostic and prognostic biomarkers for COVID-19.
Methods: Twenty-six reverse-transcription polymerase chain reaction (RT-PCR) confirmed COVID-19 patients were subjected to medical history-taking and a thorough clinical examination. Laboratory tests included complete blood count, D dimer, ferritin, and C-reactive protein (CRP). Chest CT was used to diagnose COVID-19 pneumonia. Lymphocyte subsets were compared with those in 20 healthy controls using flow cytometry.
Results: Leucopenia, relative neutrophilia, lymphopenia, eosinopenia together with marked elevation in neutrophil/lymphocyte ratio were observed in our COVID-19 patients. A marked reduction was observed in T cells, including both CD4 and CD8 cells, natural killer (NK), and natural killer T cells (NKT). Double-positive T (DPT) cells, double-negative T (DNT) cells, and B cells were elevated in the patients relative to the other lymphocyte subsets.
Conclusion: Immune-inflammatory parameters are of utmost importance in understanding the pathogenesis and in the provisional diagnosis of COVID-19. Yet, due care must be taken during their interpretation because of the vast discrepancies observed between studies even in the same locality. Further studies are needed to clarify the role of B cells, DPT, and DNT cells in the pathogenesis and control of COVID-19.

Author Biography

Khalid A. Nasif, Department of Biochemistry, Faculty of Medicine, Minia University, Minia

College of Medicine, King Khalid University, Abha, Saudi Arabia

References

Fehr AR, Perlman S. Coronaviruses: an overview of their replication and pathogenesis. Coronaviruses: Springer; 2015. p. 1-23.

Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395:507-13.

Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 2020;395:565-74.

Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol 2017;39:529-39.

Li T, Qiu Z, Zhang L, Han Y, He W, Liu Z, et al. Significant changes of peripheral T lymphocyte subsets in patients with severe acute respiratory syndrome. J Infect Dis 2004;189:648-51.

Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–infected pneumonia in Wuhan, China. JAMA 2020;323:1061-9.

Kermali M, Khalsa RK, Pillai K, Ismail Z, Harky A. The role of biomarkers in diagnosis of COVID-19–A systematic review. Life Sci 2020;254:117788.

Henry BM, De Oliveira MHS, Benoit S, Plebani M, Lippi G. Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): a meta-analysis. Clin Chem Lab Med 2020;58:1021-8.

Salehi S, Abedi A, Balakrishnan S, Gholamrezanezhad A. Coronavirus disease 2019 (COVID-19): a systematic review of imaging findings in 919 patients. Am J Roentgenol 2020;215:87-93.

Ye Z, Zhang Y, Wang Y, Huang Z, Song B. Chest CT manifestations of new coronavirus disease 2019 (COVID-19): a pictorial review. Eur Radiol 2020;30:4381-9.

Hansell DM, Bankier AA, MacMahon H, McLoud TC, Muller NL, Remy J. Fleischner Society: glossary of terms for thoracic imaging. Radiology 2008;246:697-722.

Pan F, Ye T, Sun P. Time course of lung changes on chest CT during recovery from 2019 novel coronavirus (COVID-19) pneumonia. Radiology 2020;295:715-21.

Debuc B, Smadja DM. Is COVID-19 a new hematologic disease? Stem Cell Rev Rep 2020;17:4-8.

Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis 2020;71:762-8.

Cao M, Zhang D, Wang Y, Lu Y, Zhu X, Li Y, et al. Clinical features of patients infected with the 2019 novel coronavirus (COVID-19) in Shanghai, China. MedRxiv 2020.03.04.20030395.

Velavan TP, Meyer CG. Mild versus severe COVID-19: laboratory markers. Int J Infect Dis 2020;95:304-7.

Sun Y, Koh V, Marimuthu K, Ng OT, Young B, Vasoo S, et al. Epidemiological and clinical predictors of COVID-19. Clin Infect Dis 2020;71:786-92.

Guan W-j, Ni Z-y, Hu Y, Liang W-h, Ou C-q, He J-x, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 2020;382:1708-20.

Khartabil T, Russcher H, van der Ven A, de Rijke Y. A summary of the diagnostic and prognostic value of hemocytometry markers in COVID-19 patients. Crit Rev Clin Lab Sci 2020;57:415-31.

Shoenfeld Y, Gurewich Y, Gallant L, Pinkhas J. Prednisone-induced leukocytosis: influence of dosage, method and duration of administration on the degree of leukocytosis. Am J Med 1981;71:773-8.

Actor JK. Cells and organs of the immune system. Elsevier's Integrated Review. Immunology and Microbiology. Saunders; 2012. p. 7-16.

Agraz-Cibrian JM, Giraldo DM, Mary F-M, Urcuqui-Inchima S. Understanding the molecular mechanisms of NETs and their role in antiviral innate immunity. Virus Res 2017;228:124-33.

Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science 2004;303:1532-5.

Muraro SP, De Souza GF, Gallo SW, Da Silva BK, De Oliveira SD, Vinolo MAR, et al. Respiratory Syncytial Virus induces the classical ROS-dependent NETosis through PAD-4 and necroptosis pathways activation. Sci Rep 2018;8:1-12.

Hiroki CH, Toller-Kawahisa JE, Fumagalli MJ, Colon DF, Figueiredo L, Fonseca BA, et al. Neutrophil extracellular traps effectively control acute chikungunya virus infection. Front Immunol 2020;10:3108.

Feng X, Li S, Sun Q, Zhu J, Chen B, Xiong M, et al. Immune-inflammatory parameters in COVID-19 cases: A systematic review and meta-analysis. Front Med 2020;7:301.

Yang A-P, Liu J, Tao W, Li H-m. The diagnostic and predictive role of NLR, d-NLR and PLR in COVID-19 patients. Int Immunopharmacol 2020:106504.

Yu H, Li D, Deng Z, Yang Z, Cai J, Jiang L, et al. Total protein as a biomarker for predicting coronavirus disease-2019 pneumonia. Available at SSRN: https://ssrn.com/abstract=3551289

Du Y, Tu L, Zhu P, Mu M, Wang R, Yang P, et al. Clinical features of 85 fatal cases of COVID-19 from Wuhan. A retrospective observational study. Am J Respir Crit Care Med 2020;201:1372-9.

Zhang J-J, Dong X, Cao Y-Y, Yuan Y-D, Yang Y-B, Yan Y-Q, et al. Clinical characteristics of 140 patients infected with SARS‐CoV‐2 in Wuhan, China. Allergy 2020;75:1730-41.

Flores-Torres AS, Salinas-Carmona MC, Salinas E, Rosas-Taraco AG. Eosinophils and respiratory viruses. Viral Immunol 2019;32:198-207.

Busse W, Chupp G, Nagase H, Albers FC, Doyle S, Shen Q, et al. Anti–IL-5 treatments in patients with severe asthma by blood eosinophil thresholds: Indirect treatment comparison. J Allergy Clin Immunol 2019;143:190-200.

Hassani M, Leijte G, Bruse N, Kox M, Pickkers P, Vrisekoop N, et al. Differentiation and activation of eosinophils in the human bone marrow during experimental human endotoxemia. J Leukocyte Biol 2020;108:1665-71.

Butterfield JH. Treatment of hypereosinophilic syndromes with prednisone, hydroxyurea, and interferon. Immunol Allergy Clin North Am 2007;27:493-518.

Chan M, Wong V, Wong C, Chan P, Chu C, Hui D, et al. Serum LD1 isoenzyme and blood lymphocyte subsets as prognostic indicators for severe acute respiratory syndrome. J Intern Med 2004;255:512-8.

Wang F, Nie J, Wang H, Zhao Q, Xiong Y, Deng L, et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J Infect Dis 2020;221:1762-9.

Schmidt ME, Varga SM. The CD8 T cell response to respiratory virus infections. Front in Immunol 2018;9:678.

Baazim H, Schweiger M, Moschinger M, Xu H, Scherer T, Popa A, et al. CD8+ T cells induce cachexia during chronic viral infection. Nat Immunol 2019;20:701-10.

Saeidi A, Zandi K, Cheok YY, Saeidi H, Wong WF, Lee CYQ, et al. T-cell exhaustion in chronic infections: reversing the state of exhaustion and reinvigorating optimal protective immune responses. Front Immunol 2018;9:2569.

Wang F, Hou H, Luo Y, Tang G, Wu S, Huang M, et al. The laboratory tests and host immunity of COVID-19 patients with different severity of illness. JCI Insight 2020;5:e137799.

Noulsri E, Lerdwana S, Fucharoen S, Pattanapanyasat K. Phenotypic characterization of circulating CD4/CD8 tlymphocytes in β-thalassemia patients. Asian Pac J Allergy Immunol 2014;32:261.

Waschbisch A, Sammet L, Schröder S, Lee DH, Barrantes‐Freer A, Stadelmann C, et al. Analysis of CD4+ CD8+ double‐positive T cells in blood, cerebrospinal fluid and multiple sclerosis lesions. Clin Exp Immunol 2014;177:404-11.

Nascimbeni M, Shin E-C, Chiriboga L, Kleiner DE, Rehermann B. Peripheral CD4+ CD8+ T cells are differentiated effector memory cells with antiviral functions. Blood 2004;104:478-86.

Jiang M, Guo Y, Luo Q, Huang Z, Zhao R, Liu S, et al. T cell subset counts in peripheral blood can be used as discriminatory biomarkers for diagnosis and severity prediction of COVID-19. J Infect Dis 2020;222:198-202.

Wan S, Yi Q, Fan S, Lv J, Zhang X, Guo L, et al. Characteristics of lymphocyte subsets and cytokines in peripheral blood of 123 hospitalized patients with 2019 novel coronavirus pneumonia (NCP). MedRxiv 2020.02.10.20021832.

Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020;55:102763.

Overgaard NH, Jung JW, Steptoe RJ, Wells JW. CD4+/CD8+ double‐positive T cells: more than just a developmental stage? J Leukocyte Biol 2015;97:31-8.

Reis BS, Rogoz A, Costa-Pinto FA, Taniuchi I, Mucida D. Mutual expression of the transcription factors Runx3 and ThPOK regulates intestinal CD4+ T cell immunity. Nat Immunol 2013;14:271-80.

Sullivan YB, Landay AL, Zack JA, Kitchen SG, Al‐Harthi L. Upregulation of CD4 on CD8+ T cells: CD4dimCD8bright T cells constitute an activated phenotype of CD8+ T cells. Immunology 2001;103:270-80.

Kitchen SG, Korin YD, Roth MD, Landay A, Zack JA. Costimulation of naive CD8+ lymphocytes induces CD4 expression and allows human immunodeficiency virus type 1 infection. J Virol 1998;72:9054-60.

Kitchen SG, Jones NR, LaForge S, Whitmire JK, Vu B-A, Galic Z, et al. CD4 on CD8+ T cells directly enhances effector function and is a target for HIV infection. Proc Natl Acad Sci USA 2004;101:8727-32.

Kitchen SG, Whitmire JK, Jones NR, Galic Z, Kitchen CM, Ahmed R, et al. The CD4 molecule on CD8+ T lymphocytes directly enhances the immune response to viral and cellular antigens. Proc Natl Acad Sci USA 2005;102:3794-9.

Bohner P, Chevalier MF, Cesson V, Rodrigues-Dias S-C, Dartiguenave F, Burruni R, et al. Double positive CD4+ CD8+ T cells are enriched in urological cancers and favor T Helper-2 polarization. Front Immunol 2019;10:622.

De Tullio G, Strippoli S, Angarano R, De Fazio V, Sgherza N, Negri A, et al. αβ-double negative CD4/CD8 (CD56) T cell (DNTs) in metastatic melanoma: basal frequency and behaviour during Ipilimumab treatment. Preliminary evaluations. J Transl Med 2015;13:1-3.

Quandt D, Rothe K, Scholz R, Baerwald CW, Wagner U. Peripheral CD4CD8 double positive T cells with a distinct helper cytokine profile are increased in rheumatoid arthritis. PloS One 2014;9:e93293.

AMagerus-Chatinet B-LAV, ASolary E. Human TCR alpha/beta+ CD4-CD8-double-negative T cells in patients with autoimmune lymphoproliferative syndrome express restricted Vbeta TCR diversity and are clonally related to CD8+ T cells. J Immunol 2008;181:440448.

Mathiot ND, Krueger R, French MA, Price P. Percentage of CD3+ CD4-CD8-γδ TCR-T Cells Is Increased by HIV Disease. AIDS Res Hum Retroviruses 2001;17:977-80.

Tan M, Liu Y, Zhou R, Deng X, Li F, Liang K, et al. Immunopathological characteristics of coronavirus disease 2019 cases in Guangzhou, China. Immunology 2020;160:261-8.

Mazzoni A, Salvati L, Maggi L, Capone M, Vanni A, Spinicci M, et al. Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest 2020;130:4694-703.

Quinti I, Lougaris V, Milito C, Cinetto F, Pecoraro A, Mezzaroma I, et al. A possible role for B cells in COVID-19?: Lesson from patients with Agammaglobulinemia. J Allergy Clin Immunol 2020;146:211-3.

Cron RQ, WW Chatham. The rheumatologist's role in COVID-19. J Rheumatol 2020;47:639-42.

Bekaroğl M, Arslan B. Natural killer NK cells in β thalassemia major patients. JSM Biotechnol Bioeng 2014;2:1040.

Ezer Ü, Gülderen F, Çulha VK, Akgül N, Gürbüz Ö. Immunological status of thalassemia syndrome. Pediatr Hematol Oncol 2002;19:51-8.

Godfrey DI, MacDonald HR, Kronenberg M, Smyth MJ, Van Kaer L. NKT cells: what's in a name? Nat Rev Immunol 2004;4:231-7.

Godfrey DI, Kronenberg M. Going both ways: immune regulation via CD1d-dependent NKT cells. J Clin Invest 2004;114:1379-88.

Wallace KL, Marshall MA, Ramos SI, Lannigan JA, Field JJ, Strieter RM, et al. NKT cells mediate pulmonary inflammation and dysfunction in murine sickle cell disease through production of IFN-γ and CXCR3 chemokines. Blood 2009;114:667-76.

Odak I, Barros-Martins J, Bošnjak B, Stahl K, David S, Wiesner O, et al. Reappearance of effector T cells is associated with recovery from COVID-19. EBioMedicine 2020;57:102885.

Downloads

Published

10-06-2021

Issue

Section

Original Research Articles

How to Cite

“Differential Alterations in Peripheral Lymphocyte Subsets in COVID-19 Patients: Upregulation of Double-Positive and Double-Negative T Cells”. 2021. Multidisciplinary Respiratory Medicine 16 (June). https://doi.org/10.4081/mrm.2021.758.