Defining the Role of Th17 Lineage Cells in People with COVID-19

Authors

  • Sarah Connolly Human and Translational Immunology Group, School of Medicine, Trinity College Dublin, The University of Dublin, Ireland; Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
  • Sharee A. Basdeo Human and Translational Immunology Group, School of Medicine, Trinity College Dublin, The University of Dublin, Ireland; Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland

Keywords:

Th17 Cells, COVID-19, SARS-CoV-2

Abstract

Coronavirus disease 2019 (COVID-19) is a pandemic disease which has created a serious public health threat worldwide and causes pneumonia due to infection of the host with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). There remains a key gap in the understanding of what decides the outcome between an appropriate immune response and immunopathology in COVID-19. Th17 lineage cells are a distinct population of CD4+ T helper cells which mediate protection against bacteria and fungi. Th17 cells are dysregulated in patients with severe COVID-19 and are significant contributors to the systemic cytokine storm experienced by critical patients. Th17 cells have been described to mediate damage in the lungs of COVID-19 patients by encouraging the recruitment of neutrophils, contributing to acute respiratory distress syndrome and cytokine storms, causing pulmonary fibrosis, disrupting normal alveolar architecture and oxygenation processes and ultimately leading to systemic organ damage and death. Th17 cells have also been reported to contribute to immune dysfunction in conditions associated with increased risk of disease severity in COVID-19. There is a gap in our knowledge surrounding Th17-mediated protective immunity versus aberrant uncontrolled Th17-mediated pathology in the lung. This review aims to investigate and define the mechanisms of Th17 cells in COVID-19 pathogenesis by comparing the features of Th17 cells in a healthy immune response of the lung with the severe disease state in critical COVID-19.

References

Luckheeram RV, Zhou R, Verma AD, Xia B. CD4+T cells: differentiation and functions. Clin Dev Immunol. 2012;2012:925135.

Tsai HC, Velichko S, Hung LY, Wu R. IL-17A and Th17 cells in lung inflammation: an update on the role of Th17 cell differentiation and IL-17R signaling in host defense against infection. Clin Dev Immunol. 2013;2013:267971.

Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med. 2009;361(9):888-98.

Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et

al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126(6):1121- 33.

Basdeo SA, Cluxton D, Sulaimani J, Moran B, Canavan M, Orr C, et al. Ex-Th17 (Nonclassical Th1) Cells Are Functionally Distinct from Classical Th1 and Th17 Cells and Are Not Constrained by Regulatory T Cells. J Immunol. 2017;198(6):2249-59.

Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, et al. IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science. 2015;349(6248):606-13.

Gurczynski SJ, Moore BB. IL-17 in the lung: the good, the bad, and the ugly. Am J Physiol Lung Cell Mol Physiol. 2018;314(1):L6-L16.

Chen K, Eddens T, Trevejo-Nunez G, Way EE, Elsegeiny W, Ricks DM,

et al. IL-17 Receptor Signaling in the Lung Epithelium Is Required for Mucosal Chemokine Gradients and Pulmonary Host Defense against K. pneumoniae. Cell Host Microbe. 2016;20(5):596-605.

Pociask DA, Scheller EV, Mandalapu S, McHugh KJ, Enelow RI, Fattman CL, et al. IL-22 is essential for lung epithelial repair following influenza infection. Am J Pathol. 2013;182(4):1286-96.

McAllister F, Henry A, Kreindler JL, Dubin PJ, Ulrich L, Steele C, et al. Role of IL-17A, IL-17F, and the IL-17 receptor in regulating growth-related oncogene-alpha and granulocyte colony-stimulating factor in bronchial epithelium: implications for airway inflammation in cystic fibrosis. J Immunol. 2005;175(1):404-12.

Liu H, Mi S, Li Z, Hua F, Hu ZW. Interleukin 17A inhibits autophagy through activation of PIK3CA to interrupt the GSK3B-mediated degradation of BCL2 in lung epithelial cells. Autophagy. 2013;9(5):730-42.

O’Dwyer DN, Ashley SL, Moore BB. Influences of innate immunity, autophagy, and fibroblast activation in the pathogenesis of lung fibrosis. Am J Physiol Lung Cell Mol Physiol. 2016;311(3):L590-601.

Hu M, Yang S, Yang L, Cheng Y, Zhang H. Interleukin-22 Alleviated Palmitate-Induced Endoplasmic Reticulum Stress in INS-1 Cells through Activation of Autophagy. PLoS One. 2016;11(1):e0146818.

Zhou X, Loomis-King H, Gurczynski SJ, Wilke CA, Konopka KE, Ptaschinski C, et al. Bone marrow transplantation alters lung antigen-presenting cells to promote TH17 response and the development of pneumonitis and fibrosis following gammaherpesvirus infection. Mucosal Immunol. 2016;9(3):610-20.

Way EE, Chen K, Kolls JK. Dysregulation in lung immunity - the protective and pathologic Th17 response in infection. Eur J Immunol. 2013;43(12):3116-24.

Turner DL, Farber DL. Mucosal resident memory CD4 T cells in protection and immunopathology. Front Immunol. 2014;5:331.

Clark RA. Resident memory T cells in human health and disease. Sci Transl Med. 2015;7(269):269rv1.

Connor LM, Harvie MC, Rich FJ, Quinn KM, Brinkmann V, Le Gros G, et al. A key role for lung-resident memory lymphocytes in protective immune responses after BCG vaccination. Eur J Immunol. 2010;40(9):2482-92.

Hogan RJ, Zhong W, Usherwood EJ, Cookenham T, Roberts AD, Woodland DL. Protection from respiratory virus infections can be mediated by antigen-specific CD4(+) T cells that persist in the lungs. J Exp Med. 2001;193(8):981-6.

Snyder ME, Finlayson MO, Connors TJ, Dogra P, Senda T, Bush E, et al. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci Immunol. 2019;4(33).

Amezcua Vesely MC, Pallis P, Bielecki P, Low JS, Zhao J, Harman CCD, et al. Effector T. Cell. 2019;178(5):1176-88.e15.

Chuang Y, Hung ME, Cangelose BK, Leonard JN. Regulation of the IL-10- driven macrophage phenotype under incoherent stimuli. Innate Immun. 2016;22(8):647-57.

Hou J, Shi J, Chen L, Lv Z, Chen X, Cao H, et al. M2 macrophages promote myofibroblast differentiation of LR-MSCs and are associated with pulmonary fibrogenesis. Cell Commun Signal. 2018;16(1):89.

Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, et al. Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature. 2012;484(7395):514-8.

Chang KK, Liu LB, Jin LP, Zhang B, Mei J, Li H, et al. IL-27 triggers IL-10 production in Th17 cells via a c-Maf/RORγt/Blimp-1 signal to promote the progression of endometriosis. Cell Death Dis. 2017;8(3):e2666.

Worldometer. COVID-19 Coronavirus Pandemic 2021 [Available from: https://www.worldometers.info/coronavirus/.

Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367(6483):1260-3.

Tang D, Comish P, Kang R. The hallmarks of COVID-19 disease. PLoS Pathog. 2020;16(5):e1008536.

Asrani P, Hassan MI. SARS-CoV-2 mediated lung inflammatory responses in host: targeting the cytokine storm for therapeutic interventions. Mol Cell Biochem. 2021;476(2):675-87.

Muyayalo KP, Huang DH, Zhao SJ, Xie T, Mor G, Liao AH. COVID-19 and Treg/Th17 imbalance: Potential relationship to pregnancy outcomes. Am J Reprod Immunol. 2020;84(5):e13304.

Barton LM, Duval EJ, Stroberg E, Ghosh S, Mukhopadhyay S. COVID-19 Autopsies, Oklahoma, USA. Am J Clin Pathol. 2020;153(6):725-33.

Teuwen LA, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat Rev Immunol. 2020;20(7):389-91.

Toor SM, Saleh R, Sasidharan Nair V, Taha RZ, Elkord E. T-cell responses and therapies against SARS-CoV-2 infection. Immunology. 2021;162(1):30-43.

Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor Fedratinib. J Microbiol Immunol Infect. 2020;53(3):368-70.

Zenewicz LA. IL-22: There Is a Gap in Our Knowledge. Immunohorizons. 2018;2(6):198-207.

Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420-2.

De Biasi S, Meschiari M, Gibellini L, Bellinazzi C, Borella R, Fidanza L, et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat Commun. 2020;11(1):3434.

Sadeghi A, Tahmasebi S, Mahmood A, Kuznetsova M, Valizadeh H, Taghizadieh A, et al. Th17 and Treg cells function in SARS-CoV2 patients compared with healthy controls. J Cell Physiol. 2021;236(4):2829-39.

Shahbaz S, Xu L, Sligl W, Osman M, Bozorgmehr N, Mashhouri S,

et al. The Quality of SARS-CoV-2-Specific T Cell Functions Differs

in Patients with Mild/Moderate versus Severe Disease, and T Cells Expressing Coinhibitory Receptors Are Highly Activated. J Immunol. 2021;207(4):1099-111.

Mahmoud Salehi Khesht A, Karpisheh V, Qubais Saeed B, Olegovna Zekiy A, Yapanto LM, Nabi Afjadi M, et al. Different T cell related immunological profiles in COVID-19 patients compared to healthy controls. Int Immunopharmacol. 2021;97:107828.

Li CH, Chiou HC, Lin MH, Kuo CH, Lin YC, Hung CH. Immunological map in COVID-19. J Microbiol Immunol Infect. 2021;54(4):547-56.

Zhao Y, Kilian C, Turner JE, Bosurgi L, Roedl K, Bartsch P, et al. Clonal expansion and activation of tissue-resident memory-like Th17 cells expressing GM-CSF in the lungs of severe COVID-19 patients. Sci Immunol. 2021;6(56).

Yang J, Zhong M, Zhang E, Hong K, Yang Q, Zhou D, et al. Broad phenotypic alterations and potential dysfunction of lymphocytes in individuals clinically recovered from COVID-19. J Mol Cell Biol. 2021;13(3):197-209.

Li M, Zhang Y, Lu J, Li L, Gao H, Ma C, et al. Asymptomatic COVID-19 Individuals Tend to Establish Relatively Balanced Innate and Adaptive Immune Responses. Pathogens. 2021;10(9).

Chan YH, Fong SW, Poh CM, Carissimo G, Yeo NK, Amrun SN, et al. Asymptomatic COVID-19: disease tolerance with efficient anti-viral immunity against SARS-CoV-2. EMBO Mol Med. 2021;13(6):e14045.

Khadke S, Ahmed N, Ratts R, Raju S, Gallogly M, de Lima M, et al. Harnessing the immune system to overcome cytokine storm and reduce viral load in COVID-19: a review of the phases of illness and therapeutic agents. Virol J. 2020;17(1):154.

Orlov M, Wander PL, Morrell ED, Mikacenic C, Wurfel MM. A Case for Targeting Th17 Cells and IL-17A in SARS-CoV-2 Infections. J Immunol. 2020;205(4):892-8.

Tyagi AM, Srivastava K, Mansoori MN, Trivedi R, Chattopadhyay N, Singh D. Estrogen deficiency induces the differentiation of IL-17 secreting Th17 cells: a new candidate in the pathogenesis of osteoporosis. PLoS One. 2012;7(9):e44552.

Chen RY, Fan YM, Zhang Q, Liu S, Li Q, Ke GL, et al. Estradiol inhibits Th17 cell differentiation through inhibition of RORγT transcription by recruiting the ERα/REA complex to estrogen response elements of the RORγT promoter. J Immunol. 2015;194(8):4019-28.

Li Z, Yue Y, Xiong S. Distinct Th17 inductions contribute to the gender bias in CVB3-induced myocarditis. Cardiovasc Pathol. 2013;22(5):373-82.

Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D, et al. Obesity predisposes to Th17 bias. Eur J Immunol. 2009;39(9):2629-35.

Sumarac-Dumanovic M, Stevanovic D, Ljubic A, Jorga J, Simic M, Stamenkovic-Pejkovic D, et al. Increased activity of interleukin-23/ interleukin-17 proinflammatory axis in obese women. Int J Obes (Lond). 2009;33(1):151-6.

Liu M, He P, Liu HG, Wang XJ, Li FJ, Chen S, et al. [Clinical characteristics of 30 medical workers infected with new coronavirus pneumonia]. Zhonghua Jie He He Hu Xi Za Zhi. 2020;43(0):E016.

Peng YD, Meng K, Guan HQ, Leng L, Zhu RR, Wang BY, et al. [Clinical characteristics and outcomes of 112 cardiovascular disease patients infected by 2019-nCoV]. Zhonghua Xin Xue Guan Bing Za Zhi. 2020;48(6):450-5.

Abdel-Moneim A, Bakery HH, Allam G. The potential pathogenic role of IL-17/Th17 cells in both type 1 and type 2 diabetes mellitus. Biomed Pharmacother. 2018;101:287-92.

Cortvrindt C, Speeckaert R, Moerman A, Delanghe JR, Speeckaert MM. The role of interleukin-17A in the pathogenesis of kidney diseases. Pathology. 2017;49(3):247-58.

Coto E, Gómez J, Suárez B, Tranche S, Díaz-Corte C, Ortiz A, et al. Association between the IL17RA rs4819554 polymorphism and reduced renal filtration rate in the Spanish RENASTUR cohort. Hum Immunol. 2015;76(2-3):75-8.

Ji Q, Cheng G, Ma N, Huang Y, Lin Y, Zhou Q, et al. Circulating Th1, Th2, and Th17 Levels in Hypertensive Patients. Dis Markers. 2017;2017:7146290.

Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ, et al. Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction. Hypertension. 2010;55(2):500-7.

Li Q, Ding S, Wang YM, Xu X, Shen Z, Fu R, et al. Age-associated alteration in Th17 cell response is related to endothelial cell senescence and atherosclerotic cerebral infarction. Am J Transl Res. 2017;9(11):5160-8.

Schmitt V, Rink L, Uciechowski P. The Th17/Treg balance is disturbed during aging. Exp Gerontol. 2013;48(12):1379-86.

Rathore JS, Wang Y. Protective role of Th17 cells in pulmonary infection. Vaccine. 2016;34(13):1504-14.

de Candia P, Prattichizzo F, Garavelli S, Matarese G. T Cells: Warriors of SARS-CoV-2 Infection. Trends Immunol. 2021;42(1):18-30.

Mendoza VMM. Interleukin-17: A potential therapeutic target in COVID-19. J Infect. 2020;81(2):e136-e8.

Downloads

Published

2024-03-14

How to Cite

Connolly, S., & Basdeo, S. A. (2024). Defining the Role of Th17 Lineage Cells in People with COVID-19. Trinity Student Medical Journal , 22(1), 46–52. Retrieved from https://ojs.tchpc.tcd.ie/index.php/tsmj/article/view/2819

Similar Articles

You may also start an advanced similarity search for this article.