2,792
Views
8
CrossRef citations to date
0
Altmetric
Coronaviruses

COVID-19: imbalanced cell-mediated immune response drives to immunopathology

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 2393-2404 | Received 19 May 2022, Accepted 04 Sep 2022, Published online: 09 Oct 2022

References

  • Zhang YZ, Holmes EC. A genomic perspective on the origin and emergence of SARS-CoV-2. Cell. 2020;181(2):223–227.
  • WHO Coronavirus (COVID-19) Dashboard: https://covid19.who.int/ [cited 2022 May 8]. Available from: https://covid19.who.int/
  • Matthew L, Robinson C, Morris P, et al. Impact of SARS-CoV-2 variants on inpatient clinical outcome. medRxiv [Preprint]. DOI:10.1101/2022.02.02.22270337
  • Liu J, Li S, Liu J, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. 2020;55:102763.
  • Wang J, Li Q, Yin Y, et al. Excessive neutrophils and neutrophil extracellular traps in COVID-19. Front Immunol. 2020;11:2063.
  • Zhang X, Tan Y, Ling Y, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature. 2020;583:437–440.
  • Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181(5):1036–1045.e9.
  • Cao YY, Xu X, Kitanovski S, et al. Comprehensive comparison of RNA-seq data of SARS-CoV-2, SARS-CoV and MERS-CoV infections: alternative entry routes and innate immune responses. Front Immunol. 2021;12:656433.
  • Kim YM, Shin EC. Type I and III interferon responses in SARS-CoV-2 infection. Exp Mol Med. 2021;53(5):750–760.
  • Barnes BJ, Adrover JM, Baxter-Stoltzfus A, et al. Targeting potential drivers of COVID-19: neutrophil extracellulartraps. J Exp Med. 2020;217(6):e20200652.
  • Wrapp D, Wang N, Corbett KS, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263.
  • Gibson PG, Qin L, Puah SH. COVID-19 acute respiratory distress syndrome (ARDS): clinical features and differences from typical pre-COVID-19 ARDS. Med J Aust. 2020;213(2):54–56.e1.
  • Bajaj V, Gadi N, Spihlman AP, et al. Aging, immunity, and COVID-19: how age influences the host immune response to coronavirus infections? Front Physiol. 2021;11:571416.
  • Bhattacharya S, Agarwal S, Shrimali NM, et al. Interplay between hypoxia and inflammation contributes to the progression and severity of respiratory viral diseases. Mol Aspects Med. 2021;81:101000.
  • Lee JS, Shin EC. The type I interferon response in COVID-19: implications for treatment. Nat Rev Immunol. 2020;20(10):585–586.
  • Hatton CF, Botting RA, Dueñas ME, et al. Delayed induction of type I and III interferons mediates nasal epithelial cell permissiveness to SARS-CoV-2. Nat Commun. 2021;12:7092.
  • Sa Ribero M, Jouvenet N, Dreux M, et al. Interplay between SARS-CoV-2 and the type I interferon response. PLoS Pathog. 2020;16:e1008737.
  • Huang S, Liu K, Cheng A, et al. SOCS proteins participate in the regulation of innate immune response caused by viruses. Front Immunol. 2020;11:558341.
  • Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369(6504):718–724.
  • Park A, Iwasaki A. Type I and type III interferons - induction, signaling, evasion, and application to combat COVID-19. Cell Host Microbe. 2020;27(6):870–878.
  • Coperchini F, Chiovato L, Croce L, et al. The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev. 2020;53:25–32.
  • Fox SE, Akmatbekov A, Harbert JL, et al. Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans. Lancet Respir Med. 2020;8:681–686.
  • Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–1069.
  • Du RH, Liang LR, Yang CQ, et al. Predictors of mortality for patients with COVID-19 pneumonia caused by SARS-CoV-2: a prospective cohort study. Eur Respir J. 2020;55(5):2000524.
  • Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–513.
  • Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–2629.
  • Zhou T, Su TT, Mudianto T, et al. Immune asynchrony in COVID-19 pathogenesis and potential immunotherapies. J Exp Med. 2020;217(10):e20200674.
  • Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol. 2020;20:355–362.
  • Mikacenic C, Moore R, Dmyterko V, et al. Neutrophil extracellular traps (NETs) are increased in the alveolar spaces of patients with ventilator-associated pneumonia. Crit Care. 2018;22(1):358.
  • Branzk N, Lubojemska A, Hardison SE, et al. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat Immunol. 2014;15(11):1017–1025.
  • Villanueva E, Yalavarthi S, Berthier CC, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011;187(1):538–552.
  • Veras FP, Pontelli MC, Silva CM, et al. SARS-CoV-2–triggered neutrophil extracellular traps mediate COVID-19 pathology. J Exp Med. 2020;217(12):e20201129.
  • Leppkes M, Knopf J, Naschberger E, et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine. 2020;58:102925.
  • Ackermann M, Anders HJ, Bilyy R, et al. Patients with COVID-19: in the dark-NETs of neutrophils. Cell Death Differ. 2021;28(11):3125–3139.
  • Yaqinuddin A, Kashir J. Novel therapeutic targets for SARS-CoV-2-induced acute lung injury: targeting a potential IL-1β/neutrophil extracellular traps feedback loop. Med Hypotheses. 2020;143:109906.
  • Cicco S, Cicco G, Racanelli V, et al. Neutrophil extracellular traps (NETs) and damage-associated molecular patterns (DAMPs): two potential targets for COVID-19 treatment. Mediat Inflamm. 2020;2020:7527953.
  • Zhou Z, Ren L, Zhang L, et al. Overly exuberant innate immune response to SARS-CoV-2 Infection. [2020 Mar 24]. Available from: DOI:10.2139/ssrn.3551623
  • Zhou Y, Fu B, Zheng X, et al. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl Sci Rev. 2020;7(6):998–1002.
  • Zhang D, Guo R, Lei L, et al. Frontline science: COVID-19 infection induces readily detectable morphologic and inflammation-related phenotypic changes in peripheral blood monocytes. J Leukocyte Biol. 2021;109(1):13–22.
  • Liao M, Liu Y, Yuan J, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. 2020;26:842–844.
  • Imai Y, Kuba K, Neely GG, et al. Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell. 2008;133(2):235–249.
  • Crozat K, Beutler B. TLR7: a new sensor of viral infection. Proc Natl Acad Sci U S A. 2004;101(18):6835–6836.
  • Mahmoudi S, Yaghmaei B, Sharifzadeh Ekbatani M, et al. Effects of coronavirus disease 2019 (COVID-19) on peripheral blood lymphocytes and their subsets in children: imbalanced CD4+/CD8+ T cell ratio and disease severity. Front Pediatr. 2021;9:643299.
  • Darif D, Hammi I, Kihel A, et al. The pro-inflammatory cytokines in COVID-19 pathogenesis: what goes wrong? Microb Pathog. 2021;153:104799.
  • Kim KD, Zhao J, Auh S, et al. Adaptive immune cells temper initial innate responses. Nat Med. 2007;13:1248–1252.
  • Palm NW, Medzhitov R. Not so fast: adaptive suppression of innate immunity. Nat Med. 2007;13:1142–1144.
  • Roncati L, Nasillo V, Lusenti B, et al. Signals of Th2 immune response from COVID-19 patients requiring intensive care. Ann Hematol. 2020;99(6):1419–1420.
  • Muyayalo KP, Huang DH, Zhao SJ, et al. COVID-19 and Treg/Th17 imbalance: potential relationship to pregnancy outcomes. Am J Reprod Immunol. 2020;84(5):e13304.
  • Sadeghi A, Tahmasebi S, Mahmood A, et al. Th17 and Treg cells function in SARS-CoV2 patients compared with healthy controls. J Cell Physiol. 2021;236(4):2829–2839.
  • Vick SC, Frutoso M, Mair F, et al. A regulatory T cell signature distinguishes the immune landscape of COVID-19 patients from those with other respiratory infections. Sci Adv. 2021;7(46):eabj0274.
  • Galván-Peña S, Leon J, Chowdhary K, et al. Profound Treg perturbations correlate with COVID-19 severity. Proc Natl Acad Sci U S A. 2021;118(37):e2111315118.
  • Shahbazi M, Moulana Z, Sepidarkish M, et al. Pronounce expression of Tim-3 and CD39 but not PD1 defines CD8 T cells in critical COVID-19 patients. Microb Pathog. 2021;153:104779.
  • Robbiani DF, Gaebler C, Muecksch F, et al. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature. 2020;584(7821):437–442.
  • Scheid JF, Barnes CO, Eraslan B, et al. B cell genomics behind cross-neutralization of SARS-CoV-2 variants and SARS-CoV. Cell. 2021;184(12):3205–3221.e24.
  • Thevarajan I, Nguyen THO, Koutsakos M, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453–455.
  • Long QX, Liu BZ, Deng HJ, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26(6):845–848.
  • Ni L, Ye F, Cheng ML, et al. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52(6):971–977.e3.
  • Chen J, Liu X, Zhang X, et al. Decline in neutralising antibody responses, but sustained T-cell immunity, in COVID-19 patients at 7 months post-infection. Clin Transl Immunol. 2021;10(7):e1319.
  • Ricke DO. Two different antibody-dependent enhancement (ADE) risks for SARS-CoV-2 antibodies. Front Immunol. 2021;12:640093.
  • Choe PG, Perera R, Park WB, et al. MERS-CoV antibody responses 1 year after symptom onset, South Korea, 2015. Emerg Infect Dis. 2017;23(7):1079–1084.
  • Okba NMA, Raj VS, Widjaja I, et al. Sensitive and specific detection of low-level antibody responses in mild Middle East respiratory syndrome coronavirus infections. Emerg Infect Dis. 2019;25(10):1868–1877.
  • Zhao J, Alshukairi AN, Baharoon SA, et al. Recovery from the Middle East respiratory syndrome is associated with antibody and T cell responses. Sci Immunol. 2017;2:14.
  • Li CK, Wu H, Yan H, et al. T cell responses to whole SARS coronavirus in humans. J Immunol. 2008;181(8):5490–5500.
  • Zhang Z, Mateus J, Coelho CH, et al. Humoral and cellular immune memory to four COVID-19 vaccines. Cell. 2022;185(14):2434–2451.e17.
  • Crinier A, Milpied P, Escalière B, et al. High-dimensional single-cell analysis identifies organ-specific signatures and conserved NK cell subsets in humans and mice. Immunity. 2018;49:971–986.e5.
  • Zheng M, Gao Y, Wang G, et al. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol. 2020;17(5):533–535.
  • 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;26:1070–1076.
  • Wang R, Jaw JJ, Stutzman NC, et al. Natural killer cell-produced IFN-γ and TNF-α induce target cell cytolysis through up-regulation of ICAM-1. J Leukoc Biol. 2012;91:299–309.
  • Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, et al. SARS-CoV-2 infection: the role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 2020;54:62–75. DOI:10.1016/j.cytogfr.2020.06.001
  • Chakraborty C, Sharma AR, Bhattacharya M, et al. A detailed overview of immune escape, antibody escape, partial vaccine escape of SARS-CoV-2 and their emerging variants with escape mutations. Front Immunol. 2022;13:801522.
  • Thorne LG, Bouhaddou M, Reuschl AK, et al. Evolution of enhanced innate immune evasion by SARS-CoV-2. Nature. 2022;602(7897):487–495.
  • Moss P. The T cell immune response against SARS-CoV-2. Nat Immunol. 2022;23:186–193.
  • Jordan SC, Shin BH, Gadsden TAM, et al. T cell immune responses to SARS-CoV-2 and variants of concern (Alpha and Delta) in infected and vaccinated individuals. Cell Mol Immunol. 2021;18:2554–2556.
  • Caniels TG, Bontjer I, van der Straten K, et al. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. Sci Adv. 2021;7(36):eabj5365.
  • Qin C, Zhou L, Hu Z, et al. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infect Dis. 2020;71(15):762–768.
  • Vargas A, Boivin R, Cano P, et al. Neutrophil extracellular traps are downregulated by glucocorticosteroids in lungs in an equine model of asthma. Respir Res. 2017;18(1):207.
  • Laurence J, Mulvey JJ, Seshadri M, et al. Anti-complement C5 therapy with eculizumab in three cases of critical COVID-19. Clin Immunol. 2020;219:108555.
  • Geleris J, Sun Y, Platt J, et al. Observational study of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;382:2411–2418.
  • Shao Z, Feng Y, Zhong L, et al. Clinical efficacy of intravenous immunoglobulin therapy in critical ill patients with COVID-19: a multicenter retrospective cohort study. Clin Transl Immunol. 2020;9(10):e1192.
  • Long Q, Liu BZ, Deng HJ, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26:845–848.
  • Afroja S, Sacchetti A. COVID-19 disease and the hyper-inflammatory response: are we accusing the wrong suspect? Am J Emerg Med. 2021;49:431.
  • Haji Abdolvahab M, Moradi-Kalbolandi S, Zarei M, et al. Potential role of interferons in treating COVID-19 patients. Int Immunopharmacol. 2021;90:107171.
  • Ascierto PA, Fu B, Wei H. IL-6 modulation for COVID-19: the right patients at the right time? J Immunother Cancer. 2021;9(4):e002285.
  • Kim JS, Lee JY, Yang JW, et al. Immunopathogenesis and treatment of cytokine storm in COVID-19. Theranostics. 2021;11(1):316–329.
  • Paludan SR, Mogensen TH. Innate immunological pathways in COVID-19 pathogenesis. Sci Immunol. 2022;7(67):5505.
  • Portsmore S, Tran Nguyen TN, Beacham E, et al. Combined IL-6 and JAK/STAT inhibition therapy in COVID-19-related sHLH, potential game changer. Br J Haematol. 2020;190(4):525–528.