Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1130 word(s) 1130 2021-03-04 07:05:22 |
2 format correct Meta information modification 1130 2021-03-11 02:59:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sui, Y.; Bekele, Y.; Berzofsky, J. SARS-CoV-2 infection induced Immunity. Encyclopedia. Available online: https://encyclopedia.pub/entry/7928 (accessed on 08 July 2024).
Sui Y, Bekele Y, Berzofsky J. SARS-CoV-2 infection induced Immunity. Encyclopedia. Available at: https://encyclopedia.pub/entry/7928. Accessed July 08, 2024.
Sui, Yongjun, Yonas Bekele, Jay Berzofsky. "SARS-CoV-2 infection induced Immunity" Encyclopedia, https://encyclopedia.pub/entry/7928 (accessed July 08, 2024).
Sui, Y., Bekele, Y., & Berzofsky, J. (2021, March 11). SARS-CoV-2 infection induced Immunity. In Encyclopedia. https://encyclopedia.pub/entry/7928
Sui, Yongjun, et al. "SARS-CoV-2 infection induced Immunity." Encyclopedia. Web. 11 March, 2021.
SARS-CoV-2 infection induced Immunity
Edit

SARS-CoV-2 infection-induced adaptive immunity includes humoral and cellular immune responses. Though the duration of protection is unclear so far, the induced immunity has been found to play an important role in mediating protection against subsequent  SARS-CoV-2 infections.    

SARS-CoV-2 neutralizing antibody Th1 responses

1. Introduction

After crossing the species barrier, most likely from bats, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has recently emerged to infect humans and cause severe public health problems. Since the outbreak in late December 2019, more than 85.9 million cases and 1.86 million deaths have been reported worldwide as of 5 January 2021. Effective SARS-CoV-2 vaccines and therapeutic strategies are urgently needed. While some vaccines and monoclonal antibodies have been approved for clinical use, understanding their mechanism of protection can facilitate improvements that may become necessary as long-term efficacy data become available over the next few years.

SARS-CoV-2 infects target cells via angiotensin-converting enzyme 2 (ACE2) as the primary receptor and transmembrane protease serine 2 (TMPRSS2) as an activating protease [1][2]. In the respiratory system, since ACE2 and TMPRSS2 are expressed primarily in type II pneumocytes and a fraction of secretory cells [2][3][4], SARS-CoV-2 virus productively infects these target cells in the upper and lower respiratory tracts [5][6]. The virus also can infect endothelial cells of multiple organs such as lung, gut, liver and kidneys and result in damaging blood clots in multiple organs [1][2][7][8]. In humans, the infected patients displayed various COVID-19 disease symptoms, ranging from mild to severe pneumonia, and in some cases acute respiratory distress syndrome and lethal pulmonary failure [9][10][11]. Other COVID19-related pathological manifestations in gut, heart, and brain were also observed, and led to fatalities as well as some long-term debilitating sequelae in a fraction of survivors. The mechanisms determining the variable outcomes remain elusive.

2. SARS-CoV-2 Infection-Induced Immunity That Mediates Protection against Disease

Natural SARS-CoV-2 infection results in both humoral and cellular immune responses. Humoral immunity induced by viral infection appears to play important roles in mediating protection against COVID-19 disease. SARS-CoV-2 surface glycoproteins, mainly the spike protein, as well as the internal nucleocapsid protein, are the main targets of humoral immune responses. Most PCR-confirmed SARS-CoV-2–infected persons seroconverted by 2 weeks after disease onset [12]. Virus-specific IgG, IgA, and IgM responses were detected in acute and convalescent COVID-19 patients [13][14]. Zohar et al. explored the early evolution of the humoral response in COVID-19 patients and found complicated associations between disease severity and trajectories of antibody subtypes [15]. While a rapid and potent IgG class switching is correlated with survival, a delay but ultimate maturation of IgG subclasses is associated with milder disease [15]. Interestingly, such a trend was not found for IgA and IgM subtypes, which evolve rapidly regardless of disease severity [15].

The protective potential of humoral immunity was confirmed by some early studies on transfusion of plasma from convalescent patients or animal studies using potent purified monoclonal antibody cocktails [16][17][18][19][20]. However, a recent clinical trial did not verify the clinical benefits in patients with severe COVID-19 pneumonia who received convalescent plasma [21]. A short window of time for successful administration of the plasma/antibody cocktails might be the key to explain this discrepancy, as suggested also for administration of monoclonal antibodies.

Similar to humoral immune responses, circulating SARS-CoV-2-specific CD4+ T-cell and CD8+ T-cell responses were detectable in most of SARS-CoV-2-infected patients within 1–2 weeks of symptom onset or at the convalescent stage [22][23]. Most virus specific CD4+T cell responses target spike and nucleocapsid glycoproteins as immunodominant. The responses were robust and mainly skewed towards Th1 cells, with the production of one or more of the Th1 cytokines, TNFα, IL-2, and IFNγ [22][23][24]. Though evidence of direct involvement of SARS-CoV-2-specific CD4+ T cells in mediating protection is lacking, for effective induction and long-term maintenance of antibody responses against COVID-19, a high-quality helper T cell response might be the key [25]. Several studies found that virus-specific CD4+ T cell responses were correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA against spike protein and nucleocapsid protein, live virus neutralizing antibody titers, and SARS-CoV-2 pseudovirus neutralization titers in COVID-19 patients [13][22][23][26].

Data on other coronavirus infections in animals and humans suggested that the humoral immune responses waned within a short period of time, whereas cellular immunity might persist longer. For SARS-CoV-2, while the duration of protective immunity is not yet known, a recent study suggests that IgG spike protein was relatively stable over at least 6 months, CD4+ and CD8+ T cell responses declined with a half-life of 3–5 months [27]. Previous exposure to other coronaviruses might elicit responses cross-reactive with SARS-CoV-2. In light of this, studies on pre-existing immunity against SARS-CoV-2 showed that cross-reactive T cells and antibodies are present [28][29][30]. While pre-existing T cell responses targeting a variety of viral proteins have been reported, the pre-existing antibody responses were predominantly of the IgG class and targeted the S2 subunit [28][29][30]. Whether the pre-existing immunity affects COVID-19 disease severity, or the dynamics of the current pandemic remains to be determined. Furthermore, SARS-CoV-2 -specific T cell responses were more sensitive to antigen exposure than antibody responses, as virus-specific T-cell responses were induced in antibody-seronegative individuals who had been exposed to SARS-CoV-2 [31][32].

SARS-CoV-2-specific CD8+ T cell responses were detectable in up to 70% of convalescent COVID-19 patients [13][22][23][28][31]. Virus-specific T cells displayed different phenotypes at different phases: highly activated cytotoxic phenotype at the acute stage, and polyfunctional and stem-like phenotype at the convalescent-stage [31]. Though there is some evidence suggesting that higher CD8+ T-cell responses were associated with mild disease, more studies are needed to elucidate whether virus-specific CD8+ T cells are pathogenic or protective in SARS-CoV-2 infections [23][31][33].

To identify the immune correlates of protection, though information from SARS-CoV-2 infections and vaccines are both valuable, extra caution is needed. Interpretation of the immune correlates from SARS-CoV-2–infected persons can be complicated and sometimes misleading. Like other viral infectious diseases, viral loads are a major driver of induced immune responses. Usually, patients with severe disease have high viral loads, which lead to high innate, and humoral/cellular immune responses, while the asymptomatic patients with lower viral loads induce lower immune responses. This is also true for SARS-Cov-2 infected patients. For example, vast numbers of studies have been carried out to characterize the humoral immune responses in COVID-19 acute or convalescent patients and most found that higher titers of neutralizing antibodies were induced in COVID-19 patients with severe disease than in those with mild disease [26]. The antibody responses also lasted longer in the patients with severe disease. Consistent with this, asymptomatic individuals manifested much weaker immune responses to SARS-CoV-2 infections than symptomatic ones [34]. Furthermore, in the early convalescent phase, IgG and neutralizing antibody levels in asymptomatic individuals declined much more quickly than those in the symptomatic patients [34]. However, these inverse correlations between disease severity and the magnitude of immune responses most likely indicate that severe disease, with high viral burden, leads to more robust immune responses, rather than the other way around.

References

  1. Thatcher, S.E.; Zhang, X.; Howatt, D.A.; Lu, H.; Gurley, S.B.; Daugherty, A.; Cassis, L.A. Angiotensin-converting enzyme 2 deficiency in whole body or bone marrow-derived cells increases atherosclerosis in low-density lipoprotein receptor-/- mice. Arter. Thromb. Vasc. Biol. 2011, 31, 758–765.
  2. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278.
  3. Ziegler, C.G.K.; Allon, S.J.; Nyquist, S.K.; Mbano, I.M.; Miao, V.N.; Tzouanas, C.N.; Cao, Y.; Yousif, A.S.; Bals, J.; Hauser, B.M.; et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues. Cell 2020, 181, 1016–1035.e19.
  4. Lukassen, S.; Chua, R.L.; Trefzer, T.; Kahn, N.C.; Schneider, M.A.; Muley, T.; Winter, H.; Meister, M.; Veith, C.; Boots, A.W.; et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J. 2020, 39, e105114.
  5. Wolfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Muller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020, 581, 465–469.
  6. Munster, V.J.; Feldmann, F.; Williamson, B.N.; van Doremalen, N.; Perez-Perez, L.; Schulz, J.; Meade-White, K.; Okumura, A.; Callison, J.; Brumbaugh, B.; et al. Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature 2020, 585, 268–272.
  7. Albini, A.; Di Guardo, G.; Noonan, D.M.; Lombardo, M. The SARS-CoV-2 receptor, ACE-2, is expressed on many different cell types: Implications for ACE-inhibitor- and angiotensin II receptor blocker-based cardiovascular therapies. Intern. Emerg. Med. 2020, 15, 759–766.
  8. Salamanna, F.; Maglio, M.; Landini, M.P.; Fini, M. Platelet functions and activities as potential hematologic parameters related to Coronavirus Disease 2019 (Covid-19). Platelets 2020, 31, 627–632.
  9. Zhou, Y.; Zhang, Z.; Tian, J.; Xiong, S. Risk factors associated with disease progression in a cohort of patients infected with the 2019 novel coronavirus. Ann. Palliat Med. 2020, 9, 428–436.
  10. Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422.
  11. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720.
  12. Okba, N.M.A.; Muller, M.A.; Li, W.; Wang, C.; GeurtsvanKessel, C.H.; Corman, V.M.; Lamers, M.M.; Sikkema, R.S.; de Bruin, E.; Chandler, F.D.; et al. Severe Acute Respiratory Syndrome Coronavirus 2-Specific Antibody Responses in Coronavirus Disease 2019 Patients. Emerg. Infect. Dis. 2020, 26, 1478–1488.
  13. Ni, L.; Ye, F.; Cheng, M.L.; Feng, Y.; Deng, Y.Q.; Zhao, H.; Wei, P.; Ge, J.; Gou, M.; Li, X.; et al. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 2020, 52, 971–977.e973.
  14. Padoan, A.; Sciacovelli, L.; Basso, D.; Negrini, D.; Zuin, S.; Cosma, C.; Faggian, D.; Matricardi, P.; Plebani, M. IgA-Ab response to spike glycoprotein of SARS-CoV-2 in patients with COVID-19: A longitudinal study. Clin. Chim. Acta 2020, 507, 164–166.
  15. Zohar, T.; Loos, C.; Fischinger, S.; Atyeo, C.; Wang, C.; Slein, M.D.; Burke, J.; Yu, J.; Feldman, J.; Hauser, B.M.; et al. Compromised Humoral Functional Evolution Tracks with SARS-CoV-2 Mortality. Cell 2020, 183, 1508–1519.e12.
  16. Liu, S.T.H.; Lin, H.M.; Baine, I.; Wajnberg, A.; Gumprecht, J.P.; Rahman, F.; Rodriguez, D.; Tandon, P.; Bassily-Marcus, A.; Bander, J.; et al. Convalescent plasma treatment of severe COVID-19: A propensity score-matched control study. Nat. Med. 2020, 26, 1708–1713.
  17. Salazar, E.; Christensen, P.A.; Graviss, E.A.; Nguyen, D.T.; Castillo, B.; Chen, J.; Lopez, B.V.; Eagar, T.N.; Yi, X.; Zhao, P.; et al. Treatment of Coronavirus Disease 2019 Patients with Convalescent Plasma Reveals a Signal of Significantly Decreased Mortality. Am. J. Pathol 2020, 190, 2290–2303.
  18. Liu, L.; Wang, P.; Nair, M.S.; Yu, J.; Rapp, M.; Wang, Q.; Luo, Y.; Chan, J.F.; Sahi, V.; Figueroa, A.; et al. Potent neutralizing antibodies against multiple epitopes on SARS-CoV-2 spike. Nature 2020, 584, 450–456.
  19. Tortorici, M.A.; Beltramello, M.; Lempp, F.A.; Pinto, D.; Dang, H.V.; Rosen, L.E.; McCallum, M.; Bowen, J.; Minola, A.; Jaconi, S.; et al. Ultrapotent human antibodies protect against SARS-CoV-2 challenge via multiple mechanisms. Science 2020, 370, 950–957.
  20. Baum, A.; Ajithdoss, D.; Copin, R.; Zhou, A.; Lanza, K.; Negron, N.; Ni, M.; Wei, Y.; Mohammadi, K.; Musser, B.; et al. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science 2020, 370, 1110–1115.
  21. Simonovich, V.A.; Burgos Pratx, L.D.; Scibona, P.; Beruto, M.V.; Vallone, M.G.; Vazquez, C.; Savoy, N.; Giunta, D.H.; Perez, L.G.; Sanchez, M.D.L.; et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N. Engl. J. Med. 2020.
  22. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020.
  23. Peng, Y.; Mentzer, A.J.; Liu, G.; Yao, X.; Yin, Z.; Dong, D.; Dejnirattisai, W.; Rostron, T.; Supasa, P.; Liu, C.; et al. Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020, 21, 1336–1345.
  24. Neidleman, J.; Luo, X.; Frouard, J.; Xie, G.; Gill, G.; Stein, E.S.; McGregor, M.; Ma, T.; George, A.F.; Kosters, A.; et al. SARS-CoV-2-specific T cells exhibit phenotypic features of robust helper function, lack of terminal differentiation, and high proliferative potential. Cell Rep. Med. 2020.
  25. Hellerstein, M. What are the roles of antibodies versus a durable, high quality T-cell response in protective immunity against SARS-CoV-2? Vaccine X 2020, 6, 100076.
  26. Wang, Y.; Zhang, L.; Sang, L.; Ye, F.; Ruan, S.; Zhong, B.; Song, T.; Alshukairi, A.N.; Chen, R.; Zhang, Z.; et al. Kinetics of viral load and antibody response in relation to COVID-19 severity. J. Clin. Investig. 2020, 130, 5235–5244.
  27. Dan, J.M.; Mateus, J.; Kato, Y.; Hastie, K.M.; Yu, E.D.; Faliti, C.E.; Grifoni, A.; Ramirez, S.I.; Haupt, S.; Frazier, A.; et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. BioRxiv 2020.
  28. Le Bert, N.; Tan, A.T.; Kunasegaran, K.; Tham, C.Y.L.; Hafezi, M.; Chia, A.; Chng, M.H.Y.; Lin, M.; Tan, N.; Linster, M.; et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584, 457–462.
  29. Braun, J.; Loyal, L.; Frentsch, M.; Wendisch, D.; Georg, P.; Kurth, F.; Hippenstiel, S.; Dingeldey, M.; Kruse, B.; Fauchere, F.; et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020, 587, 270–274.
  30. Ng, K.W.; Faulkner, N.; Cornish, G.H.; Rosa, A.; Harvey, R.; Hussain, S.; Ulferts, R.; Earl, C.; Wrobel, A.G.; Benton, D.J.; et al. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 2020, 370, 1339–1343.
  31. Sekine, T.; Perez-Potti, A.; Rivera-Ballesteros, O.; Stralin, K.; Gorin, J.B.; Olsson, A.; Llewellyn-Lacey, S.; Kamal, H.; Bogdanovic, G.; Muschiol, S.; et al. Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19. Cell 2020, 183, 158–168.e114.
  32. Floriane Gallais, A.V.; Wendling, M.; Nazon, C.; Partisani, M.; Sibilia, J.; Candon, S.; Fafi-Kremer, S. Intrafamilial Exposure to SARS-CoV-2 Induces Cellular Immune Response without Seroconversion. MedRxiv 2020.
  33. Schulien, I.; Kemming, J.; Oberhardt, V.; Wild, K.; Seidel, L.M.; Killmer, S.; Daul, F.; Salvat Lago, M.; Decker, A.; Luxenburger, H.; et al. Characterization of pre-existing and induced SARS-CoV-2-specific CD8(+) T cells. Nat. Med. 2020, 27, 78–85.
  34. Long, Q.X.; Tang, X.J.; Shi, Q.L.; Li, Q.; Deng, H.J.; Yuan, J.; Hu, J.L.; Xu, W.; Zhang, Y.; Lv, F.J.; et al. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat. Med. 2020, 26, 1200–1204.
More
Information
Subjects: Pathology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 482
Entry Collection: COVID-19
Revisions: 2 times (View History)
Update Date: 11 Mar 2021
1000/1000
Video Production Service