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 + 3082 word(s) 3082 2021-10-11 06:08:57 |
2 Format change -2 word(s) 3080 2021-11-30 02:34:19 |

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.
Mangge, H. Immune Responses against SARS-CoV-2—Questions and Experiences. Encyclopedia. Available online: https://encyclopedia.pub/entry/16512 (accessed on 27 July 2024).
Mangge H. Immune Responses against SARS-CoV-2—Questions and Experiences. Encyclopedia. Available at: https://encyclopedia.pub/entry/16512. Accessed July 27, 2024.
Mangge, Harald. "Immune Responses against SARS-CoV-2—Questions and Experiences" Encyclopedia, https://encyclopedia.pub/entry/16512 (accessed July 27, 2024).
Mangge, H. (2021, November 29). Immune Responses against SARS-CoV-2—Questions and Experiences. In Encyclopedia. https://encyclopedia.pub/entry/16512
Mangge, Harald. "Immune Responses against SARS-CoV-2—Questions and Experiences." Encyclopedia. Web. 29 November, 2021.
Immune Responses against SARS-CoV-2—Questions and Experiences
Edit

Understanding immune reactivity against SARS-CoV-2 is essential for coping with the COVID-19 pandemic. Herein, we discuss experiences and open questions about the complex immune responses to SARS-CoV-2. Some people react excellently without experiencing any clinical symptoms, they do not get sick, and they do not pass the virus on to anyone else (“sterilizing” immunity). Others produce antibodies, develop memory T cells, and do not get COVID-19 but transmit the virus to others (“protective” immunity). Some people get sick but recover. A varying percentage develops respiratory failure, systemic symptoms, clotting disorders, cytokine storms, or multi-organ failure; they subsequently decease. Some develop long COVID, a new, controversioal pathologic entity similar to fatigue syndrome or autoimmunity. In reality, COVID-19 is considered more of a systemic immune–vascular disease than a pulmonic disease, involving many tissues and the central nervous system. To fully comprehend the complex clinical manifestations, a profound understanding of the immune responses to SARS-CoV-2 is a good way to improve clinical management of COVID-19. Although neutralizing antibodies are an established approach to recognize an immune status, cellular immunity plays at least an equivalent or an even more important role. However, reliable methods to estimate the SARS-CoV-2-specific T cell capacity are not available for clinical routines. This deficit is important because an unknown percentage of people may exist with good memory T cell responsibility but a low number of or completely lacking peripheral antibodies against SARS-CoV-2. Apart from natural immune responses, vaccination against SARS-CoV-2 turned out to be very effective and much safer than naturally acquired immunity. Nevertheless, besides unwanted side effects of the currently available vector and mRNA preparations, concerns remain whether these vaccines will be strong enough to defeat the pandemic. Altogether, discussing important questions, and try to give answers based on the current knowledge and preliminary data from our laboratories. 

SARS-CoV-2 inflammation T and B cell responses immunity development of the COVID-19 pandemic

1. Introduction

The innate and the adaptive immune system cooperate and coincide for an effective immune response to a severe acute respiratory syndrome infection of coronavirus 2 (SARS-CoV-2). In the best-case scenario, the person becomes immune against the pathogen without clinical symptoms. However, in the worst-case scenario, deficiency or over-activation of the immune response could lead to severe illnesses accompanied by cytokine storms, respiratory failure, and death. SARS-CoV-2 uses a spike (S) protein to infect human cells via the angiotensin-2 (ACE2) receptor. Observational studies of SARS-CoV-2 infection and vaccine trials [1][2] provide evidence that robust neutralizing antibody titers and virus-specific T cell responses against the S protein effectively protect against severe COVID-19 [3][4]. Thus, this protein became a main target with which to stimulate the immune system by vaccination. The favorable risk–benefit ratio of vaccination certainly make this intervention an effective medicinal approach. Nevertheless, the worldwide vaccination campaign has not defeated the pandemic so far. In the following paper we will discuss questions that are still open concerning the immune response to SARS-CoV-2 infection, vaccination, and recently emerging adverse reactions based on unwanted effects of the vaccine-related immune activation [5][6][7][8][9].

2. Questions Addressing the Human Immune Answer to Infection with SARS-CoV-2

2.1. Mild Versus Severe Disease Course: How Different Is the Immune Response?

A pronounced initial immune response and higher SARS-CoV-2 copy numbers at diagnosis usually correlate with the severity of disease [8]. In fatal cases, the immune response itself is pathogenic. This can occur through the production of antibodies [10] that damage tissues [11][12], or through inducing excessive inflammation, resulting in a cytokine storm [13][14]. The immunological measurements of the immune response to SARS-CoV-2 are concentrated so far on total IgG antibody levels, with some groups also reporting IgA, IgM, and neutralizing antibody levels. Only occasionally, memory B and T cell levels are also included in the assessments. This is a considerable deficit because the cellular immune capacity is an important element with which to control SARS-CoV-2. Better immunological metrics must be developed, and reliable biomarkers to diagnose earlier courses with a subsequent damaging immune response are missing as of now. Figure 1 outlines in detail the important facets of the immune response found in mild compared to severe disease. The clinical manifestation of COVID-19 frequently shows smooth transitions between these two extreme phenotypes. It remains unknown which main determinants are responsible for a shift to a bad course. Advanced age [15], cardiovascular disease (CVD), and obesity [16] can also include the genetic factors that could be involved in causing severe COVID-19 [15]. Nevertheless, a minor subset of young and middle-aged individuals also develop massive inflammatory responses and need intensive care treatment with mechanical ventilation [14]. Several studies have tried to find out mediators which indicate a switch to a hyper-inflammatory reaction pattern [13][17][18][19][20]. Genetic predisposition may play an important role in this context [21]. Furthermore, late COVID immune responses are an emerging problem. Some individuals with a mild initial course of COVID-19 suffer from chronic symptoms with a duration of >2 months after the initial infection, called long COVID syndrome [22][23]. The symptoms resemble other postinfectious syndromes following outbreaks of Ebola [24] and chikungunya [25]. There is some overlap with myalgic encephalomyelitis [26], with dysregulated autonomic nervous system components and perturbed immune parameters (https://www.biorxiv.org/content/10.1101/2020.02.20.958249v2, accessed on 28 February 2020). The currently extensive impact of COVID-19 offers a great chance for science to improve the understanding of this long-lasting postinfection syndrome, which is probably based on a disturbed neuro-endocrino-immunological axis.
Figure 1. The involvement of different classes of immune cells mediating different courses of COVID-19 during development of the disease, modified from ref. [27]. Abbreviations: CCR = C-C chemokine receptor. CTLA-4 = cytotoxic T-lymphocyte protein 4. CXCR = C-X-C chemokine receptor. HLA-DR = HLA DR isotype. IL = interleukin. ISGs = interferon-stimulated genes. NETs = neutrophil extracellular traps. PD-1 = programmed cell death 1. TNF = tumor necrosis factor. Tregs = regulatory T cells. Modified from [27].

2.1.1. Grey Area

In the early phase of COVID-19, activated peripheral blood CD4+ and CD8+ T lymphocyte fractions arise in the circulation. CD4+ T cells show an increased level of IL-2 and IL-17 production. T regulatory cell (Tregs), neutrophil, and classic monocyte levels are also increased. Numbers of B cells and monocytes remain unchanged and increase later on in the course. The CD4+ and CD8+ T effector and memory lymphocytes are activated (CD38+, HLA-DR+) and show exhaustion-associated markers (PD-1, CD54). The chemokine receptor expression pattern shows decreased CCR6, CCR4, CXCR3, and CXCR4 antigens.

2.1.2. Blue Area

A subsequent favorable clinical course is associated with increased numbers of circulating dendritic cells, recovery of T cell numbers by polyfunctional and follicular helper T cell fractions, decreased neutrophils, and an increased number of intermediate HLA-DR-expressing monocytes. The humoral antibody response starts via activated and expanded B cells.

2.1.3. Red Area

Bad disease courses show decreased levels of dendritic and natural killer cell fractions and increased levels of immature neutrophils. Monocytes downregulate HLA-DR expression and upregulate IL-1β, TNF, and interferon-stimulated genes (ISGs). Although low in absolute numbers, CD8+ T cells upregulate granzyme B and perforin secretion. While the number of plasma blasts increases, specific antibodies are consistently produced. The main point is that myeloid cells show pro-inflammatory behavior with an egress of immature neutrophils from the bone marrow and an enhanced formation of neutrophil extracellular nets (NETs). NET formation is an important contributor to the development of organ and endothelial injury in conjunction with activation of the complement and coagulation cascades and a so-called cytokine storm.

2.2. How Is an Immune Response after Infection with the Virus Currently Measured?

Neutralizing antibodies to SARS-CoV-2 are measured because they detect unique components of the virus. The virus has four main structural proteins: the spike (S) glycoprotein, the small envelope (E) glycoprotein, the membrane (M) glycoprotein, and the nucleocapsid (N) protein. Memory T cells and memory B cells also recognize these components, but they are difficult to measure. Notably, the presence of antibodies does not always predict the presence of specific T cells or memory B cells [28][29]. Interestingly, the CD4+ T cell response equally targets several SARS-CoV-2 proteins, whereas the CD8+ T cell response in turn targets the nucleoprotein. This observation indicates the importance of including the nucleoprotein as a potential antigen target in future vaccines [30]. Nevertheless, SARS-CoV-2 S and N peptides are not potently immunogenic, and none of the single peptides could universally induce robust T cell responses, suggesting the necessity of using a multi-epitope strategy for COVID-19 vaccine design [31].

2.3. How Do Innate and Adaptive Immune Responses Contribute to the Course of COVID-19?

(A)
Innate immune response (recognition, interferon, and inflammasome activation)
After entering target cells via the ACE2 receptor, SARS-CoV-2 is detected by pattern-recognition receptors that resemble Toll-like receptors 3, 7, 8, and 9, as well as by the viral-infection sensors RIG-I and MDA5 [14][32]. The recognition stimulates the type I interferon (IFN) response and activates IFN-dependent genes [14][33]. The release of danger-associated molecular patterns (DAMP) contribute to the activation of the NLRP3 inflammasome [34] and other inflammasome complexes. The NLRP3 inflammasome induces caspase-1-dependent cleavage and the release of the key proinflammatory cytokines interleukin-1ß (IL-ß) and IL-18, and this correlates with COVID-19 disease severity [14][35]. Furthermore, gasdermin-D-mediated pyroptotic cell death is triggered [14][36]. Due to cell death, lactate dehydrogenase (LDH) is produced. These data underline that inflammasome activation is an important feature of COVID-19 which is reflected by high LDH blood levels [14][37]. Similar to SARS-CoV and MERS-CoV, SARS-CoV-2 is able to inhibit type I IFN responses in infected cells [38][39][40][41]. This allows the virus to replicate itself and to induce stronger tissue damage, and therefore an overwhelming immune response (see Figure 2, severe SARS-CoV-2 disease). Hence, the immune system struggles to limit viral replication while managing dying and dead cells [14]. Immune cells begin to also flow into the lungs, where they produce high amounts of proinflammatory cytokines, a process that escalates the situation [14]. The imbalanced immune reaction caused by the impaired type I IFN response contributes significantly to the overall severity of acute COVID-19 [13][41][42][43] by inducing a prolonged and ineffective innate immune response (Figure 2). This is also emphasized by new results coming from the COVID Human Genetic Effort, which found that inborn errors in the type I IFN pathway [44], or the presence of neutralizing autoantibodies to type I IFNs [11][45], were over-represented among individuals who developed life-threatening COVID-19. If these imbalanced or impaired innate responses also contribute to the development of MIS-C and long COVID remains to be determined [14]. In any case, an overwhelming inflammasome activation is an important component of severe COVID-19 [37]. This pathway also triggers a coagulation cascade which explains coagulopathy and severe thrombotic events in patients with severe COVID-19 [36][46]. Figure 2 shows the different kinetics shown in an average and in a severe SARS-CoV-2 infection. During a simple SARS-CoV-2 infection, the innate immune response phase gains rapid control of the viral load and is replaced by an effective and adaptive immune response based on adequate antibodies and T cell control. During a severe infection, the balance between innate and adaptive immune responses is essentially disturbed. An ineffective innate immune response with a probably weak type I IFN response does not gain control over the virus, and therefore prolongs and overlaps with an ineffective adaptive immune response. An ineffective adaptive immune response is characterized by a very weak and delayed adaptive T cell response. This situation allows the virus to expand, which usually leads to an overwhelming systemic inflammatory reaction.
Figure 2. The kinetics of the innate and adaptive immune responses in simple versus severe SARS-CoV-2 infections. Grey area: innate immune response. Blue area: adaptive immune response. Adopted from [47].
(B)
Adaptive immune response (Antibody production, seroconversion, T cell memory development)
The adaptive immune responses induced by SARS-CoV-2 infection largely follow the expected patterns observed in other comparable viral infections. Thus, more than 90% of infected individuals seroconvert a few weeks after initial infection [48][49], and anti-spike IgG antibodies are associated with protection from reinfection. T cell responses to the SARS-CoV-2 spike protein parallel B cell responses to the similar protein in nearly all COVID-19 cases [50] (Figure 2). Additionally, unexposed people occasionally show T cell reactivity to SARS-CoV-2 due to cross-reactive immunity to common cold coronaviruses [50]. The so-called antibody-dependent enhancement (ADE) has been supposed as another possible mechanism of severe COVID-19. These antibodies can facilitate Fc-receptor-mediated endocytosis of the virus and enhance viral replication resulting in massive hyperinflammation. ADE has been proved in dengue [51] and MERS [52] but clear evidence for its involvement in severe SARS-CoV-2 infections as well as SARS-CoV-2 second infections is lacking so far (https://doi.org/10.1016/S1473-3099(20)30783-0, accessed on 2 May 2021). Furthermore, to what extent this putative mechanism may also influence the balance between innate and adaptive immune responses in severe COVID-19 as shown in Figure 2 is not known so far.

2.4. How Strongly Does the Immune Response to SARS-CoV-2 Differ between Individuals?

Immune responses can vary markedly. Some people generate a very effective immune response. They cannot be infected again, and they will not give the virus to anyone else (so called ‘sterilizing’ immunity). Other people produce antibodies and are protected from disease, but not from future infection by SARS-CoV-2 [28]. These people still pass the virus. The number of antibodies produced after infection, the quality of those antibodies (how good they are at preventing infection) and the number and quality of the T cell response generated vary markedly between individuals. It is not known in detail so far for how long and how strong this immunity is over a longer period of time. With the rapidly growing proportion of the world’s population having been infected with SARS-CoV2, the degree to which this group is protected from reinfection is increasingly of interest. The waning of neutralizing antibody responses over the first year after infection makes reinfections more possible [53]. However, B cell and T cell memory responses induced by primary infection suggest that reinfection severity, and potentially transmission, may be mitigated over the longer term [53]. The potential of higher levels of neutralizing antibodies to be induced by vaccination suggests that reinfection could be further reduced by vaccination of those who have previously been infected [53]. Undoubtedly, a better understanding of all those factors, which influence immunity, is urgently needed to help achieve long-term immune control of the SARS-CoV-2 pandemic.
(A)
The influence of age and lifestyle
Age is an important factor. Usually, the risk for severe COVID-19 increases abruptly above age 70 [54][55][56]. Nevertheless, this may not be true for all elderly people. Some studies reported that antibody responses to infection do not vary with age in adults [57] and were even higher in older patients [58][59]. This was recently confirmed in 217 participants obtained from the Austrian Ischgl cohort by a 7–8 months (follow up) after infection [60]. Lifestyle factors associating with increased low-grade inflammation may explain these discrepancies. Preexisting obesity, hypertension, chronic obstructive pulmonary disease, cardiovascular disease (CVD) and smoking associate stronger with severe COVID-19 courses [61]. Notably, smoking induces, besides other negative effects, the expression of angiotensin-converting enzyme 2 (ACE2), which allows SARS-CoV-2 to enter cells [62]. Furthermore, an increased neutrophil to lymphocyte ratio (NLR) is a surrogate marker for systemic inflammation and is associated with poor prognosis in COVID-19 [63]. The NLR increases with the degree of obesity especially in context with metabolic syndrome and type 2 diabetes [64]. Older individuals with such conditions show two characteristics of severe COVID-19, such as failure in developing sufficient antiviral immune responses and a tendency to develop uncontrolled exacerbating responses to infections resulting in hyperinflammation and acute respiratory distress syndrome [14]. Older individuals also have weaker type I IFN responses, which further aggravate situations [65]. Also, additional “inflammaging” markers like NLPR3 activation [37], IL-6, IL-12 and IL-1ß secretion [66], and danger-related molecular patterns, including high mobility group box 1 (HMGB1) [67] have shown to be predictive for a severe COVID-19 course in elderly. The majority of young people have mild COVID-19 disease [68]. This is interesting because, although newborns and young children produce lower amounts of type I IFN upon stimulation through the virus and show a reduced breadth of antibody responses to SARS-CoV-2 proteins, they experience mild COVID-19 [58][69]. Possible explanations may be given to this by protective action taken by cross-reactive antibodies to common-cold coronaviruses and constitutive differences of the immune system [70]. Furthermore, the immune system of young children is more accustomed to face novel challenges, while the elderly mainly rely on memory driven immune responses [14]. This may explain the more benign clinical courses in the young.
(B)
The influence of gender
Men have a much greater risk of severe COVID-19 courses but women more frequently develop long COVID [71]. It is interesting that, in this context, women elicit stronger type I IFN responses after stimulation with TLR7 ligands [72], that they develop stronger vaccine responses and that they have better survival rates for a number of acute infections than men [73]. These sex differences are not age dependent and, thus, can also be seen before puberty between boys and girls, which indicates rather genetic differences. Interestingly, TLR7, a common virus sensor, is expressed on the X chromosome, suggesting a possible gene-dosage effect between the sexes [74]. Furthermore, neutralizing autoantibodies to type I IFN are much more frequently found in male patients with COVID-19 [14]. Future research is warranted to elucidate the reason for this phenomenon. The thymus involutes more rapidly in boys than in girls which may explain a weaker T cell control in men [75]. Otherwise, MIS-C is quite evenly distributed between boys and girls [76]. It is also important to which extent social factors and differing exposure play a part in sex differences [14].
(C)
The influence of immune deficiency
People with immunodeficiency, autoimmune diseases, or those who take immunosuppressive medication respond less to SARS-CoV-2 than young healthy people [77]. However, the data are also contradictory for this. One systematic review found no statistically significant increased risk of severe COVID-19 in immunosuppressed persons [78] whereas in cases with solid-organ transplants and cancer patients an increased risk was seen [79]. Especially, cancer patients treated with checkpoint inhibitors may be at a high risk to develop severe COVID-19 [80]. On the other hand, it is also possible that the host immune response against SARS-CoV-2 can cause an anticancer effect in certain constellations. Challenor et al. [81] reported one case with classical Hodgkin lymphoma with stage III disease that went into remission without corticosteroids or immunochemotherapy. Thus, the SARS-CoV-2 triggered immune activation may mediate an anti-tumor immune response as seen in other infections in the context of high-grade non-Hodgkin lymphoma [82]. Future research is needed to clarify the outcome of cancer patients who overcome COVID-19 disease. Particularly tumor patients with good checkpoint inhibitor effects that successfully overcome COVID-19 disease should be subject to research [83]. Furthermore, the effects of immunosuppressive medications, especially methotrexate and rituximab, on decreasing serological responses must be determined in future studies. Concerning successful vaccination against SARS-CoV-2 in rheumatic diseases, methotrexate may be held for up to 2 weeks after the vaccination, and rituximab a few weeks after the vaccination until further clinical trials can answer this question [84]

References

  1. Kim, D.S.; Rowland-Jones, S.; Gea-Mallorqui, E. Will SARS-CoV-2 Infection Elicit Long-Lasting Protective or Sterilising Immunity? Implications for Vaccine Strategies (2020). Front. Immunol. 2020, 11, 571481.
  2. Poland, G.A.; Ovsyannikova, I.G.; Kennedy, R.B. SARS-CoV-2 immunity: Review and applications to phase 3 vaccine candidates. Lancet 2020, 396, 1595–1606.
  3. Sahin, U.; Muik, A.; Derhovanessian, E.; Vogler, I.; Kranz, L.M.; Vormehr, M.; Baum, A.; Pascal, K.; Quandt, J.; Maurus, D.; et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature 2020, 586, 594–599.
  4. Awadasseid, A.; Wu, Y.; Tanaka, Y.; Zhang, W. Current advances in the development of SARS-CoV-2 vaccines. Int. J. Biol. Sci. 2021, 17, 8–19.
  5. Franchini, M.; Liumbruno, G.M.; Pezzo, M. COVID-19 vaccine-associated immune thrombosis and thrombocytopenia (VITT): Diagnostic and therapeutic recommendations for a new syndrome. Eur. J. Haematol. 2021, 107, 173–180.
  6. Abu Mouch, S.; Roguin, A.; Hellou, E.; Ishai, A.; Shoshan, U.; Mahamid, L.; Zoabi, M.; Aisman, M.; Goldschmid, N.; Berar Yanay, N. Myocarditis following COVID-19 mRNA vaccination. Vaccine 2021, 39, 3790–3793.
  7. Gargano, J.W.; Wallace, M.; Hadler, S.C.; Langley, G.; Su, J.R.; Oster, M.E.; Broder, K.R.; Gee, J.; Weintraub, E.; Shimabukuro, T.; et al. Use of mRNA COVID-19 Vaccine After Reports of Myocarditis Among Vaccine Recipients: Update from the Advisory Committee on Immunization Practices—United States, June 2021. MMWR Morb. Mortal. Wkly. Rep. 2021, 70, 977–982.
  8. Liu, R.; Yi, S.; Zhang, J.; Lv, Z.; Zhu, C.; Zhang, Y. Viral Load Dynamics in Sputum and Nasopharyngeal Swab in Patients with COVID-19. J. Dent. Res. 2020, 99, 1239–1244.
  9. Matheny, M.; Maleque, N.; Channell, N.; Eisch, A.R.; Auld, S.C.; Banerji, A.; Druey, K.M. Severe Exacerbations of Systemic Capillary Leak Syndrome After COVID-19 Vaccination: A Case Series. Ann. Intern. Med. 2021.
  10. Wang, E.Y.; Team, Y.I.; Mao, T.; Klein, J.; Dai, Y.; Huck, J.D.; Jaycox, J.R.; Liu, F.; Zhou, T.; Israelow, B.; et al. Diverse Functional Autoantibodies in Patients with COVID-19. Nature 2021, 595, 283–288.
  11. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Beziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370.
  12. Zuo, Y.; Estes, S.K.; Ali, R.A.; Gandhi, A.A.; Yalavarthi, S.; Shi, H.; Sule, G.; Gockman, K.; Madison, J.A.; Zuo, M.; et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci. Transl. Med. 2020, 12.
  13. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469.
  14. Brodin, P. Immune determinants of COVID-19 disease presentation and severity. Nat. Med. 2021, 27, 28–33.
  15. Farshbafnadi, M.; Zonouzi, S.K.; Sabahi, M.; Dolatshahi, M.; Aarabi, M.H. Aging & COVID-19 susceptibility, disease severity, and clinical outcomes: The role of entangled risk factors. Exp. Gerontol. 2021, 154, 111507.
  16. Amin, M.T.; Fatema, K.; Arefin, S.; Hussain, F.; Bhowmik, D.R.; Hossain, M.S. Obesity, a major risk factor for immunity and severe outcomes of COVID-19. Biosci. Rep. 2021, 41, BSR20210979.
  17. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; Hlh Across Speciality Collaboration, U.K. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034.
  18. Cao, X. COVID-19: Immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020, 20, 269–270.
  19. Mathew, D.; Giles, J.R.; Baxter, A.E.; Oldridge, D.A.; Greenplate, A.R.; Wu, J.E.; Alanio, C.; Kuri-Cervantes, L.; Pampena, M.B.; D’Andrea, K.; et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 2020, 369, eabc8511.
  20. Rodriguez, L.; Pekkarinen, P.T.; Lakshmikanth, T.; Tan, Z.; Consiglio, C.R.; Pou, C.; Chen, Y.; Mugabo, C.H.; Nguyen, N.A.; Nowlan, K.; et al. Systems-Level Immunomonitoring from Acute to Recovery Phase of Severe COVID-19. Cell Rep. Med. 2020, 1, 100078.
  21. Pairo-Castineira, E.; Clohisey, S.; Klaric, L.; Bretherick, A.D.; Rawlik, K.; Pasko, D.; Walker, S.; Parkinson, N.; Fourman, M.H.; Russell, C.D.; et al. Genetic mechanisms of critical illness in COVID-19. Nature 2021, 591, 92–98.
  22. Dennis, A.; Wamil, M.; Alberts, J.; Oben, J.; Cuthbertson, D.J.; Wootton, D.; Crooks, M.; Gabbay, M.; Brady, M.; Hishmeh, L.; et al. Multiorgan impairment in low-risk individuals with post-COVID-19 syndrome: A prospective, community-based study. BMJ Open 2021, 11, e048391.
  23. Ludvigsson, J.F. Case report and systematic review suggest that children may experience similar long-term effects to adults after clinical COVID-19. Acta Paediatr. 2021, 110, 914–921.
  24. Clark, D.V.; Kibuuka, H.; Millard, M.; Wakabi, S.; Lukwago, L.; Taylor, A.; Eller, M.A.; Eller, L.A.; Michael, N.L.; Honko, A.N.; et al. Long-term sequelae after Ebola virus disease in Bundibugyo, Uganda: A retrospective cohort study. Lancet Infect. Dis. 2015, 15, 905–912.
  25. Guillot, X.; Ribera, A.; Gasque, P. Chikungunya-Induced Arthritis in Reunion Island: A Long-Term Observational Follow-Up Study Showing Frequently Persistent Joint Symptoms, Some Cases of Persistent Chikungunya Immunoglobulin M Positivity, and No Anticyclic Citrullinated Peptide Seroconversion After 13 Years. J. Infect. Dis. 2020, 222, 1740–1744.
  26. Hickie, I.; Davenport, T.; Wakefield, D.; Vollmer-Conna, U.; Cameron, B.; Vernon, S.D.; Reeves, W.C.; Lloyd, A.; Dubbo Infection Outcomes Study, G. Post-infective and chronic fatigue syndromes precipitated by viral and non-viral pathogens: Prospective cohort study. BMJ 2006, 333, 575.
  27. Osuchowski, M.F.; Winkler, M.S.; Skirecki, T.; Cajander, S.; Shankar-Hari, M.; Lachmann, G.; Monneret, G.; Venet, F.; Bauer, M.; Brunkhorst, F.M.; et al. The COVID-19 puzzle: Deciphering pathophysiology and phenotypes of a new disease entity. Lancet Respir. Med. 2021, 9, 622–642.
  28. Lumley, S.F.; O’Donnell, D.; Stoesser, N.E.; Matthews, P.C.; Howarth, A.; Hatch, S.B.; Marsden, B.D.; Cox, S.; James, T.; Warren, F.; et al. Antibody Status and Incidence of SARS-CoV-2 Infection in Health Care Workers. N. Engl. J. Med. 2021, 384, 533–540.
  29. 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. Science 2021, 371.
  30. Cohen, K.W.; Linderman, S.L.; Moodie, Z.; Czartoski, J.; Lai, L.; Mantus, G.; Norwood, C.; Nyhoff, L.E.; Edara, V.V.; Floyd, K.; et al. Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells. medRxiv 2021.
  31. Zhao, J.; Wang, L.; Schank, M.; Dang, X.; Lu, Z.; Cao, D.; Khanal, S.; Nguyen, L.N.; Nguyen, L.N.T.; Zhang, J.; et al. SARS-CoV-2 specific memory T cell epitopes identified in COVID-19-recovered subjects. Virus Res. 2021, 304, 198508.
  32. Lim, Y.X.; Ng, Y.L.; Tam, J.P.; Liu, D.X. Human Coronaviruses: A Review of Virus-Host Interactions. Diseases 2016, 4, 26.
  33. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066.
  34. Nieto-Torres, J.L.; Verdia-Baguena, C.; Jimenez-Guardeno, J.M.; Regla-Nava, J.A.; Castano-Rodriguez, C.; Fernandez-Delgado, R.; Torres, J.; Aguilella, V.M.; Enjuanes, L. Severe acute respiratory syndrome coronavirus E protein transports calcium ions and activates the NLRP3 inflammasome. Virology 2015, 485, 330–339.
  35. Rodrigues, T.S.; de Sa, K.S.G.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Goncalves, A.V.; Perucello, D.B.; Andrade, W.A.; Castro, R.; et al. Inflammasomes are activated in response to SARS-CoV-2 infection and are associated with COVID-19 severity in patients. J. Exp. Med. 2021, 218.
  36. Wu, C.; Lu, W.; Zhang, Y.; Zhang, G.; Shi, X.; Hisada, Y.; Grover, S.P.; Zhang, X.; Li, L.; Xiang, B.; et al. Inflammasome Activation Triggers Blood Clotting and Host Death through Pyroptosis. Immunity 2019, 50, 1401–1411.
  37. Han, Y.; Zhang, H.; Mu, S.; Wei, W.; Jin, C.; Tong, C.; Song, Z.; Zha, Y.; Xue, Y.; Gu, G. Lactate dehydrogenase, an independent risk factor of severe COVID-19 patients: A retrospective and observational study. Aging 2020, 12, 11245–11258.
  38. Zust, R.; Cervantes-Barragan, L.; Habjan, M.; Maier, R.; Neuman, B.W.; Ziebuhr, J.; Szretter, K.J.; Baker, S.C.; Barchet, W.; Diamond, M.S.; et al. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 2011, 12, 137–143.
  39. Spiegel, M.; Pichlmair, A.; Martinez-Sobrido, L.; Cros, J.; Garcia-Sastre, A.; Haller, O.; Weber, F. Inhibition of Beta interferon induction by severe acute respiratory syndrome coronavirus suggests a two-step model for activation of interferon regulatory factor 3. J. Virol. 2005, 79, 2079–2086.
  40. Miorin, L.; Kehrer, T.; Sanchez-Aparicio, M.T.; Zhang, K.; Cohen, P.; Patel, R.S.; Cupic, A.; Makio, T.; Mei, M.; Moreno, E.; et al. SARS-CoV-2 Orf6 hijacks Nup98 to block STAT nuclear import and antagonize interferon signaling. Proc. Natl. Acad. Sci. USA 2020, 117, 28344–28354.
  41. Arunachalam, P.S.; Wimmers, F.; Mok, C.K.P.; Perera, R.; Scott, M.; Hagan, T.; Sigal, N.; Feng, Y.; Bristow, L.; Tak-Yin Tsang, O.; et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 2020, 369, 1210–1220.
  42. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.
  43. Vabret, N.; Britton, G.J.; Gruber, C.; Hegde, S.; Kim, J.; Kuksin, M.; Levantovsky, R.; Malle, L.; Moreira, A.; Park, M.D.; et al. Immunology of COVID-19: Current State of the Science. Immunity 2020, 52, 910–941.
  44. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020, 370.
  45. Bastard, P.; Levy, R.; Henriquez, S.; Bodemer, C.; Szwebel, T.A.; Casanova, J.L. Interferon-beta Therapy in a Patient with Incontinentia Pigmenti and Autoantibodies against Type I IFNs Infected with SARS-CoV-2. J. Clin. Immunol. 2021, 41, 931–933.
  46. Middeldorp, S.; Coppens, M.; van Haaps, T.F.; Foppen, M.; Vlaar, A.P.; Muller, M.C.A.; Bouman, C.C.S.; Beenen, L.F.M.; Kootte, R.S.; Heijmans, J.; et al. Incidence of venous thromboembolism in hospitalized patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1995–2002.
  47. Sette, A.; Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021, 184, 861–880.
  48. Gudbjartsson, D.F.; Norddahl, G.L.; Melsted, P.; Gunnarsdottir, K.; Holm, H.; Eythorsson, E.; Arnthorsson, A.O.; Helgason, D.; Bjarnadottir, K.; Ingvarsson, R.F.; et al. Humoral Immune Response to SARS-CoV-2 in Iceland. N. Engl. J. Med. 2020, 383, 1724–1734.
  49. Wajnberg, A.; Amanat, F.; Firpo, A.; Altman, D.R.; Bailey, M.J.; Mansour, M.; McMahon, M.; Meade, P.; Mendu, D.R.; Muellers, K.; et al. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science 2020, 370, 1227–1230.
  50. 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, 181, 1489–1501.
  51. Tetro, J.A. Is COVID-19 receiving ADE from other coronaviruses? Microbes Infect. 2020, 22, 72–73.
  52. Wan, Y.; Shang, J.; Sun, S.; Tai, W.; Chen, J.; Geng, Q.; He, L.; Chen, Y.; Wu, J.; Shi, Z.; et al. Molecular Mechanism for Antibody-Dependent Enhancement of Coronavirus Entry. J. Virol. 2020, 94, e02015-19.
  53. Cromer, D.; Juno, J.A.; Khoury, D.; Reynaldi, A.; Wheatley, A.K.; Kent, S.J.; Davenport, M.P. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat. Rev. Immunol. 2021, 21, 395–404.
  54. Smorenberg, A.; Peters, E.J.; van Daele, P.; Nossent, E.J.; Muller, M. How does SARS-CoV-2 targets the elderly patients? A review on potential mechanisms increasing disease severity. Eur. J. Intern. Med. 2021, 83, 1–5.
  55. Soiza, R.L.; Scicluna, C.; Thomson, E.C. Efficacy and safety of COVID-19 vaccines in older people. Age Ageing 2021, 50, 279–283.
  56. Takahashi, T.; Ellingson, M.K.; Wong, P.; Israelow, B.; Lucas, C.; Klein, J.; Silva, J.; Mao, T.; Oh, J.E.; Tokuyama, M.; et al. Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature 2020, 588, 315–320.
  57. Kowitdamrong, E.; Puthanakit, T.; Jantarabenjakul, W.; Prompetchara, E.; Suchartlikitwong, P.; Putcharoen, O.; Hirankarn, N. Antibody responses to SARS-CoV-2 in patients with differing severities of coronavirus disease 2019. PLoS ONE 2020, 15, e0240502.
  58. Weisberg, S.P.; Connors, T.J.; Zhu, Y.; Baldwin, M.R.; Lin, W.H.; Wontakal, S.; Szabo, P.A.; Wells, S.B.; Dogra, P.; Gray, J.; et al. Distinct antibody responses to SARS-CoV-2 in children and adults across the COVID-19 clinical spectrum. Nat. Immunol. 2021, 22, 25–31.
  59. Wang, X.; Guo, X.; Xin, Q.; Pan, Y.; Hu, Y.; Li, J.; Chu, Y.; Feng, Y.; Wang, Q. Neutralizing Antibody Responses to Severe Acute Respiratory Syndrome Coronavirus 2 in Coronavirus Disease 2019 Inpatients and Convalescent Patients. Clin. Infect. Dis. 2020, 71, 2688–2694.
  60. Pichler, D.; Baumgartner, M.; Kimpel, J.; Rossler, A.; Riepler, L.; Bates, K.; Fleischer, V.; von Laer, D.; Borena, W.; Wurzner, R. Marked increase in avidity of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) antibodies 7-8 months after infection is not diminished in old age. J. Infect. Dis. 2021, 224, 764–770.
  61. Huang, D.; Lian, X.; Song, F.; Ma, H.; Lian, Z.; Liang, Y.; Qin, T.; Chen, W.; Wang, S. Clinical features of severe patients infected with 2019 novel coronavirus: A systematic review and meta-analysis. Ann. Transl. Med. 2020, 8, 576.
  62. Smith, J.C.; Sausville, E.L.; Girish, V.; Yuan, M.L.; Vasudevan, A.; John, K.M.; Sheltzer, J.M. Cigarette Smoke Exposure and Inflammatory Signaling Increase the Expression of the SARS-CoV-2 Receptor ACE2 in the Respiratory Tract. Dev. Cell 2020, 53, 514–529.
  63. Lagunas-Rangel, F.A. Neutrophil-to-lymphocyte ratio and lymphocyte-to-C-reactive protein ratio in patients with severe coronavirus disease 2019 (COVID-19): A meta-analysis. J. Med. Virol. 2020, 92, 1733–1734.
  64. Yilmaz, H.; Ucan, B.; Sayki, M.; Unsal, I.; Sahin, M.; Ozbek, M.; Delibasi, T. Usefulness of the neutrophil-to-lymphocyte ratio to prediction of type 2 diabetes mellitus in morbid obesity. Diabetes Metab. Syndr. 2015, 9, 299–304.
  65. Molony, R.D.; Nguyen, J.T.; Kong, Y.; Montgomery, R.R.; Shaw, A.C.; Iwasaki, A. Aging impairs both primary and secondary RIG-I signaling for interferon induction in human monocytes. Sci. Signal. 2017, 10.
  66. McGonagle, D.; Sharif, K.; O’Regan, A.; Bridgewood, C. The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun. Rev. 2020, 19, 102537.
  67. Kang, R.; Chen, R.; Zhang, Q.; Hou, W.; Wu, S.; Cao, L.; Huang, J.; Yu, Y.; Fan, X.G.; Yan, Z.; et al. HMGB1 in health and disease. Mol. Aspects Med. 2014, 40, 1–116.
  68. Brodin, P. Why is COVID-19 so mild in children? Acta Paediatr. 2020, 109, 1082–1083.
  69. Kollmann, T.R.; Levy, O.; Montgomery, R.R.; Goriely, S. Innate immune function by Toll-like receptors: Distinct responses in newborns and the elderly. Immunity 2012, 37, 771–783.
  70. Saso, A.; Kampmann, B. Vaccine responses in newborns. Semin. Immunopathol. 2017, 39, 627–642.
  71. Torjesen, I. Covid-19: Middle aged women face greater risk of debilitating long term symptoms. BMJ 2021, 372, n829.
  72. Berghofer, B.; Frommer, T.; Haley, G.; Fink, L.; Bein, G.; Hackstein, H. TLR7 ligands induce higher IFN-alpha production in females. J. Immunol. 2006, 177, 2088–2096.
  73. Klein, S.L.; Jedlicka, A.; Pekosz, A. The Xs and Y of immune responses to viral vaccines. Lancet Infect. Dis. 2010, 10, 338–349.
  74. Webb, K.; Peckham, H.; Radziszewska, A.; Menon, M.; Oliveri, P.; Simpson, F.; Deakin, C.T.; Lee, S.; Ciurtin, C.; Butler, G.; et al. Sex and Pubertal Differences in the Type 1 Interferon Pathway Associate With Both X Chromosome Number and Serum Sex Hormone Concentration. Front. Immunol. 2018, 9, 3167.
  75. Pido-Lopez, J.; Imami, N.; Aspinall, R. Both age and gender affect thymic output: More recent thymic migrants in females than males as they age. Clin. Exp. Immunol. 2001, 125, 409–413.
  76. Whittaker, E.; Bamford, A.; Kenny, J.; Kaforou, M.; Jones, C.E.; Shah, P.; Ramnarayan, P.; Fraisse, A.; Miller, O.; Davies, P.; et al. Clinical Characteristics of 58 Children With a Pediatric Inflammatory Multisystem Syndrome Temporally Associated With SARS-CoV-2. JAMA 2020, 324, 259–269.
  77. Pulendran, B.; Li, S.; Nakaya, H.I. Systems vaccinology. Immunity 2010, 33, 516–529.
  78. Gao, Y.; Chen, Y.; Liu, M.; Shi, S.; Tian, J. Impacts of immunosuppression and immunodeficiency on COVID-19: A systematic review and meta-analysis. J. Infect. 2020, 81, e93–e95.
  79. Fung, M.; Babik, J.M. COVID-19 in Immunocompromised Hosts: What We Know So Far. Clin. Infect. Dis. 2021, 72, 340–350.
  80. Robilotti, E.V.; Babady, N.E.; Mead, P.A.; Rolling, T.; Perez-Johnston, R.; Bernardes, M.; Bogler, Y.; Caldararo, M.; Figueroa, C.J.; Glickman, M.S.; et al. Determinants of COVID-19 disease severity in patients with cancer. Nat. Med. 2020, 26, 1218–1223.
  81. Challenor, S.; Tucker, D. SARS-CoV-2-induced remission of Hodgkin lymphoma. Br. J. Haematol. 2021, 192, 415.
  82. Buckner, T.W.; Dunphy, C.; Fedoriw, Y.D.; van Deventer, H.W.; Foster, M.C.; Richards, K.L.; Park, S.I. Complete spontaneous remission of diffuse large B-cell lymphoma of the maxillary sinus after concurrent infections. Clin. Lymphoma Myeloma Leuk 2012, 12, 455–458.
  83. Kahraman, S.; Akinci, M.B.; Sendur, M.A.N.; Yalcin, B. Can the host immune response against SARS-CoV2 also cause an anticancer effect? Med. Oncol. 2021, 38, 90.
  84. Sonani, B.; Aslam, F.; Goyal, A.; Patel, J.; Bansal, P. COVID-19 vaccination in immunocompromised patients. Clin. Rheumatol. 2021, 40, 797–798.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 408
Revisions: 2 times (View History)
Update Date: 30 Nov 2021
1000/1000
Video Production Service