Next Article in Journal
Manganese Ferrite Nanoparticles Enhance the Sensitivity of Hepa1-6 Hepatocellular Carcinoma to Radiation by Remodeling Tumor Microenvironments
Next Article in Special Issue
Intracellular Redox-Modulated Pathways as Targets for Effective Approaches in the Treatment of Viral Infection
Previous Article in Journal
Impact of Different Physical Exercises on the Expression of Autophagy Markers in Mice
Previous Article in Special Issue
Epstein–Barr Virus-Positive Mucocutaneous Ulcer: A Unique and Curious Disease Entity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Immunogenetics in COVID-19

1
Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, 90134 Palermo, Italy
2
Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, 70124 Bari, Italy
3
Institute of Occupational, Social and Environmental Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally.
Int. J. Mol. Sci. 2021, 22(5), 2636; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052636
Submission received: 5 January 2021 / Revised: 26 February 2021 / Accepted: 3 March 2021 / Published: 5 March 2021

Abstract

:
Coronavirus disease 2019 (COVID-19) is induced by SARS-CoV-2 and may arise as a variety of clinical manifestations, ranging from an asymptomatic condition to a life-threatening disease associated with cytokine storm, multiorgan and respiratory failure. The molecular mechanism behind such variability is still under investigation. Several pieces of experimental evidence suggest that genetic variants influencing the onset, maintenance and resolution of the immune response may be fundamental in predicting the evolution of the disease. The identification of genetic variants behind immune system reactivity and function in COVID-19 may help in the elaboration of personalized therapeutic strategies. In the frenetic look for universally shared treatment plans, those genetic variants that are common to other diseases/models may also help in addressing future research in terms of drug repurposing. In this paper, we discuss the most recent updates about the role of immunogenetics in determining the susceptibility to and the history of SARS-CoV-2 infection. We propose a narrative review of available data, speculating about lessons that we have learnt from other viral infections and immunosenescence, and discussing what kind of aspects of research should be deepened in order to improve our knowledge of how host genetic variability impacts the outcome for COVID-19 patients.

1. Introduction

Since the emergence of the first documented cases in December 2019, it soon became evident that subjects infected by SARS-CoV-2 experience a disease promptly named coronavirus disease 2019 (COVID-19) with dramatic interindividual variability in its clinical manifestations. In fact, COVID-19 may arise as an asymptomatic state as well as a panel of respiratory conditions ranging from mild flu-like symptoms (fever, fatigue and dry cough) to a severe respiratory disease (including pneumonia and dyspnoea) that may require hospitalization and may be exacerbated by cytokine storm, acute respiratory distress syndrome (ARDS), multi-organ and respiratory failure, with potentially fatal consequences [1,2,3,4]. In contrast to the SARS-CoV (-1)-caused SARS pandemic (2002/2003), recorded as the first pandemic of the 21st century, the SARS-CoV-2-caused COVID-19 pandemic was not stopped by public health preventive measures in its first season 2019/20. Instead, it enforced rapid development of vaccines. This was never accomplished for SARS-1 [5,6].
Epidemiological data demonstrated that clinically relevant variables like sex, age and comorbidities, as well as viral genome variants were not enough in explaining why subjects with apparently similar clinical history and pre-existent status develop such a different group of symptoms [2,4]. Thus, other factors must contribute to the pathogenesis of the disease, determining which patients will develop an asymptomatic infection and how serious the disease will be in symptomatic subjects [4,7]. Defining these factors will be especially advantageous to detect patients who may benefit from an early hospitalization and therapeutic intervention [2,3]. The attention was immediately captivated by those host genetic variants ruling the expression and characteristics of viral receptors and host enzymes involved in viral entry [1,3,7,8,9,10,11,12,13]. However, this represents only one of the elements that must be considered in the complex interplay involving the virus, the host respiratory tract–lung tissues, and the host immune system [2,7,14].
Host immune responses and immune-related symptoms are extremely variable between patients who have effective control of SARS-CoV-2, i.e., asymptomatic, and patients who are unable to control the virus, i.e., affected by severe COVID-19. This suggests that host immune dysregulation contributes to pathogenesis in some cases. However, it is not known if the development of a severe form of the disease is ruled by ether immune hyperactivity or by a failure to resolve the inflammatory response due to the ongoing viral replication and immune dysregulation. The correlation among cytokine levels, nasopharyngeal viral load and declining viral load in moderate cases suggests that the immune response is associated with the viral burden [15,16].
However, the genetic basis of the immune response, the focus of immunogenetics, may account for a notable part of the (still) unexplained interindividual disease variability and may provide the key to stratifying patients on the basis of the risk of developing severe symptoms according to the presence/absence of genetic variants [17]. In line with this hypothesis, a number of studies are currently ongoing in order to explore the genetic characteristics of COVID-19 patients as regards those genes that are involved in the setting/maintenance/control/switching off of the immune response [14,18].

2. AB0 Groups

The AB0 molecules represent complex membrane antigens widely expressed both on the surface of red blood cells (RBC) and many other cells, extending the importance and the clinical significance of the AB0 system beyond transfusion medicine. Historically, the AB0 phenotype was one of the first markers involved in cancer susceptibility. Evidence has since then accumulated that AB0 blood antigens could play a key role in various human diseases, although the data are not clear [19,20]. On this basis, it is not surprising that several studies have studied the associations between COVID-19 and the AB0 system.
The suspicion that the AB0 locus should be involved with the increased risk of developing a severe form of the disease arose from the observation that: (i) the frequency of blood group 0 among COVID-19 patients was lower vs. control subjects, whereas the frequency of blood group A among COVID-19 patients was higher vs. control subjects; (ii) the risk of infection was lower in blood group 0 subjects vs. non-0 blood groups; on the contrary, blood group A was associated with a higher risk of infection vs. non-A blood groups (Table 1) [21,22,23,24,25,26,27,28].
However, controversies still exist about the association of blood groups with the severity of the disease, with some reports documenting that blood group A is associated with an increased risk of death vs. non-A blood groups and blood group AB is associated with an increased risk of intubation and death vs. blood group 0, while other reports failed in reproducing the same observations or even in demonstrating an association between any blood group and disease severity (expressed variably as hospitalization, intensive care unit—ICU—admission, intubation, required proning, extracorporeal membrane oxygenation—ECMO) (Table 1) [21,23,26,27,30,31]. Obviously, the adoption of universal criteria to define the severity of COVID-19 would be extremely beneficial in terms of proper stratification of patients and reproducibility of the results.
A genome wide association study (GWAS) performed by The Severe Covid-19 GWAS Group on Italian and Spanish subjects demonstrated that the association of severe COVID-19 expressed as respiratory failure with rs657152 (A or C single nucleotide polymorphism (SNP)) at locus 9q34.2 (coinciding with the AB0 locus) was significant at the genome wide level, even after correction for sex and age. Moreover, blood-group-specific analysis corrected for age and gender showed a higher risk of experiencing respiratory failure in blood group A than in other blood groups and a protective effect in blood group 0 in comparison with other blood groups, but no association was detected with disease severity expressed as the need for mechanical ventilation (Table 1) [28].
In a subsequent study, the authors reported that the rs657152 SNP is in almost complete linkage disequilibrium (LD; D’ = 0.996, r2 = 0.97) with rs8176719 (c.259-1_259insG), whose deletion is the main determinant of group 0, allele AB0*O.01.01. However, no differences in the genotype and allele frequencies were detected comparing age matched COVID-19 patients who required hospitalization with healthy controls and comparing severe with non-severe COVID-19 patients (with severity defined as the need of critical care support—high-flow oxygen, positive-pressure ventilation, vasoactive drugs) [31].
A further source of heterogeneity may be represented by the choice of the pool of the analyzed patients. In a cohort of Italian transplanted and waiting for organ transplantation patients, blood group A was more frequent in COVID-19+ (45.5%) than COVID-19- patients (39.0%) but no difference in blood group A distribution was observed comparing dead and alive patients (Table 1); however, the authors do not provide readers with further details about the cause of death—if it was determined by COVID-19 related complications or not [29].
About possible mechanistic considerations, it would be worth deepening the observation that serum levels of inflammatory marker soluble E-selectin are higher in 0/0 individuals, whereas a single nucleotide polymorphism in A1 allele is associated with low levels of this inflammatory marker. Furthermore, subjects homozygous for G allele in rs657152 SNP, corresponding to blood type 0 carriers, showed higher interleukin-6 (IL-6) circulating levels respect to non-0 carriers [19]. Moreover, ABH oligosaccharide structures have been identified on the N-linked oligo-saccharide chains of von Willebrand factor (VWF) located in the A1 domain, which contains the binding site for platelet glycoprotein Ib. The VWF levels are approximately 25% higher in individuals who have a blood group other than 0, and it might depend on endothelial A, B glycosyltransferase enzymes generating A and B antigens on the existing VWF “H” oligosaccharides. In turn, this addition to VWF might influence its blood level, and hence plasma levels of factor VIII and finally coagulation [19,20]. However, these literature data appear to conflict with the results of association studies, putting the accent on the fact that well-controlled inflammation is not “per se” a negative phenomenon, but rather the necessary response of the immune system to pathogenic viruses or bacteria [32]. Instead, the hyper inflammation response may be revealed to be dangerous (see below cytokine storm). This hypothesis is strengthened by the data of the high healing rates in the centenarians, known to have a good control of the inflammatory response [33].
Finally, the association could rely on the increase in natural antibodies influencing the history of SARS-CoV-2 infection [21,23,26,28,34,35].

3. HLA

The human leukocyte antigen (HLA) system codes for cell membrane proteins responsible for regulating the immune system. HLA genes are highly polymorphic; the different alleles are involved in fine-tuning the acquired immune responses. HLA classes have different functions. Class I (HLA-A, -B and -C) antigens present peptides from inside the cell on the cell surface. If the cell is infected by a virus, peptides of viral origin will be presented so that the cell can be lysed by CD8+ cytotoxic lymphocytes. Class II (HLA-DRA, HLA-DRB1, HLA-DRB3–5, HLA-DQA, HLA-DQB, HLA-DPA and HLA-DPB) molecules present peptides from outside the cell (which is an Antigen Presenting Cell) to CD4+ helper lymphocytes, which in turn stimulate B lymphocytes to produce antibodies against that specific antigen. Class III encodes the components of the complement system and the proinflammatory cytokine tumor necrosis factor (TNF-α). The high polymorphism of class I and II molecules affects the peptide binding groove, since it varies the amino acid sequences that can be housed within the groove. Different HLA alleles exhibit different peptide binding repertoires. Therefore, it is not surprising that different infectious diseases are associated with different HLA antigens, which are responsible for different humoral or cellular immune responses against different viral epitopes [17].
Furthermore, regulation of class II gene expression has been claimed to play a role in susceptibility/resistance to HLA-associated diseases [36]. In cancer, HLA class I gene expression was frequently down-regulated at both protein and mRNA levels and hypermethylation of the promoter regions of the HLA-A, -B and -C genes is a major mechanism of transcriptional inactivation [37]. This mechanism allowing evasion from the cytotoxic response has been shown to be present in cell line infection by the Epstein-Barr Virus (EBV) [38]. However, to the best of our knowledge no study has demonstrated this possibility for the immune responses to SARS-CoV-2 yet. The association between HLA alleles and COVID-19 infection and severity was assayed in a number of studies using laboratory, ecological, and in silico approaches. This kind of intense research was pushed by the mentioned fundamental role of the HLA in immune response setting and in susceptibility to infections [14,39,40,41,42], and also by the reports documenting that monocytes exhibit a decrease in HLA-DR expression inversely correlated with severity—the more HLA-DR decreases, the more the severity increases. Interpreting this piece of data in the light of other clinical and molecular characteristics of SARS-CoV-2 infected subjects, the authors concluded that HLA-DR levels may mirror the balance between the inflammatory and immunosuppressive status of COVID-19 patients [43,44].

3.1. Incidence and Susceptibility

Using an ecological approach, it was demonstrated that B*44 and C*01 alleles (whose prevalence is higher in northern Italy) showed a positive log-linear correlation with COVID-19 incidence rate measured in close proximity to the date of the national outbreak, keeping their positive association with COVID-19 incidence in multivariable regression analysis also adjusted for regions (Table 2).
On the contrary, HLA-B*14, B*18, and B*49 alleles (more frequent in southern Italy) showed an inverse log-linear correlation with COVID-19 incidence rate, but these associations lost their significance in the multiple regression model [39]. The authors underlined the fact that the product of HLA-C*01 allele represents the ligand of Natural Killer (NK) inhibitor receptors Killer Cell Immunoglobulin-Like Receptor (KIR) 2DL2 and KIR2DL3 [49,50], thus suggesting a possible role for HLA-C*01 carrying in establishing the entity of the immune response [39] (see below). In a Chinese study, HLA-B*15:27 and HLA-C*07:29 were more frequent in COVID-19 patients vs. control subjects, with the results staying significant after correction. However, the small sample size may hamper the proper interpretation of the results, as the authors warn (Table 2) [45]. An in silico study reported that among the compared alleles HLA-B*46:01 had the smallest predicted binding capacity for SARS-CoV-2 peptides and extremely low binding affinity measured for human coronavirus conserved peptides (defined as dissociation constant <500 nM), thus suggesting a possible role for HLA-B*46:01 in determining disease susceptibility [51,52], but this association was not confirmed by a following study on Hong Kong Chinese patients (Table 2) [46]. Furthermore, serotype HLA-B22 was found more frequently in COVID-19 Hong Kong Chinese patients vs. non-age matched controls but the association was not significant when the comparison was made against age matched controls [46]. Finally, it was demonstrated that countries where HLA-A*02:01 is the most frequent in the population had lower numbers of COVID-19 cases out of 106 population vs. countries where HLA-A*24:02 or HLA-A*11:01 are the most frequent alleles, as confirmed at two different time points (April 2020 p = 0.009 and August 2020 p = 0.013) [53].

3.2. Disease Severity

As regards the association of HLA alleles with disease severity, published papers report conflicting results, mainly due to the lack of shared criteria for disease severity classification, and the differences in the choice of compared groups of subjects. No significant correlation was found between HLA-B27 status and COVID-19 severity evaluated on an arbitrary scale ranging from mild to life-threatening disease by a team of researchers studying northern American patients (Table 2) [48]. These results were independently confirmed by a report on Hong Kong Chinese patients finding no significant association between HLA-B27 serotype and lymphopenia or disease severity expressed as mild (mild symptoms up to mild pneumonia) or severe (dyspnoea, hypoxia, or >50% lung involvement on imaging)/critical (respiratory failure, shock, or multi-organ system dysfunction) (Table 2) [46]. Instead, another study performed on a cohort of COVID-19 patients with severe (respiratory impairment, requiring non-invasive ventilation) and extremely severe (respiratory failure, requiring invasive ventilation and ICU admission) disease showed that the frequency of HLA-B*27:07, -DRB1*15:01, -DQB1*06:02 was higher in the studied COVID-19 patients vs. a reference group representing the local Italian population (Table 2) [41]. On the contrary, the assessment of allelic distribution at the HLA loci (Classes I and II) in a group of Italian and Spanish patients showed no association with the need for mechanical ventilation (Table 2) [28]. Comparing a group of mild (non-experimenting pneumonia) and severe (including severe and critically ill subjects according to the Chinese Center for Disease Control and Prevention -CDC- criteria) Chinese patients, HLA-C*14:02, -B*51:01, and -A*11:01 were associated with the severity of the disease in logistic regression analysis also adjusted for age and gender, with the three alleles in strong linkage disequilibrium, thus representing a haplotype. As regards Class II HLA alleles, -DRB1*14:04, -DRB1*01:01, -DQA1*01:01 represented severity risk alleles, whereas -DPB1*03:01 and -DRB1*12:01 were protective alleles (Table 2) [10]. A possible correlation between in silico predictions and observed symptoms was provided by Iturrieta-Zuazo et al. The authors demonstrated that mild or asymptomatic patients (mild group) exhibited a significant greater number of viral peptides, making a comparison with both hospitalized patients (moderate group) and subjects requiring ICU admission and supportive care (severe group) by HLA locus (HLA-A, -B, -C) and by HLA Class I genotype (for HLA Class I, p < 0.0001 estimated on the number of tightly (dissociation constant <50 nM) and loosely (dissociation constant < 500 nM) binding peptides, in both severe vs. mild and moderate vs. mild) [54].

3.3. Mortality

Regarding the association between HLA alleles and death rates, data are extremely varied. A Spanish study failed in detecting differences in HLA allele distribution comparing COVID-19 patients and healthy controls. However, the authors noticed that the Acute Physiology and Chronic Health Evaluation (APACHE)-II score, Sepsis-related Organ Failure Assessment (SOFA) score, and frequency of HLA-A*11, HLA-C*01 and HLA-DQB1*04 were higher in non-surviving vs. surviving patients. In their binomial logistic regression models, all the three alleles preserved their association with mortality after controlling for SOFA, and HLA-A*11 and HLA-C*01 kept their association after controlling for APACHE-II (Table 2) [40]. In Mexico, a statistically significant correlation was documented between HLA-DRB1*01:01 allele and the predicted fatality rate in hospitalized patients (R = −0.44, p = 0.02) [55]. Similarly, analyzing the HLA haplotype distribution in three Italian zones (northern, central and southern regions), the haplotype HLA-A*01:01g-B*08:01g-C*07:01g-DRB1*03:01g showed a positive correlation and the haplotype HLA-A*02:01g-B*18:01g-C*07:01g-DRB1*11:04g and a negative correlation with both COVID-19 incidence (number of cases/100,000 inhabitants) and mortality (number of deaths/100,000 inhabitants) at three out of four the analyzed timepoints (the first time point was reported to have no significant results because it was too premature) [56]. HLA-A*01:01, -B*08:01, -C*07:01,-DRB1*03:01, -DQA1*05:01 and -DQB1*02:01 is a common Caucasoid haplotype called 8.1 ancestral haplotype (AH). Intriguingly, numerous genetic studies reported that individuals with 8.1 AH have a higher risk of specific autoimmune disorders than those without these alleles [57]. It is worth nothing that bivariate correlation analysis among the regional frequencies of HLA-A*01:01g-B*08:01g-C*07:01g-DRB1*03:01g haplotype and incidence and mortality showed a cluster distribution, with the northern regions having the highest haplotype frequencies and the highest incidence and mortality, the central regions recording intermediate and the southern regions exhibiting the lowest values. The opposite frame was reported for the haplotype HLA-A*02:01g-B*18:01g-C*07:01g-DRB1*11:04g [56]. However, the alleles analyzed in this study HLA-A*02:01, HLA-B*08:01, HLA-A*01:01, HLA-B*18:01, HLA-C*07:01 are predicted to exhibit a good viral antigen presenting capacity, with HLA-A*02:01 offering the highest numbers of both tightly (dissociation constant < 50 nM, peptide number 267) and loosely (dissociation constant < 500 nM, peptide number 795) SARS-CoV-2 peptides [51,56], thus making the relationship between COVID-19 death rates and in silico estimated HLA antigen presenting capacity less clear. Curiously, in countries where HLA-A*02:01 is the most frequent in the population, a significant correlation exists with mortality (expressed as deaths out of 106 population) vs. countries where HLA-A*24:02 or HLA-A*11:01 are the most frequent alleles, as confirmed at two different time points (April 2020 p = 0.003 and August 2020 p < 0.001, respectively), excluding from further analysis that the number of recorded deaths accounts for the simple increase in COVID-19 cases regardless of the HLA genotype. The authors also predicted the functional performance of the three analyzed alleles, with HLA-A*02:01 having the lowest viral antigen-presenting capacity in comparison with HLA-A*24:02 and HLA-A*11:01, completely contradicting a previous paper reporting that the number of loosely+tightly binding peptides was higher for HLA-A*02:01 vs. both HLA-A*11:01 and HLA-A*24:02 [51,53,54].

3.4. Transplanted Patients

Transplanted patients, for whom HLA typing is speedily available, may represent an important source of information. In a cohort of Italian transplanted or waiting for organ transplantation patients, HLA-DRB1*08 was associated with a higher risk of infection and with a higher risk of death, with both observations keeping their significance in adjusted logistic regression analysis. Importantly, in an in silico prediction, none of the DRB1*08 alleles were able to bind with high affinity any of the viral peptides included in the simulation, raising questions about the functional performance of HLA-DRB1*08 in the case of SARS-CoV-2 infection (Table 1) [29]. Similarly, research performed in the UK showed that in a cohort of COVID-19+ transplanted (kidney or hematopoietic stem cells) and on the waiting list for solid organ transplantation patients, HLA-DQB1*06 was significantly associated with the risk of infection vs. a group of individuals on the renal transplant waiting list representing the local population (52.5% vs. 36%, respectively), as summarized in Table 2 [47].

3.5. Functional and Mechanistic Considerations

One report explored the association of HLA types with the immunophenotypic and functional characteristics of immune cells. HLA-A*02:01-restricted SARS-CoV-2-reactive CD8+ T cells can be detected at higher levels in COVID-19 patients vs. uninfected subjects, but their frequency is lower than that recorded for influenza or EBV-specific memory CD8+ T cells in SARS-CoV-2 uninfected subjects. In addition, in COVID-19 convalescent subjects these HLA-A*02:01-restricted SARS-CoV-2-reactive CD8+ T cells expressed granzymes and/or perforin, but were negative for CD38, HLA-DR, PD-1, and CD71 activation markers, while all these markers where positive in acute phase patients [58].
The lack of uniformity among the recalled reports could be easily explained. Indeed, several methodological problems can elucidate the discrepancies observed in studies concerning the association of HLA with diseases as follows: (I) insufficient sample sizes to detect differences in HLA antigen frequencies; (II) inadequate inclusion with inappropriate mixing of data (cohort effect) and inappropriate control matching, i.e., lack of proper selection from the same target population; (III) the different genetic backgrounds of the studied population; (IV) the lack of Bonferroni’s correction for multiple comparisons [59]. Differences in susceptibility/resistance to the same disease in populations can then be linked to the presence of different HLA subtypes in the various populations [60]. Moreover, caution should be adopted in the attempt to justify the observed correlation between HLA allele geographical distribution and COVID-19 incidence, prevalence and related mortality. In fact, in silico analysis of predicted HLA allele binding affinity must be validated in vitro together with the evaluation of the effect of the calculated binding affinity for each HLA allele on the increase in sustained immune responses and on cytokine synthesis control.
Concerning the meaning of the observed associations of HLA with COVID-19, it has to be pointed out that, in the last few years, it is becoming clear that Class I antigens can play a role as ligand for KIRs. Therefore, several observed associations of HLA class I antigen with COVID-19 might be explained by their role as ligands for KIR [61] (see above).
Finally, recent studies have investigated the role of nonclassical HLA class I HLA-E in the control of viral diseases. With only two alleles described, HLA-E shows a very low level of allelic variation. HLA-E is thus considered to play a role in both innate and adaptive immunity, by interacting with NK cells as well as presenting peptides to antigen-specific CD8+ T cells. The HLA-E alleles’ association with different viral infections seems to be discrepant. Each of the alleles can be advantageous for an individual or the entire population against a particular virus. Therefore, it has been assumed that the presence of both alleles in the gene pool can be beneficial for the survival of the population [62,63]. Thus, HLA-E should be an interesting new player in the field of immunology. However, to best of our knowledge no study has been performed on COVID-19 patients.

4. Other Immune Response Genes

COVID-19 severity and mortality may depend on attenuation or suppression of cytokines’ elicited pathways. In fact, SARS-CoV-2 blocks the proper synthesis of interferon-α (IFN-α) and interferon-β (IFN-β), thus impairing type I interferon signaling and leading to a suboptimal immune response [64,65]. The suppression of IFN-β synthesis is a mechanism shared with SARS-CoV, which exhibits a number of other ways to attenuate response to type I interferons, including interference with interferon receptor turnover or with the expression of interferon induced genes. However, these mechanisms have not been investigated deeply in SARS-CoV-2 infection yet [65]. Anyway, uncontrolled cytokine release and signaling may also cause life-threatening consequences.
Cytokine storm is an umbrella term encompassing a wide range of clinical and laboratory abnormalities. However, all cases involve elevated circulating cytokine levels, acute systemic inflammatory symptoms, and secondary organ dysfunction. A critical question concerns the factors that contribute to the severe cytokine storm-like phenotype observed in a small fraction of COVID-19 patients. Coexisting conditions such as hypertension, diabetes, and obesity are associated with more severe cases of COVID-19, possibly because of the pre-existing chronic inflammatory state or a lower threshold for the development of organ dysfunction from the immune response. Another prominent feature of COVID-19 severity is the association with advanced age [1,2,66,67]. Below the role of comorbidities, which tend to become more frequent during the aging process, a potential contributor to COVID-19 severity in older people may be represented by the subtle chronic inflammatory status called inflammaging (one of the three hallmarks of immunosenescence—an increase in serum cytokines impairing the effectivity of immune responses) and by the age-related progressive reduction in the ability to trigger effective antibody and cellular responses against infections and vaccinations [66,68,69,70,71]. Thus, immunosenescence represents a further potential contributor to the insurgence of fatal complications in COVID-19 [8,62,63]. Lymphopenia and accumulation of immune cells showing immunophenotypic and functional signs of exhaustion were commonly documented in COVID-19 patients, correlating with age and severity [43,68,72,73,74,75,76,77,78,79,80,81,82,83,84]. This strong resemblance to immunosenescence makes COVID-19 a possible scenario to deepen the knowledge about the molecular basis affecting normal immune cell metabolism/lifespan/turnover and to determine the importance of genetic variants and epigenetic modifications behind the appearance of inflammaging and immunosenescence [66,85,86,87,88]. Furthermore, the improvement of our understanding of the relationship between immunogenetics and immunosenescence (including inflammaging) would be crucial in the design of a therapeutic strategy specifically for older COVID-19 patients [66,68,69,89].
Several data suggest that cytokine storm may contribute to the pathogenesis of COVID-19. Serum cytokine levels that are elevated in patients with COVID-19-associated cytokine storm include IL-1β, IL-6, interferon γ-induced protein-10 (IP-10), TNF-α, interferon-γ (IFN-γ), macrophage inflammatory protein (MIP) -1α and -1β, and vascular endothelial growth factor (VEGF) [3,7,52,90,91,92,93].
Available data suggest that cytokine receptor antagonists might have a positive impact on survival of COVID-19 patients. IL-1 receptor antagonist Anakinra showed to be beneficial in terms of the need for mechanical ventilation and risk of death, despite larger studies being necessary to confirm if this agent is safe to be used alone or requires combination with other drugs such as glucocorticoids, and to estimate the exact entity of adverse effects like bacteremia. On the other hand, despite the association of high IL-6 levels with shorter survival and the evidence that laboratory findings of hyperinflammation (example, elevated values of C-reactive protein—CRP) and tissue damage predict worsening outcomes in COVID-19, data for anti–IL-6 receptor antibody therapies lack a uniform confirmation and/or are still insufficient, and are thus reported as conflicting in terms of mortality and hospitalization. This it is not surprising, since IL-1, IL-6 and other cytokines are potentially critical for both a healthy response to SARS-CoV-2 and a detrimental cytokine storm. Thus, completely blocking cytokine signaling might actually impair clearance of SARS-CoV-2, increase the risk of secondary infections, and lead to worse outcomes, as seen with influenza virus [16,94,95,96,97,98,99,100]. However, studies deeply dissecting the genetic basis of the insurgence of this phenomenon or the possible role of cytokine receptors in COVID-19 severity are still missing.

4.1. Type I Interferons and Players of Their Molecular Pathways

Synthesis and release of type I interferons together with their signaling by binding to IFN-α receptor 1 (IFNAR1) and IFNAR2 are all fundamental steps in the defense process raised by viral RNA or DNA. These molecular pathways involve a number of actors, including toll like receptors (TLR), IFN regulatory factors (IRF), signal transducers and activator of transcription (STAT) 1 and 2, TIR-domain containing adaptor inducing IFN-β (TICAM1/TRIF), TANK binding kinase 1 (TBK10), TNF Receptor Associated Factor 3 (TRAF3) and Unc-93 homolog B1 (UNC93B1) [101,102,103,104,105,106,107,108,109,110,111,112].
A notable effort was made to analyze 12 autosomal loci (STAT1 on chromosome 2, TLR 3 on chromosome 4, IRF7 and UNC93B1 on chromosome 11, TBK1 and STAT2 on chromosome 12, IRF9 and TRAF3 on chromosome 14, TICAM1/TRIF and IRF3 on chromosome 19, IFNAR1 and IFNAR2 on chromosome 21) and one X linked locus (NF-κB essential modulator—NEMO/IKBKG) in COVID-19 patients with life-threatening pneumonia vs. mild and asymptomatic COVID-19 cases (control group) [113,114]. The studied loci were connected to viral encephalitis, complications after measles, mumps, and rubella (MMR) vaccination and ARDS/critical influenza pneumonia [113,115,116]. As regards the 12 autosomal loci, a significant enrichment in what are predicted to be loss-of-function (pLOF) variants in patients vs. controls was detected under an autosomal-dominant (AD) mode of inheritance (Table 3).
Using overexpression systems, 24 variants (including the pLOF ones) in TLR3, UNC93B1, IRF7, IRF3, TICAM1/TRIF, TBK1, IFNAR1, IFNAR2 were demonstrated to be deleterious, since they were loss-of-expression, LOF or severely hypomorphic; of these 24 variants, four were autosomal-recessive (AR) deficiencies (homozygosity or compound heterozygosity for IRF7; homozygosity for IFNAR1) and 19 AD deficiencies (TLR3, TICAM1, TBK1, IRF3, UNC93B1, IRF7, IFNAR1, and IFNAR2). AR and AD IRF7-deficiency was associated with reduced levels of IRF7 expression on phytohemagglutinin (PHA) stimulated T cells. Plasmacytoid dendritic cells isolated from AR IRF7-deficient patients did not produce detectable type I or III IFNs on SARS-CoV-2 infection. Similarly, PHA stimulated T cells from a patient with AR IFNAR1 deficiency had impaired IFNAR1 expression and response to type I IFNs. Then, the authors checked the functional consequences of IRF7 and IFNAR1 defects on SARS-CoV-2 susceptibility in vitro, taking into account that angiotensin-converting enzyme 2 (ACE2) works as a receptor for SARS-CoV-2, and that transmembrane protease serine protease 2 (TMPRSS2) processes SARS-CoV-2 S protein and activates viral entry [2,89]. Thus, they noticed that TLR3−/−, TLR3+/−, IRF7−/−, and IFNAR1−/− fibroblasts previously transduced with ACE2 and TMPRSS2 were more susceptible to SARS-CoV-2 infection in vitro vs. cells from control subjects but were rescued by transduction of wild type IRF7 or IFNAR1. All these data suggest that genetic defects in the pathway of type I IFNs may be key determinant of COVID-19 severity [113].
Comforting these results, a recent study detected, validated and replicated the association between COVID-19 severity (i.e., admission to critical care) and rs2236757 (chr21q22.1) in the interferon receptor gene IFNAR2 and rs10735079 in the gene cluster coding for interferon-inducible 2’-5’-Oligoadenylate Synthetase (OAS) 1, 2, and 3 (chr12q24.13) comparing severe COVID-19 patients with controls from the population [117] (Table 3). OAS genes encodes for enzymes that synthetize 2′,5′-oligoadenylate, leading to RNase L activation and dsRNA degradation [124]. Exon 3 and 3′ untranslated region (UTR) variants in OAS1 are reported to confer increased susceptibility to and protection against SARS, respectively [115].
Interferon-induced transmembrane protein 3 (IFITM3) is an endosomal antiviral protein which is upregulated as a consequence of type I and type II interferon signaling as well as by cytokines like IL-6. The rs12252(C) allele in IFITM3 (chr. 11p15.5) was associated with influenza severity and mortality, and faster progression of HIV infection towards AIDS [125,126,127,128]. As regards COVID-19, the rs12252 (C) allele is considered a risk variant [120], especially as regards the homozygosity for the C allele, i.e., the (CC) genotype, in symptomatic cases [118,129]. As discussed in the previous sections, the adoption of uniform criteria to describe disease severity and the careful choice of the compared groups/populations are essential in order to obtain reproducible results. Comparing COVID-19 mild (fever, respiratory symptoms, and pneumonia at imaging) patients with severe (respiratory distress, blood oxygen saturation <93%, ratio of arterial oxygen pressure to fraction of inspired oxygen <300 mm Hg, respiratory failure with mechanical ventilation, shock, or other organ failure requiring intensive care in the intensive care unit) cases, an association between homozygosity for the C allele (CC vs. CT/TT) and disease severity was detected in logistic regression analysis adjusted for age (Table 3) [118]. Another study failed in demonstrating a significant association between carrying the C allele and disease severity intended as critical care support (including high-flow oxygen, positive-pressure ventilation or vasoactive drugs), but showed that carrying the C allele was associated with an increased risk of hospitalization comparing COVID-19 patients vs. controls—recruited before the pandemics—whose status about COVID-19 was unknown, with results keeping their significance in multiple logistic regression adjusted for age and sex (Table 3). No specific statistical analysis for the homozygosity for the C allele was carried [119].
Protein Activator of the Interferon-Induced Protein Kinase (PRKRA) together with IFN-induced, double-stranded RNA-activated protein kinase (PKR) is a long-time described mediator of interferon antiviral activity [130,131,132]. By whole exome sequencing and unbiased collapsing gene analysis, an Italian study identified PRKRA as one of the two protective genes (see below) comparing hospitalized COVID-19 patients with controls of unknown status. In this context, the deleterious variant was more frequent in COVID-19 patients than in controls [120].

4.2. Other Cytokines, Chemokines and Their Signaling Pathways

Data about promoter polymorphism -308 (G/A) (rs1800629) of TNF-α depict its involvement in the immune response set up and maintenance in different clinical scenarios, but the explanation of the related molecular mechanism is still pending. The minor allele A was described as associated with the risk of inflammatory disorders, sepsis, chronic obstructive pulmonary disease and asthma, whereas it confers protection against Dengue fever and has no effect on Kawasaki disease [133,134,135,136,137,138]. In addition, it exhibits a relation with increased serum TNF-α levels in asthma and not uniformly in cancer patients, but an analogous influence on transcription was not detected in other contexts like Alzheimer’s disease and generalized vitiligo [139,140,141,142,143]. A hospital-based case-control study comparing COVID-19 patients with controls described the frequency of −308 (G/A) TNF-α polymorphisms. The AA genotype and the A allele were more frequent in COVID-19 subjects vs. the control group (p = 0.019 and p = 0.005, respectively). Moreover, the AA genotype was associated with age > 60 years, increased degree lymphopenia, high CRP and serum ferritin (Table 3). The AA genotype was more frequent in severe (with any of the following: tachypnoea with a respiratory rate more than 30 cycle/min; PaO2 less than 300 mmHg; oxygen saturation below 93 at rest; shock; respiratory failure or other organ dysfunction) vs. mild cases (Table 3), with no GG genotype detected among severe patients. All dead subjects in this study carried the AA genotype [121].
Chemokine receptors are undoubtedly a keystone of immune cell trafficking; in particular, C-C Motif Chemokine Receptor 9 (CCR9) has a documented role in mediating immune cell localization in inflammatory disorders, C-X-C Motif Chemokine Receptor 6 (CXCR6) rules memory T cell recruiting in the airways, and X-C Motif Chemokine Receptor 1(XCR1), with its ligand X-C Motif Chemokine Ligand 1 (XCL1), has effects on the establishment of cytotoxic immune responses [144,145,146,147,148,149,150,151]. Thus, it would sound logical to suppose that any polymorphism impacting the transcription levels of these three receptors may affect the ability to mount an adequate immune response, also against airway pathogens. A GWAS revealed that the rs11385942 insertion–deletion GA or G variant at locus 3p21.31 was associated with COVID-19 related respiratory failure. The homozygosity for the risk allele GA was encountered in younger subjects vs. heterozygosity or homozygosity for the non-risk allele (59 years median age—interquartile range 49 to 68- vs. 66 years median age—interquartile range, 56 to 75; p = 0.005). The association locus included genes coding for chemokine receptors CCR9, CXCR6, and XCR1 (plus the genes SLC6A20, LZTFL1, and FYCO1), and the risk allele GA of rs11385942 accounted for a reduction in the expression of CXCR6. Moreover, the frequency of the risk allele was associated with a higher risk of receiving mechanical ventilation, also after correction for sex and age (Table 3) [28]. More studies are necessary to deepen these results, also establishing a causal relationship between expression of chemokine receptors and history of the SARS-CoV-2 infection. Supporting this urgency, an Italian study focused on hospitalized COVID-19 patients looking for variants involved in viral infection, susceptibility or protection by whole exome sequencing; the authors detected variant rs1799864 of C-C Motif Chemokine Receptor 2 (CCR2) in 8 out of 35 patients and rs1800940 of CCR5 (both associated with protection against HIV) in 1 out of 35 patients [120,152,153]. Furthermore, in lung tissues, transcriptome-wide association demonstrated that predicted low levels of CXCR6 together with low expression of CCCR3 and high expression of CCR2 are associated with severe COVID-19 (defined as admission to critical care) vs. controls [117,154].
There is a general paucity of data regarding interleukin signaling pathways; however, interesting information may be extrapolated by more general studies. Products of the neighboring transmembrane protein 189 (TMEM189) and ubiquitin-conjugating enzyme E2 variant 1 (UBE2V1) genes are involved in IL-1 pathway, with variants in UBE2V1 also associated with HIV-1 acquisition risk [155,156,157,158]. Using a genome wide association approach on COVID-19 patients divided into a mild (asymptomatic+mild+moderate subjects) group and a severe (severe+critically ill subjects) group according to Chinese CDC criteria, a report documented that A allele frequency of intronic rs6020298 in TMEM189–UBE2V1 (chr. 20q13.13) showed a significant association with both mild and severe conditions (Table 3) and a severity score estimated on the basis of age, gender and laboratory assessment (linear regression p = 1.1 × 10–6, β = 0.35). A similar association is shown by SNPs in linkage disequilibrium with rs6020298 (r2 > 0.8). The rs6020298 variant is an expression quantitative locus (eQTL) for TMEM189, with the A allele increasing TMEM189 expression [10].
A number of results are already available to address future research in this direction, in the form of papers awaiting the peer review process.
One preprint compares the frequency of rs1800796 and rs1800795 of IL-6, rs2228145 of IL-6R, rs1800896 and rs1800871 of IL-10, rs2275913 of IL-17A and rs76378 of IL-17F as recorded in previous studies with the prevalence/106 population and mortality/106 population of COVID-19 in China, Japan, India, Iran, Spain, Italy, Mexico, Netherlands, Sweden, Turkey, Finland, Brazil, Czechia, Russia, Poland, suggesting a possible role for rs2275913 of IL-17A in COVID-19 prevalence and mortality [159,160,161,162]. This piece of data is particular intriguing, assuming that the analyzed polymorphism is reported as associated with susceptibility to cutaneous and airway infections, ARDS and asthma [163,164,165,166,167,168,169].
Another preprint leading with supervariants involved in COVID-19 mortality focuses on the possible role of rs60811869, an eQTL of gene of the E3 ubiquitin ligase WD Repeat And SOCS Box Containing 1 (WSB1), in determining death rates among COVID-19 patients [170,171]. WSB1 is a particularly promising candidate, being involved in hormone homeostasis and hypoxia, as well as in the maturation of IL-21 receptor with IL-21 being a key player in humoral immunity and in the formation of the immune memory [172,173,174,175,176].

4.3. Other Genetic Determinants of Establishment/Maintenance/Resolution of the Immune Response and Antigen Presentation

Among dipeptidyl peptidase (DPP) family members, DPP9 on chromosome 19 seems to be implicated in macrophage differentiation and apoptosis [177,178]. In addition, it is involved in idiopathic pulmonary fibrosis [179]. Variant rs2109069 in the DPP9 gene is associated with disease severity in a cohort of UK patients (Table 3) [117]. Instead, DPP7 (9q34.3) is essential for the maintenance of quiescence in lymphocytes [180,181,182] and its expression is associated with influenza vaccination response [183]. Using a pedigree approach, a study demonstrated that a 1-bp insertion in DPP7 (rs11391519), disrupting gene transcription, was present in asymptomatic subjects [10].
Looking for monogenic effects determining COVID-19 severity, the same group reported the rs143359233 splice acceptor variant in golgin subfamily A 3 (GOLGA3, chr. 12q24.33) in critically ill patients (defined as subjects with one of the following conditions: respiratory failure and requesting mechanical ventilation, shock, failure of other organs and requesting intensive care monitoring) [10,184]. This is an interesting piece of data, since golgin family members are involved in nuclear transport, and another variant in GOLGA3 (rs12282) is associated with response to smallpox vaccine [185,186]. Intriguingly, this does not seem to be the only member of the golgin family involved in defining the history of SARS-CoV-2 infection. In fact, through the single variant association test to compare COVID-19 hospitalized cases and controls, missense variant rs200975425 in golgin A8 family member B (GOLGA8B in15q14) emerged as associated with infection susceptibility [10,187,188,189].
Resident macrophages engaged in the fight against SARS-CoV-2 may be influenced by genetic variants ruling polarization—thus tissue macrophages’ functions. Macrophage stimulating 1 receptor (MST1R in 3p21.31) is expressed on macrophages and involved in their polarization [190,191,192,193,194,195]. In unrelated samples Wang et al. identified a loss of function variants in MST1R (hg38 chr3:49896789_C/T) in asymptomatic and mild subjects comparing them with moderate, severe and critically ill patients (all categories defined according to the Chinese CDC criteria) [10].
Antigen presentation by HLA-DR may be a further potential pattern whose molecular defects may lead to an incomplete or insufficient response in COVID-19 patients. Expression of lysosomal localized Lysosomal Protein Transmembrane 4 β (LAPTM4B in 8q22.1), a lysosomal regulator of autophagy and an essential partner for endosomal transporters is finely regulated, since its overexpression causes enlargement of lysosomes [26,196,197,198,199]. LAPTM4B is the second gene identified by an Italian study for which deleterious variants were reported at higher frequencies in COVID-19 patients than in controls (Table 3) [120].
Other candidate genes may be involved in immune cell activation at various levels, as the pro-inflammatory allele ε4 (apoliprotein E in 19q13.32) ApoE gene is known to be involved in the pathophysiology of age-related inflammatory diseases as well as in susceptibility to infectious diseases [200,201]. In a logistic regression model adjusted also for age and sex, the ApoE ε4ε4 homozygous genotype was found to be associated with an increased risk of developing COVID-19 vs. ε3ε3 homozygotes (Table 3), preserving its statistical significance after the exclusion of participants related to the third degree or closer, and patients carrying ε4 with dementia, hypertension, cardiovascular disease, and type 2 diabetes. In this study, disease positivity for COVID-19 was interpreted as a synonym of severity, given that testing was restricted to hospitalized symptomatic patients [14,122]. The same authors later expanded their analysis and noticed that carrying ε4 represents a susceptibility factor (Table 3), but only the results for ε4ε4 genotype kept their statistical significance after the exclusion of patients carrying ε4 with dementia, hypertension, cardiovascular disease, and type 2 diabetes. Similarly, comparing COVID-19 patients who died with subjects from the UK biobank excluding COVID-19+ surviving subjects, the ApoE ε4ε4 homozygous genotype was found to be significantly associated with an increased risk of death vs. ε3ε3 homozygotes (Table 3) even after the exclusion of patients carrying e4 with dementia, hypertension, cardiovascular disease, and type 2 diabetes [123].
Figure 1 summarizes immunogenetic factors discussed in the present review [202,203,204,205,206,207,208,209,210,211,212,213].

5. Discussion

The continuously updated epidemiological and molecular data tell a story of a pandemic still lacking universally shared treatment protocols [2,214,215]. It is pretty evident that a number of factors contribute to determine SARS-CoV-2 infection susceptibility and COVID-19 severity (Figure 1), and that such a high number of new infections/hospitalized cases put extreme pressure on the global web of health care systems, creating the premises for the urgent need for a shared scheme to face the pandemic [2,214,216]. Together with public health measures to control the spread of the infection, stratifying patients according to the risk of developing a life-threatening disease could be a winning strategy to ensure the highest reachable survival rates while waiting for the completion of a global vaccination plan or the introduction of SARS-CoV-2 specific antiviral drugs [2,89,215]. The elaboration of an algorithm to calculate the probability of requiring hospitalization and to predict the efficacy and safety of early treatment/targeted treatment is an attractive perspective. Current efforts in this sense are focused on a number of predictors among clinical and laboratory evidence [1,217]. As demonstrated by experimental data, in this process the impact of host genetics, and—even better—immunogenetics should be surely included, together with gender related immunogenetic differences.
Although SARS-CoV-2 may infect people regardless of age, ethnic group or sex, male older subjects have been identified as a high-risk group regarding the clinical outcome of the disease, both for developing severe pneumonia with respiratory distress and death [1,2,67]. The reason behind the increased severity of the disease in the male older population is currently an unanswered question. A sex-specific GWAS performed in the UK showed no sex specific association [117]. However, it has been demonstrated that a more rapid aging of the immune system occurs in men than women. In fact, the strength and types of immune responses are different between men and women. Oestrogens promote while androgens suppress immune responses in the case of infections, vaccination and autoimmunity. On these grounds, men are more susceptible to many infections, while women suffer more from infectious diseases with enhanced immunopathological impact as well as from autoimmune diseases [71]. Peripheral blood mononuclear cells of men and women significantly differ after the age of 65, contradicting expectations related to declining sex hormones. Annotation of sex-biased loci reveal that older women have higher genomic activity for adaptive cells and older men have higher activity for monocytes and inflammation [218,219].
In the case of COVID-19, some authors proposed that polymorphism in ACE2 gene (located on chromosome X) coding and regulatory regions may partially explain such a sex-connected variability [11]. However, other factors may be involved, possibly including biochemical patterns ruling natural defenses against viruses (for example the sex related differences in H2S synthesis) as well as genes involved on immune responses located on chromosome X like CD40 ligand (CD40L), TLR7, TLR8, forkhead box p3 (FOXP3) and C-X-C Motif Chemokine Receptor 3 (CXCR3) [219,220,221,222,223,224,225]. Supporting this hypothesis, a study performed on two unrelated families (each one with a pair of male brothers with severe COVID-19) identified two variants in TLR7: (I) a pLOF variant NM_016562.3 c.2129_2132del, p.(Gln710Argfs*18) in the first family and (II) a missense variant NM_016562.3 c.2383G>T, p.(Val795Phe) in the second family. These variants significantly impaired the increase in TRL7 transcription, the upregulation of type I IFN-related genes interferon β 1 (IFNB1), IRF7 and ubiquitin-like modifier ISG15 (ISG15) in the TLR7 pathway, and the IFN-γ production after stimulation with imiquimod [226,227,228].
The sex effect may also be the result of an interplay between non-immune oestrogen/progesterone/androgen functions and other immune causes. Intriguingly, a significant association was described between blood type A and frequency of COVID-19 in the female subgroup (p = 0.02, OR = 1.56, 95% CI = 1.08–2.27) but not in the male subgroup of analyzed subjects (p = 0.51, OR = 1.14, 95%CI = 0.78–1.67) in a cohort of gender-stratified Chinese controls and COVID19+ patients [24].
In addition, the role of ethnic dependent differences in the frequency of variants in immunity ruling genes would be worth deeper investigation. In this sense, the choice of technical strategies to investigate the immunogenetic diversity is crucial in order to obtain the deepest pieces of information and to predict the course of the disease. GWAS is surely a helpful approach in the identification of those ethnic groups showing an increased susceptibility to SARS-CoV-2 infection/life threatening COVID-19. GWAS are frequently performed on nationally available pools of patients [28,117,171]. It would be highly recommendable that the impact of ethnicity related genetic diversity is always deeply investigated [107], even when the chosen patients appear to be homogeneous for their ethnic background [28,171]. All GWAS data should be confirmed on larger/cross national scales, stratifying the obtained results on the basis of ethnic differences of the studied populations, thus in terms of genetic background.
Future analysis should include genes that have not been studied yet, such as, for example, KIRs and immunoglobulin G heavy chain (GM). KIRs, expressed on the membrane of NK cells and a minority of T lymphocytes, regulate the killing function of these cells by interacting with specific amino acid motifs (public epitopes) carried by some HLA class I molecules and expressed on their targets. The KIR gene complex is characterized by multiple gene-content haplotypes, i.e., it is polygenic and some genes are polymorphic. KIRs are able to detect cells infected by viruses and transformed cells by binding the different class I allelic variants. Most inhibitory KIRs specifically recognize sets of HLA class I alleles. Specific combinations of KIRs with their cognate HLA ligands have been associated with onset and severity of infectious diseases [229,230,231]. This supports the role for HLA class I diversity in the innate immune response in addition to the documented acquired immune response. GM allotypes contribute to the inter-individual differences in the magnitude of immune responsiveness. GM allotypes have been shown to be associated with immune responsiveness to several major infectious pathogens and with survival of epidemics [231].
Furthermore, another type of immune response control also deserves in-depth studies in relation to COVID-19: the so-called trained immunity. Indeed, exposure to selected vaccines, such as bacille Calmette—Guérin (BCG) or microbial components, can increase the baseline tone of innate immunity and trigger pathogen-agnostic antimicrobial resistance. Such epigenetics training is directly relevant to resistance against infectious diseases, including COVID-19 and several trials to determine whether BCG can help prevent or ameliorate COVID-19 are under way [232].

6. Conclusions

Experimental data underline the contribution of immunogenetics in determining the susceptibility to SARS-CoV-2 infection as well as severity and mortality of COVID-19. Some of the involved genes and loci have already been analyzed in literature for their involvement in other infectious processes (as happened for example for AB0 locus, HLA genes, sequences regulating the expression of/coding for cytokines and chemokines); others are less investigated and may open the space for mechanistic studies (for example, Golgin and DPP genes). The most evident conclusion is that available reports are not always reproducible, and no definitive conclusions may be inferred since a lack of uniformity hampers the elaboration of a definitive cause–effect relationship. This situation may depend on differences in the experimental settings and adopted criteria to stratify the analyzed population, as well as on other factors influencing the genetic background of the immune response, such as environment and lifestyle (also accounting for epigenetics modulation).
A huge comprehension of the impact of genetic/epigenetic variants on immune mechanisms would offer the chance to: (i) identify those molecular pathways that are impaired in the setting of the immune response against SARS-CoV-2, leading to a more severe disease and to a more difficult resolution of the infection; (ii) concentrate the efforts on the design of a personalized approach for those patients presenting variants associated with a notable risk of altered lymphocyte distribution, uncontrolled or suboptimal immune response and cytokine storm syndrome; (iii) detect molecular defects common to other diseases that may be considered as possible targets in terms of drug repurposing [10,12,89,233,234,235,236,237]. This specific pandemic situation requires a global coordinated approach to face the disease in a short time with the most limited loss of human lives. To reach such an ambitious and necessary goal, a priority will be the setting of shared criteria to stratify patients according to their clinical presentation (i.e., COVID-19 severity) and the prompt validation of in silico predictions, both of these representing a powerful tool to guarantee reproducibility of results and proper interpretation of epidemiological and experimental data.

Author Contributions

Conceptualization, A.A., G.A. and C.C.; writing—original draft preparation, F.P.; writing—review and editing, A.A., G.A., C.C. and D.A.G.; all authors provided suggestions and comments on the manuscript. F.P. generated figure. All authors have read and agreed to the published version of the manuscript.

Funding

“This research received no external funding”. Original work performed by G.C., C.C., G.A., A.A. is funded by Improved Vaccination Strategies for Older Adults granted by European Commission (Horizon 2020 ID 848166).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACE2Angiotensin-converting enzyme 2
ADAutosomal-dominant
AHAncestral haplotype
AIDSAcquired immunodeficiency syndrome
APACHEAcute Physiology and Chronic Health Evaluation
ApoEApoliprotein E
ARAutosomal-recessive
ARDSAcute respiratory distress syndrome
CD40LCD40 ligand
CDCCenters for Disease Control and Prevention
CCR2C-C Motif Chemokine Receptor 2
CCR3C-C Motif Chemokine Receptor 3
CCR5C-C Motif Chemokine Receptor 5
CCR9C-C Motif Chemokine Receptor 9
COVID-19Coronavirus disease 19
CRPC-reactive protein
CXCR3C-X-C Motif Chemokine Receptor 3
CXCR6C-X-C Motif Chemokine Receptor 6
DPPDipeptidyl peptidase
EBVEpstein-Barr Virus
ECMOExtracorporeal membrane oxygenation
eQTLExpression quantitative locus
FOXP3Forkhead box p3
GMImmunoglobulin G heavy chain
GOLGA3Golgin subfamily A 3
GOLGA8BGolgin A8 family member B
GWASGenome wide association study
HIVHuman immunodeficiency virus
HLAHuman Leukocyte Antigens
ICUIntensive care unit
IFITM3Interferon-induced transmembrane protein 3
IFNB1Interferon β 1
IFN-αInterferon-α
IFN-βInterferon-β
IFN-γInterferon-γ
IFNAR1IFN-α receptor 1
IFNAR2IFN-α receptor 2
IL-1βInterleukin-1β
IL-6Interleukin-6
IL-10Interleukin-10
IL-17AInterleukin-17A
IL-17FInterleukin-17F
IL-21Interleukin-21
IP-10Interferon γ-induced protein-10
IRFIFN regulatory factors
ISG15Ubiquitin-like modifier ISG15
KIRKiller Immunoglobulin Receptors
KIR2DL2Killer Cell Immunoglobulin-Like Receptor 2DL2
KIR2DL3Killer Cell Immunoglobulin-Like Receptor 2DL3
LAPTM4BLysosomal Protein Transmembrane 4 β
MIP-1αMacrophage inflammatory protein 1α
MIP-1βMacrophage inflammatory protein 1β
MMRMeasles, mumps, and rubella
MST1RMacrophage stimulating 1 receptor
NEMO/IKBKGNF-κB essential modulator
NKNatural Killer
OAS2’-5’-Oligoadenylate synthetase
PHAPhytohemagglutinin
PKRIFN-induced, double-stranded RNA-activated protein kinase
pLOFPredicted to be loss-of-function
PRKRAProtein Activator of The Interferon-Induced Protein Kinase
RBCRed blood cells
SNPSingle nucleotide polymorphism
SOFASepsis-related Organ Failure Assessment
STAT1Signal transducer and activator of transcription 1
STAT2Signal transducer and activator of transcription 2
TBK1TANK binding kinase 1
TICAM1/TRIFTIR-domain containing adaptor inducing IFN-β
TLRToll like receptor
TMEM189Transmembrane protein 189
TMPRSS2Transmembrane protease serine protease 2
TNF-αTumor necrosis factor-α
TRAF3TNF Receptor Associated Factor 3
UBE2V1Ubiquitin-conjugating enzyme E2 variant 1
UNC93B1Unc-93 homolog B1
UTRUntranslated region
VEGFVascular endothelial growth factor
VWFvon Willebrand factor
WSB1WD Repeat And SOCS Box Containing 1
XCL1X-C Motif Chemokine Ligand 1
XCR1X-C Motif Chemokine Receptor 1

References

  1. Guo, G.; Ye, L.; Pan, K.; Chen, Y.; Xing, D.; Yan, K.; Chen, Z.; Ding, N.; Li, W.; Huang, H.; et al. New Insights of Emerging SARS-CoV-2: Epidemiology, Etiology, Clinical Features, Clinical Treatment, and Prevention. Front. Cell Dev. Biol. 2020, 410. PMCID: PMC7256189. [Google Scholar] [CrossRef] [PubMed]
  2. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2020, 1–14, PMCID: PMC7537588. [Google Scholar] [CrossRef] [PubMed]
  3. Hu, B.; Huang, S.; Yin, L. The cytokine storm and COVID-19. J. Med. Virol. 2020. PMCID: PMC7361342. [Google Scholar] [CrossRef] [PubMed]
  4. Tizaoui, K.; Zidi, I.; Lee, K.H.; Ghayda, R.A.; Hong, S.H.; Li, H.; Smith, L.; Koyanagi, A.; Jacob, L.; Kronbichler, A.; et al. Update of the current knowledge on genetics, evolution, immunopathogenesis, and transmission for coronavirus disease 19 (COVID-19). Int. J. Biol. Sci. 2020, 16, 2906–2923, PMCID: PMC7545713. [Google Scholar] [CrossRef] [PubMed]
  5. Groneberg, D.A.; Hilgenfeld, R.; Zabel, P. Molecular mechanisms of severe acute respiratory syndrome (SARS). Respir. Res. 2005, 6, 8, PMCID: PMC548145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Groneberg, D.A.; Poutanen, S.M.; Low, D.E.; Lode, H.; Welte, T.; Zabel, P. Treatment and vaccines for severe acute respiratory syndrome. Lancet Infect. Dis. 2005, 5, 147–155, PMCID:PMC7106466. [Google Scholar] [CrossRef] [PubMed]
  7. Mortaz, E.; Tabarsi, P.; Varahram, M.; Folkerts, G.; Adcock, I.M. The Immune Response and Immunopathology of COVID-19. Front. Immunol. 2020, 11, 2037, PMCID: PMC7479965. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, L.; Zheng, S. Understand variability of COVID-19 through population and tissue variations in expression of SARS-CoV-2 host genes. Inf. Med. Unlocked 2020, 21, 100443, PMCID: PMC7550072. [Google Scholar] [CrossRef] [PubMed]
  9. Srivastava, A.; Bandopadhyay, A.; Das, D.; Pandey, R.K.; Singh, V.; Khanam, N.; Srivastava, N.; Singh, P.P.; Dubey, P.K.; Pathak, A.; et al. Genetic Association of ACE2 rs2285666 Polymorphism With COVID-19 Spatial Distribution in India. Front. Genet. 2020, 11, 564741, PMCID: PMC7545580. [Google Scholar] [CrossRef] [PubMed]
  10. Wang, F.; Huang, S.; Gao, R.; Zhou, Y.; Lai, C.; Li, Z.; Xian, W.; Qian, X.; Li, Z.; Huang, Y.; et al. Initial whole-genome sequencing and analysis of the host genetic contribution to COVID-19 severity and susceptibility. Cell Discov. 2020, 6, 83. [Google Scholar] [CrossRef]
  11. Barash, A.; Machluf, Y.; Ariel, I.; Dekel, Y. The Pursuit of COVID-19 Biomarkers: Putting the Spotlight on ACE2 and TMPRSS2 Regulatory Sequences. Front. Med. 2020, 7, 582793, PMCID: PMC7661736. [Google Scholar] [CrossRef] [PubMed]
  12. Choudhary, S.; Sreenivasulu, K.; Mitra, P.; Misra, S.; Sharma, P. Role of Genetic Variants and Gene Expression in the Susceptibility and Severity of COVID-19. Ann. Lab. Med. 2021, 41, 129–138, PMCID: PMC7591285. [Google Scholar] [CrossRef] [PubMed]
  13. Chiappelli, F. CoViD-19 Susceptibility. Bioinformation 2020, 16, 501–504, PMCID: PMC7505245. [Google Scholar] [CrossRef] [PubMed]
  14. Anastassopoulou, C.; Gkizarioti, Z.; Patrinos, G.P.; Tsakris, A. Human genetic factors associated with susceptibility to SARS-CoV-2 infection and COVID-19 disease severity. Hum. Genom. 2020, 14, 40, PMCID: PMC7578581. [Google Scholar] [CrossRef] [PubMed]
  15. 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, PMCID: PMC7477538. [Google Scholar] [CrossRef] [PubMed]
  16. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef] [PubMed]
  17. Martin, M.P.; Carrington, M. Immunogenetics of viral infections. Curr. Opin. Immunol. 2005, 17, 510–516. [Google Scholar] [CrossRef] [PubMed]
  18. McCoy, K.; Peterson, A.; Tian, Y.; Sang, Y. Immunogenetic Association Underlying Severe COVID-19. Vaccines 2020, 8, 700. [Google Scholar] [CrossRef] [PubMed]
  19. Rizzo, C.; Caruso, C.; Vasto, S. Possible role of ABO system in age-related diseases and longevity: A narrative review. Immun. Ageing 2014, 11, 16, PMCID: PMC4265994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Vasto, S.; Caruso, C.; Castiglia, L.; Duro, G.; Monastero, R.; Rizzo, C. Blood group does not appear to affect longevity a pilot study in centenarians from Western Sicily. Biogerontology 2011, 12, 467–471. [Google Scholar] [CrossRef] [PubMed]
  21. Zhao, J.; Yang, Y.; Huang, H.; Li, D.; Gu, D.; Lu, X.; Zhang, Z.; Liu, L.; Liu, T.; Liu, Y.; et al. Relationship between the ABO Blood Group and the COVID-19 Susceptibility. Clin. Infect. Dis. 2020, ciaa1150, PMCID: PMC7454371. [Google Scholar] [CrossRef] [PubMed]
  22. Kaser, A. Genetic Risk of Severe Covid-19. N. Engl. J. Med. 2020, 383, 1590–1591, PMCID: PMC7583681. [Google Scholar] [CrossRef] [PubMed]
  23. Zietz, M.; Zucker, J.; Tatonetti, N.P. Associations between blood type and COVID-19 infection, intubation, and death. Nat. Commun. 2020, 11, 5761, PMCID: PMC7666188. [Google Scholar] [CrossRef] [PubMed]
  24. Fan, Q.; Zhang, W.; Li, B.; Li, D.J.; Zhang, J.; Zhao, F. Association between ABO Blood Group System and COVID-19 Susceptibility in Wuhan. Front. Cell Infect. Microbiol. 2020, 10, 404, PMCID: PMC7385064. [Google Scholar] [CrossRef] [PubMed]
  25. Muñiz-Diaz, E.; Llopis, J.; Parra, R.; Roig, I.; Ferrer, G.; Grifols, J.; Millán, A.; Ene, G.; Ramiro, L.; Maglio, L.; et al. Relationship between the ABO blood group and COVID-19 susceptibility, severity and mortality in two cohorts of patients. Blood Transfus. 2020. [Google Scholar] [CrossRef] [PubMed]
  26. Wu, B.B.; Gu, D.Z.; Yu, J.N.; Yang, J.; Shen, W.Q. Association between ABO blood groups and COVID-19 infection, severity and demise: A systematic review and meta-analysis. Infect. Genet. Evol. 2020, 84, 104485, PMCID: PMC7391292. [Google Scholar] [CrossRef] [PubMed]
  27. Barnkob, M.B.; Pottegård, A.; Støvring, H.; Haunstrup, T.M.; Homburg, K.; Larsen, R.; Hansen, M.B.; Titlestad, K.; Aagaard, B.; Møller, B.K.; et al. Reduced prevalence of SARS-CoV-2 infection in ABO blood group O. Blood Adv. 2020, 4, 4990–4993, PMCID: PMC7594382. [Google Scholar] [CrossRef] [PubMed]
  28. Severe Covid-19 GWAS Group; Ellinghaus, D.; Degenhardt, F.; Bujanda, L.; Buti, M.; Albillos, A.; Invernizzi, P.; Fernández, J.; Prati, D.; Baselli, G.; et al. Genomewide Association Study of Severe Covid-19 with Respiratory Failure. N. Engl. J. Med. 2020, 383, 1522–1534, PMCID: PMC7315890. [Google Scholar] [CrossRef]
  29. Amoroso, A.; Magistroni, P.; Vespasiano, F.; Bella, A.; Bellino, S.; Puoti, F.; Alizzi, S.; Vaisitti, T.; Boros, S.; Grossi, P.A.; et al. HLA and AB0 Polymorphisms May Influence SARS-CoV-2 Infection and COVID-19 Severity. Transplantation 2020. [Google Scholar] [CrossRef] [PubMed]
  30. Latz, C.A.; DeCarlo, C.; Boitano, L.; Png, C.Y.M.; Patell, R.; Conrad, M.F.; Eagleton, M.; Dua, A. Blood type and outcomes in patients with COVID-19. Ann. Hematol. 2020, 99, 2113–2118, PMCID: PMC7354354. [Google Scholar] [CrossRef] [PubMed]
  31. Coto, E.; Albaiceta, G.M.; Clemente, M.G.; Gómez, J. Lack of association between SNPsrs8176719(O blood group) and COVID-19: Data from Spanish age matched patients and controls. Transfusion 2020. [Google Scholar] [CrossRef] [PubMed]
  32. Licastro, F.; Candore, G.; Lio, D.; Porcellini, E.; Colonna-Romano, G.; Franceschi, C.; Caruso, C. Innate immunity and inflammation in ageing: A key for understanding age-related diseases. Immun. Ageing 2005, 2, 8, PMCID: PMC1166571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lio, D.; Scola, L.; Giarratana, R.M.; Candore, G.; Colonna-Romano, G.; Caruso, C.; Balistreri, C.R. SARS CoV2 infection _The longevity study perspectives. Ageing Res. Rev. 2021, 67, 101299, PMCID: PMC7885677. [Google Scholar] [CrossRef] [PubMed]
  34. Cooling, L. Blood Groups in Infection and Host Susceptibility. Clin. Microbiol. Rev. 2015, 28, 801–870, PMCID: PMC4475644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Liumbruno, G.M.; Franchini, M. Beyond immunohaematology: The role of the ABO blood group in human diseases. Blood Transfus. 2013, 11, 491–499, PMCID: PMC3827391. [Google Scholar] [CrossRef] [PubMed]
  36. Handunnetthi, L.; Ramagopalan, S.V.; Ebers, G.C.; Knight, J.C. Regulation of major histocompatibility complex class II gene expression, genetic variation and disease. Genes Immun. 2010, 11, 99–112, PMCID: PMC2987717. [Google Scholar] [CrossRef] [PubMed]
  37. Nie, Y.; Yang, G.-y.; Song, Y.; Zhao, X.; So, C.; Liao, J.; Wang, L.-D.; Yang, C.S. DNA hypermethylation is a mechanism for loss of expression of the HLA class I genes in human esophageal squamous cell carcinomas. Carcinogenesis 2001, 22, 1615–1623. [Google Scholar] [CrossRef] [Green Version]
  38. Singh, S.; Banerjee, S. Downregulation of HLA-ABC expression through promoter hypermethylation and downmodulation of MIC-A/B surface expression in LMP2A-positive epithelial carcinoma cell lines. Sci. Rep. 2020, 10, 5415. [Google Scholar] [CrossRef] [PubMed]
  39. Correale, P.; Mutti, L.; Pentimalli, F.; Baglio, G.; Saladino, R.E.; Sileri, P.; Giordano, A. HLA-B*44 and C*01 Prevalence Correlates with Covid19 Spreading across Italy. Int. J. Mol. Sci. 2020, 21, 5205, PMCID: PMC7432860. [Google Scholar] [CrossRef] [PubMed]
  40. Lorente, L.; Martín, M.M.; Franco, A.; Barrios, Y.; Cáceres, J.J.; Solé-Violán, J.; Perez, A.; Marcos, Y.; Ramos, J.A.; Ramos-Gómez, L.; et al. HLA genetic polymorphisms and prognosis of patients with COVID-19. Med. Intensiva. 2021, 45, 96–103, PMCID: PMC7474921. [Google Scholar] [CrossRef] [PubMed]
  41. Novelli, A.; Andreani, M.; Biancolella, M.; Liberatoscioli, L.; Passarelli, C.; Colona, V.L.; Rogliani, P.; Leonardis, F.; Campana, A.; Carsetti, R.; et al. HLA allele frequencies and susceptibility to COVID-19 in a group of 99 Italian patients. HLA 2020, 96, 610–614, PMCID: PMC7461491. [Google Scholar] [CrossRef] [PubMed]
  42. Kachuri, L.; Francis, S.S.; Morrison, M.; Wendt, G.A.; Bossé, Y.; Cavazos, T.B.; Rashkin, S.R.; Ziv, E.; Witte, J.S. The landscape of host genetic factors involved in infection to common viruses and SARS-CoV-2. medRxiv 2020, 12, 93, Update in Genome Med. 2020, 12, 93. PMCID: PMC7273301. [Google Scholar] [CrossRef]
  43. Benlyamani, I.; Venet, F.; Coudereau, R.; Gossez, M.; Monneret, G. Monocyte HLA-DR Measurement by Flow Cytometry in COVID-19 Patients: An Interim Review. Cytom. A 2020. [Google Scholar] [CrossRef] [PubMed]
  44. Zmijewski, J.W.; Pittet, J.F. Human Leukocyte Antigen-DR Deficiency and Immunosuppression-Related End-Organ Failure in SARS-CoV2 Infection. Anesth. Analg. 2020, 131, 989–992, PMCID: PMC7386673. [Google Scholar] [CrossRef]
  45. Wang, W.; Zhang, W.; Zhang, J.; He, J.; Zhu, F. Distribution of HLA allele frequencies in 82 Chinese individuals with coronavirus disease-2019 (COVID-19). HLA 2020, 96, 194–196, PMCID: PMC7276866. [Google Scholar] [CrossRef]
  46. Yung, Y.L.; Cheng, C.K.; Chan, H.Y.; Xia, J.T.; Lau, K.M.; Wong, R.S.M.; Wu, A.K.L.; Chu, R.W.; Wong, A.C.C.; Chow, E.Y.D.; et al. Association of HLA-B22 serotype with SARS-CoV-2 susceptibility in Hong Kong Chinese patients. HLA 2020. [Google Scholar] [CrossRef] [PubMed]
  47. Poulton, K.; Wright, P.; Hughes, P.; Savic, S.; Welberry Smith, M.; Guiver, M.; Morton, M.; van Dellen, D.; Tholouli, E.; Wynn, R.; et al. A role for human leucocyte antigens in the susceptibility to SARS-Cov-2 infection observed in transplant patients. Int. J. Immunogenet. 2020, 47, 324–328, PMCID: PMC7361549. [Google Scholar] [CrossRef]
  48. Rosenbaum, J.T.; Hamilton, H.; Weisman, M.H.; Reveille, J.D.; Winthrop, K.L.; Choi, D. The Effect of HLA-B27 on Susceptibility and Severity of COVID-19. J. Rheumatol. 2020. [Google Scholar] [CrossRef] [PubMed]
  49. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=KIR2DL2 (accessed on 26 February 2021).
  50. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=KIR2DL3&keywords=KIR2DL3 (accessed on 26 February 2021).
  51. Nguyen, A.; David, J.K.; Maden, S.K.; Wood, M.A.; Weeder, B.R.; Nellore, A.; Thompson, R.F. Human Leukocyte Antigen Susceptibility Map for Severe Acute Respiratory Syndrome Coronavirus 2. J. Virol. 2020, 94, e00510-20, PMCID: PMC7307149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Olwenyi, O.A.; Dyavar, S.R.; Acharya, A.; Podany, A.T.; Fletcher, C.V.; Ng, C.L.; Reid, S.P.; Byrareddy, S.N. Immuno-epidemiology and pathophysiology of coronavirus disease 2019 (COVID-19). J. Mol. Med. 2020, 98, 1369–1383, PMCID: PMC7431311. [Google Scholar] [CrossRef]
  53. Tomita, Y.; Ikeda, T.; Sato, R.; Sakagami, T. Association between HLA gene polymorphisms and mortality of COVID-19: An in silico analysis. Immun. Inflamm. Dis. 2020, 8, 684–694, PMCID: PMC7654404. [Google Scholar] [CrossRef]
  54. Iturrieta-Zuazo, I.; Rita, C.G.; García-Soidán, A.; de Malet Pintos-Fonseca, A.; Alonso-Alarcón, N.; Pariente-Rodríguez, R.; Tejeda-Velarde, A.; Serrano-Villar, S.; Castañer-Alabau, J.L.; Nieto-Gañán, I. Possible role of HLA class-I genotype in SARS-CoV-2 infection and progression: A pilot study in a cohort of Covid-19 Spanish patients. Clin. Immunol. 2020, 219, 108572, PMCID: PMC7428760. [Google Scholar] [CrossRef] [PubMed]
  55. Romero-López, J.P.; Carnalla-Cortés, M.; Pacheco-Olvera, D.L.; Ocampo-Godínez, J.M.; Oliva-Ramírez, J.; Moreno-Manjón, J.; Bernal-Alferes, B.; López-Olmedo, N.; García-Latorre, E.; Domínguez-López, M.L.; et al. A bioinformatic prediction of antigen presentation from SARS-CoV-2 spike protein revealed a theoretical correlation of HLA-DRB1*01 with COVID-19 fatality in Mexican population: An ecological approach. J. Med. Virol. 2020. PMCID: PMC7537233. [Google Scholar] [CrossRef]
  56. Pisanti, S.; Deelen, J.; Gallina, A.M.; Caputo, M.; Citro, M.; Abate, M.; Sacchi, N.; Vecchione, C.; Martinelli, R. Correlation of the two most frequent HLA haplotypes in the Italian population to the differential regional incidence of Covid-19. J. Transl. Med. 2020, 18, 352, PMCID: PMC7491019. [Google Scholar] [CrossRef] [PubMed]
  57. Gambino, C.M.; Aiello, A.; Accardi, G.; Caruso, C.; Candore, G. Autoimmune diseases and 8.1 ancestral haplotype: An update. HLA 2018, 92, 137–143. [Google Scholar] [CrossRef] [PubMed]
  58. Habel, J.R.; Nguyen, T.H.O.; van de Sandt, C.E.; Juno, J.A.; Chaurasia, P.; Wragg, K.; Koutsakos, M.; Hensen, L.; Jia, X.; Chua, B.; et al. Suboptimal SARS-CoV-2-specific CD8+ T cell response associated with the prominent HLA-A*02:01 phenotype. Proc. Natl. Acad. Sci. USA 2020, 117, 24384–24391, PMCID: PMC7533701. [Google Scholar] [CrossRef]
  59. Caruso, C.; Candore, G.; Romano, G.C.; Lio, D.; Bonafè, M.; Valensin, S.; Franceschi, C. Immunogenetics of longevity. Is major histocompatibility complex polymorphism relevant to the control of human longevity? A review of literature data. Mech. Ageing Dev. 2001, 122, 445–462. [Google Scholar] [CrossRef] [PubMed]
  60. Player, M.A.; Barracchini, K.C.; Simonis, T.B.; Rivoltini, L.; Arienti, F.; Castelli, C.; Mazzocchi, A.; Belli, F.; Parmiani, G.; Marincola, F.M. Differences in frequency distribution of HLA-A2 subtypes between North American and Italian white melanoma patients: Relevance for epitope specific vaccination protocols. J. Immunother. Emphas. Tumor Immunol. 1996, 19, 357–363. [Google Scholar] [CrossRef] [PubMed]
  61. Rizzo, C.; Accardi, G.; Caruso, C. Genetic variation in human leukocyte antigen and susceptibility to acute myeloid leukemia. Acta Haematol. 2015, 133, 162–163. [Google Scholar] [CrossRef] [PubMed]
  62. Joosten, S.A.; Sullivan, L.C.; Ottenhoff, T.H. Characteristics of HLA-E Restricted T-Cell Responses and Their Role in Infectious Diseases. J. Immunol. Res. 2016, 2016, 2695396, PMCID: PMC5028793. [Google Scholar] [CrossRef] [Green Version]
  63. Kanevskiy, L.; Erokhina, S.; Kobyzeva, P.; Streltsova, M.; Sapozhnikov, A.; Kovalenko, E. Dimorphism of HLA-E and its Disease Association. Int. J. Mol. Sci. 2019, 20, 5496, PMCID: PMC6862560. [Google Scholar] [CrossRef] [Green Version]
  64. Lopez, L.; Sang, P.C.; Tian, Y.; Sang, Y. Dysregulated Interferon Response Underlying Severe COVID-19. Viruses 2020, 12, 1433. [Google Scholar] [CrossRef] [PubMed]
  65. Schreiber, G. The Role of Type I Interferons in the Pathogenesis and Treatment of COVID-19. Front. Immunol. 2020, 11, 595739, PMCID: PMC7561359. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, Y.; Klein, S.L.; Garibaldi, B.T.; Li, H.; Wu, C.; Osevala, N.M.; Li, T.; Margolick, J.B.; Pawelec, G.; Leng, S.X. Aging in COVID-19: Vulnerability, immunity and intervention. Ageing Res. Rev. 2020, 65, 101205, PMCID: PMC7604159. [Google Scholar] [CrossRef] [PubMed]
  67. Angioni, R.; Sánchez-Rodríguez, R.; Munari, F.; Bertoldi, N.; Arcidiacono, D.; Cavinato, S.; Marturano, D.; Zaramella, A.; Realdon, S.; Cattelan, A.; et al. Age- severity matched cytokine profiling reveals specific signatures in Covid-19 patients. Cell Death Dis. 2020, 11, 957, PMCID: PMC7646225. [Google Scholar] [CrossRef] [PubMed]
  68. Hazeldine, J.; Lord, J.M. Immunesenescence: A Predisposing Risk Factor for the Development of COVID-19? Front. Immunol. 2020, 11, 573662, PMCID: PMC7573102. [Google Scholar] [CrossRef]
  69. Pietrobon, A.J.; Teixeira, F.M.E.; Sato, M.N. Immunosenescence and Inflammaging: Risk Factors of Severe COVID-19 in Older People. Front. Immunol. 2020, 11, 579220, PMCID: PMC7656138. [Google Scholar] [CrossRef]
  70. Minciullo, P.L.; Catalano, A.; Mandraffino, G.; Casciaro, M.; Crucitti, A.; Maltese, G.; Morabito, N.; Lasco, A.; Gangemi, S.; Basile, G. Inflammaging and Anti-Inflammaging: The Role of Cytokines in Extreme Longevity. Arch. Immunol. Ther. Exp. 2016, 64, 111–126. [Google Scholar] [CrossRef] [PubMed]
  71. Aiello, A.; Farzaneh, F.; Candore, G.; Caruso, C.; Davinelli, S.; Gambino, C.M.; Ligotti, M.E.; Zareian, N.; Accardi, G. Immunosenescence and Its Hallmarks: How to Oppose Aging Strategically? A Review of Potential Options for Therapeutic Intervention. Front. Immunol. 2019, 10, 2247, PMCID: PMC6773825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bouadma, L.; Wiedemann, A.; Patrier, J.; Surénaud, M.; Wicky, P.H.; Foucat, E.; Diehl, J.L.; Hejblum, B.P.; Sinnah, F.; de Montmollin, E.; et al. Immune Alterations in a Patient with SARS-CoV-2-Related Acute Respiratory Distress Syndrome. J. Clin. Immunol. 2020, 40, 1082–1092, PMCID: PMC7443154. [Google Scholar] [CrossRef] [PubMed]
  73. Jurado, A.; Martín, M.C.; Abad-Molina, C.; Orduña, A.; Martínez, A.; Ocaña, E.; Yarce, O.; Navas, A.M.; Trujillo, A.; Fernández, L.; et al. COVID-19: Age, Interleukin-6, C-reactive protein, and lymphocytes as key clues from a multicentre retrospective study. Immun Ageing. 2020, 17, 22, PMCID: PMC7426672. [Google Scholar] [CrossRef]
  74. Files, J.K.; Boppana, S.; Perez, M.D.; Sarkar, S.; Lowman, K.E.; Qin, K.; Sterrett, S.; Carlin, E.; Bansal, A.; Sabbaj, S.; et al. Sustained cellular immune dysregulation in individuals recovering from SARS-CoV-2 infection. J. Clin. Investig. 2020, 140491. [Google Scholar] [CrossRef] [PubMed]
  75. Maucourant, C.; Filipovic, I.; Ponzetta, A.; Aleman, S.; Cornillet, M.; Hertwig, L.; Strunz, B.; Lentini, A.; Reinius, B.; Brownlie, D.; et al. Natural killer cell immunotypes related to COVID-19 disease severity. Sci. Immunol. 2020, 5, eabd6832, PMCID: PMC7665314. [Google Scholar] [CrossRef]
  76. Akbari, H.; Tabrizi, R.; Lankarani, K.B.; Aria, H.; Vakili, S.; Asadian, F.; Noroozi, S.; Keshavarz, P.; Faramarz, S. The role of cytokine profile and lymphocyte subsets in the severity of coronavirus disease 2019 (COVID-19): A systematic review and meta-analysis. Life Sci. 2020, 258, 118167, PMCID: PMC7387997. [Google Scholar] [CrossRef] [PubMed]
  77. Cacciapuoti, S.; De Rosa, A.; Gelzo, M.; Megna, M.; Raia, M.; Pinchera, B.; Pontarelli, A.; Scotto, R.; Scala, E.; Scarano, F.; et al. Immunocytometric analysis of COVID patients: A contribution to personalized therapy? Life Sci. 2020, 261, 118355, PMCID: PMC7456265. [Google Scholar] [CrossRef]
  78. Cantenys-Molina, S.; Fernández-Cruz, E.; Francos, P.; Lopez Bernaldo de Quirós, J.C.; Muñoz, P.; Gil-Herrera, J. Lymphocyte subsets early predict mortality in a large series of hospitalized COVID-19 patients in Spain. Clin. Exp. Immunol. 2020. [Google Scholar] [CrossRef] [PubMed]
  79. Li, M.; Guo, W.; Dong, Y.; Wang, X.; Dai, D.; Liu, X.; Wu, Y.; Li, M.; Zhang, W.; Zhou, H.; et al. Elevated Exhaustion Levels of NK and CD8+ T Cells as Indicators for Progression and Prognosis of COVID-19 Disease. Front. Immunol. 2020, 11, 580237, PMCID: PMC7591707. [Google Scholar] [CrossRef]
  80. Liu, J.; Li, H.; Luo, M.; Liu, J.; Wu, L.; Lin, X.; Li, R.; Wang, Z.; Zhong, H.; Zheng, W.; et al. Lymphopenia predicted illness severity and recovery in patients with COVID-19: A single-center, retrospective study. PLoS ONE 2020, 15, e0241659, PMCID: PMC7673513. [Google Scholar] [CrossRef] [PubMed]
  81. Lu, Q.; Wang, Z.; Yin, Y.; Zhao, Y.; Tao, P.; Zhong, P. Association of Peripheral Lymphocyte and the Subset Levels With the Progression and Mortality of COVID-19: A Systematic Review and Meta-Analysis. Front. Med. 2020, 7, 558545, PMCID: PMC7546210. [Google Scholar] [CrossRef] [PubMed]
  82. Osman, M.; Faridi, R.M.; Sligl, W.; Shabani-Rad, M.T.; Dharmani-Khan, P.; Parker, A.; Kalra, A.; Tripathi, M.B.; Storek, J.; Cohen Tervaert, J.W.; et al. Impaired natural killer cell counts and cytolytic activity in patients with severe COVID-19. Blood Adv. 2020, 4, 5035–5039, PMCID: PMC7594380. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, W.; Li, L.; Liu, J.; Chen, L.; Zhou, F.; Jin, T.; Jiang, L.; Li, X.; Yang, M.; Wang, H. The characteristics and predictive role of lymphocyte subsets in COVID-19 patients. Int. J. Infect. Dis. 2020, 99, 92–99, PMCID: PMC7398035. [Google Scholar] [CrossRef]
  84. Zingaropoli, M.A.; Perri, V.; Pasculli, P.; Dezza, F.C.; Nijhawan, P.; Savelloni, G.; La Torre, G.; D’Agostino, C.; Mengoni, F.; Lichtner, M.; et al. Major reduction of NKT cells in patients with severe COVID-19 pneumonia. Clin. Immunol. 2020, 222, 108630, PMCID: PMC7661928. [Google Scholar] [CrossRef]
  85. Naumova, E.; Ivanova, M.; Pawelec, G. Immunogenetics of ageing. Int. J. Immunogenet. 2011, 38, 373–381. [Google Scholar] [CrossRef] [PubMed]
  86. Johnson, N.D.; Conneely, K.N. The role of DNA methylation and hydroxymethylation in immunosenescence. Ageing Res. Rev. 2019, 51, 11–23. [Google Scholar] [CrossRef] [PubMed]
  87. Keenan, C.R.; Allan, R.S. Epigenomic drivers of immune dysfunction in aging. Aging Cell 2019, 18, e12878, PMCID: PMC6351880. [Google Scholar] [CrossRef] [Green Version]
  88. Ostan, R.; Bucci, L.; Capri, M.; Salvioli, S.; Scurti, M.; Pini, E.; Monti, D.; Franceschi, C. Immunosenescence and immunogenetics of human longevity. Neuroimmunomodulation 2008, 15, 224–240. [Google Scholar] [CrossRef] [PubMed]
  89. Atlante, S.; Mongelli, A.; Barbi, V.; Martelli, F.; Farsetti, A.; Gaetano, C. The epigenetic implication in coronavirus infection and therapy. Clin. Epigenet. 2020, 12, 156, PMCID: PMC7576975. [Google Scholar] [CrossRef] [PubMed]
  90. Sugiyama, M.; Kinoshita, N.; Ide, S.; Nomoto, H.; Nakamoto, T.; Saito, S.; Ishikane, M.; Kutsuna, S.; Hayakawa, K.; Hashimoto, M.; et al. Serum CCL17 level becomes a predictive marker to distinguish between mild/moderate and severe/critical disease in patients with COVID-19. Gene 2021, 766, 145145, PMCID: PMC7489253. [Google Scholar] [CrossRef] [PubMed]
  91. Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages. Nat. Rev. Immunol. 2020, 20, 355–362, Erratum in 2020, 20, 448, doi:10.1038/s41577-020-0353-y. PMCID: PMC7201395. [Google Scholar] [CrossRef] [PubMed]
  92. Rahmati, M.; Moosavi, M. Cytokine-targeted therapy in severely ill COVID-19 patients: Options and cautions. EJMO 2020, 4, 179–181. [Google Scholar] [CrossRef]
  93. Neumann, J.; Prezzemolo, T.; Vanderbeke, L.; Roca, C.P.; Gerbaux, M.; Janssens, S.; Willemsen, M.; Burton, O.; Van Mol, P.; Van Herck, Y.; et al. Increased IL-10-producing regulatory T cells are characteristic of severe cases of COVID-19. Clin. Transl. Immunol. 2020, 9, e1204, PMCID: PMC7662088. [Google Scholar] [CrossRef]
  94. Darif, D.; Hammi, I.; Kihel, A.; El Idrissi Saik, I.; Guessous, F.; Akarid, K. The pro-inflammatory cytokines in COVID-19 pathogenesis: What goes wrong? Microb Pathog. 2021, 18, 104799, PMCID: PMC7889464. [Google Scholar] [CrossRef]
  95. Pontali, E.; Volpi, S.; Signori, A.; Antonucci, G.; Castellaneta, M.; Buzzi, D.; Montale, A.; Bustaffa, M.; Angelelli, A.; Caorsi, R.; et al. Efficacy of early anti-inflammatory treatment with high doses IV Anakinra with or without glucocorticoids in patients with severe COVID-19 pneumonia. J. Allergy Clin. Immunol. 2021. PMCID: PMC7865089. [Google Scholar] [CrossRef] [PubMed]
  96. Borie, R.; Savale, L.; Dossier, A.; Ghosn, J.; Taillé, C.; Visseaux, B.; Jebreen, K.; Diallo, A.; Tesmoingt, C.; Morer, L.; et al. Glucocorticoids with low-dose anti-IL1 anakinra rescue in severe non-ICU COVID-19 infection: A cohort study. PLoS ONE 2020, 15, e0243961, PMCID: PMC7743937. [Google Scholar] [CrossRef]
  97. Bozzi, G.; Mangioni, D.; Minoia, F.; Aliberti, S.; Grasselli, G.; Barbetta, L.; Castelli, V.; Palomba, E.; Alagna, L.; Lombardi, A.; et al. Anakinra combined with methylprednisolone in patients with severe COVID-19 pneumonia and hyperinflammation: An observational cohort study. J. Allergy Clin. Immunol. 2021, 147, 561–566.e4, PMCID: PMC7674131. [Google Scholar] [CrossRef] [PubMed]
  98. Khan, F.A.; Stewart, I.; Fabbri, L.; Moss, S.; Robinson, K.; Smyth, A.R.; Jenkins, G. Systematic review and meta-analysis of anakinra, sarilumab, siltuximab and tocilizumab for COVID-19. Thorax 2021. [Google Scholar] [CrossRef]
  99. The REMAP-CAP Investigators; Gordon, A.C.; Mouncey, P.R.; Al-Beidh, F.; Rowan, K.M.; Nichol, A.D.; Arabi, Y.M.; Annane, D.; Beane, A.; van Bentum-Puijk, W.; et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19—Preliminary report. medRxiv 2021. Available online: https://www.medrxiv.org/content/10.1101/2021.01.07.21249390v1 (accessed on 26 February 2021). [CrossRef]
  100. Lauder, S.N.; Jones, E.; Smart, K.; Bloom, A.; Williams, A.S.; Hindley, J.P.; Ondondo, B.; Taylor, P.R.; Clement, M.; Fielding, C.; et al. Interleukin-6 limits influenza-induced inflammation and protects against fatal lung pathology. Eur. J. Immunol. 2013, 43, 2613–2625, PMCID: PMC3886386. [Google Scholar] [CrossRef] [Green Version]
  101. Mazewski, C.; Perez, R.E.; Fish, E.N.; Platanias, L.C. Type I Interferon (IFN)-Regulated Activation of Canonical and Non-Canonical Signaling Pathways. Front. Immunol. 2020, 11, 606456, PMCID: PMC7719805. [Google Scholar] [CrossRef]
  102. Carty, M.; Guy, C.; Bowie, A.G. Detection of Viral Infections by Innate Immunity. Biochem. Pharmacol. 2020, 183, 114316. [Google Scholar] [CrossRef] [PubMed]
  103. Bourdon, M.; Manet, C.; Montagutelli, X. Host genetic susceptibility to viral infections: The role of type I interferon induction. Genes Immun. 2020, 1–15, PMCID: PMC7677911. [Google Scholar] [CrossRef]
  104. Lee, B.L.; Barton, G.M. Trafficking of endosomal Toll-like receptors. Trends Cell Biol. 2014, 24, 360–369, PMCID: PMC4037363. [Google Scholar] [CrossRef] [Green Version]
  105. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IFNAR1&keywords=IFNAR1 (accessed on 26 February 2021).
  106. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IFNAR2&keywords=IFNAR2 (accessed on 26 February 2021).
  107. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=STAT1&keywords=stat1 (accessed on 26 February 2021).
  108. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=STAT2&keywords=stat2 (accessed on 26 February 2021).
  109. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=TICAM1&keywords=TICAM1 (accessed on 26 February 2021).
  110. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=TBK1&keywords=TBK1 (accessed on 26 February 2021).
  111. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=TRAF3&keywords=traf3 (accessed on 26 February 2021).
  112. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=UNC93B1&keywords=UNC93B1 (accessed on 26 February 2021).
  113. 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, eabd4570. [Google Scholar] [CrossRef] [PubMed]
  114. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IKBKG&keywords=nemo (accessed on 26 February 2021).
  115. Elhabyan, A.; Elyaacoub, S.; Sanad, E.; Abukhadra, A.; Elhabyan, A.; Dinu, V. The role of host genetics in susceptibility to severe viral infections in humans and insights into host genetics of severe COVID-19: A systematic review. Virus Res. 2020, 289, 198163, PMCID: PMC7480444. [Google Scholar] [CrossRef] [PubMed]
  116. Di Maria, E.; Latini, A.; Borgiani, P.; Novelli, G. Genetic variants of the human host influencing the coronavirus-associated phenotypes (SARS, MERS and COVID-19): Rapid systematic review and field synopsis. Hum. Genom. 2020, 14, 30, PMCID: PMC7484929. [Google Scholar] [CrossRef]
  117. 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 2020. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, Y.; Qin, L.; Zhao, Y.; Zhang, P.; Xu, B.; Li, K.; Liang, L.; Zhang, C.; Dai, Y.; Feng, Y.; et al. Interferon-Induced Transmembrane Protein 3 Genetic Variant rs12252-C Associated With Disease Severity in Coronavirus Disease 2019. J. Infect. Dis. 2020, 222, 34–37, PMCID: PMC7197559. [Google Scholar] [CrossRef]
  119. Gómez, J.; Albaiceta, G.M.; Cuesta-Llavona, E.; García-Clemente, M.; López-Larrea, C.; Amado-Rodríguez, L.; López-Alonso, I.; Melón, S.; Alvarez-Argüelles, M.E.; Gil-Peña, H.; et al. The Interferon-induced transmembrane protein 3 gene (IFITM3) rs12252 C variant is associated with COVID-19. Cytokine 2020, 137, 155354. [Google Scholar] [CrossRef] [PubMed]
  120. Benetti, E.; Giliberti, A.; Emiliozzi, A.; Valentino, F.; Bergantini, L.; Fallerini, C.; Anedda, F.; Amitrano, S.; Conticini, E.; Tita, R.; et al. Clinical and molecular characterization of COVID-19 hospitalized patients. PLoS ONE 2020, 15, e0242534, PMCID: PMC7673557. [Google Scholar] [CrossRef] [PubMed]
  121. Saleh, A.; Sultan, A.; Elashry, M.A.; Farag, A.; Mortada, M.I.; Ghannam, M.A.; Saed, A.M.; Ghoneem, S. Association of TNF-α G-308 a Promoter Polymorphism with the Course and Outcome of COVID-19 Patients. Immunol. Investig. 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  122. Kuo, C.L.; Pilling, L.C.; Atkins, J.L.; Masoli, J.A.H.; Delgado, J.; Kuchel, G.A.; Melzer, D. APOE e4 Genotype Predicts Severe COVID-19 in the UK Biobank Community Cohort. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 2231–2232, PMCID: PMC7314139. [Google Scholar] [CrossRef] [PubMed]
  123. Kuo, C.L.; Pilling, L.C.; Atkins, J.L.; Masoli, J.A.H.; Delgado, J.; Kuchel, G.A.; Melzer, D. ApoE e4e4 Genotype and Mortality With COVID-19 in UK Biobank. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1801–1803, PMCID: PMC7337688. [Google Scholar] [CrossRef] [PubMed]
  124. Schwartz, S.L.; Conn, G.L. RNA regulation of the antiviral protein 2′-5′-oligoadenylate synthetase. Wiley Interdiscip. Rev. RNA 2019, 10, e1534, PMCID: PMC6585406. [Google Scholar] [CrossRef] [PubMed]
  125. Available online: https://www.omim.org/entry/605579?search=ifitm3&highlight=ifitm3 (accessed on 26 February 2021).
  126. Wellington, D.; Laurenson-Schafer, H.; Abdel-Haq, A.; Dong, T. IFITM3: How genetics influence influenza infection demographically. Biomed. J. 2019, 42, 19–26, PMCID: PMC6468115. [Google Scholar] [CrossRef]
  127. Lee, J.; Robinson, M.E.; Ma, N.; Artadji, D.; Ahmed, M.A.; Xiao, G.; Sadras, T.; Deb, G.; Winchester, J.; Cosgun, K.N.; et al. IFITM3 functions as a PIP3 scaffold to amplify PI3K signalling in B cells. Nature 2020, 588, 491–497. [Google Scholar] [CrossRef] [PubMed]
  128. Bedford, J.G.; O’Keeffe, M.; Reading, P.C.; Wakim, L.M. Rapid interferon independent expression of IFITM3 following T cell activation protects cells from influenza virus infection. PLoS ONE 2019, 14, e0210132, PMCID: PMC6334895. [Google Scholar] [CrossRef]
  129. Thevarajan, I.; Nguyen, T.H.O.; Koutsakos, M.; Druce, J.; Caly, L.; van de Sandt, C.E.; Jia, X.; Nicholson, S.; Catton, M.; Cowie, B.; et al. Breadth of concomitant immune responses prior to patient recovery: A case report of non-severe COVID-19. Nat. Med. 2020, 26, 453–455, PMCID: PMC7095036. [Google Scholar] [CrossRef] [Green Version]
  130. Online Mendelian Inheritance in Man, OMIM®. Available online: https://www.omim.org/entry/603424?search=PRKRA&highlight=prkra (accessed on 26 February 2021).
  131. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=PRKRA&keywords=PRKRA (accessed on 26 February 2021).
  132. Patel, R.C.; Sen, G.C. PACT, a protein activator of the interferon-induced protein kinase, PKR. EMBO J. 1998, 17, 4379–4390, PMCID: PMC1170771. [Google Scholar] [CrossRef]
  133. Al-Meghaiseeb, E.S.; Al-Robayan, A.A.; Al-Otaibi, M.M.; Arfin, M.; Al-Asmari, A.K. Association of tumor necrosis factor-α and -β gene polymorphisms in inflammatory bowel disease. J. Inflamm. Res. 2016, 9, 133–140, PMCID: PMC4918894. [Google Scholar] [CrossRef] [Green Version]
  134. Zhang, Y.; Cui, X.; Ning, L.; Wei, D. The effects of tumor necrosis factor-α (TNF-α) rs1800629 and rs361525 polymorphisms on sepsis risk. Oncotarget 2017, 8, 111456–111469, PMCID: PMC5762335. [Google Scholar] [CrossRef] [Green Version]
  135. Shi, C.; Zhao, H. Association between Tumor Necrosis Factor-308 G/A Polymorphism and Chronic Obstructive Pulmonary Disease Risk in Chinese Population: Evidence from a Meta-Analysis. Clin. Lab. 2019, 65. [Google Scholar] [CrossRef] [PubMed]
  136. Kang, S.W.; Kim, S.K.; Han, Y.R.; Hong, D.; Chon, J.; Chung, J.H.; Hong, S.J.; Park, M.S.; Ban, J.Y. Promoter Polymorphism (-308G/A) of Tumor Necrosis Factor-Alpha (TNF-α) Gene and Asthma Risk: An Updated Meta-Analysis. Genet. Test. Mol. Biomark. 2019, 23, 363–372. [Google Scholar] [CrossRef] [PubMed]
  137. Santos, N.C.D.; Gomes, T.N.; Góis, I.A.F.; Oliveira, J.S.; Coelho, L.F.L.; Ferreira, G.P.; Silva, F.R.P.D.; Pereira, A.C.T.D.C. Association of single nucleotide polymorphisms in TNF-α (-308G/A and -238G/A) to dengue: Case-control and meta-analysis study. Cytokine 2020, 134, 155183. [Google Scholar] [CrossRef] [PubMed]
  138. Ferdosian, F.; Dastgheib, S.A.; Hosseini-Jangjou, S.H.; Nafei, Z.; Lookzadeh, M.H.; Noorishadkam, M.; Mirjalili, S.R.; Neamatzadeh, H. Association of TNF-α rs1800629, CASP3 rs72689236 and FCGR2A rs1801274 Polymorphisms with Susceptibility to Kawasaki Disease: A Comprehensive Meta-Analysis. Fetal Pediatr. Pathol. 2019, 1–17. [Google Scholar] [CrossRef] [PubMed]
  139. Nasser, M.Z.; Ezzat, D.A. Association of-308G/A Polymorphism and Serum Level of TNF-α with Bronchial asthma in Children. Egypt. J. Immunol. 2018, 25, 117–124. [Google Scholar] [PubMed]
  140. Thriveni, K.; Raju, A.; Ramaswamy, G.; Krishnamurthy, S. Impact of gene polymorphism of TNF-α rs 1800629 and TNF-β rs 909253 on plasma levels of South Indian breast cancer patients. Indian J. Cancer. 2018, 55, 179–183. [Google Scholar] [CrossRef] [PubMed]
  141. Gutiérrez-Hurtado, I.A.; Puebla-Pérez, A.M.; Delgado-Saucedo, J.I.; Figuera, L.E.; Zúñiga-González, G.M.; Gomez-Mariscal, K.; Ronquillo-Carreón, C.A.; Gallegos-Arreola, M.P. Association between TNF-α-308G>A and -238G>A gene polymorphisms and TNF-α serum levels in Mexican colorectal cancer patients. Genet. Mol. Res. 2016, 15. [Google Scholar] [CrossRef] [PubMed]
  142. Culjak, M.; Perkovic, M.N.; Uzun, S.; Strac, D.S.; Erjavec, G.N.; Leko, M.B.; Simic, G.; Tudor, L.; Konjevod, M.; Kozumplik, O.; et al. The Association between TNF-alpha, IL-1 alpha and IL-10 with Alzheimer’s Disease. Curr. Alzheimer Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  143. Ahmed, R.; Sharif, D.; Jaf, M.; Amin, D.M. Effect of TNF-α -308G/A (rs1800629) Promoter Polymorphism on the Serum Level of TNF-α Among Iraqi Patients with Generalized Vitiligo. Clin. Cosmet. Investig. Dermatol. 2020, 13, 825–835, PMCID: PMC7671505. [Google Scholar] [CrossRef] [PubMed]
  144. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CCR9&keywords=ccr9 (accessed on 26 February 2021).
  145. Pathak, M.; Lal, G. The Regulatory Function of CCR9+ Dendritic Cells in Inflammation and Autoimmunity. Front. Immunol. 2020, 11, 536326, PMCID: PMC7566413. [Google Scholar] [CrossRef]
  146. Biswas, S.; Bryant, R.V.; Travis, S. Interfering with leukocyte trafficking in Crohn’s disease. Best Pract. Res. Clin. Gastroenterol. 2019, 38–39, 1016. [Google Scholar] [CrossRef] [PubMed]
  147. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CXCR6&keywords=cxcr6 (accessed on 26 February 2021).
  148. Wein, A.N.; McMaster, S.R.; Takamura, S.; Dunbar, P.R.; Cartwright, E.K.; Hayward, S.L.; McManus, D.T.; Shimaoka, T.; Ueha, S.; Tsukui, T.; et al. CXCR6 regulates localization of tissue-resident memory CD8 T cells to the airways. J. Exp. Med. 2019, 216, 2748–2762, PMCID: PMC6888981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=XCR1&keywords=XCR1 (accessed on 26 February 2021).
  150. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=XCL1&keywords=xcl1 (accessed on 26 February 2021).
  151. Lei, Y.; Takahama, Y. XCL1 and XCR1 in the immune system. Microbes Infect. 2012, 14, 262–267. [Google Scholar] [CrossRef] [PubMed]
  152. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CCR2&keywords=ccr2 (accessed on 26 February 2021).
  153. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CCR5&keywords=ccr5 (accessed on 26 February 2021).
  154. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CCR3&keywords=ccr3 (accessed on 26 February 2021).
  155. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=PEDS1-UBE2V1 (accessed on 26 February 2021).
  156. Online Mendelian Inheritance in Man, OMIM®. Available online: https://www.omim.org/entry/610994?search=tmem189&highlight=tmem189 (accessed on 26 February 2021).
  157. Online Mendelian Inheritance in Man, OMIM®. Available online: https://www.omim.org/entry/602995?search=ube2v1&highlight=ube2v1 (accessed on 26 February 2021).
  158. Mackelprang, R.D.; Bamshad, M.J.; Chong, J.X.; Hou, X.; Buckingham, K.J.; Shively, K.; deBruyn, G.; Mugo, N.R.; Mullins, J.I.; McElrath, M.J.; et al. Whole genome sequencing of extreme phenotypes identifies variants in CD101 and UBE2V1 associated with increased risk of sexually acquired HIV-1. PLoS Pathog. 2017, 13, e1006703, Erratum in 2019, 15, e1007588. PMCID: PMC5690691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IL10&keywords=il10 (accessed on 26 February 2021).
  160. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IL17A&keywords=il17a (accessed on 26 February 2021).
  161. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IL17F&keywords=il17f (accessed on 26 February 2021).
  162. Karcıoğlu Batur, L.; Hekim, N. Correlations of IL-6, IL-6R, IL-10 and IL-17 gene polymorphisms with the prevalence of COVID-2019 infection and its mortality rate. medRxiv 2020. Available online: https://www.researchsquare.com/article/rs-82662/v1 (accessed on 26 February 2021). [CrossRef]
  163. Gonçalves de Albuquerque, S.D.C.; da Costa Oliveira, C.N.; Vaitkevicius-Antão, V.; Silva, A.C.; Luna, C.F.; de Lorena, V.M.B.; de Paiva-Cavalcanti, M. Study of association of the rs2275913 IL-17A single nucleotide polymorphism and susceptibility to cutaneous leishmaniasis caused by Leishmania braziliensis. Cytokine 2019, 123, 154784. [Google Scholar] [CrossRef] [PubMed]
  164. Rolandelli, A.; Hernández Del Pino, R.E.; Pellegrini, J.M.; Tateosian, N.L.; Amiano, N.O.; de la Barrera, S.; Casco, N.; Gutiérrez, M.; Palmero, D.J.; García, V.E. The IL-17A rs2275913 single nucleotide polymorphism is associated with protection to tuberculosis but related to higher disease severity in Argentina. Sci. Rep. 2017, 7, 40666, PMCID: PMC5241634. [Google Scholar] [CrossRef] [Green Version]
  165. Yu, Z.G.; Wang, B.Z.; Li, J.; Ding, Z.L.; Wang, K. Association between interleukin-17 genetic polymorphisms and tuberculosis susceptibility: An updated meta-analysis. Int. J. Tuberc. Lung Dis. 2017, 21, 1307–1313. [Google Scholar] [CrossRef] [PubMed]
  166. Xie, M.; Cheng, B.; Ding, Y.; Wang, C.; Chen, J. Correlations of IL-17 and NF-κB gene polymorphisms with susceptibility and prognosis in acute respiratory distress syndrome in a chinese population. Biosci. Rep. 2019, 39, BSR20181987, PMCID: PMC6367126. [Google Scholar] [CrossRef] [Green Version]
  167. Zhai, C.; Li, S.; Feng, W.; Shi, W.; Wang, J.; Wang, Q.; Chai, L.; Zhang, Q.; Yan, X.; Li, M. Association of interleukin-17a rs2275913 gene polymorphism and asthma risk: A meta-analysis. Arch. Med. Sci. 2018, 14, 1204–1211, PMCID: PMC6209699. [Google Scholar] [CrossRef]
  168. Holster, A.; Teräsjärvi, J.; Lauhkonen, E.; Törmänen, S.; Helminen, M.; Koponen, P.; Korppi, M.; Peltola, V.; He, Q.; Nuolivirta, K. IL-17A gene polymorphism rs2275913 is associated with the development of asthma after bronchiolitis in infancy. Allergol. Int. 2018, 67, 109–113. [Google Scholar] [CrossRef] [PubMed]
  169. Du, J.; Han, J.C.; Zhang, Y.J.; Qi, G.B.; Li, H.B.; Zhang, Y.J.; Cai, S. Single-Nucleotide Polymorphisms of IL-17 Gene Are Associated with Asthma Susceptibility in an Asian Population. Med. Sci. Monit. 2016, 22, 780–787, PMCID: PMC4793684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=WSB1&keywords=wsb1v (accessed on 26 February 2021).
  171. Hu, J.; Li, C.; Wang, S.; Li, T.; Zhang, H. Genetic variants are identified to increase risk of COVID-19 related mortality from UK Biobank data. medRxiv 2020. Available online: https://www.medrxiv.org/content/10.1101/2020.11.05.20226761v1 (accessed on 26 February 2021). [CrossRef]
  172. Haque, M.; Kendal, J.K.; MacIsaac, R.M.; Demetrick, D.J. WSB1: From homeostasis to hypoxia. J. Biomed. Sci. 2016, 23, 61, PMCID: PMC4992216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=IL21R&keywords=il-21 (accessed on 26 February 2021).
  174. Nara, H.; Onoda, T.; Rahman, M.; Araki, A.; Juliana, F.M.; Tanaka, N.; Asao, H. WSB-1, a novel IL-21 receptor binding molecule, enhances the maturation of IL-21 receptor. Cell Immunol. 2011, 269, 54–59. [Google Scholar] [CrossRef] [PubMed]
  175. Tangye, S.G.; Ma, C.S. Regulation of the germinal center and humoral immunity by interleukin-21. J. Exp. Med. 2020, 217, e20191638, PMCID: PMC7037251. [Google Scholar] [CrossRef] [PubMed]
  176. Ren, H.M.; Lukacher, A.E. IL-21 in Homeostasis of Resident Memory and Exhausted CD8 T Cells during Persistent Infection. Int. J. Mol. Sci. 2020, 21, 6966, PMCID: PMC7554897. [Google Scholar] [CrossRef] [PubMed]
  177. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/608258 (accessed on 26 February 2021).
  178. Waumans, Y.; Baerts, L.; Kehoe, K.; Lambeir, A.M.; De Meester, I. The Dipeptidyl Peptidase Family, Prolyl Oligopeptidase, and Prolyl Carboxypeptidase in the Immune System and Inflammatory Disease, Including Atherosclerosis. Front. Immunol. 2015, 6, 387, PMCID: PMC4528296. [Google Scholar] [CrossRef] [Green Version]
  179. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=DPP9&keywords=dpp9 (accessed on 26 February 2021).
  180. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=DPP7&keywords=dpp7 (accessed on 26 February 2021).
  181. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/610537?search=dpp7&highlight=dpp7 (accessed on 26 February 2021).
  182. Mele, D.A.; Bista, P.; Baez, D.V.; Huber, B.T. Dipeptidyl peptidase 2 is an essential survival factor in the regulation of cell quiescence. Cell Cycle 2009, 8, 2425–2434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. HIPC-CHI Signatures Project Team; HIPC-I Consortium. Multicohort analysis reveals baseline transcriptional predictors of influenza vaccination responses. Sci Immunol. 2017, 2, eaal4656, PMCID: PMC5800877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Online Mendelian Inheritance in Man, OMIM®. Available online: https://www.omim.org/entry/602581?search=golga3&highlight=golga3 (accessed on 26 February 2021).
  185. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=GOLGA3&keywords=golga3 (accessed on 26 February 2021).
  186. Ovsyannikova, I.G.; Kennedy, R.B.; O’Byrne, M.; Jacobson, R.M.; Pankratz, V.S.; Poland, G.A. Genome-wide association study of antibody response to smallpox vaccine. Vaccine 2012, 30, 4182–4189, PMCID: PMC3367131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/609619?search=golga8b&highlight=golga8b (accessed on 26 February 2021).
  188. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=GOLGA8B&keywords=golga8b (accessed on 26 February 2021).
  189. Available online: https://www.iaf.urmc.rochester.edu/results?search=rs200975425; https://www.iaf.urmc.rochester.edu/about.html (accessed on 26 February 2021).
  190. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/600168?search=mst1r&highlight=mst1r (accessed on 26 February 2021).
  191. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=MST1R&keywords=MST1R (accessed on 26 February 2021).
  192. Chaudhuri, A. Regulation of Macrophage Polarization by RON Receptor Tyrosine Kinase Signaling. Front. Immunol. 2014, 5, 546, PMCID: PMC4215628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Wilson, C.B.; Ray, M.; Lutz, M.; Sharda, D.; Xu, J.; Hankey, P.A. The RON receptor tyrosine kinase regulates IFN-gamma production and responses in innate immunity. J. Immunol. 2008, 181, 2303–2310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Morrison, A.C.; Wilson, C.B.; Ray, M.; Correll, P.H. Macrophage-stimulating protein, the ligand for the stem cell-derived tyrosine kinase/RON receptor tyrosine kinase, inhibits IL-12 production by primary peritoneal macrophages stimulated with IFN-gamma and lipopolysaccharide. J. Immunol. 2004, 172, 1825–1832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Wang, M.H.; Zhou, Y.Q.; Chen, Y.Q. Macrophage-stimulating protein and RON receptor tyrosine kinase: Potential regulators of macrophage inflammatory activities. Scand. J. Immunol. 2002, 56, 545–553. [Google Scholar] [CrossRef] [PubMed]
  196. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/613296?search=laptm4b&highlight=laptm4b (accessed on 26 February 2021).
  197. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=LAPTM4B&keywords=LAPTM4B (accessed on 26 February 2021).
  198. Vergarajauregui, S.; Martina, J.A.; Puertollano, R. LAPTMs regulate lisosomal function and interact with mucolipin 1: New clues for understanding mucolipidosis type IV. J. Cell Sci. 2011, 124, 459–468, PMCID: PMC3022000. [Google Scholar] [CrossRef] [Green Version]
  199. Milkereit, R.; Persaud, A.; Vanoaica, L.; Guetg, A.; Verrey, F.; Rotin, D. LAPTM4b recruits the LAT1-4F2hc Leu transporter to lysosomes and promotes mTORC1 activation. Nat. Commun. 2015, 6, 7250, PMCID: PMC4455107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/107741?search=apoe&highlight=apoe (accessed on 26 February 2021).
  201. Mahley, R.W.; Weisgraber, K.H.; Huang, Y. Apolipoprotein E: Structure determines function, from atherosclerosis to Alzheimer’s disease to AIDS. J. Lipid Res. 2009, 50, S183–S188, PMCID: PMC2674716. [Google Scholar] [CrossRef] [Green Version]
  202. Kumar, N.; Janmohamed, K.; Nyhan, K.; Martins, S.S.; Cerda, M.; Hasin, D.; Scott, J.; Pates, R.; Ghandour, L.; Wazaify, M.; et al. Substance use and substance use disorder, in relation to COVID-19: Protocol for a scoping review. Syst. Rev. 2021, 10, 48, PMCID: PMC7857102. [Google Scholar] [CrossRef] [PubMed]
  203. Brickhill-Atkinson, M.; Hauck, F.R. Impact of COVID-19 on Resettled Refugees. Prim. Care 2021, 48, 57–66, PMCID: PMC7538065. [Google Scholar] [CrossRef]
  204. Fronteira, I.; Sidat, M.; Magalhães, J.P.; de Barros, F.P.C.; Delgado, A.P.; Correia, T.; Daniel-Ribeiro, C.T.; Ferrinho, P. The SARS-CoV-2 pandemic: A syndemic perspective. One Health 2021, 12, 100228, PMCID: PMC7887445. [Google Scholar] [CrossRef]
  205. Emeny, R.T.; Carpenter, D.O.; Lawrence, D.A. Health disparities: Intracellular consequences of social determinants of health. Toxicol. Appl. Pharmacol. 2021, 115444. [Google Scholar] [CrossRef] [PubMed]
  206. Bourdrel, T.; Annesi-Maesano, I.; Alahmad, B.; Maesano, C.N.; Bind, M.A. The impact of outdoor air pollution on COVID-19: A review of evidence from in vitro, animal, and human studies. Eur. Respir. Rev. 2021, 30, 200242, PMCID: PMC7879496. [Google Scholar] [CrossRef]
  207. Engin, A.B.; Engin, E.D.; Engin, A. The effect of environmental pollution on immune evasion checkpoints of SARS-CoV-2. Environ. Toxicol. Pharmacol. 2021, 81, 103520, PMCID: PMC7580701. [Google Scholar] [CrossRef]
  208. Suzuki, T.; Hidaka, T.; Kumagai, Y.; Yamamoto, M. Environmental pollutants and the immune response. Nat. Immunol. 2020, 21, 1486–1495. [Google Scholar] [CrossRef] [PubMed]
  209. Quinete, N.; Hauser-Davis, R.A. Drinking water pollutants may affect the immune system: Concerns regarding COVID-19 health effects. Environ. Sci. Pollut. Res. Int. 2021, 28, 1235–1246, PMCID: PMC7644792. [Google Scholar] [CrossRef] [PubMed]
  210. Zahra, A.; Sisu, C.; Silva, E.; De Aguiar Greca, S.C.; Randeva, H.S.; Chatha, K.; Kyrou, I.; Karteris, E. Is There a Link between Bisphenol A (BPA), a Key Endocrine Disruptor, and the Risk for SARS-CoV-2 Infection and Severe COVID-19? J. Clin. Med. 2020, 9, 3296, PMCID: PMC7602132. [Google Scholar] [CrossRef]
  211. Wang, P.; Liu, L.; Iketani, S.; Luo, Y.; Guo, Y.; Wang, M.; Yu, J.; Zhang, B.; Kwong, P.D.; Graham, B.S.; et al. Increased Resistance of SARS-CoV-2 Variants B.1.351 and B.1.1.7 to Antibody Neutralization. BioRxiv 2021. PMCID: PMC7852271. [Google Scholar] [CrossRef]
  212. Ortuso, F.; Mercatelli, D.; Guzzi, P.H.; Giorgi, F.M. Structural genetics of circulating variants affecting the SARS-CoV-2 spike/human ACE2 complex. J. Biomol. Struct. Dyn. 2021, 1–11, PMCID: PMC7885719. [Google Scholar] [CrossRef] [PubMed]
  213. Brookman, S.; Cook, J.; Zucherman, M.; Broughton, S.; Harman, K.; Gupta, A. Effect of the new SARS-CoV-2 variant B.1.1.7 on children and young people. Lancet Child. Adolesc. Health 2021. [Google Scholar] [CrossRef]
  214. Available online: https://covid19.who.int/ (accessed on 2 February 2021).
  215. Abduljalil, J.M.; Abduljalil, B.M. Epidemiology, genome, and clinical features of the pandemic SARS-CoV-2: A recent view. New Microbes New Infect. 2020, 35, 100672, PMCID: PMC7171182. [Google Scholar] [CrossRef] [PubMed]
  216. Yao, Y.; Lawrence, D.A. Susceptibility to COVID-19 in populations with health disparities: Posited involvement of mitochondrial disorder, socioeconomic stress, and pollutants. J. Biochem. Mol. Toxicol. 2020, e22626. [Google Scholar] [CrossRef] [PubMed]
  217. Gupta, R.K.; Harrison, E.M.; Ho, A.; Docherty, A.B.; Knight, S.R.; van Smeden, M.; Abubakar, I.; Lipman, M.; Quartagno, M.; Pius, R.; et al. Development and validation of the ISARIC 4C Deterioration model for adults hospitalised with COVID-19: A prospective cohort study. Lancet Respir. Med. 2021. PMCID: PMC7832571. [Google Scholar] [CrossRef] [PubMed]
  218. Márquez, E.J.; Chung, C.-H.; Marches, R.; Rossi, R.J.; Nehar-Belaid, D.; Eroglu, A.; Mellert, D.J.; Kuchel, G.A.; Banchereau, J.; Ucar, D. Sexual-dimorphism in human immune system aging. Nat. Commun. 2020, 11, 751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Márquez, E.J.; Trowbridge, J.; Kuchel, G.A.; Banchereau, J.; Ucar, D. The lethal sex gap: COVID-19. Immun. Ageing 2020, 17, 13, PMCID: PMC7240166. [Google Scholar] [CrossRef] [PubMed]
  220. Conti, P.; Younes, A. Coronavirus COV-19/SARS-CoV-2 affects women less than men: Clinical response to viral infection. J. Biol. Regul. Homeost. Agents 2020, 34, 339–343. [Google Scholar] [CrossRef] [PubMed]
  221. Gadi, N.; Wu, S.C.; Spihlman, A.P.; Moulton, V.R. What’s Sex Got to Do With COVID-19? Gender-Based Differences in the Host Immune Response to Coronaviruses. Front. Immunol. 2020, 11, 2147, PMCID: PMC7485092. [Google Scholar] [CrossRef] [PubMed]
  222. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/300386 (accessed on 26 February 2021).
  223. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/300292?search=foxp3&highlight=foxp3 (accessed on 26 February 2021).
  224. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CXCR3&keywords=cxcr3 (accessed on 26 February 2021).
  225. Dattilo, M. The role of host defences in Covid 19 and treatments thereof. Mol. Med. 2020, 26, 90, PMCID: PMC7522454. [Google Scholar] [CrossRef]
  226. Van der Made, C.I.; Simons, A.; Schuurs-Hoeijmakers, J.; van den Heuvel, G.; Mantere, T.; Kersten, S.; van Deuren, R.C.; Steehouwer, M.; van Reijmersdal, S.V.; Jaeger, M.; et al. Presence of Genetic Variants Among Young Men With Severe COVID-19. JAMA 2020, 324, 1–11, PMCID: PMC7382021. [Google Scholar] [CrossRef] [PubMed]
  227. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/147640?search=IFNB1&highlight=ifnb1 (accessed on 26 February 2021).
  228. Online Mendelian Inheritance in Man, OMIM®. Available online: https://omim.org/entry/147571?search=isg15&highlight=isg15 (accessed on 26 February 2021).
  229. Aiello, A.; Accardi, G.; Candore, G.; Caruso, C.; Colomba, C.; Di Bona, D.; Duro, G.; Gambino, C.M.; Ligotti, M.E.; Pandey, J.P. Role of Immunogenetics in the Outcome of HCMV Infection: Implications for Ageing. Int. J. Mol. Sci. 2019, 20, 685, PMCID: PMC6386818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Aiello, A.; Candore, G.; Accardi, G.; Caruso, C.; Colomba, C.; Duro, G.; Gambino, C.M.; Ligotti, M.E.; Di Bona, D. Translation of Basic Research into Clinics: Killer Immunoglobulin-like Receptors Genes in Autoimmune and Infectious Diseases. Curr. Pharm. Des. 2018, 24, 3113–3122. [Google Scholar] [CrossRef] [PubMed]
  231. Caruso, C.; Pandey, J.P.; Puca, A.A. Genetics of exceptional longevity: Possible role of GM allotypes. Immun. Ageing 2018, 15, 25, PMCID: PMC6219196. [Google Scholar] [CrossRef] [PubMed]
  232. Mantovani, A.; Netea, M.G. Trained Innate Immunity, Epigenetics, and Covid-19. N. Engl. J. Med. 2020, 383, 1078–1080. [Google Scholar] [CrossRef] [PubMed]
  233. Khalaf, K.; Papp, N.; Chou, J.T.; Hana, D.; Mackiewicz, A.; Kaczmarek, M. SARS-CoV-2: Pathogenesis, and Advancements in Diagnostics and Treatment. Front. Immunol. 2020, 11, 570927, PMCID: PMC7573101. [Google Scholar] [CrossRef] [PubMed]
  234. Tandon, N.; Luxami, V.; Tandon, R.; Paul, K. Recent Approaches of Repositioning and Traditional Drugs for the Treatment of COVID-19 Pandemic Outbreak. Mini Rev. Med. Chem. 2020. [Google Scholar] [CrossRef] [PubMed]
  235. Borcherding, N.; Jethava, Y.; Vikas, P. Repurposing Anti-Cancer Drugs for COVID-19 Treatment. Drug Des. Dev. Ther. 2020, 14, 5045–5058, PMCID: PMC7680713. [Google Scholar] [CrossRef] [PubMed]
  236. Singh, T.U.; Parida, S.; Lingaraju, M.C.; Kesavan, M.; Kumar, D.; Singh, R.K. Drug repurposing approach to fight COVID-19. Pharm. Rep. 2020, 1–30, PMCID: PMC7474498. [Google Scholar] [CrossRef] [PubMed]
  237. Benavides-Cordoba, V. Drug Repositioning for COVID-19. Colomb. Med. 2020, 51, e4279, PMCID: PMC7518729. [Google Scholar] [CrossRef]
Figure 1. Scheme of factors influencing susceptibility to and severity of COVID-19. “Lifestyle” refers to habits (in terms of quality of nutrition, physical activity, drug consumption, smoking) and to socio-economic parameters (access to the health care system, level of education, access to high quality medical information), i.e., the social determinants of health. “Environment” (intended as exposure to contaminants, pollution, and non-ionizing radio frequency, but also as exposure to multiple pathogens) is relevant since it mechanistically modifies the immune response [202,203,204,205,206,207,208,209,210]. Viral genetic variants and their effects on virulence/COVID-19 severity are also a matter of intense research [211,212,213].
Figure 1. Scheme of factors influencing susceptibility to and severity of COVID-19. “Lifestyle” refers to habits (in terms of quality of nutrition, physical activity, drug consumption, smoking) and to socio-economic parameters (access to the health care system, level of education, access to high quality medical information), i.e., the social determinants of health. “Environment” (intended as exposure to contaminants, pollution, and non-ionizing radio frequency, but also as exposure to multiple pathogens) is relevant since it mechanistically modifies the immune response [202,203,204,205,206,207,208,209,210]. Viral genetic variants and their effects on virulence/COVID-19 severity are also a matter of intense research [211,212,213].
Ijms 22 02636 g001
Table 1. List of the associations between blood groups and variants at the AB0 locus with COVID-19 susceptibility and severity.
Table 1. List of the associations between blood groups and variants at the AB0 locus with COVID-19 susceptibility and severity.
Group/LocusVariantOutcomepOR (95% CI)RR (95%CI)Refs.
A
Susceptibility0.0271.21 (1.02–1.43) [21]
Susceptibility* 0.04* 1.33 (1.02–1.73) [24]
Susceptibility0.00241.23 (1.08–1.41) [25]
Susceptibility<0.001 1.09 (1.02–1.13)[27]
Susceptibility (1)<0.0011.249 (1.114–1.440) [26]
Susceptibility (2)0.031.3 (1.02–1.66) [29]
Respiratory failure* 1.48×10−4* 1.45 (1.20–1.75) [28]
Mortality0.0081.482 (1.113–1.972) [21]
AB
Susceptibility* 0.035* 1.37 (1.02–1.83) [30]
B
Susceptibility* 0.004* 1.28 (1.08–1.52) [30]
0
Susceptibility<0.0010.67 (0.60–0.75) [21]
Susceptibility0.00060.787 (0.69–0.90) [25]
Susceptibility<0.001 0.87 (0.82–0.91)[27]
Susceptibility* 0.0070.84 (0.75–0.95) [30]
Susceptibility (1)<0.0010.699 (0.635–0.770) [26]
Respiratory failure* 1.06×10−5* 0.65 (0.53–0.79) [28]
Mortality0.0140.660 (0.479–0.911) [21]
Locus AB0
rs657152 (A)Respiratory failure* 5.35×10−7* 1.39 (1.22–1.59) [28]
Group/locus, AB0 blood group or locus; variant, name of the polymorphism at the indicated locus (allele); outcome, type of assayed outcome; p, p value (corrected p values are preceded by a *); OR (95% CI), odds ratio (95% confidence interval) for the analysed blood group or variant (adjusted OR are preceded by a *); RR (95% CI), relative risk (95% confidence interval) for the analysed blood group or variant; Ref, references. (1) meta-analysis; (2) in transplanted patients.
Table 2. Association between human leukocyte antigen (HLA) class I and II allele/serotype/haplotype with COVID-19 incidence, susceptibility and severity.
Table 2. Association between human leukocyte antigen (HLA) class I and II allele/serotype/haplotype with COVID-19 incidence, susceptibility and severity.
VariableAllele/SerotypepOR (95%CI)Regression CoefficientGrowth Rate (95%CI)Refs.
IncidenceB*440.05 0.14841.16 (1–1.35%)[39]
C*010.042 0.17471.19 (1.01–1.41%)[39]
SusceptibilityB*15:270.0303.59 (1.72–7.50) § [45]
B46n.s. [46]
B220.0321.71 (1.23–2.38) § [46]
C*07:290.025130.20 (5.28–3211) § [45]
DQB1*060.0468 #1.96 (1.19–3.22) [47]
DRB1*080.010 #1.814 (1.151–2.860) # [29]
SeverityAn.s. [28]
A*11:010.0082.33 [10]
B22n.s. [46]
B27n.s. [46,48]
B*27:07* 0.004 [41]
B46n.s. [46]
B*51:010.0073.38 [10]
Cn.s. [28]
C*14:020.0034.75 [10]
DPB1*03:010.0370.09 [10]
DQA1*01:010.0396.05 [10]
DQB1n.s. [28]
DQB1*06:020.016 [41]
DRB1n.s. [28]
DRB1*01:010.0213.7 [10]
DRB1*12:010.0450.18 [10]
DRB1*14:040.0115.1 [10]
DRB1*15:010.048 [37]
A*110.04 (1)7.693 (1.063–55.65) (1) [40]
0.02 (2)11.858 (1.524–92.273) (2) [40]
MortalityC*010.04 (1)11.182 (1.053–118.7) (1) [40]
0.02 (2)17.604 (1.629–190.211) (2) [40]
DQB1*040.03 (1)9.963 (1.235–80.358) (1) [40]
DRB1*080.01 #8.6 (1.7–43.9) # [29]
Variable, type of variable considered in the statistical analysis; allele/serotype, HLA alleles or serotypes; p, p value (corrected p values are preceded by a *); OR (95% CI), odds ratio (95% confidence interval) for the analyzed allele/serogroup/haplotype; regression coefficient, regression coefficient at regression analysis; growth rate (95% CI), growth rate (95% confidence interval) for the analyzed allele/serogroup; Ref, references; n.s. not significant. Alleles are reported in italics. The listed statistics, coefficients and p values are corrected. Unadjusted statistics are indicated by a §. # in transplanted patients; (1) after controlling for Sepsis-related Organ Failure Assessment (SOFA)—see text for details. (2) after controlling for the Acute Physiology and Chronic Health Evaluation (APACHE)-II—see text for details.
Table 3. List of genetic variants influencing immune responses associated with COVID-19 susceptibility, severity and mortality.
Table 3. List of genetic variants influencing immune responses associated with COVID-19 susceptibility, severity and mortality.
Gene/LocusVariant/
Position
Reference/
Other Allele
Altered/
Risk Allele
Protein VariantTypepOR (95%CI)OutcomeRefs.
TLR3§ 187003852ATAp.Ser339fspLOF0.018.28
(1.04–65.64)
severity[113]
TLR3§ 187005146GAp.Trp769 *pLOF
UNC93B1§ 67770598CAp.Glu96 *pLOF
TBK1§ 64875731CTp.Arg308 *pLOF
IRF7§ 615095ACp.Arg7fspLOF
IRF7§ 614300GAp.Gln185 *pLOF
IRF7§ 613966CGGGCTGGGGCCCGCp.Pro246fspLOF
IRF7§ 613353GGCp.Pro364fspLOF
IFNAR2§ 34621038AGATTGTTGGTTTTAp.Glu140fspLOF
IFNAR2rs2236757GA 4.99 × 10−81.28severity[117]
OAS3rs10735079GA 1.65 × 10−81.29severity[117]
IFITM3rs12252TC # 0.0093# 6.37severity[118]
0.0251.93
(1.09–3.46)
severity[119]
PRKRA I226N 0.02 severity[120]
TNF-αrs1800629GA ° <0.001 age > 60[121]
° <0.001 lymphopenia[121]
° 0.009 high CRP[121]
° <0.001 high ferritin[121]
° <0.001 severity[121]
0.045 severity[121]
3p21.31rs11385942GGA 1.15 × 10−101.77
(1.48–2.11)
respiratory failure[28]
0.0031.56
(1.17–2.01)
mechanical ventilation[28]
TMEM189UBE2V1rs6020298GA 4.1 × 10–61.2severity[10]
DPP9rs2109069GA 3.98 × 10−121.36severity[117]
GOLGA8Brs200975425CT 9.4 × 10–105.4susceptibility[10]
LAPTM4B P219L 0.029 [120]
P220L
I109F
P50T
ApoE e3e4 1.19 × 10–62.31
(1.65–3.24)
severity[122]
^ 0.009^ 1.20
(1.05–1.37)
severity[123]
3.24 × 10−92.24
(1.72–2.93)
severity[123]
1.22 × 10–64.29
(2.38–7.72)
mortality[123]
Gene/locus, analyzed gene or locus; variant/position, variant name, or position of the variant on reference human genome; reference/other allele., reference allele or other allele; altered/risk allele, altered allele or risk allele; protein variant, changes in the aminoacidic sequence; p, p value (all the reported p values are corrected); OR (95% CI), odds ratio (95% confidence interval); outcome, type of the assayed outcome; Ref, references. § GRCh37; # homozygosity for the risk allele; ° homozygosity for the risk allele vs. homozygosity for the other allele; † homozygosity for the risk allele vs. heterozygosity for the risk allele; ^ heterozygosity for the risk allele vs. homozygosity for the other allele.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pojero, F.; Candore, G.; Caruso, C.; Di Bona, D.; Groneberg, D.A.; Ligotti, M.E.; Accardi, G.; Aiello, A. The Role of Immunogenetics in COVID-19. Int. J. Mol. Sci. 2021, 22, 2636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052636

AMA Style

Pojero F, Candore G, Caruso C, Di Bona D, Groneberg DA, Ligotti ME, Accardi G, Aiello A. The Role of Immunogenetics in COVID-19. International Journal of Molecular Sciences. 2021; 22(5):2636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052636

Chicago/Turabian Style

Pojero, Fanny, Giuseppina Candore, Calogero Caruso, Danilo Di Bona, David A. Groneberg, Mattia E. Ligotti, Giulia Accardi, and Anna Aiello. 2021. "The Role of Immunogenetics in COVID-19" International Journal of Molecular Sciences 22, no. 5: 2636. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052636

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop