Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Association between HLA-C alleles and COVID-19 severity in a pilot study with a Spanish Mediterranean Caucasian cohort

Abstract

The clinical presentations of COVID-19 may range from an asymptomatic or mild infection to a critical or fatal disease. Several host factors such as elderly age, male gender, and previous comorbidities seem to be involved in the most severe outcomes, but also an impaired immune response that causes a hyperinflammatory state but is unable to clear the infection. In order to get further understanding about this impaired immune response, we aimed to determine the association of specific HLA alleles with different clinical presentations of COVID-19. Therefore, we analyzed HLA Class I and II, as well as KIR gene sequences, in 72 individuals with Spanish Mediterranean Caucasian ethnicity who presented mild, severe, or critical COVID-19, according to their clinical characteristics and management. This cohort was recruited in Madrid (Spain) during the first and second pandemic waves between April and October 2020. There were no significant differences in HLA-A or HLA-B alleles among groups. However, despite the small sample size, we found that HLA-C alleles from group C1 HLA-C*08:02, -C*12:03, or -C*16:01 were more frequently associated in individuals with mild COVID-19 (43.8%) than in individuals with severe (8.3%; p = 0.0030; pc = 0.033) and critical (16.1%; p = 0.0014; pc = 0.0154) disease. C1 alleles are supposed to be highly efficient to present peptides to T cells, and HLA-C*12:03 may present a high number of verified epitopes from abundant SARS-CoV-2 proteins M, N, and S, thereby being allegedly able to trigger an efficient antiviral response. On the contrary, C2 alleles are usually poorly expressed on the cell surface due to low association with β2-microglobulin (β2M) and peptides, which may impede the adequate formation of stable HLA-C/β2M/peptide heterotrimers. Consequently, this pilot study described significant differences in the presence of specific HLA-C1 alleles in individuals with different clinical presentations of COVID-19, thereby suggesting that HLA haplotyping could be valuable to get further understanding in the underlying mechanisms of the impaired immune response during critical COVID-19.

Introduction

In 2019, the severe acute respiratory syndrome (SARS) coronavirus type 2 (SARS-CoV-2) emerged, becoming the etiological agent of the major pandemic in this century. More than 296 million people have passed the coronavirus disease 2019 (COVID-19) worldwide so far, with approximately 5.4 million deaths [1]. These numbers are rapidly increasing due to new emergent variants such as Omicron [2]. Not all the individuals respond similarly to SARS-CoV-2 infection and, therefore COVID-19 may have different presentations, ranging from an asymptomatic infection to a critical or fatal disease [3]. Although it is still uncertain why some individuals develop the most severe forms of the disease, an anomalous innate immune response that ends in a cytokine storm seems to be a major cause for a poor prognosis [4,5]. Accordingly, high levels of interleukin (IL)-7, IL-6, ferritin, or C reactive protein (CRP) have been detected in the plasma of individuals with severe COVID-19 [6,7]. On the contrary, these patients usually show low levels of cytokines related to T-cell antiviral activity, such as IL-2, IL-12 or interferon (IFN)-γ [8], as well as reduced antiviral responses mediated by IFN type-I and -III [9,10]. Consequently, this defective immune response would not only be unable to clear the infection by SARS-CoV-2, but patients may even experience the reactivation of latent herpesvirus infections [11]. For a long time, the only treatment available for the hyper inflammation state derived from SARS-CoV-2 infection has been corticosteroids such as dexamethasone [12], but recently, the Food and Drug Administration (FDA) has approved the oral administration of nirmatrelvir and ritonavir for the emergency treatment of mild-to-moderate COVID-19 in children and adults who present high risk for progression to severe COVID-19, as well as oral molnupiravir for adults [13,14].

Within the host factors that may influence the susceptibility to develop the most severe forms of COVID-19, the human leukocyte antigen (HLA) complex has been appointed as a potential candidate [15]. The HLA complex is formed by highly polymorphic genes located on chromosome 6 that encode glycoproteins that regulate the immune response by presenting exogenous and endogenous peptides to T lymphocytes and Natural Killer (NK) cells [16]. HLA genes are inherited as a group known as haplotype [17]. Classical HLA class I glycoproteins A, B, and C are expressed on the surface of all nucleated cells in association with the invariant β2 microglobulin (β2M), a protein encoded in chromosome 15 that stabilizes the tertiary structure of these molecules [18,19]. On the other hand, HLA class II glycoproteins DR, DQ, DM, DO, and DP are located only on the surface of professional antigen presenting cells (APCs) such as macrophages, B cells and dendritic cells [20]. HLA class I glycoproteins mostly present intracellular small peptides to CD8+ T cells, such as those derived from a viral infection [18,21], whereas HLA class II glycoproteins mostly present exogenous peptides to CD4+ T cells, which would induce a humoral response through the activation of specific B cells [22,23]. HLA-C molecules are also recognized by NK cells, thereby acting mostly as ligands for the Killer Immunoglobulin-like Receptors (KIRs) that are expressed on the cell surface [24] and participate in the activation or inhibition of these cells [25].

An efficient presentation of peptides by HLA molecules to the cells of the immune system is essential to initiate a potent and effective response against pathogens such as SARS-CoV-2 in order to swiftly clear the infection. However, due to the high polymorphism of HLA, not all molecules are equally effective to generate an efficient response. More than 24,000 HLA alleles have been reported so far, mostly within HLA class I [26]. In the Spanish population, HLA-B shows the greatest allele number, whereas HLA-A and -C show the major polymorphism [27]. HLA-B and -C alleles are hierarchically classified according to the amino acid residues located at positions 77 and 80–83 of the peptide-binding pocket. Therefore, HLA-B is traditionally subdivided into -Bw6 (Ser77 and Asn80) and -Bw4 epitopes [28] that are characterized by at least seven different patterns of amino acid exchanges in these positions, whereas HLA-C alleles are subdivided into groups C1 and C2, which are mutually exclusive. Alleles from group C1 contain Ser77 and Asn80 and are ligands for the inhibitory KIR2DL2 and -2DL3, as well as for the activating KIR2DS2, whereas alleles from group C2 contain Asn77 and Lys80 [29] and are ligands for the inhibitory KIR2DL1 and activating KIR2DS1 [30]. As a major characteristic, HLA-C molecules show a distinctly lower expression on the cell surface in comparison with HLA-A and -B molecules, most likely due to low mRNA expression, poor assembly with β2M, or restricted peptide binding that leads to the accumulation of misfolded intermediates in the endoplasmic reticulum [31].

The potential role of HLA molecules in the different susceptibility to infectious diseases has been described for several alleles [32,33]. In fact, the expression of specific HLA-B and -C alleles have been associated with higher resistance to infection by the human immunodeficiency virus type 1 (HIV-1) [3436] and with the spontaneous clearance of hepatitis C virus (HCV). Moreover, the presence or absence of specific HLA class I and II molecules appear to be essential for the immune response against respiratory infections such as tuberculosis [37,38], influenza [39], or the first SARS-CoV. In the latter case, the expression of specific HLA alleles such as HLA-B*07:03 and -B*46:01 may confer a higher predisposition to develop severe SARS [40,41], whereas the presence of HLA-C*15:02 may have a protective effect [42]. Regarding the specific association of HLA alleles with COVID-19, healthy individuals carrying HLA-B*44 and/or -C*01 (group C1) may be more susceptible to be infected with SARS-CoV-2 and to spread the infection within the population [43]. Other HLA-C alleles from group HLA-C1 have been described to show a higher degree of recognition of SARS-CoV-2 peptides [44], and HLA-C*01 has also been correlated with a higher mortality during COVID-19 [45] in the Spanish population.

During the first COVID-19 pandemic peak (April to May 2020), Spain was one of the European countries more severely affected, with approximately 100 excess deaths per 100,000 people [46] and a case fatality rate 2.3% higher than the average rate in the neighboring countries [47]. The causes for this nearly 38% of relative increased death have not been completely elucidated yet. Therefore, in this pilot study, we analyzed the potential association between the different clinical presentations of COVID-19 and the presence of specific HLA and KIR alleles in a cohort of Spanish Mediterranean Caucasian individuals who were recruited during the first wave of SARS-CoV-2. This information could be useful to identify predictive biomarkers for COVID-19 severity and to further understand the underlying pathogenesis of this emergent disease.

Materials and methods

Patients

Blood samples were obtained from 72 individuals with Spanish ethnicity (Mediterranean Caucasian origin) who developed different presentations of COVID-19. All patients were recruited in Madrid (Spain) after the first pandemic peak and at the beginning of the second wave, between April and October 2020. The inclusion criteria were to have a positive SARS-CoV-2 qRT-PCR assay in nasopharyngeal smear and to be older than 18 years old at the moment of sampling. Twenty-four participants were asymptomatic or showed mild symptoms of COVID-19 and hospitalization was not required, whereas 48 patients were hospitalized due to the severity of the symptoms. Hospitalized patients were classified as severe (n = 17) or critical (n = 31), based on COVID-19 evolution during hospitalization. Patients with critical COVID-19 were those who needed intensive care, invasive mechanical ventilation, and/or died due to complications of the disease. Main demographic and clinical characteristics of all individuals recruited for this study are summarized in Table 1.

thumbnail
Table 1. Demographic and clinical data of participants with different presentations of COVID-19 who were recruited for the study.

https://doi.org/10.1371/journal.pone.0272867.t001

Ethical statement

The individuals with the most severe forms of COVID-19 were recruited from Hospital Universitario Ramón y Cajal, Hospital Universitario Puerta de Hierro, and Hospital de El Escorial (Comunidad de Madrid, Spain). Participants with mild COVID-19 were recruited at the Primary Healthcare Center Doctor Pedro Lain Entralgo (Alcorcón, Madrid, Spain). All patients gave informed written consent to participate in the study or witnessed oral consent with written consent by a representative to avoid handling contaminated documents. The confidentiality and anonymity of all participants were secured by Current Spanish and European Data Protection Acts. Protocol for this study (CEI PI 32_2020-v2) was prepared in accordance with the Helsinki Declaration and previously reviewed and approved by the Ethics Committees of Instituto de Salud Carlos III (IRB IORG0006384) and all participating hospitals.

DNA isolation and HLA genotyping

Whole genomic DNA was isolated from blood using QIAamp DNA Blood Mini Kit (Qiagen Iberia, Madrid, Spain). Intermediate HLA genotyping resolution (four digits, the corresponding “P” for every 2nd field allele typing was discarded to clarify the text) was performed with 2μg of genomic DNA using LinkSēq™ HLA ABCDRDQB1 384 kit (One Lambda, Thermo Fisher Scientific España, Madrid, Spain), which allows testing human class I classical HLA-A, -B, -C alleles and class II–DR, -DQ and–DP alleles, including Bw4 and Bw6 genotypes. qPCR was performed using QuantStudio 5 Real-Time PCR System (Thermo Fisher Scientific), according to manufacturer’s instructions and using the following conditions: 95°C, 2 min; 36 cycles: 95°, 15 seconds; 64°C, 1 min. A dissociation profile was added using the following conditions: 95°C, 15 seconds; 65°C, 30 seconds. The dissociation data corresponding to the melt curve were collected per well between 65°C and 95°C using QuantStudio™ Design & Analysis Software (Thermo Fisher Scientific). These data were exported to SureTyper software (One Lambda) for the interpretation and identification of HLA alleles.

HLA haplotyping

Statistical analysis was performed with Arlequin v3.5 software [48]. In brief, this program calculates HLA allele frequencies, Hardy-Weinberg equilibrium (HWE), which assumes random mating within the population, and the linkage disequilibrium between n alleles at n different loci and their level of significance (p) for 2x2 comparisons to establish the 2, 3 and 4 loci associations. In addition, the most frequent complete extended haplotypes were deduced from: 1) 2-loci HLA haplotype frequencies, 2) from the previously described haplotypes in other populations; and 3) from the haplotypes if they appeared in two or more individuals and the alternative haplotype was well defined [49].

KIR genotyping

KIR genotyping was performed with 360 ng of genomic DNA using LinkSeq KIR Typing kit (One Lambda, Thermo Fisher Scientific). This kit allows testing all 15 human KIR genes and 2 pseudogenes, including both full length and deleted forms of 3DP1 and 2DS4 and allele-specific variants in 3DL1, 2DL1. qPCR was performed using StepOne Real-Time PCR System (Thermo Fisher Scientific) according to manufacturer’s instructions and using the following conditions: 95°C, 2 min; 36 cycles: 95°, 15 seconds; 64°C, 1 min. A dissociation profile was added (95°C, 15 seconds; 65°C, 30 seconds) and dissociation data corresponding to the melt curve of the amplification were collected per well between 65°C and 95°C using StepOne Plus software v2.3. These data were exported to SureTyper software (One Lambda) for interpretation and identification of KIR genotypes.

HLA class I epitope prediction and analysis

HLA class I epitopes between 8–12 residues for the whole reference SARS-CoV-2 proteome (Wuhan-1; RefSeq: NC_045512.2) were predicted for all HLA class I alleles observed in the patient cohort using the NetMHCpan-4.1 server that may predict the binding of the viral peptides to any known MHC molecule using artificial neural networks [50]. Binding epitopes were considered those with rank ≤ 0.5 and score ≥ 0.5. Redundant epitopes for the same allele and sharing the same peptide core but distinct total lengths and lower scores, were ignored. Experimentally validated epitopes were downloaded from the Immune Epitope Database and Analysis Resource (Last accession: 19/03/2021) [51], using the following search terms: Epitopes: "Any epitopes"; Assay: "T Cell", "MHC Ligand" and outcome: "Positive"; MHC Restriction: "MHC Class I"; Host: "Human"; Disease "COVID-19 [52]". Predicted epitopes were considered as validated when there was a validated epitope that shared perfect matching protein coordinates. Distribution of HLA alleles in mild, severe and critical COVID-19 was represented with GraphPad Prism software (GraphPad Software Inc.).

HLA-C alleles 3D modelling

The three-dimensional (3D) structure of HLA-C*07:02 was obtained from the Protein Data Bank (PDB) (PDB code: 6PA1) [53] to be used as a template, and it was visualized using the PyMOL Molecular Graphics System, 2.0 (Schrödinger, LLC). Mutagenesis in silico was performed in this template to obtain model 3D structures for consensus sequences, including key pocket residues of HLA-C*08:02 (group C1) and HLA-C*07:02 (group C2) alleles in order to determine the modifications produced in the binding pocket due to amino acid exchanges. Amino acids at positions 9, 11, 24, 77, 80, 90, 94, and 95 were selected as key residues in the structure of HLA-C pockets and the amino acid sequences of HLA-C alleles, as well as the corresponding multiple alignments, were obtained from the Immuno Polymorphism database (IPD)- Immunogenetics (IMGT)/HLA Database (EMBL-EBI, Wellcome Genome Campus, Hinxton, UK) [26].

Statistical analysis

Statistical significance between groups was determined by comparing the data obtained in the groups of individuals with severe and critical presentations of COVID-19 with the data obtained from the individuals who had mild or asymptomatic COVID-19. Categorical data were expressed as percentages, and the median was used to describe tendency. Statistical significance for two-row by two-column (2x2) contingency table was determined by Fisher’s exact test or chi-square test using Graph Pad Prism 8.0 (Graph Pad Software Inc., San Diego, CA). Alleles with counts less than five in every group were combined into a common group before computing the test. Significance levels were corrected by Bonferroni correction. For each HLA haplotype the corrected P value (Pc) was calculated by multiplying the P value obtained with the two-tailed chi-square or Fisher’s exact test by the number of tested allelic combinations. pc<0.05 were considered statistically significant in all comparisons.

Results

Patients’ characteristics

Seventy-two patients of Spanish Mediterranean Caucasian origin who were diagnosed with COVID-19 by positive qRT-PCR were recruited for this study between April and October 2020. The most relevant demographic and clinical characteristics of all participants are summarized in Table 1 and all detailed characteristics for non-hospitalized and hospitalized individuals are shown in S1 and S2 Tables, respectively. The individuals were classified into three groups depending on the medical assistance they received and according to the progression of the disease. Twenty-four patients received primary health assistance from Primary Healthcare Centers, and they were homebound until the resolution of the symptoms or a negative qRT-PCR test for SARS-CoV-2 (henceforth, Mild COVID-19). 58.3% of these individuals were females with a median age at diagnosis of 47.5 years (IQR 32.8–58.8). Median days from clinical onset to sample acquisition was 88.5 days (IQR 79.8–95.0). Forty-eight patients were hospitalized due to COVID-19 severity. Of these individuals, 31 were categorized as critical and admitted to the Intensive Care Unit (ICU) (henceforth, Critical COVID-19), whereas the rest of hospitalized individuals (n = 17) presented severe forms of COVID-19 but without requiring admission to the ICU (henceforth, Severe COVID-19). Most severe and critical patients were males (64.7% and 64.5%, respectively), with median age at diagnosis of 72.0 (IQR 68.0–86.5) and 65.0 (IQR 58.0–72.0) years, respectively. Median number of days from clinical onset to sample acquisition was 23.0 (IQR 9.3–39.3) and 35.0 (IQR 20.0–53.0) days in severe and critical, respectively. Median length of hospitalization was 17.0 (IQR 8.5–28.5) and 48.0 (IQR 32.0–81.0) days in severe and critical patients, respectively. Median length of hospitalization at the ICU was 43.5 days (IQR 21.3–59.0) in critical patients. Fourteen individuals with critical COVID-19 (45.2%) expired due to complications of the disease. The most common symptoms showed by individuals with mild COVID-19 were asthenia (75%), fever (58.3%), and cough (58.3%). Most individuals with mild COVID-19 did not develop pneumonia (87.5%), whereas pneumonia (82.4%) and cough (52.9%) were the most frequent symptoms in severe patients and pneumonia (83.9%), fever (74.2%), and cough (67.7%) were the most frequent symptoms in critical individuals. Among the potential risk factors for developing severe forms of COVID-19, dyslipidemia was present in 25.0%, 29.4%, and 45.2% of individuals with mild, severe, and critical COVID-19, respectively, whereas arterial hypertension was present in 16.7%, 64.7%, and 45.2%, respectively.

HLA allele frequencies

HLA class I and II allele frequencies were analyzed in individuals with mild, severe and critical COVID-19 and the statistical significance of the comparison between the groups with severe and critical COVID-19 and the group with mild COVID-19 was calculated by Fisher’s exact test or chi-square test 2x2 and corrected by Bonferroni correction (Table 2). The most common HLA-C alleles in our cohort were HLA-C*04:01 and -C*05:01, but no significant differences were found for the frequency of these alleles in the comparison between groups. However, the presence of HLA-C*08:02 was significantly higher in individuals with mild COVID-19 (18.8%) in comparison with individuals with critical COVID-19 (1.6%; p = 0.0024; pc = 0.0240). The most common HLA-B alleles in our cohort were HLA-B*44:03 and -B*18:01, but no significant differences were found for the frequency of these alleles between groups. The frequency of HLA-B rare alleles was higher in individuals with severe COVID-19 (82.4%) regarding those with mild COVID-19 (54.2%; p = 0.0080; pc = 0.0400), but no significance in the comparison between mild and critical COVID-19 was achieved. The most frequent HLA-A allele in our cohort was HLA-A*02:01, whereas the most frequent HLA class II alleles were DRB1*07:01 and *03:01, but no significant differences were observed in the comparison between groups for these alleles.

thumbnail
Table 2. HLA class I and II allele frequencies in individuals with mild, severe and critical COVID-19 who were recruited for this pilot study.

The statistical significance of the comparisons between the allele frequencies of groups of individuals with severe and critical COVID-19 and the group of individuals with mild COVID-19 was calculated using Fisher’s exact test or chi-square test 2x2. Significance levels were corrected by Bonferroni correction. Significant pc <0.05 are highlighted in bold font.

https://doi.org/10.1371/journal.pone.0272867.t002

The frequency of associations between specific HLA-A, -B and -C alleles that may interact with KIRs was also analyzed and the statistical significance of the comparison between the groups with severe and critical COVID-19 and the group with mild COVID-19 was calculated by Fisher’s exact test or chi-square 2x2 (Table 3). The frequency of associations between HLA-C*08 or -C*12 alleles was significantly higher in individuals with mild COVID-19 (33.3%) than in individuals with critical COVID-19 (8.1%) (p = 0.0008; pc = 0.0088). Moreover, specific HLA-C*08:02 or -C*12:03 alleles were more frequent in 31.3% of individuals with mild COVID-19, in comparison with 3.2% (p = 0.0001; pc = 0.0010) of individuals with critical disease. These differences were still significant in the comparison between groups when the frequency of the association between alleles HLA-C*08:02, HLA-C*12:03 or HLA-C*16:01 was analyzed, being more frequent in individuals with mild COVID-19 (43.8%) than in individuals with severe (8.3%; p = 0.0030; pc = 0.0330) or critical COVID-19 (16.1%; p = 0.0014; pc = 0.0154).

thumbnail
Table 3. Frequency of the associations between specific HLA class I alleles that may interact with KIR in individuals with mild, severe and critical COVID-19.

The statistical significance of the comparisons between the frequencies of these allele combinations in the groups of individuals with severe and critical COVID-19 and the group of individuals with mild COVID-19 was calculated using Fisher’s exact test or chi-square 2x2. Significance levels were corrected by Bonferroni correction. Significant pc <0.05 are highlighted in bold font.

https://doi.org/10.1371/journal.pone.0272867.t003

HLA haplotype frequencies

HLA complex is highly stable and allows little recombination between genes. Therefore, the most frequent HLA haplotypes were deduced from the two HLA loci haplotype frequencies [49]. The extended HLA haplotypes that were more frequent in our cohort are summarized in Table 4. No significant differences were observed in the comparison between groups. Although we also observed a different haplotype pattern in the frequency of HLA-B genotypes -Bw4 and -Bw6 amongst the different groups of patients, these results did not reach statistical significance between the groups (S1 Fig).

thumbnail
Table 4. Most common HLA haplotypes observed in individuals with mild, severe and critical COVID-19.

The statistical significance of the comparisons between the frequencies of these haplotypes in the groups of individuals with severe and critical COVID-19 and the group of individuals with mild COVID-19 was calculated using Fisher’s exact test 2x2. Significance levels were corrected by Bonferroni correction. Significant pc <0.05 are highlighted in bold font.

https://doi.org/10.1371/journal.pone.0272867.t004

Association between KIR genes and alleles and clinical presentations of COVID-19

The association between specific KIR genes and alleles and the different presentations of COVID-19 was analyzed. Due to the low quantity of DNA available in some samples, KIR genotyping could only be performed in 55 patients of our cohort (mild, n = 18; severe, n = 17; critical, n = 20). No significant differences were observed between the frequencies of thirty alleles of fifteen KIR loci and two pseudogenes in the groups of individuals with severe and critical COVID-19 in comparison with the group of individuals with mild COVID-19 (Table 5).

thumbnail
Table 5. KIR genes and alleles identified in individuals with mild, severe and critical COVID-19.

The statistical significance of the comparisons between the frequencies of KIR genes and alleles in the groups of individuals with severe and critical COVID-19 and the group of individuals with mild COVID-19 was calculated using Fisher’s exact test 2x2. Significance levels were corrected by Bonferroni correction.

https://doi.org/10.1371/journal.pone.0272867.t005

Association between HLA-C alleles and COVID-19 presentations

Some HLA-C alleles can be divided into two large groups according to the amino acids located at positions 77 and 80 when they are considered as ligands for KIRs. Alleles from group C1 contain Ser77 and Asn80 (HLA-C*01; *03; *08; *12; *14, and *16), whereas alleles from group C2 contain Asn77 and Lys80 (HLA-C*02; *04; *05; *06; *07; *15; *17, and *18) [29]. When the frequency of hetero- and homozygosis of HLA-C1 and/or -C2 alleles was analyzed in the individuals from our cohort, we did not find statistical significance in the comparison between groups (S2 Fig).

In order to determine whether the amino acid exchanges at positions 9, 11, 24, 77, 80, 90, 94, and 95, which are characteristic of groups C1 and C2, may differentially influence the binding-pocket structure in HLA-C alleles, we used the 3D structure of HLA-C*07:02 [53] as a template to perform mutagenesis in silico in these positions (marked as grey residues in Fig 1A and 1B). These positions were changed to Tyr9, Ser11, Ala24, Ser77, Asn80, Ala90, Thr94, and Leu95, as are in HLA-C*08:02 allele, which belongs to group C1 and its frequency was significantly higher in individuals with mild COVID-19 (Fig 1A, left image), as well as to Asp9, Ala11, Ser24, Asn77, Lys80, Asp90, Ile94, and Ile95, as are in HLA-C*07:01 allele, which belongs to group C2 and its frequency was significantly higher in individuals with severe COVID-19 (Fig 1A, right graph). We observed that these amino acid exchanges induced a conformational modification in the binding pocket structure due to changes in the number and/or position of hydrogen-type bonds (red lines), which could affect both residue-residue and residue-antigenic amino acids interactions in the pocket (green residues) (Fig 1B), as well as the molecule stability.

thumbnail
Fig 1. Comparative structure of peptide-binding pockets of HLA-C1 and -C2 alleles.

(A) 3D modelling of HLA-C*07:02 (C1) as a template to perform mutagenesis in silico in positions 9, 11, 24, 77, 80, 90, 94, and 95 (grey residues) of the binding pocket, according to the amino acids presented by HLA-C*08:02 allele (group C1) (Tyr9, Ser11, Ala24, Ser77, Asn80, Ala90, Thr94, and Leu95) (left image) or by HLA-C*07:01 allele (group C2) (Asp9, Ala11, Ser24, Asn77, Lys80, Asp90, Ile94, and Ile95) (right image). (B) Hydrogen-type bonds between amino acids are represented with red lines and both residue-residue and residue-antigenic amino acids interactions in the pocket are represented in green.

https://doi.org/10.1371/journal.pone.0272867.g001

The combination between HLA-C alleles and their specific KIR alleles was also analyzed in our cohort and no statistically significant differences were found in the comparison between groups (S3 Fig).

SARS-CoV-2 epitope analysis

Inter-allelic differences associated with COVID-19 severity may reflect differences in the whole number of SARS-CoV-2-epitope repertoires. Therefore, epitopes for each significant HLA class I allele were predicted and further verified if they were associated with experimentally confirmed ligands, as described above (see Methods). Since presentation of HLA class I epitopes is largely determined by protein amount [54], the analysis was focused in the three SARS-CoV-2 proteins more abundant: M, N and S [55]. Predictive values reinforced the data observed in this cohort. The HLA-C*12:03 allele, linked to mild COVID-19 presentation in our study, contained the second highest number of verified epitopes, i.e., 65 (p <0.001, Fisher exact test), among 20 HLA-C alleles (26 ± 19 epitopes, mean ± standard deviation) in abundant SARS-CoV-2 proteins. In contrast, HLA-C*04:01 and -C*07:01, both linked to severe and/or critical COVID-19 presentations in our cohort, ranked 16 and 17th on this scale with only eight of these epitopes each (p = 0.0030). Conflicting results were obtained for HLA-B*14:01, with only one validated abundant epitope, and for HLA-C*08:02, with two epitopes, despite these alleles were also linked to mild symptoms.

Discussion

Spain was one of the most severely affected European countries during the first pandemic peak of COVID-19 that occurred between March and June 2020 [56]. At this time, the mortality rate during hospitalization was approximately 30%, and even higher after the admission at the ICU, which was approximately 50% [57]. The underlying mechanisms that influenced the poor prognosis of COVID-19 in Spain during this first pandemic wave are still not fully understood. It is already known that some host factors are essential to conditionate the development of a severe or critical presentation of the disease, such as male gender, to be over 65 years of age, and to have previous comorbidities [58]. However, other host factors could also be responsible for the development of an exacerbated inflammatory response [59] that was ineffectual to clear the infection due to an impaired cytotoxic response [8].

The HLA complex has an essential role in the development of an effective immune response against pathogens, thereby influencing the clinical outcomes of infectious diseases, such as the ones caused by human coronaviruses [3941,60]. The high polymorphism presented by HLA molecules may help explain the great variability of immune responses that are developed by the individuals within a community, including the higher susceptibility to progress to a critical form of COVID-19. In fact, the lower incidence of SARS-CoV-2 in Africa has been related to genetic factors such as the occurrence of different HLA alleles in comparison with other regions [61]. Therefore, the variability in HLA may be also considered an essential host factor that impacts the progression of COVID-19.

In this pilot study, we analyzed whether the presence of specific HLA molecules may influence the different outcomes of COVID-19 in a cohort of individuals with Spanish Mediterranean Caucasian ethnicity who developed different forms of the disease at the beginning of the pandemic in 2020. The Spanish population presents distinctive allelic distributions and haplotypic associations that are consistent with multiple migrations of different ethnicities that occurred throughout history in Spain [62] and that show a relative homogenous distribution fitting the HWE model [27]. According to previous reports [63], most individuals of our cohort who were hospitalized with severe and critical COVID-19 were males (63.5%) with more than 65-year-old, whereas most individuals who developed asymptomatic or mild COVID-19 were females (61.5%) with 47.5-year-old on average.

As expected, HLA-B was the locus that showed the highest number of alleles in our cohort [27]. The presence of specific HLA-B alleles has been associated with increased susceptibility to different viral infections and therefore, it may be considered a potential predictive biomarker of COVID-19 severity [32,64]. However, although we found HLA-B rare alleles which presence was significantly different between the groups of individuals with mild and severe presentations of COVID-19 (p = 0.0080; pc = 0.0400), this significance was not extended to individuals with critical COVID-19, which reduced its biological importance. On the other hand, HLA-A*02:01 was the most common allele in all groups of our cohort, which was expected for a population of Spanish origin [49,65], but no significant differences were found in the comparisons of HLA-A alleles frequencies between the groups with different presentations of COVID-19. Regarding HLA-C locus, it showed the highest significance in the different frequency between groups, being HLA-C*04:01 and -C*05:01 the most common HLA-C alleles in all groups. The frequency of HLA-C*08:02 was significantly higher in individuals of our cohort with mild COVID-19 (18.8%) in comparison with individuals with critical COVID-19 (1.6%; p = 0.0024; pc = 0.0240). The frequency of this allele HLA-C*08:02 in the Spanish population is 5.3% [27], which meant that it was 3.5-fold more prevalent in the group of individuals with mild COVID-19 of our cohort. Moreover, 43.8% of the individuals with mild COVID-19 presented an association of HLA-C*08:02 or -C*12:03 or -C*16:01 alleles, all of them classified as alleles from group C1, which was 2.4-fold more frequent than in the general Spanish population. In comparison, individuals with severe and critical COVID-19 showed a reduced frequency of the associations between these alleles, being 8.3% (p = 0.0030; pc = 0.033) and 16.1% (p = 0.0014; pc = 0.0154), respectively. This may indicate a protective effect of alleles from group C1 against the infection by SARS-CoV-2. Moreover, due to the close distance between HLA-B and -C loci, their expression may be considered together [66] and accordingly, B*14:01-C*08:02 haplotype was one of the most frequent extended haplotypes in the group of mild COVID-19, as well as in the general Spanish population [27,67]. In fact, B*14:01-C*08:02 haplotype was 8.6-fold less frequent in individuals with critical COVID-19, in comparison with the group with mild disease, which also may indicate a potential protective role for this extended haplotype. Although the presence of C2 alleles such as HLA-C*06 or -C*07 [29] and, specifically HLA-C*07:01 or -C*06:02, was significantly increased in individuals with severe COVID-19 (35.3%) in comparison with participants with mild COVID-19 (6.3%; p = 0.0012; pc = 0.0132), this difference was not significant in the comparison between individuals with critical and mild COVID-19.

The protection exerted by specific HLA alleles may be related to a more efficient antigenic presentation to T lymphocytes, but also to an effective NK cell activation. The activation of NK cells is modulated by a subtle balance between stimulatory and inhibitory signals that are received by several receptors expressed on the cell surface, including KIRs [68]. Consequently, the interaction between HLA and KIR molecules may influence NK cell activation and, consequently, the clearance of viral infections [69]. In this regard, previous reports predict that some C2 alleles such as HLA-C*06:02 and–C*07:01 would bind weakly to SARS-CoV-2 antigens, allowing a poorer disease progression [70], most likely due to a deficient antigen presentation and NK cell activation. These results are in accordance with our prediction that C2 alleles such as HLA-C*07:01 and -C*04:01 would be able to present a reduced number of epitopes from the most abundant SARS-CoV-2 proteins M, N, and S. However, these models need to be confirmed with functional analyses due to they may not always provide confirmatory results. This is the case of C1 alleles such as HLA-C*08 and-C*12, which have also been related to a better cytotoxic response against SARS-CoV-2 [70] and, accordingly, the presence of HLA-C*08:02 and -C*12:03 was significantly higher in individuals with mild COVID-19 from our cohort. Our predictive SARS-CoV-2 epitope analyses confirmed that HLA-C*12:03 allele was able to present a high number of verified epitopes, whereas it could not be confirmed for HLA-C*08:02, at least for the epitopes from proteins M, N, and S. Therefore, we cannot rule out that HLA-C*08:02 may present efficiently epitopes from other less abundant proteins.

The HLA-C molecules may participate in the activation or inhibition of NK cells by acting as ligands for KIRs [25]. To our knowledge, the contribution of the association between specific HLA-C alleles and KIRs to the clinical presentations of COVID-19 has not been determined yet. Peptides presented by HLA-C alleles from group C1 that shows Asn80 in the heavy chain (HLA-CAsn80) would modulate the activation of NK cells through the interaction with inhibitory KIR2DL2 and KIR2DL3, but also with the activating KIR2DS2 [71]. Due to C1 alleles HLA-C*08:02, -C*12:03, and -C*16:01 were significantly more frequent in the group with mild COVID-19 and they have been associated to a protective effect during COVID-19, this may suggest that NK cells from these patients could be more efficient to clear the infection by SARS-CoV-2. In fact, a higher presence of C1 alleles has also been described in patients able to clear spontaneously the infection with hepatitis C virus [72]. On the contrary, the presence of C2 alleles (HLA-CLys80) such as HLA-C*07:01 and -C*06:02, which are ligands for the inhibitory KIR2DL1 and activating KIR2DS1 [30], would theoretically lead to lower NK inhibition and higher activation would be more beneficial for the clearance of viral infections [73]. Therefore, we hypothesized that KIR2DL1/HLA-C group C2 interactions would confer strong inhibitory responses [74,75], at least in those individuals with critical COVID-19 from our cohort that were homozygous for HLA-C group C2 alleles and of whom we previously described that they show an impaired cytotoxic response against SARS-CoV-2 mostly based on high levels of NK, NKT, and CD8+ T cells that display immune exhaustion markers and poor cytotoxic functionality [8].

We also determined that changes of amino acid residues at essential positions in the binding pocket of HLA-C alleles of group C1 (HLA-C*08:02, which was significantly more expressed in individuals with mild COVID-19) and C2 (HLA-C*07:01) modified the groove structure, changing the positions of hydrogen-type bonds and amino acids interactions within residues between both molecules. All these changes would undoubtedly affect the molecule stability, thereby influencing the quality of the immune response induced by these molecules. Moreover, it has been described that alleles of group C1 such as HLA-C*08 display greater stability than alleles of group C2 such as HLA-C*07 [76], most likely due to the reduced ability of HLA-C*07 to present a large range of peptides, which is essential to stabilize the trimeric complexes of HLA-C/β2M/peptide on the cell surface [77]. In fact, HLA-C*07 is considered a low expressed variant with low β2M/peptide binding stability [76] and individuals with this HLA-C allotype usually present cells that express in the surface a large pool of HLA-C free heavy chains, unable to present peptides to the T cells [78]. On the contrary, individuals with more stable HLA-C allotypes such as HLA-C*08 present cells that express on the surface a greater quantity of HLA-C/β2M/peptide heterotrimers able to induce an efficient antiviral response. Therefore, the presence of more stable HLA-C alleles of group C1 in individuals with mild COVID-19 could contribute to their more efficient immune response, which was characterized by a higher cytotoxic activity against SARS-CoV-2 infected cells [8].

In conclusion, the great variability in the clinical presentations of COVID-19 appears to be consequence of several host factors, such as age, gender, and the presence of comorbidities. However, other factors such as an impaired ability to present viral peptides to the immune cells due to unstable peptide-HLA molecule conformations may also be considered, mostly because an impaired cellular immune response has also been related with a poor prognosis during COVID-19. In this observational pilot study, we determined that a small cohort of Spanish Mediterranean Caucasian individuals with a mild or asymptomatic presentations of COVID-19 expressed HLA alleles with potentially protective effect such as highly stable HLA-C alleles of group C1 such as HLA-C*08:02, -C*12:03, and -C*16:01. On the contrary, 47% of individuals with critical COVID-19 were homozygous for HLA-C2 alleles, which are more unstable and could be associated to a poorer disease prognosis due to their lower efficiency to present SARS-CoV-2 peptides to CD8+ T cells and/or activate NK cells.

One main limitation of our study was the relatively small size of the cohort. Despite of this, we observed statistical significance in the comparisons between groups about the frequency of the associations between HLA-C alleles of group C1. In addition, the size of our cohort is similar to other studies previously published [44,45,79,80] that also validate a correlation between HLA-C and an increased COVID-19 severity in the Spanish population [44,45]. Therefore, these data may help us to get further understanding of the underlying mechanisms of COVID-19 pathogenesis, although they must be taken with caution until they are confirmed in a larger cohort. Importantly, to our knowledge, this is the first study that describes a relationship between the presence of specific HLA-C alleles and the different presentations and prognosis of COVID-19. Further analyses will be necessary to determine the actual role of HLA-C locus as a predictive biomarker for COVID-19 progression.

Supporting information

S1 Fig. Distribution of Bw4 and Bw6 genotypes in our cohort of individuals with COVID-19.

HLA-B alleles are divided into HLA-Bw6 (Ser77 and Asn80) and -Bw4 (characterized by at least seven different patterns of amino acid changes at positions 77 and 80–83), based on their serological reaction. Statistical significance was calculated using Fisher’s exact test. Significance levels were corrected by Bonferroni correction.

https://doi.org/10.1371/journal.pone.0272867.s001

(TIF)

S2 Fig. Analysis of the frequency of HLA-C1 and -C2 alleles in the cohort.

Distribution of alleles from HLA-C1 (HLA-C Asn80) and -C2 (HLA-C Lys80) groups in the individuals with mild, severe and critical COVID-19. The numbers above the bars indicate the percentage of HLA-C alleles’ frequency for each group of individuals. The statistical significance of the comparisons between the allele frequencies of groups of individuals with severe and critical COVID-19 and the group of individuals with mild COVID-19 was calculated using Fisher’s exact test 2x2. Significance levels were corrected by Bonferroni correction.

https://doi.org/10.1371/journal.pone.0272867.s002

(TIF)

S3 Fig. Interactions between HLA-CAsn80 and HLA-CLys80 and specific KIR alleles.

Interactions between HLA-C alleles from group C1 (HLA-CAsn80) and KIR2DL2, KIR2DL3 and KIR2DS2 (A), or between HLA-C alleles from group C2 (HLA-CLys80) and KIR2DL1 and KIR2DS1 (B) were evaluated in individuals with different presentations of COVID-19. Statistical significance was calculated using Fisher’s exact test. Significance levels were corrected by Bonferroni correction.

https://doi.org/10.1371/journal.pone.0272867.s003

(TIF)

S1 Table. Demographic and clinical characteristics of homebound patients with mild COVID-19 who were recruited for this study at the Primary Healthcare Center Laín Entralgo (Alcorcón, Madrid, Spain).

https://doi.org/10.1371/journal.pone.0272867.s004

(DOCX)

S2 Table. Demographic and clinical characteristics of hospitalized patients with severe and critical COVID-19 who were recruited for this study at the Hospital Universitario Ramon y Cajal, Hospital Universitario Puerta de Hierro and Hospital de El Escorial (Madrid, Spain).

https://doi.org/10.1371/journal.pone.0272867.s005

(DOCX)

Acknowledgments

We greatly appreciate all the individuals who participate in this study. We thank Daniel Fuertes (School of Telecommunications Engineering, Universidad Politécnica, Madrid, Spain) and Dr Amanda Fernández-Rodriguez (National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain) for the revision of all statistical analyses performed in this study. We also thank the excellent secretarial assistance of Ms. Olga Palao. The full membership list of the Contributing members of the Multidisciplinary Group of Study of COVID-19 (MGS-COVID) is as follows:

1Centro de Salud Doctor Pedro Laín Entralgo, Alcorcón, Spain

2Hematology Service, Hospital Universitario Ramón y Cajal, Madrid, Spain

3Infectious Diseases Service, Hospital Universitario Ramón y Cajal, Madrid, Spain

4Neumology Service, Hospital Universitario Puerta de Hierro, Majadahonda, Spain

5Intensive Medicine Service, Hospital Universitario Ramón y Cajal, Madrid, Spain

#Contributing members of the Multidisciplinary Group of Study of COVID-19 (MGS-COVID) (in alphabetical order):

Esther Alonso Herrador1, Pablo Amich Alemany1, Victoria Bosch Martos1, Sandra Chamorro2, Belén Comeche3, Lorena Cordova Castaño1, Susana Domínguez-Mateos1, Aurora Expósito Mora1, Valle Falcones4, María Mercedes Gea Martinez1, Alberto Gomez Bonilla1, María Victoria Leon Gomez1, Gema Lora Rey1, Alejandro Luna de Abia2, Patricia Mínguez4, Maria Luisa Muñoz Balsa1, Javier Pérez Gonzalez1, Sandra Pérez-Santos1, Jose Sanchez Hernández1, Cruz Soriano5, Andrea Vinssac Rayado1.

References

  1. 1. (WHO) TWHO. WHO Coronavirus (COVID-19) Dashboard 2022 [Available from: https://covid19.who.int/ (accessed on 06 January 2022).
  2. 2. Kandeel M, Mohamed MEM, Abd El-Lateef HM, Venugopala KN, El-Beltagi HS. Omicron variant genome evolution and phylogenetics. J Med Virol. 2021. pmid:34888894
  3. 3. Fauci AS, Lane HC, Redfield RR. Covid-19—Navigating the Uncharted. N Engl J Med. 2020;382(13):1268–9. pmid:32109011
  4. 4. Barton LM, Duval EJ, Stroberg E, Ghosh S, Mukhopadhyay S. COVID-19 Autopsies, Oklahoma, USA. Am J Clin Pathol. 2020;153(6):725–33. pmid:32275742
  5. 5. Yao XH, Li TY, He ZC, Ping YF, Liu HW, Yu SC, et al. [A pathological report of three COVID-19 cases by minimal invasive autopsies]. Zhonghua Bing Li Xue Za Zhi. 2020;49(5):411–7. pmid:32172546
  6. 6. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–13. pmid:32007143
  7. 7. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. pmid:31986264
  8. 8. Vigón L, Fuertes D, García-Pérez J, Torres M, Rodríguez-Mora S, Mateos E, et al. Impaired Cytotoxic Response in PBMCs From Patients With COVID-19 Admitted to the ICU: Biomarkers to Predict Disease Severity. Front Immunol. 2021;12:665329. pmid:34122423
  9. 9. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell. 2020;181(5):1036–45.e9. pmid:32416070
  10. 10. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130(5):2620–9. pmid:32217835
  11. 11. Vigón L, García-Pérez J, Rodríguez-Mora S, Torres M, Mateos E, Castillo de la Osa M, et al. Impaired Antibody-Dependent Cellular Cytotoxicity in a Spanish Cohort of Patients With COVID-19 Admitted to the ICU. Front Immunol. 2021;12:742631. pmid:34616404
  12. 12. Group RC, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med. 2021;384(8):693–704. pmid:32678530
  13. 13. Parums DV. Editorial: Current Status of Oral Antiviral Drug Treatments for SARS-CoV-2 Infection in Non-Hospitalized Patients. Med Sci Monit. 2022;28:e935952. pmid:34972812
  14. 14. Couzin-Frankel J. Antiviral pills could change pandemic’s course. Science. 2021;374(6569):799–800. pmid:34762459
  15. 15. Migliorini F, Torsiello E, Spiezia F, Oliva F, Tingart M, Maffulli N. Association between HLA genotypes and COVID-19 susceptibility, severity and progression: a comprehensive review of the literature. Eur J Med Res. 2021;26(1):84. pmid:34344463
  16. 16. Janeway CA TP Jr, Walport M, Shlomchik MJ. The major histocompatibility complex and its functions. Immunobiology: The Immune System in Health and Disease: New York: Garland Science; 2001.
  17. 17. Shiina T, Hosomichi K, Inoko H, Kulski JK. The HLA genomic loci map: expression, interaction, diversity and disease. J Hum Genet. 2009;54(1):15–39. pmid:19158813
  18. 18. Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. Structure of the human class I histocompatibility antigen, HLA-A2. Nature. 1987;329(6139):506–12. pmid:3309677
  19. 19. Challa-Malladi M, Lieu YK, Califano O, Holmes AB, Bhagat G, Murty VV, et al. Combined genetic inactivation of beta2-Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell. 2011;20(6):728–40. pmid:22137796
  20. 20. Cruz-Tapias P CJ, Anaya JM. Major histocompatibility complex: Antigen processing and presentation 2013. Available from: https://www.ncbi.nlm.nih.gov/books/NBK459467/.
  21. 21. Joffre OP, Segura E, Savina A, Amigorena S. Cross-presentation by dendritic cells. Nat Rev Immunol. 2012;12(8):557–69. pmid:22790179
  22. 22. Allard M, Oger R, Benlalam H, Florenceau L, Echasserieau K, Bernardeau K, et al. Soluble HLA-I/peptide monomers mediate antigen-specific CD8 T cell activation through passive peptide exchange with cell-bound HLA-I molecules. J Immunol. 2014;192(11):5090–7. pmid:24752447
  23. 23. Leddon SA, Sant AJ. Generation of MHC class II-peptide ligands for CD4 T-cell allorecognition of MHC class II molecules. Curr Opin Organ Transplant. 2010;15(4):505–11. pmid:20616724
  24. 24. Hamerman JA, Ogasawara K, Lanier LL. NK cells in innate immunity. Curr Opin Immunol. 2005;17(1):29–35. pmid:15653307
  25. 25. Uhrberg M, Valiante NM, Shum BP, Shilling HG, Lienert-Weidenbach K, Corliss B, et al. Human diversity in killer cell inhibitory receptor genes. Immunity. 1997;7(6):753–63. pmid:9430221
  26. 26. Robinson J, Barker DJ, Georgiou X, Cooper MA, Flicek P, Marsh SGE. IPD-IMGT/HLA Database. Nucleic Acids Res. 2020;48(D1):D948–D55. pmid:31667505
  27. 27. Montero-Martín G, Mallempati KC, Gangavarapu S, Sánchez-Gordo F, Herrero-Mata MJ, Balas A, et al. High-resolution characterization of allelic and haplotypic HLA frequency distribution in a Spanish population using high-throughput next-generation sequencing. Hum Immunol. 2019;80(7):429–36. pmid:30763600
  28. 28. Müller CA, Engler-Blum G, Gekeler V, Steiert I, Weiss E, Schmidt H. Genetic and serological heterogeneity of the supertypic HLA-B locus specificities Bw4 and Bw6. Immunogenetics. 1989;30(3):200–7. pmid:2777338
  29. 29. Biassoni R, Falco M, Cambiaggi A, Costa P, Verdiani S, Pende D, et al. Amino acid substitutions can influence the natural killer (NK)-mediated recognition of HLA-C molecules. Role of serine-77 and lysine-80 in the target cell protection from lysis mediated by "group 2" or "group 1" NK clones. J Exp Med. 1995;182(2):605–9. pmid:7629517
  30. 30. Faridi RM, Agrawal S. Killer immunoglobulin-like receptors (KIRs) and HLA-C allorecognition patterns implicative of dominant activation of natural killer cells contribute to recurrent miscarriages. Human Reproduction. 2010;26(2):491–7. pmid:21159685
  31. 31. Blais ME, Dong T, Rowland-Jones S. HLA-C as a mediator of natural killer and T-cell activation: spectator or key player? Immunology. 2011;133(1):1–7. pmid:21355865
  32. 32. Blackwell JM, Jamieson SE, Burgner D. HLA and infectious diseases. Clin Microbiol Rev. 2009;22(2):370–85, Table of Contents. pmid:19366919
  33. 33. Matzaraki V, Kumar V, Wijmenga C, Zhernakova A. The MHC locus and genetic susceptibility to autoimmune and infectious diseases. Genome Biol. 2017;18(1):76. pmid:28449694
  34. 34. Bailey JR, Williams TM, Siliciano RF, Blankson JN. Maintenance of viral suppression in HIV-1-infected HLA-B*57+ elite suppressors despite CTL escape mutations. J Exp Med. 2006;203(5):1357–69. pmid:16682496
  35. 35. Descours B, Avettand-Fenoel V, Blanc C, Samri A, Mélard A, Supervie V, et al. Immune responses driven by protective human leukocyte antigen alleles from long-term nonprogressors are associated with low HIV reservoir in central memory CD4 T cells. Clin Infect Dis. 2012;54(10):1495–503. pmid:22441653
  36. 36. Apps R, Qi Y, Carlson JM, Chen H, Gao X, Thomas R, et al. Influence of HLA-C expression level on HIV control. Science. 2013;340(6128):87–91. pmid:23559252
  37. 37. Ladel CH, Daugelat S, Kaufmann SH. Immune response to Mycobacterium bovis bacille Calmette Guérin infection in major histocompatibility complex class I- and II-deficient knock-out mice: contribution of CD4 and CD8 T cells to acquired resistance. Eur J Immunol. 1995;25(2):377–84. pmid:7875199
  38. 38. Rolph MS, Raupach B, Köbernick HH, Collins HL, Pérarnau B, Lemonnier FA, et al. MHC class Ia-restricted T cells partially account for beta2-microglobulin-dependent resistance to Mycobacterium tuberculosis. Eur J Immunol. 2001;31(6):1944–9. pmid:11433392
  39. 39. Dutta M, Dutta P, Medhi S, Borkakoty B, Biswas D. Polymorphism of HLA class I and class II alleles in influenza A(H1N1)pdm09 virus infected population of Assam, Northeast India. J Med Virol. 2018;90(5):854–60. pmid:29315655
  40. 40. Ng MH, Lau KM, Li L, Cheng SH, Chan WY, Hui PK, et al. Association of human-leukocyte-antigen class I (B*0703) and class II (DRB1*0301) genotypes with susceptibility and resistance to the development of severe acute respiratory syndrome. J Infect Dis. 2004;190(3):515–8. pmid:15243926
  41. 41. Lin M, Tseng HK, Trejaut JA, Lee HL, Loo JH, Chu CC, et al. Association of HLA class I with severe acute respiratory syndrome coronavirus infection. BMC Med Genet. 2003;4:9. pmid:12969506
  42. 42. Wang SF, Chen KH, Chen M, Li WY, Chen YJ, Tsao CH, et al. Human-leukocyte antigen class I Cw 1502 and class II DR 0301 genotypes are associated with resistance to severe acute respiratory syndrome (SARS) infection. Viral Immunol. 2011;24(5):421–6. pmid:21958371
  43. 43. Correale P, Mutti L, Pentimalli F, Baglio G, Saladino RE, Sileri P, et al. HLA-B*44 and C*01 Prevalence Correlates with Covid19 Spreading across Italy. Int J Mol Sci. 2020;21(15). pmid:32717807
  44. 44. Iturrieta-Zuazo I, Rita CG, Garcia-Soidan A, de Malet Pintos-Fonseca A, Alonso-Alarcon N, Pariente-Rodriguez R, et al. 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. pmid:32810602
  45. 45. Lorente L, Martin MM, Franco A, Barrios Y, Caceres JJ, Sole-Violan J, et al. [HLA genetic polymorphisms and prognosis of patients with COVID-19]. Med Intensiva (Engl Ed). 2021;45(2):96–103. pmid:32988645
  46. 46. Kontis V, Bennett JE, Rashid T, Parks RM, Pearson-Stuttard J, Guillot M, et al. Magnitude, demographics and dynamics of the effect of the first wave of the COVID-19 pandemic on all-cause mortality in 21 industrialized countries. Nat Med. 2020;26(12):1919–28. pmid:33057181
  47. 47. Sanitarias. CdCdAyE. Actualización no 180, 07.08.2020, SITUACIÓN EN ESPAÑA 2020 [Available from: https://www.mscbs.gob.es/profesionales/saludPublica/ccayes/alertasActual/nCov/documentos/Actualizacion_180_COVID-19.pdf.
  48. 48. Excoffier L, Lischer HE. Arlequin suite ver 3.5: a new series of programs to perform population genetics analyses under Linux and Windows. Mol Ecol Resour. 2010;10(3):564–7. pmid:21565059
  49. 49. Arnaiz-Villena A, Martínez-Laso J, Gómez-Casado E, Díaz-Campos N, Santos P, Martinho A, et al. Relatedness among Basques, Portuguese, Spaniards, and Algerians studied by HLA allelic frequencies and haplotypes. Immunogenetics. 1997;47(1):37–43. pmid:9382919
  50. 50. Reynisson B, Alvarez B, Paul S, Peters B, Nielsen M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 2020;48(W1):W449–W54. pmid:32406916
  51. 51. Vita R, Mahajan S, Overton JA, Dhanda SK, Martini S, Cantrell JR, et al. The Immune Epitope Database (IEDB): 2018 update. Nucleic Acids Res. 2019;47(D1):D339–D43. pmid:30357391
  52. 52. Available from: https://www.ebi.ac.uk/ols/ontologies/doid/terms?obo_id=DOID:0080600.
  53. 53. Moradi S, Rossjohn J., Vivian J.P. Killer cell immunoglobulin-like receptor 2DL2 in complex with HLA-C*07:02. 2019.
  54. 54. Fortier MH, Caron E, Hardy MP, Voisin G, Lemieux S, Perreault C, et al. The MHC class I peptide repertoire is molded by the transcriptome. J Exp Med. 2008;205(3):595–610. pmid:18299400
  55. 55. Grenga L, Gallais F, Pible O, Gaillard JC, Gouveia D, Batina H, et al. Shotgun proteomics analysis of SARS-CoV-2-infected cells and how it can optimize whole viral particle antigen production for vaccines. Emerg Microbes Infect. 2020;9(1):1712–21. pmid:32619390
  56. 56. The Lancet Public H. COVID-19 in Spain: a predictable storm? Lancet Public Health. 2020;5(11):e568. pmid:33075295
  57. 57. Casas-Rojo JM, Anton-Santos JM, Millan-Nunez-Cortes J, Lumbreras-Bermejo C, Ramos-Rincon JM, Roy-Vallejo E, et al. Clinical characteristics of patients hospitalized with COVID-19 in Spain: Results from the SEMI-COVID-19 Registry. Rev Clin Esp (Barc). 2020;220(8):480–94.
  58. 58. Mallapaty S. The coronavirus is most deadly if you are older and male—new data reveal the risks. Nature. 2020;585(7823):16–7. pmid:32860026
  59. 59. Anastassopoulou C, Gkizarioti Z, Patrinos GP, Tsakris A. Human genetic factors associated with susceptibility to SARS-CoV-2 infection and COVID-19 disease severity. Hum Genomics. 2020;14(1):40. pmid:33092637
  60. 60. Bardeskar NS, Mania-Pramanik J. HIV and host immunogenetics: unraveling the role of HLA-C. HLA. 2016;88(5):221–31. pmid:27620973
  61. 61. Tavasolian F, Rashidi M, Hatam GR, Jeddi M, Hosseini AZ, Mosawi SH, et al. HLA, Immune Response, and Susceptibility to COVID-19. Front Immunol. 2020;11:601886. pmid:33488597
  62. 62. Adams SM, Bosch E, Balaresque PL, Ballereau SJ, Lee AC, Arroyo E, et al. The genetic legacy of religious diversity and intolerance: paternal lineages of Christians, Jews, and Muslims in the Iberian Peninsula. Am J Hum Genet. 2008;83(6):725–36. pmid:19061982
  63. 63. Jin JM, Bai P, He W, Wu F, Liu XF, Han DM, et al. Gender Differences in Patients With COVID-19: Focus on Severity and Mortality. Front Public Health. 2020;8:152. pmid:32411652
  64. 64. Leite MM, Gonzalez-Galarza FF, Silva BCCD, Middleton D, EJMD. Predictive immunogenetic markers in COVID-19. Hum Immunol. 2021;82(4):247–54. pmid:33546902
  65. 65. Martinez-Laso J, De Juan D, Martinez-Quiles N, Gomez-Casado E, Cuadrado E, Arnaiz-Villena A. The contribution of the HLA-A, -B, -C and -DR, -DQ DNA typing to the study of the origins of Spaniards and Basques. Tissue Antigens. 1995;45(4):237–45. pmid:7638859
  66. 66. Olaisen B, Teisberg P, Jonassen R, Thorsby E, Gedde-Dahl T Jr., Gene order and gene distances in the HLA region studied by the haplotype method. Ann Hum Genet. 1983;47(4):285–92. pmid:6418058
  67. 67. Santos S, Balas A, Lillo R, Garcia-Sanchez F, Merino JL, Vicario JL. HLA-B14 subtyping by semi-nested PCR-SSP and haplotype distribution in a Spanish population. Tissue Antigens. 1997;50(6):671–4. pmid:9458126
  68. 68. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–58. pmid:23516982
  69. 69. Rizzo R, Gentili V, Rotola A, Bortolotti D, Cassai E, Di Luca D. Implication of HLA-C and KIR alleles in human papillomavirus infection and associated cervical lesions. Viral Immunol. 2014;27(9):468–70. pmid:25188020
  70. 70. Barquera R, Collen E, Di D, Buhler S, Teixeira J, Llamas B, et al. Binding affinities of 438 HLA proteins to complete proteomes of seven pandemic viruses and distributions of strongest and weakest HLA peptide binders in populations worldwide. HLA. 2020;96(3):277–98. pmid:32475052
  71. 71. Fadda L, Borhis G, Ahmed P, Cheent K, Pageon SV, Cazaly A, et al. Peptide antagonism as a mechanism for NK cell activation. Proc Natl Acad Sci U S A. 2010;107(22):10160–5. pmid:20439706
  72. 72. Khakoo SI, Thio CL, Martin MP, Brooks CR, Gao X, Astemborski J, et al. HLA and NK cell inhibitory receptor genes in resolving hepatitis C virus infection. Science. 2004;305(5685):872–4. pmid:15297676
  73. 73. Kulkarni S, Martin MP, Carrington M. The Yin and Yang of HLA and KIR in human disease. Semin Immunol. 2008;20(6):343–52. pmid:18635379
  74. 74. Winter CC, Gumperz JE, Parham P, Long EO, Wagtmann N. Direct binding and functional transfer of NK cell inhibitory receptors reveal novel patterns of HLA-C allotype recognition. J Immunol. 1998;161(2):571–7. pmid:9670929
  75. 75. Ahlenstiel G, Martin MP, Gao X, Carrington M, Rehermann B. Distinct KIR/HLA compound genotypes affect the kinetics of human antiviral natural killer cell responses. J Clin Invest. 2008;118(3):1017–26. pmid:18246204
  76. 76. Sibilio L, Martayan A, Setini A, Monaco EL, Tremante E, Butler RH, et al. A single bottleneck in HLA-C assembly. J Biol Chem. 2008;283(3):1267–74. pmid:17956861
  77. 77. Kaur G, Gras S, Mobbs JI, Vivian JP, Cortes A, Barber T, et al. Structural and regulatory diversity shape HLA-C protein expression levels. Nat Commun. 2017;8:15924. pmid:28649982
  78. 78. Parolini F, Biswas P, Serena M, Sironi F, Muraro V, Guizzardi E, et al. Stability and Expression Levels of HLA-C on the Cell Membrane Modulate HIV-1 Infectivity. J Virol. 2018;92(1).
  79. 79. 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(2):194–6. pmid:32424945
  80. 80. Abdelhafiz AS, Ali A, Fouda MA, Sayed DM, Kamel MM, Kamal LM, et al. HLA-B*15 predicts survival in Egyptian patients with COVID-19. Hum Immunol. 2021. pmid:34607724