Next Article in Journal
Ectopic Overexpression of Pineapple Transcription Factor AcWRKY31 Reduces Drought and Salt Tolerance in Rice and Arabidopsis
Next Article in Special Issue
Evaluation of the Impact of Pregnancy-Associated Factors on the Quality of Wharton’s Jelly-Derived Stem Cells Using SOX2 Gene Expression as a Marker
Previous Article in Journal
Centronuclear Myopathy Caused by Defective Membrane Remodelling of Dynamin 2 and BIN1 Variants
Previous Article in Special Issue
Leydig Cells in Immunocastrated Polish Landrace Pig Testis: Differentiation Status and Steroid Enzyme Expression Status
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Beyond GWAS—Could Genetic Differentiation within the Allograft Rejection Pathway Shape Natural Immunity to COVID-19?

1
Biostatistics Group, Department of Genetics, Wrocław University of Environmental and Life Sciences, 51-631 Wroclaw, Poland
2
Department of Cattle Breeding, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
3
Central Clinical Hospital of Ministry of the Interior and Administration in Warsaw, 02-507 Warsaw, Poland
4
Department of Haematology, Transplantation and Internal Medicine, University Clinical Centre of the Medical University of Warsaw, 02-091 Warsaw, Poland
5
MNM Diagnostics, Małe Garbary 9, 61-756 Poznan, Poland
6
Department of Neurology and Neurophysiology, University of Zurich, Balgrist University Hospital, 8008 Zurich, Switzerland
7
Department of Infectious Diseases, Medical University of Lublin, 20-059 Lublin, Poland
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 6272; https://doi.org/10.3390/ijms23116272
Submission received: 19 April 2022 / Revised: 26 May 2022 / Accepted: 30 May 2022 / Published: 3 June 2022

Abstract

:
COVID-19 infections pose a serious global health concern so it is crucial to identify the biomarkers for the susceptibility to and resistance against this disease that could help in a rapid risk assessment and reliable decisions being made on patients’ treatment and their potential hospitalisation. Several studies investigated the factors associated with severe COVID-19 outcomes that can be either environmental, population based, or genetic. It was demonstrated that the genetics of the host plays an important role in the various immune responses and, therefore, there are different clinical presentations of COVID-19 infection. In this study, we aimed to use variant descriptive statistics from GWAS (Genome-Wide Association Study) and variant genomic annotations to identify metabolic pathways that are associated with a severe COVID-19 infection as well as pathways related to resistance to COVID-19. For this purpose, we applied a custom-designed mixed linear model implemented into custom-written software. Our analysis of more than 12.5 million SNPs did not indicate any pathway that was significant for a severe COVID-19 infection. However, the Allograft rejection pathway (hsa05330) was significant (p = 0.01087) for resistance to the infection. The majority of the 27 SNP marking genes constituting the Allograft rejection pathway were located on chromosome 6 (19 SNPs) and the remainder were mapped to chromosomes 2, 3, 10, 12, 20, and X. This pathway comprises several immune system components crucial for the self versus non-self recognition, but also the components of antiviral immunity. Our study demonstrated that not only single variants are important for resistance to COVID-19, but also the cumulative impact of several SNPs within the same pathway matters.

1. Introduction

COVID-19 infections pose a serious global health concern, so it remains crucial to identify the biomarkers for the susceptibility to and resistance against this disease that could help in a rapid risk assessment and reliable decisions being made on patients’ treatment and their potential hospitalisation. The knowledge of genomic variants that are associated with the susceptibility to and resistance against COVID-19 infections is essential for understanding the metabolic pathways and regulatory factors related to different presentations of the disease and, in consequence, for developing new drug targets [1].
Several studies investigated factors associated with severe COVID-19 outcomes that can be either environmental, population based, or genetic. For instance, a geographic and population variation in the disease course and outcome has demonstrated that individuals representing Black, Hispanic, or Asian ethnicities have a higher risk of death compared to Caucasians [2,3]. The identified risk factors of COVID-19 infection are advanced age, male sex, and comorbidities (especially: renal disease, oncological pathologies, chronic respiratory disease, and cardiovascular disease—excluding hypertension and dementia), but they do not fully explain the wide spectrum of disease manifestations [2,4,5,6]. Moreover, the viral load can play a role in the COVID-19 severity, as demonstrated for mortality, especially in combination with advanced age [7,8]. On the other hand, many studies have supported the hypothesis that there are patients who have a natural resistance to COVID-19 infection. For instance, children or adolescents were barely affected [9] and they showed antibody responses up to four months after infection [10]. Thus, some children appear to be able to either repel the infection without the need to strongly engage adaptive immunity or are resistant to infection.
It was also demonstrated that the genetics of the host plays an important role in the various immune responses and, therefore, in the different clinical presentations of COVID-19 infection. Risk and protective variants were associated with multiple loci on chromosomes 3, 9, 6, 12, 19, and 21 [11,12] including a segment on chromosome 9 that defines the ABO blood groups [13], a segment on chromosome 12 that contains OAS1, OAS2, and OAS3 [14], and an isoform p41 of CD74, which is a part of the MHC-II that blocks the endosomal entry pathway of the virus [15]. Further significant genes were ACE2 located on chromosome X and human leukocyte antigens (HLA).
The main aim of our study was to use variant descriptive statistics from GWAS and variant genomic annotations to identify metabolic pathways that are associated with a severe COVID-19 infection as well as pathways related to resistance to COVID-19. For this purpose, we applied a custom-designed mixed linear model implemented in custom-written software.

2. Results

The raw number of SNPs amounted to 43,469,928; out of these, after filtering, 9,767,423 SNPs remained for GWAS analysis (Figure 1). The most significant SNP from GWAS was located on chromosome 7 (p = 7.10547 × 10−9) within the intron of RAPGEF5 gene (ENSG00000136237); however, this gene is not assigned to any KEGG pathway. In total, 12,736 SNPs were included in the pathway model. Out of the 288 KEGG pathway effects predicted, none were significant in the SEVERE analysis, while the Allograft rejection pathway (hsa05330) was significant (p = 0.01087) in the RESISTANT analysis. All 27 SNPs, each representing one gene constituting hsa05330, were significant in GWAS (Table 1, Figure 2). The majority of them were located on chromosome 6 (19 SNPs) and the remainder were mapped to chromosomes 2, 3, 10, 12, 20, and X. The summary of read depth quality underlying each variant in each sample is summarised in Supplementary Figure S1. Interestingly, the comparison of p-values of all SNPs from GWAS with p-values of the subset of SNPs included in the KEGG model showed that genic SNPs have lower p-values than intergenic SNPs (Figure 3).

3. Discussion

An important property of the human immune system is natural resistance, defined as the inability of pathogens, viruses included, to infect the host or as an ability of the host to limit the disease burden after infection [16]. Two mechanisms of natural resistance have been suggested [17]. The first mechanism comprises genetic variants in genes important for entry mechanisms, especially in the receptors. The second mechanism is the rapid elimination of the pathogen by host resistance mechanisms. As for the host’s survival, the balance between resistance mechanisms aiming to eliminate host pathogens and tolerance mechanisms aiming to avoid collateral damage induced by inflammation is crucial; natural resistance is mostly related to the immune system, either to its robustness or on the genetic level [17].
In our study, we used GWAS and custom-designed models to identify molecular pathways related to different presentations of COVID-19 infections and to describe significant genetic variants within these pathways. Interestingly, the comparison between the full dataset (all SNPs analysed in GWAS) and the subset used in the estimation of KEGG pathway effects, demonstrated that the majority of most significant p-values were assigned to intergenic SNPs (Figure 3), which can only be used as markers for a potential association with genes, but not as causal mutations. Moreover, although the most common cause of such a significant association is linkage, it is not exclusive proof that an associated SNP always points to the closest located gene as the functional cause of a phenotype (see, e.g., [18]). It was demonstrated that SNP density across the human genome is reduced in conserved regions [19]. However, both exonic and intronic regions are constrained to the same degree and they contain a reduced density of SNPs in comparison to intergenic regions [19]. Therefore, the custom-designed models for the KEGG pathways’ analysis used in this study allowed us to consider additional aspects of the genetic background in COVID-19 infections that were not identified in single SNP GWAS analyses.
Our analysis did not indicate any significant pathway in the case of a severe COVID-19 phenotype. However, in the case of a resistant phenotype, the Allograft rejection pathway (hsa05330) was identified as significant, with 27 SNPs marking genes constituting this pathway. The Allograft rejection pathway (Figure 4), associated with resistance to COVID-19 infection, comprises several immune system components crucial for self versus non-self recognition, but also the components of antiviral immunity [20]. Molecules involved in this pathway play pivotal roles in many other physiological and pathological processes, including allergies or even an anticancer response [20]. For all those processes the interplay between at least two cells is crucial, one of which remains the immune cell. The majority of the significant SNPs from the Allograft rejection pathway marked genes from the HLA complex (18 SNPs on chromosome 6, Table 1) that encode proteins forming the major histocompatibility complex (MHC), and several of them were already indicated as proteins protective against COVID-19 infection [21].
Considering the genomic location of the 27 SNPs from the Allograft rejection pathway (Table 1), five SNPs were identified in CD (cluster of differentiation—classification determinant of immune cells) protein-coding genes, as depicted in the table below (Table 2). CD40 is a member of the TNF superfamily, constitutively expressed on B cells and APC cells [22,23]. CD40 binds its ligand CD40L, which is transiently expressed on T cells and other non-immune cells under inflammatory conditions [24]. CD28 is expressed on T cells, providing signals required for T cell activation and survival, and CD80 and CD86 are its ligands [25,26,27,28]. The activity of CD80–CD28 complex stimulates the activation of transcription factors NF-κB, promoting IL-2 production [22,26,29,30]. CD80 is also a ligand for cytotoxic T-lymphocyte antigen 4 (CTLA-4, also known as CD152), which remains constitutively expressed on most of the T cells, for example, on Tregs, in which CTLA-4 expression increases upon activation [25,27,29]. CD28 competes with CTLA-4 for binding to CD80 and CD86 [26,29,31]. Interestingly, CD80 and CD86 proteins may act as receptors for some adenoviruses [22]. Malfunctioning CD80 molecules are also involved in some pathological conditions, such as lupus erythematosus. CD86 is also associated with myocarditis and gallbladder squamous cell carcinoma [22].
One SNP on chromosome 12 pointed out interferon IFN-gamma, which is a cytokine produced mostly by activated T cells and NK cells and that can induce MHC expression, which makes it an important factor in transplantation. IFN plays a dual role in COVID-19 infections, on one side aggravating the symptoms, and on the other alleviating the disease course, depending on the disease stage and types of the interferon involved and on the personal predispositions [31]. This occurred in some patients shown to be associated with higher mortality [31], whereas in others poor IFN responses were associated with a more severe disease course [32,33]. In immunosuppressed patients, IFN has a beneficial effect and was applied as an adjuvant treatment [34]. It was shown in mice models that a lack of IFNg led to the failure of microcirculation and necrosis of transplants, which suggested that IFNg modulates allogenic responses and could have a protective role in the early stages of the response to an organ allograft. On the other hand, IFNg promoted graft vessel disease at the later stages [35]. Therefore, the effect of IFNg on an organ allograft depended on the time after transplantation and graft type [36]. The pivotal role of IFNg in allograft rejection has been suggested also in humans. High rates of primary and secondary rejection after hematopoietic stem cell transplantation were reported in a cohort of eight HLA-identical paediatric patients [37]. Already at day +3 after hematopoietic stem cell transplantation in 15 children experiencing graft rejection, the levels of IFNg were significantly higher than in the control group, and IFNg has been suggested to be a marker of early graft rejection [38].
Unsurprisingly, the outcome of COVID-19 has been reported to be more severe in patients with co-existing pathologies, especially those associated with an impaired immune system [39,40]. The complex relationship existing between the immune system component, cancer, and COVID-19 brings about the possibility of continuing immune checkpoint inhibitors treatment among COVID-19 positive cancer patients, as well as the use of immunotherapy for the treatment of severe COVID-19 infection. In fact, several attempts have already been made, some of which are undoubtedly very promising [40,41,42]. It is known now that cancer patients have an increased risk of developing severe COVID-19 infection often leading to hospitalization and intensive care [43,44]. Given the fact that the SARS-CoV-2 virus might potentially trigger immune system over-reactivity in some patients, T cell exhaustion has also been observed in a subset of patients. Immune checkpoint inhibitors remain a useful tool used to modulate the immune reaction [43]. It is worth emphasizing that some COVID-19 patients exhibit lymphocytopenia and suffer from T cell exhaustion, a phenomenon very common in advanced cancer disease, which—in the case of COVID-19 infection—may lead to viral sepsis and an increased mortality rate [39,42,43]. It has been observed that in cancer patients, especially among immunocompromised individuals, treatment with the immune checkpoint inhibitors may restore their antitumoral immune response [42]. The use of immune checkpoint inhibitors in COVID-19 cases has been reviewed well in [42].
Despite the promising results, we must bear in mind the fact that severe COVID-19 might also be triggered by immune system aberrations, such as inborn errors of the interferon pathways [45]. The human immune system remains extremely complex and involves many genes and genomic elements, including those genes encoding cytokines: individuals that lack specific cytokines, such as interferons, or possessing gene alleles potentially impairing immune system functioning, can be more susceptible to certain infectious diseases, including COVID-19. In these people, immunotherapy should be administered with particular caution, including cancer patients.
The immunological synapse is an important element in the Allograft rejection pathway. The structure of the immunological synapse, i.e., the gap between the immune cell and another cell, such as antigen-presenting cells, APC, together with its complex molecular interactions and canonical, means the step-manner organisation has been known as “the bull’s eye structure”, with the central complex made of MHC-TCR (major histocompatibility complex–T cell receptor), leading to it providing the first signal [29,46,47,48,49]. The interaction between MHC and TCR controls the specificity and accuracy of the immune response, as the TCR genes undergo several complicated rearrangements similar to those known from antibody genes [46,48]. Furthermore, a central element of the synapse might be a directed secretion of soluble molecules into the synaptic cleft [46,50]. All other molecules, such as the LFA-1-ICAM adhesion complex, or checkpoint molecules, form distal rings and clusters around the MHC-TCR complex. The rings are required to modulate the response by providing secondary signals since the interaction between MHC and TCR is insufficient [46,49,51]. If no secondary signal is provided, the T cell becomes anergic or even undergoes apoptosis [49]. Summing up, at least two signals are required to activate a T cell: the first is an antigen recognition by the T cell receptor and MHC complex, the second is provided by the multitude of co-stimulatory and co-inhibitory receptors and ligands interactions, all of which are crucial in the allograft rejection pathway cell to cell interconnections [52]. Interestingly, studies have reported the relationship between anti-COVID-19 immunisation and acute corneal graft rejection, regardless of the vaccine or graft type [53,54,55,56]. Moreover, acute allograft rejection after immunisation was observed in the case of kidney and liver transplants [57]. However, a direct causative effect is hard to prove and needs further studies.
Furthermore, two SNPs were found on chromosome 10 (Table 1). The first of those SNPs was located within the Fas receptor (CD95), which plays an important role in the maintenance of immune tolerance. Fas-induced apoptosis (induced by Fas–FasL interactions) is involved in the cytotoxic activity of T cells and NK cells [58,59]. Studies conducted in mice models showed that mutations in Fas or FasL that inactivated their function led to disturbances in the immune response to infections with multiple different viruses, such as the influenza virus [60], herpes simplex viruses, mouse hepatitis virus or, mousepox virus [61]. Moreover, it was shown that Fas could play a role in the destruction of graft tissue. Although mechanisms for allograft injury remain unclear, the contribution of Fas and Fas ligand (FasL) was verified in the case of different commonly transplanted organs (e.g., liver, lungs, kidneys) that were probably targeted by FasL-expressing cytotoxic T lymphocytes [62]. In humans, it was shown in vivo that an increased sCD95 is related to the rejection in liver-transplanted recipients [63,64].
The most significant SNP from GWAS, marking the RAPGEF5 gene, with an outstanding p-value of 7.10547 × 10−9, was not assigned to any KEGG pathway. RAPGEF5 encodes Rap guanine nucleotide exchange factor 5 protein and belongs to the Ras family of GTPases that plays an important role in cell growth, differentiation, and malignant transformation [65]. The gene has not been associated with severe COVID infection in previous GWAS.

4. Material and Methods

4.1. Sample Collection

Blood samples were collected from 1235 individuals across Poland between April 2020 and April 2021. In our analysis, a subset of 1076 samples from unrelated individuals was used. For all individuals, basic clinical data including age, gender, BMI, and comorbidities (diabetes, hypertension, ischemic heart disease, stroke, heart failure, cancer, kidney disease, hepatitis B, chronic obstructive pulmonary disease) were collected. For some participants, additional data on genetic disorders, flu, tuberculosis, and measles vaccination status, smoking habits, as well as hepatitis C infection were ascertained. Only individuals without diagnosed severe health disorders (till the moment of sample collection), such as cancer, were qualified for this study. Within this cohort, the SEVERE group (N = 235) was composed of patients with severe, life-threatening outcomes of COVID-19 infection including respiratory insufficiency, requiring intensive medical care and artificial ventilation, and NEWS (National Early Warning Score) less than 5 [66]. The RESISTANT group (N = 306) was composed of volunteers who did not contract the disease or develop any symptoms despite being highly exposed to COVID-19. This group had multiple antibody blood-based tests conducted to confirm the lack of anti-SARS-CoV-2 antibodies.
Detailed information about the cohort, including demographic and clinical features, can be found in our previous paper by Kaja et al. [67].

4.2. Ethical Policy

All participants of this study provided informed consent before the collection of blood samples and clinical data. The ethical approval was granted by the Ethics Committee of the Central Clinical Hospital of the Ministry of Interior and Administration in Warsaw (decisions 41/2020 from 3 April 2020 and 125/2020 from 1 July 2020). The study complied with the 1964 Helsinki declaration and its later amendments and adhered to the highest data security standards 140 of ISO 27001 and the General Data Protection Regulation (GDPR) act.

4.3. Total Quality Management

The project was carried out following the Total Quality Management (TQM) methodology, which ensures the quality of results and analyzes the risk and possible difficulties of the planned methodology. TQM involves defining all critical points of the procedures: reference ranges for collected biological material, material preparation, DNA isolation, DNA concentration and quality, genome sequencing, and quality control of the data. The legal and ethical transparency of the entire project was ensured, including confidentiality, integrity, and impartiality of the data.

4.4. Whole Genome Sequencing

Whole genomes of 1076 unrelated participants were sequenced in this study. Four mL of K-EDTA peripheral blood from participants were collected according to the standardised Quality Management System protocol. Genomic DNA was isolated from the peripheral blood leukocytes using a QIAamp DNA Blood Mini Kit, Blood/Cell DNA Mini Kit (Syngen Biotech, Wrocław, Poland) and Xpure Blood Kit (A&A Biotechnology, Gdańsk, Poland) according to the manufacturers’ protocols. The concentration and purity of isolated DNA were measured using the NanoDropTM spectrophotometer and the quality of the DNA was evaluated using gel electrophoresis. The sequencing library was prepared by Macrogen Europe (Amsterdam, The Netherlands) using TruSeq DNA PCR-free kit (Illumina Inc., San Diego, CA, USA) and 550 bp inserts. The quality of DNA libraries was measured using 2100 Bioanalyzer, Agilent Technologies, Santa Clara, CA, USA. The Whole Genome Sequencing (WGS) was performed on the Illumina NovaSeq 6000 platform using 150 bp paired-end reads, yielding a mean depth of coverage of 35.26X in the cohort.

4.5. Pre-Processing of Whole Genome Sequence Data

The quality of sequenced reads was assessed using FastQC v0.11.7 (github.com/s-andrews/FastQC) and reads were subsequently mapped to the GRCh38 human reference genome using Speedseq framework v.0.1.2 [68], encompassing alignment with BWA-MEM 0.7.10 [69], using SAMBLASTER v0.1.22 [70] duplicate removal, and Sambamba v0.5.9 [71] for sorting and indexing. Mapping coverage was calculated for reads with MQ > 0 using Mosdepth v0.2.4 [71]. Single nucleotide variants in the nuclear genome were detected using DeepVariant v0.8.0 [72] and jointly genotyped with GLnexus v1.2.6-0-g4d057dc [73]. Next, multiallelic variant calls were decomposed into monoallelic and normalised using BCFtools v1.9 [74]. The raw SNP-set was edited by excluding SNPs that did not meet the following criteria: SNP call rate > 95%, p-value for HWE test > 0.0001, Minor Allele Frequency > 0.001, and being biallelic. Filtering was performed using PLINK 1.9 [75].

4.6. Phenotype Encoding

The original classification of the infection outcome was divided into five categories: control, resistant, benign, mild, and severe. However, in GWAS, binary phenotypes were defined. For the analysis of the resistance to COVID-19 infection (hereafter termed RESISTANT analysis) the resistant group was coded as “1”, the control group was removed from the analysis, and all the other groups were coded as “0”. For the analysis of the susceptibility to severe COVID-19 infection (hereafter termed SEVERE analysis) the severe group was coded as “1”, the control group was removed from the analysis, and all the other groups were coded as “0”.

4.7. Genome-Wide Association Study

The mixed linear model, simultaneously fitting effects of all 9,767,423 SNPs for the 1076 individuals, was applied for GWAS. In this model the genetic similarity between individuals was partitioned into a component that is due to the variation in SNP genotypes among patients and a “rest” component, that was not assessed by SNPs and thus represents a pure polygenic part of the total phenotypic variation. The model is given by
y = μ + X β + Z 1 u + Z 2 a + e ,
where y (1076) represents a vector of binary phenotypes representing the SEVERE or the RESISTANT phenotype, μ (1076) is a general mean, β (3) is a vector of fixed effects represented by sex and age at data sampling with a corresponding design matrix X (1076 × 3), u (1076) is a vector of random additive polygenic effects of individuals assessed by the variation in SNP genotypes, with a corresponding design matrix Z 1 (1076 × 9,767,423), while a (1076) is a vector of the rest component of random additive polygenic variation among individuals that were not assessed by SNPs, with a corresponding design matrix Z 2 (1076 × 1076), e (1076) is a vector of residuals with e ~ N ( 0 , I σ ^ e 2 ) , where I is an identity matrix and σ ^ e 2 represents the residual variance. The additive genetic covariance between patients ( A ) was calculated based on their kinship coefficients defining a ~ N ( 0 , A σ ^ a 2 ) as well as on the similarity of patients’ SNP genotypes— u ~ N ( 0 , G σ ^ a 2 ) , where G is given by
G = M M T 2 i = 1 N S N P * p i q i ,
where M i j 2 2 p i , 1 2 p i , 2 p i , respectively, stand for homozygous, heterozygous, and alternative homozygous genotypes at ith SNP of jth individual, p i / q i represent frequency of the reference/alternative allele for ith SNP, and N S N P * is a total number of SNPs considered in the model (here, 9,767,423).
In the next step the additive effects of particular SNPs ( v ) were calculated based on patient SNP additive component ( u ) using the back-solve method proposed by [76]
v = 1 k N S N P * 1 + 1 k M A 1 M T 1 1 k M A 1 u ^ ,
where k is a tuning parameter, here k = 0.2 . The significance of ith SNP ( v i ) was assessed by the Wald test: v i σ ^ a 2 , and the resulting nominal p-values were transformed to False Discovery Rates to account for the multiple testing. The above GWAS model was implemented by the GTCA software [76], while the back-solving of SNP effects and hypotheses testing were conducted based on custom-written scripts in R [77] and Python.

4.8. Estimation of KEGG Pathway Effects

SNPs were annotated to genes and KEGG pathways using David software [77]. The downstream analysis was carried out separately for RESISTANT and SEVERE phenotypes. In this context of statistical modelling, as compared to GWAS, the goal is to estimate the joint effects of multiple genes (here represented by SNPs) acting functionally together within a metabolic pathway. Individual effects of many of the genes would have been disregarded in a conventional GWAS as nonsignificant, but they may play a role in a joint metabolic response to infection.
The estimation of KEGG pathway effects was based on the following mixed linear model
v = μ + W t + ε ,
where, v (12,736) is the vector of absolute values of SNP effects estimated from GWAS that were mapped to genes; μ (12,736) is the general mean; t (288) is the random effect KEGG pathway effect with a pre-imposed normal distribution defined by N 0 , V σ t 2 ; W (12,736 × 288) is the corresponding incidence matrix for t assigning SNPs to KEGG pathways; ε (12,736) is a vector of residuals distributed as N 0 , I σ ε 2 . The covariance matrix between KEGG pathways ( V ) was expressed by the Jaccard similarity coefficient J i , j = K N , where K represents the number of genes shared between KEGG i and j, while N represents the total number of genes involved in KEGG i and j. Variance components were assumed as known, amounting to σ t 2 = 0.3 σ y 2 and σ e 2 = 1 σ y 2 . Note that to avoid confounding, within each gene, only one SNP with the highest effect from GWAS was selected for the analysis. The mixed model equations (Henderson C.R. 1984 University of Guelph) were used to obtain solutions for μ and t
μ ^ t ^ = 1 T R 1 1 1 T R 1 W W R 1 1 W T R 1 W + G * 1 1 1 T R 1 v W T R 1 v   ,   where   R = I σ ^ ε 2   and   G * = V σ ^ t 2 .
To maximise the computational performance of the estimation ( μ ^ ) and prediction ( t ^ ) process, a custom Python program implementing the NumPy library was used [78]. Since all calculations were carried out on a high-performance server, the NumPy library was also used to set the array indexing and ordering, which further improved the computing time as compared to a native Python application. Each element of t ^ was assessed for significance ( H 0 : t i 0   v s .   H 1 :   t i > 0 ) by calculating the probability of obtaining a more extreme value using the N 0 , σ t 2 density function.

5. Conclusions

The GWAS studies provide a reliable method to profile patients susceptible to a severe course of COVID-19 by, for instance, the identification of putative genes associated with COVID-19 severity [11,79]. Moreover, the Next-Generation Sequencing (NGS) evaluations provided a full characterization of the entire genome of SARS-CoV-2, and nowadays it is used not only for the discovery of novel molecular variants of this virus, but also to detect novel emerging strains that might pose a threat to public health. The identification of new mutations in the genome of the SARS-CoV-2 virus is also crucial for the development of novel vaccines [80].
Here, we used GWAS analysis to identify metabolic pathways that are associated with a severe COVID-19 infection as well as pathways related to resistance to COVID-19. The main finding of this study were:
(1)
the Allograft rejection pathway (hsa05330) was significant for the resistance to the COVID-19 infection;
(2)
27 SNPs marking genes constituting the Allograft rejection pathway, and the majority of these were located on chromosome 6 (19 SNPs), while the remainder were mapped to chromosomes 2, 3, 10, 12, 20, and X.
(3)
the Allograft rejection pathway comprises several immune system components crucial for the self versus non-self recognition, and also the components of antiviral immunity;
(4)
no significant metabolic pathway was indicated in the case of susceptibility to COVID-19 and its severe course.
Our study showed that not only are single variants important, but also the cumulative impact of several SNPs within the same pathway seems to matter in the case of severe COVID-19 disease. This evidence supports the notion of a multifactorial background of a disease course. Moreover, as previously indicated by us and others, accumulating evidence suggests inborn errors of the immune system components, especially IFN and TNF superfamily members, are crucial when it comes to the COVID-19 severe course.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23116272/s1.

Author Contributions

Conceptualization, J.S. (Joanna Stojak), P.D., J.S. (Joanna Szyda); methodology, J.S. (Joanna Szyda), P.D., J.S. (Joanna Stojak), M.S. (Mateusz Sypniewski), T.S., K.K., D.S, P.S., J.L.; software, J.S. (Joanna Szyda), D.S.; formal analysis, J.S. (Joanna Szyda), T.S., K.K., D.S; data curation, J.S. (Joanna Stojak), P.D., Z.J.K.; writing—original draft preparation, J.S. (Joanna Stojak), P.D., J.S. (Joanna Szyda), M.S. (Mateusz Sypniewski), T.S., M.S. (Maria Stępień), S.B., M.M.; writing—review and editing, J.S. (Joanna Stojak), P.D., J.S. (Joanna Szyda), M.M; visualization, P.D., K.K., D.S.; funding acquisition, Z.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially funded by the National Centre for Research and Development project “Szpitale Jednoimienne/02/2020”, “Development of an innovative diagnostic test to assess the course of COVID-19 and post-death complications with the aid of whole genome analysis”, as well as The Medical Research Agency project 2020/ABM/COVID19/0022 “A clinical trial in the search for genetic markers responsible for the intensity of the course of the COVID-19 disease, with particular emphasis on patients with accompanying cardiopulmonary diseases”. The publication cost was financed by Wroclaw University of Environmental and Life Sciences.

Institutional Review Board Statement

All participants, or guardians/parents for the participants under 18, provided informed consent before collection of blood samples and filling in the clinical data form, which included a questionnaire about country of origin and chronic diseases. The ethical approval for the study was obtained from the Ethics Committee of the Central Clinical Hospital of the Ministry of Interior and Administration in Warsaw (decision nr: 41/2020 from 3 April 2020 and 125/2020 from 1 July 2020). The study complies with the 1964 Helsinki declaration and its later amendments and adhered to the highest data security standards of ISO 27001 and the General Data Protection Regulation (GDPR) act.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The dataset is freely available for researchers here https://1000polishgenomes.com/ (accessed on 4 April 2022) on a reasonable request (https://www.mdpi.com/1422-0067/23/9/4532 (accessed on 4 April 2022)).

Acknowledgments

The authors are grateful to Wojciech Górski for his kind help in preparing Figure 4 for the manuscript and Bernt Guldbrandtsen for an enlightening discussion on the mixed linear models. Calculations have been carried out using resources provided by the Wroclaw Centre for Networking and Supercomputing. The authors thank two anonymous reviewers for their comments that allowed this manuscript to be improved substantially.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mosharaf, M.P.; Reza, M.S.; Kibria, M.K.; Ahmed, F.F.; Kabir, M.H.; Hasan, S.; Mollah, M.N.H. Computational identification of host genomic biomarkers highlighting their functions, pathways and regulators that influence SARS-CoV-2 infections and drug repurposing. Sci. Rep. 2022, 12, 4279. [Google Scholar] [CrossRef]
  2. Zerbo, O.; Lewis, N.; Fireman, B.; Goddard, K.; Skarbinski, J.; Sejvar, J.J.; Azziz-Baumgartner, E.; Klein, N.P. Population-based assessment of risks for severe COVID-19 disease outcomes. Influenza Other Respir. Viruses 2022, 16, 159–165. [Google Scholar] [CrossRef] [PubMed]
  3. Singh, P.P.; Srivastava, A.; Sultana, G.N.N.; Khanam, N.; Pathak, A.; Suravajhala, P.; Singh, R.; Shrivastava, P.; van Driem, G.; Thangaraj, K.; et al. The major genetic risk factor for severe COVID-19 does not show any association among South Asian populations. Sci. Rep. 2021, 11, 12346. [Google Scholar] [CrossRef] [PubMed]
  4. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  5. Maximiano Sousa, F.; Roelens, M.; Fricker, B.; Thiabaud, A.; Iten, A.; Cusini, A.; Flury, D.; Buettcher, M.; Zukol, F.; Balmelli, C.; et al. CH-Sur Study Group Risk factors for severe outcomes for COVID-19 patients hospitalised in Switzerland during the first pandemic wave, February to August 2020: Prospective observational cohort study. Swiss Med. Wkly. 2021, 151, w20547. [Google Scholar] [CrossRef]
  6. Sandoval, M.; Nguyen, D.T.; Vahidy, F.S.; Graviss, E.A. Risk factors for severity of COVID-19 in hospital patients age 18–29 years. PLoS ONE 2021, 16, e0255544. [Google Scholar] [CrossRef] [PubMed]
  7. Satlin, M.J.; Zucker, J.; Baer, B.R.; Rajan, M.; Hupert, N.; Schang, L.M.; Pinheiro, L.C.; Shen, Y.; Sobieszczyk, M.E.; Westblade, L.F.; et al. Changes in SARS-CoV-2 viral load and mortality during the initial wave of the pandemic in New York City. PLoS ONE 2021, 16, e0257979. [Google Scholar] [CrossRef]
  8. Dadras, O.; Afsahi, A.M.; Pashaei, Z.; Mojdeganlou, H.; Karimi, A.; Habibi, P.; Barzegary, A.; Fakhfouri, A.; Mirzapour, P.; Janfaza, N.; et al. The relationship between COVID-19 viral load and disease severity: A systematic review. Immun. Inflamm. Dis. 2022, 10, e580. [Google Scholar] [CrossRef] [PubMed]
  9. Duarte-Salles, T.; Vizcaya, D.; Pistillo, A.; Casajust, P.; Sena, A.G.; Lai, L.Y.H.; Prats-Uribe, A.; Ahmed, W.-U.-R.; Alshammari, T.M.; Alghoul, H.; et al. Thirty-Day Outcomes of Children and Adolescents With COVID-19: An International Experience. Pediatrics 2021, 148, e2020042929. [Google Scholar] [CrossRef]
  10. Garrido, C.; Hurst, J.H.; Lorang, C.G.; Aquino, J.N.; Rodriguez, J.; Pfeiffer, T.S.; Singh, T.; Semmes, E.C.; Lugo, D.J.; Rotta, A.T.; et al. Asymptomatic or mild symptomatic SARS-CoV-2 infection elicits durable neutralizing antibody responses in children and adolescents. JCI Insight 2021, 6, e150909. [Google Scholar] [CrossRef]
  11. 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. [Google Scholar] [CrossRef]
  12. Pairo-Castineira, E.; Clohisey, S.; Klaric, L.; Bretherick, A.D.; Rawlik, K.; Pasko, D.; Walker, S.; Parkinson, N.; Fourman, M.H.; Russell, C.D.; et al. Genetic mechanisms of critical illness in COVID-19. Nature 2021, 591, 92–98. [Google Scholar] [CrossRef] [PubMed]
  13. Zeberg, H.; Pääbo, S. The major genetic risk factor for severe COVID-19 is inherited from Neanderthals. Nature 2020, 587, 610–612. [Google Scholar] [CrossRef] [PubMed]
  14. Zeberg, H.; Pääbo, S. A genomic region associated with protection against severe COVID-19 is inherited from Neandertals. Proc. Natl. Acad. Sci. USA 2021, 118, e2026309118. [Google Scholar] [CrossRef]
  15. Bruchez, A.; Sha, K.; Johnson, J.; Chen, L.; Stefani, C.; McConnell, H.; Gaucherand, L.; Prins, R.; Matreyek, K.A.; Hume, A.J.; et al. MHC class II transactivator CIITA induces cell resistance to Ebola virus and SARS-like coronaviruses. Science 2020, 370, 241–247. [Google Scholar] [CrossRef] [PubMed]
  16. Schneider, D.S.; Ayres, J.S. Two ways to survive infection: What resistance and tolerance can teach us about treating infectious diseases. Nat. Rev. Immunol. 2008, 8, 889–895. [Google Scholar] [CrossRef] [PubMed]
  17. Netea, M.G.; Domínguez-Andrés, J.; van de Veerdonk, F.L.; van Crevel, R.; Pulendran, B.; van der Meer, J.W.M. Natural resistance against infections: Focus on COVID-19. Trends Immunol. 2022, 43, 106–116. [Google Scholar] [CrossRef]
  18. Cardon, L.R.; Palmer, L.J. Population stratification and spurious allelic association. Lancet 2003, 361, 598–604. [Google Scholar] [CrossRef]
  19. Davidson, S.; Starkey, A.; MacKenzie, A. Evidence of uneven selective pressure on different subsets of the conserved human genome; implications for the significance of intronic and intergenic DNA. BMC Genom. 2009, 10, 614. [Google Scholar] [CrossRef] [Green Version]
  20. Siu, J.H.Y.; Surendrakumar, V.; Richards, J.A.; Pettigrew, G.J. T cell Allorecognition Pathways in Solid Organ Transplantation. Front. Immunol. 2018, 9, 2548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. 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, 13. [Google Scholar] [CrossRef] [Green Version]
  22. Weizmann Institute of Science Gene Cards. Available online: https://genecards.weizmann.ac.il/v3/ (accessed on 4 April 2022).
  23. Mahoney, K.M.; Rennert, P.D.; Freeman, G.J. Combination cancer immunotherapy and new immunomodulatory targets. Nat. Rev. Drug Discov. 2015, 14, 561–584. [Google Scholar] [CrossRef] [PubMed]
  24. Elgueta, R.; Benson, M.J.; de Vries, V.C.; Wasiuk, A.; Guo, Y.; Noelle, R.J. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol. Rev. 2009, 229, 152–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. McHugh, R.S.; Ahmed, S.N.; Wang, Y.C.; Sell, K.W.; Selvaraj, P. Construction, purification, and functional incorporation on tumor cells of glycolipid-anchored human B7-1 (CD80). Proc. Natl. Acad. Sci. USA 1995, 92, 8059–8063. [Google Scholar] [CrossRef] [Green Version]
  26. Bromley, S.K.; Iaboni, A.; Davis, S.J.; Whitty, A.; Green, J.M.; Shaw, A.S.; Weiss, A.; Dustin, M.L. The immunological synapse and CD28-CD80 interactions. Nat. Immunol. 2001, 2, 1159–1166. [Google Scholar] [CrossRef]
  27. Krummel, M.F.; Allison, J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [Green Version]
  28. Pentcheva-Hoang, T.; Egen, J.G.; Wojnoonski, K.; Allison, J.P. B7-1 and B7-2 Selectively Recruit CTLA-4 and CD28 to the Immunological Synapse. Immunity 2004, 21, 401–413. [Google Scholar] [CrossRef] [Green Version]
  29. Yokosuka, T.; Saito, T. The Immunological Synapse, TCR Microclusters, and T Cell Activation. In Immunological Synapse; Springer: Berlin/Heidelberg, Germany, 2010; pp. 81–107. [Google Scholar]
  30. Kong, K.-F.; Yokosuka, T.; Canonigo-Balancio, A.J.; Isakov, N.; Saito, T.; Altman, A. A motif in the V3 domain of the kinase PKC-θ determines its localization in the immunological synapse and functions in T cells via association with CD28. Nat. Immunol. 2011, 12, 1105–1112. [Google Scholar] [CrossRef] [Green Version]
  31. Galbraith, M.D.; Kinning, K.T.; Sullivan, K.D.; Araya, P.; Smith, K.P.; Granrath, R.E.; Shaw, J.R.; Baxter, R.; Jordan, K.R.; Russell, S.; et al. Specialized interferon action in COVID-19. Proc. Natl. Acad. Sci. USA 2022, 119, e2116730119. [Google Scholar] [CrossRef]
  32. Ziegler, C.G.K.; Miao, V.N.; Owings, A.H.; Navia, A.W.; Tang, Y.; Bromley, J.D.; Lotfy, P.; Sloan, M.; Laird, H.; Williams, H.B.; et al. Impaired local intrinsic immunity to SARS-CoV-2 infection in severe COVID-19. Cell 2021, 184, 4713–4733.e22. [Google Scholar] [CrossRef]
  33. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.-H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Béziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef] [PubMed]
  34. Van Laarhoven, A.; Kurver, L.; Overheul, G.J.; Kooistra, E.J.; Abdo, W.F.; van Crevel, R.; Duivenvoorden, R.; Kox, M.; ten Oever, J.; Schouten, J.; et al. Interferon gamma immunotherapy in five critically ill COVID-19 patients with impaired cellular immunity: A case series. Med 2021, 2, 1163–1170.e2. [Google Scholar] [CrossRef]
  35. Hidalgo, L.G.; Halloran, P.F. Role of IFN-gamma in allograft rejection. Crit. Rev. Immunol. 2002, 22, 317–349. [Google Scholar] [CrossRef]
  36. Halloran, P.F.; Afrouzian, M.; Ramassar, V.; Urmson, J.; Zhu, L.-F.; Helms, L.M.H.; Solez, K.; Kneteman, N.M. Interferon-γ Acts Directly on Rejecting Renal Allografts to Prevent Graft Necrosis. Am. J. Pathol. 2001, 158, 215–226. [Google Scholar] [CrossRef]
  37. Roesler, J.; Horwitz, M.E.; Picard, C.; Bordigoni, P.; Davies, G.; Koscielniak, E.; Levin, M.; Veys, P.; Reuter, U.; Schulz, A.; et al. Hematopoietic stem cell transplantation for complete IFN-γ receptor 1 deficiency: A multi-institutional survey. J. Pediatr. 2004, 145, 806–812. [Google Scholar] [CrossRef]
  38. Merli, P.; Caruana, I.; De Vito, R.; Strocchio, L.; Weber, G.; Del Bufalo, F.; Buatois, V.; Montanari, P.; Cefalo, M.G.; Pitisci, A.; et al. Role of interferon-γ in immune-mediated graft failure after allogeneic hematopoietic stem cell transplantation. Haematologica 2019, 104, 2314–2323. [Google Scholar] [CrossRef] [PubMed]
  39. Renu, K.; Prasanna, P.L.; Valsala Gopalakrishnan, A. Coronaviruses pathogenesis, comorbidities and multi-organ damage—A review. Life Sci. 2020, 255, 117839. [Google Scholar] [CrossRef] [PubMed]
  40. Van de Veerdonk, F.L.; Giamarellos-Bourboulis, E.; Pickkers, P.; Derde, L.; Leavis, H.; van Crevel, R.; Engel, J.J.; Wiersinga, W.J.; Vlaar, A.P.J.; Shankar-Hari, M.; et al. A guide to immunotherapy for COVID-19. Nat. Med. 2022, 28, 39–50. [Google Scholar] [CrossRef]
  41. Garassino, M.C.; Whisenant, J.G.; Huang, L.C.; Trama, A.; Torri, V.; Agustoni, F.; Baena, J.; Banna, G.; Berardi, R.; Bettini, A.C.; et al. COVID-19 in patients with thoracic malignancies (TERAVOLT): First results of an international, registry-based, cohort study. Lancet Oncol. 2020, 21, 914–922. [Google Scholar] [CrossRef]
  42. Vivarelli, S.; Falzone, L.; Torino, F.; Scandurra, G.; Russo, G.; Bordonaro, R.; Pappalardo, F.; Spandidos, D.A.; Raciti, G.; Libra, M. Immune-checkpoint inhibitors from cancer to COVID-19: A promising avenue for the treatment of patients with COVID-19 (Review). Int. J. Oncol. 2021, 58, 145–157. [Google Scholar] [CrossRef]
  43. Vivarelli, S.; Falzone, L.; Grillo, C.M.; Scandurra, G.; Torino, F.; Libra, M. Cancer Management during COVID-19 Pandemic: Is Immune Checkpoint Inhibitors-Based Immunotherapy Harmful or Beneficial? Cancers 2020, 12, 2237. [Google Scholar] [CrossRef]
  44. Qian, W.; Ye, Y.; Zuo, L.; Song, T.; Xu, Q.; Wang, Y.; Qian, J.; Tian, Y. Immune checkpoint inhibitors use and effects on prognosis of COVID-19 infection: A systematic review and meta-analysis. Immunotherapy 2021, 13, 1271–1282. [Google Scholar] [CrossRef]
  45. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.; 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]
  46. Dustin, M.L. The Immunological Synapse. Cancer Immunol. Res. 2014, 2, 1023–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Grakoui, A.; Bromley, S.K.; Sumen, C.; Davis, M.M.; Shaw, A.S.; Allen, P.M.; Dustin, M.L. The Immunological Synapse: A Molecular Machine Controlling T Cell Activation. Science 1999, 285, 221–227. [Google Scholar] [CrossRef] [Green Version]
  48. Schubert, D.A.; Gordo, S.; Sabatino, J.J.; Vardhana, S.; Gagnon, E.; Sethi, D.K.; Seth, N.P.; Choudhuri, K.; Reijonen, H.; Nepom, G.T.; et al. Self-reactive human CD4 T cell clones form unusual immunological synapses. J. Exp. Med. 2012, 209, 335–352. [Google Scholar] [CrossRef]
  49. Khalil, D.N.; Smith, E.L.; Brentjens, R.J.; Wolchok, J.D. The future of cancer treatment: Immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 2016, 13, 273–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Dustin, M.L.; Colman, D.R. Neural and Immunological Synaptic Relations. Science 2002, 298, 785–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Tseng, S.-Y.; Waite, J.C.; Liu, M.; Vardhana, S.; Dustin, M.L. T Cell-Dendritic Cell Immunological Synapses Contain TCR-dependent CD28-CD80 Clusters That Recruit Protein Kinase Cθ. J. Immunol. 2008, 181, 4852–4863. [Google Scholar] [CrossRef] [Green Version]
  52. Turnis, M.E.; Andrews, L.P.; Vignali, D.A.A. Inhibitory receptors as targets for cancer immunotherapy. Eur. J. Immunol. 2015, 45, 1892–1905. [Google Scholar] [CrossRef]
  53. Balidis, M.; Mikropoulos, D.; Gatzioufas, Z.; de Politis, P.B.; Sidiropoulos, G.; Vassiliadis, V. Acute corneal graft rejection after anti-severe acute respiratory syndrome-coronavirus-2 vaccination: A report of four cases. Eur. J. Ophthalmol. 2021, 112067212110640. [Google Scholar] [CrossRef] [PubMed]
  54. Wasser, L.M.; Roditi, E.; Zadok, D.; Berkowitz, L.; Weill, Y. Keratoplasty Rejection After the BNT162b2 messenger RNA Vaccine. Cornea 2021, 40, 1070–1072. [Google Scholar] [CrossRef] [PubMed]
  55. Phylactou, M.; Li, J.-P.O.; Larkin, D.F.P. Characteristics of endothelial corneal transplant rejection following immunisation with SARS-CoV-2 messenger RNA vaccine. Br. J. Ophthalmol. 2021, 105, 893–896. [Google Scholar] [CrossRef] [PubMed]
  56. Ravichandran, S.; Natarajan, R. Corneal graft rejection after COVID-19 vaccination. Indian J. Ophthalmol. 2021, 69, 1953. [Google Scholar] [CrossRef] [PubMed]
  57. Vyhmeister, R.; Enestvedt, C.K.; VanSandt, M.; Schlansky, B. Steroid-Resistant Acute Cellular Rejection of the Liver After Severe Acute Respiratory Syndrome Coronavirus 2 mRNA Vaccination. Liver Transplant. 2021, 27, 1339–1342. [Google Scholar] [CrossRef]
  58. Ramaswamy, M.; Clel, S.Y.; Cruz, A.C.; Siegel, R.M. Many Checkpoints on the Road to Cell Death:Regulation of Fas–FasL Interactions and Fas Signaling in Peripheral Immune Responses. In Death Receptors and Cognate Ligands in Cancer; Springer: Berlin/Heidelberg, Germany, 2009; pp. 17–47. [Google Scholar]
  59. Chan, F.K.-M.; Chun, H.J.; Zheng, L.; Siegel, R.M.; Bui, K.L.; Lenardo, M.J. A Domain in TNF Receptors That Mediates Ligand-Independent Receptor Assembly and Signaling. Science 2000, 288, 2351–2354. [Google Scholar] [CrossRef] [PubMed]
  60. Topham, D.J.; Tripp, R.A.; Doherty, P.C. CD8+ T cells clear influenza virus by perforin or Fas-dependent processes. J. Immunol. 1997, 159, 5197–5200. [Google Scholar] [PubMed]
  61. Bień, K.; Sokołowska, J.; Bąska, P.; Nowak, Z.; Stankiewicz, W.; Krzyzowska, M. Fas/FasL Pathway Participates in Regulation of Antiviral and Inflammatory Response during Mousepox Infection of Lungs. Mediat. Inflamm. 2015, 2015, 281613. [Google Scholar] [CrossRef]
  62. Martinez, O.M.; Krams, S.M. Involvement of Fas-Fas Ligand Interactions in Graft Rejection. Int. Rev. Immunol. 1999, 18, 527–546. [Google Scholar] [CrossRef]
  63. Wang, Y.-L. Correlation of CD95 and soluble CD95 expression with acute rejection status of liver transplantation. World J. Gastroenterol. 2005, 11, 1700. [Google Scholar] [CrossRef] [PubMed]
  64. Rivero, M.; Crespo, J.; Mayorga, M.; Fabrega, E.; Casafont, F.; Pons-Romero, F. Involvement of the Fas system in liver allograft rejection. Am. J. Gastroenterol. 2002, 97, 1501–1506. [Google Scholar] [CrossRef] [PubMed]
  65. Ichiba, T.; Hoshi, Y.; Eto, Y.; Tajima, N.; Kuraishi, Y. Characterization of GFR, a novel guanine nucleotide exchange factor for Rap1. FEBS Lett. 1999, 457, 85–89. [Google Scholar] [CrossRef] [Green Version]
  66. NIH National Early Warning Score. Available online: https://www.mdcalc.com/national-early-warning-score-news#why-use (accessed on 10 March 2022).
  67. Kaja, E.; Lejman, A.; Sielski, D.; Sypniewski, M.; Gambin, T.; Dawidziuk, M.; Suchocki, T.; Golik, P.; Wojtaszewska, M.; Mroczek, M.; et al. The Thousand Polish Genomes-A Database of Polish Variant Allele Frequencies. Int. J. Mol. Sci. 2022, 23, 4532. [Google Scholar] [CrossRef]
  68. Chiang, C.; Layer, R.M.; Faust, G.G.; Lindberg, M.R.; Rose, D.B.; Garrison, E.P.; Marth, G.T.; Quinlan, A.R.; Hall, I.M. SpeedSeq: Ultra-fast personal genome analysis and interpretation. Nat. Methods 2015, 12, 966–968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv 2013, arXiv:1303.3997. [Google Scholar]
  70. Faust, G.G.; Hall, I.M. SAMBLASTER: Fast duplicate marking and structural variant read extraction. Bioinformatics 2014, 30, 2503–2505. [Google Scholar] [CrossRef] [Green Version]
  71. Pedersen, B.; Quinlan, A. Mosdepth: Quick coverage calculation for genomes and exomes. Bioinformatics 2018, 34, 867–868. [Google Scholar] [CrossRef]
  72. Poplin, R.; Chang, P.-C.; Alexander, D.; Schwartz, S.; Colthurst, T.; Ku, A.; Newburger, D.; Dijamco, J.; Nguyen, N.; Afshar, P.T.; et al. A universal SNP and small-indel variant caller using deep neural networks. Nat. Biotechnol. 2018, 36, 983–987. [Google Scholar] [CrossRef]
  73. Yun, T.; Li, H.; Chang, P.-C.; Lin, M.F.; Carroll, A.; McLean, C.Y. Accurate, scalable cohort variant calls using DeepVariant and GLnexus. Bioinformatics 2020, 36, 5582–5589. [Google Scholar] [CrossRef]
  74. Danecek, P.; Bonfield, J.K.; Liddle, J.; Marshall, J.; Ohan, V.; Pollard, M.O.; Whitwham, A.; Keane, T.; McCarthy, S.A.; Davies, R.M.; et al. Twelve years of SAMtools and BCFtools. GigaScience 2021, 10, giab008. [Google Scholar] [CrossRef]
  75. Purcell, S.; Neale, B.; Todd-Brown, K.; Thomas, L.; Ferreira, M.A.R.; Bender, D.; Maller, J.; Sklar, P.; de Bakker, P.I.W.; Daly, M.J.; et al. PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses. Am. J. Hum. Genet. 2007, 81, 559–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liu, Z.; Goddard, M.E.; Hayes, B.J.; Reinhardt, F.; Reents, R. Technical note: Equivalent genomic models with a residual polygenic effect. J. Dairy Sci. 2016, 99, 2016–2025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Dennis, G.; Sherman, B.T.; Hosack, D.A.; Yang, J.; Gao, W.; Lane, H.C.; Lempicki, R.A. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4, R60. [Google Scholar] [CrossRef] [Green Version]
  78. Harris, C.R.; Millman, K.J.; van der Walt, S.J.; Gommers, R.; Virtanen, P.; Cournapeau, D.; Wieser, E.; Taylor, J.; Berg, S.; Smith, N.J.; et al. Array programming with NumPy. Nature 2020, 585, 357–362. [Google Scholar] [CrossRef]
  79. Thiecke, M.J.; Yang, E.J.; Burren, O.S.; Ray-Jones, H.; Spivakov, M. Prioritisation of Candidate Genes Underpinning COVID-19 Host Genetic Traits Based on High-Resolution 3D Chromosomal Topology. Front. Genet. 2021, 12, 745672. [Google Scholar] [CrossRef] [PubMed]
  80. Falzone, L.; Gattuso, G.; Tsatsakis, A.; Spandidos, D.A.; Libra, M. Current and innovative methods for the diagnosis of COVID-19 infection (Review). Int. J. Mol. Med. 2021, 47, 100. [Google Scholar] [CrossRef]
Figure 1. Manhattan plot (a) for 9,767,423 SNPs analysed in GWAS. SNPs significant in the Allograft rejection pathway (hsa05330) are marked by green dots. The inside plot (b) shows the subset of 12,736 genic SNPs used for the prediction of KEGG pathway effects. The red line indicates the threshold for genome-wide significance (p < 5 × 10−8) and the blue line for suggestive associations (p < 1 × 10−5).
Figure 1. Manhattan plot (a) for 9,767,423 SNPs analysed in GWAS. SNPs significant in the Allograft rejection pathway (hsa05330) are marked by green dots. The inside plot (b) shows the subset of 12,736 genic SNPs used for the prediction of KEGG pathway effects. The red line indicates the threshold for genome-wide significance (p < 5 × 10−8) and the blue line for suggestive associations (p < 1 × 10−5).
Ijms 23 06272 g001
Figure 2. Additive genetic effects of 27 SNPs constituting hsa05330 KEGG pathway on the resistance to COVID-19 infection.
Figure 2. Additive genetic effects of 27 SNPs constituting hsa05330 KEGG pathway on the resistance to COVID-19 infection.
Ijms 23 06272 g002
Figure 3. SNP significance of all 9,767,423 SNPs (GWAS model) and the subset of 12,736 (KEGG model).
Figure 3. SNP significance of all 9,767,423 SNPs (GWAS model) and the subset of 12,736 (KEGG model).
Ijms 23 06272 g003
Figure 4. The Allograft rejection pathway (Figure adapted from the KEGG database. Copyright: Paula Dobosz & Wojciech Górski 2022.) with the most significant SNPs from each gene indicated. If the molecule appears on the figure more than once, relevant SNPs have been depicted only once to maintain the clarity of the scheme. Novel SNPs are indicated by their position in bp.
Figure 4. The Allograft rejection pathway (Figure adapted from the KEGG database. Copyright: Paula Dobosz & Wojciech Górski 2022.) with the most significant SNPs from each gene indicated. If the molecule appears on the figure more than once, relevant SNPs have been depicted only once to maintain the clarity of the scheme. Novel SNPs are indicated by their position in bp.
Ijms 23 06272 g004
Table 1. SNPs representing genes within the Allograft rejection pathway (hsa05330).
Table 1. SNPs representing genes within the Allograft rejection pathway (hsa05330).
ChromosomePosition (bp)Gene IDGene NameMutationGenomic AnnotationSNP IDMedian DPSD
2203709432ENSG00000178562CD28G>Aintronrs984981241284.145465441
3119551547ENSG00000121594CD80G>Aintronnovel264.063210675
3122057921ENSG00000114013CD86G>Aintronrs186115804305.143266314
629725272ENSG00000204642HLA-FC>Texonrs374197706336.119863999
629827086ENSG00000204632HLA-GT>Cintronrs538982928336.636027363
629945505ENSG00000206503HLA-AC>T3′UTRrs7462624503210.17326727
630493031ENSG00000204592HLA-ET>C3′UTRrs192326720295.059734473
631271736ENSG00000204525HLA-CC>Texonrs41548913398.517438111
631356411ENSG00000234745HLA-BG>Aexonrs151341222439.575215658
631576590ENSG00000232810TNFaC>Tintronrs763838774284.933491701
632441500ENSG00000204287HLA-DRAT>Gintronrs1338070938356.726123187
632528966ENSG00000198502HLA-DRB5C>Tintronrs11685666893912.87413207
632583693ENSG00000196126HLA-DRB1T>TCintronnovel318.339663173
632643698ENSG00000196735HLA-DQA1A>GExon of a non-coding transcriptrs1459153928367.155148333
632664633ENSG00000179344HLA-DQB1C>Tintronrs1184841282367.686703581
632746600ENSG00000237541HLA-DQA2GAGA>G3′UTRnovel295.463038801
632813318ENSG00000241106HLA-DOBCAG>Cintronnovel254.576624558
632935042ENSG00000242574HLA-DMBG>Aintronrs779280356254.040525561
632964115ENSG00000204257HLA-DMAT>Cintronnovel274.697217326
633009100ENSG00000204252HLA-DOAG>AintronNovel335.700484494
633078874ENSG00000231389HLA-DPA1G>Aintronrs146322130285.622143954
633086775ENSG00000223865HLA-DPB1CTGTT>C3′UTRnovel318.032764829
1070599302ENSG00000180644PRF1G>Aintronnovel314.807858254
1088956063ENSG00000026103FAST>Cintronnovel284.145442918
1268156786ENSG00000111537IFNGC>Tintronrs745989394284.229532615
2046121566ENSG00000101017CD40G>Aintronnovel263.808906036
X136659930ENSG00000102245CD40LC>T3′UTRrs879041317126.017283757
Table 2. Cluster of differentiation proteins with significant impact on the allograft rejection pathway, identified as being significant in the current project.
Table 2. Cluster of differentiation proteins with significant impact on the allograft rejection pathway, identified as being significant in the current project.
Cluster of Differentiation ProteinFunctionReferences
CD28is one of the proteins expressed on T cells, providing costimulatory signals required for T cell activation and survival; provides a potent signal for the production of various interleukins, especially IL-6; molecules CD80 and CD86 are its ligands; the activity of CD80–CD28 complex stimulates the activation of transcription factors NF-κB, promoting IL-2 production[25,26,27,28,29]
CD40is a costimulatory protein, a member of the TNF superfamily, constitutively expressed on B cells and antigen-presenting cells; CD40 binds its ligand CD40L, which is transiently expressed on T cells and other non-immune cells under inflammatory conditions; essential in mediating a broad variety of immune and inflammatory responses including T cell-dependent immunoglobulin class switching, germinal centre formation memory B cell development, to name just a few[22,23]
CD80is an immunoglobulin, also a ligand for cytotoxic T-lymphocyte antigen 4 (CTLA-4, also known as CD152), which remains constitutively expressed on most of the T cells; present at APCs and their receptors present on the T cells; present specifically on dendritic cells, activated B cells, and macrophages, but also T cells; malfunctioning CD80 molecules are also involved in some pathological conditions, such as lupus erythematosus[22,25,26,27,28,29]
CD86is a costimulatory protein, immunoglobulin, constitutively expressed on dendritic cells, pancreatic Langerhans cells, macrophages, B cells (including memory B cells), and on other antigen-presenting cells; provides costimulatory signals crucial for T cell activation and survival; it is also associated with myocarditis and gallbladder squamous cell carcinoma[22,25,26,27,28,29]
CD152also known as CTLA-4 (cytotoxic T-lymphocyte-associated protein); it is a receptor that functions as an immune checkpoint protein and downregulates immune responses; it is constitutively expressed on regulatory T cells but found to be upregulated in conventional T cells after activation, being a phenomenon particularly significant in cancers, thus, being important as a background of immunotherapy utilising checkpoint inhibitors[25,27,29]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Szyda, J.; Dobosz, P.; Stojak, J.; Sypniewski, M.; Suchocki, T.; Kotlarz, K.; Mroczek, M.; Stępień, M.; Słomian, D.; Butkiewicz, S.; et al. Beyond GWAS—Could Genetic Differentiation within the Allograft Rejection Pathway Shape Natural Immunity to COVID-19? Int. J. Mol. Sci. 2022, 23, 6272. https://doi.org/10.3390/ijms23116272

AMA Style

Szyda J, Dobosz P, Stojak J, Sypniewski M, Suchocki T, Kotlarz K, Mroczek M, Stępień M, Słomian D, Butkiewicz S, et al. Beyond GWAS—Could Genetic Differentiation within the Allograft Rejection Pathway Shape Natural Immunity to COVID-19? International Journal of Molecular Sciences. 2022; 23(11):6272. https://doi.org/10.3390/ijms23116272

Chicago/Turabian Style

Szyda, Joanna, Paula Dobosz, Joanna Stojak, Mateusz Sypniewski, Tomasz Suchocki, Krzysztof Kotlarz, Magdalena Mroczek, Maria Stępień, Dawid Słomian, Sławomir Butkiewicz, and et al. 2022. "Beyond GWAS—Could Genetic Differentiation within the Allograft Rejection Pathway Shape Natural Immunity to COVID-19?" International Journal of Molecular Sciences 23, no. 11: 6272. https://doi.org/10.3390/ijms23116272

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