Association of Toll-like receptor 7 variants with life-threatening COVID-19 disease in males ============================================================================================ * Chiara Fallerini * Sergio Daga * Stefania Mantovani * Elisa Benetti * Aurora Pujol * Nicola Picchiotti * Agatha Schluter * Laura Planas-Serra * Jesús Troya * Margherita Baldassarri * Francesca Fava * Serena Ludovisi * Francesco Castelli * Maria Eugenia Quiros-Roldan * Massimo Vaghi * Stefano Rusconi * Matteo Siano * Maria Bandini * Simone Furini * Francesca Mari * GEN-COVID Multicenter Study * Alessandra Renieri * Mario U. Mondelli * Elisa Frullanti ## ABSTRACT **Background** COVID-19 clinical presentation ranges from asymptomatic to fatal outcome. This variability is due in part to host genome specific mutations. Recently, two families in which COVID-19 segregates like an X-linked recessive monogenic disorder environmentally conditioned by SARS-CoV-2 have been reported leading to identification of loss-of-function variants in *TLR7*. **Objective** We sought to determine whether the two families represent the tip of the iceberg of a subset of COVID-19 male patients. **Methods** We compared male subjects with extreme phenotype selected from the Italian GEN-COVID cohort of 1178 SARS-CoV-2-infected subjects (<60y, 79 severe cases versus 77 control cases). We applied the LASSO Logistic Regression analysis, considering only rare variants on the young male subset, picking up *TLR7* as the most important susceptibility gene. **Results** Rare *TLR7* missense variants were predicted to impact on protein function in severely affected males and in none of the asymptomatic subjects. We then investigated a similar white European cohort in Spain, confirming the impact of *TRL7* variants. A gene expression profile analysis in peripheral blood mononuclear cells after stimulation with TLR7 agonist demonstrated a reduction of mRNA level of TLR7, IRF7, ISG15, IFN-□ and IFN-γ in COVID-19 patients compared with unaffected controls demonstrating an impairment in type I and II INF responses. **Conclusion** Young males with *TLR7* loss-of-function mutations and severe COVID-19 in the two reported families represent only a fraction of a broader and complex host genome situation. Specifically, missense mutations in the X-linked recessive *TLR7* disorder may significantly contribute to disease susceptibility in up to 4% of severe COVID-19. **Clinical Implication** In this new yet complex scenario, our observations provide the basis for a personalized interferon-based therapy in patients with rare *TLR7* variants. **CAPSULE SUMMARY** Our results in large cohorts from Italy and Spain showed that X-linked recessive TLR7 disorder may represent the cause of disease susceptibility to COVID-19 in up to 4% of severely affected young male cases. Key Words * TLR7 * COVID-19 * LASSO Logistic Regression analysis ## INTRODUCTION Coronavirus disease 2019 (COVID-19), a potentially severe systemic disease caused by coronavirus SARS-CoV-2, is characterized by a highly heterogeneous phenotypic variability, with the large majority of infected individuals experiencing only mild or even no symptoms. However, severe cases can rapidly evolve towards a critical respiratory distress syndrome and multiple organ failure1. COVID-19 still represents an enormous challenge for the world’s healthcare systems ten months after the first appearance in December 2019 in Wuhan, Huanan, Hubei Province of China. Symptoms of COVID-19 range from fever, cough, sore throat, congestion, anosmia and fatigue to shortness of breath, interstitial pneumonia followed by respiratory distress and septic shock1. Although older age and the presence of cardiovascular or metabolic comorbidities have been identified as risk factors predisposing to severe disease2, these factors alone do not fully explain the differences in the severity. Male patients show more severe clinical manifestations than females with higher prevalence of hospitalizations (16% versus 12%), ICU admissions (3% versus 2%), and deaths (6% versus 5%), indicating that gender may influence the outcome of the disease3. These findings suggest a role of host predisposing genetic factors in the pathogenesis of the disease which may be responsible for different clinical outcomes as a result of different antiviral defence mechanisms as well as specific receptor permissiveness to virus and immunogenicity. More than that, recent evidences suggest also a fundamental role of IFN genes in genetic bases of COVID-19; in particular, recently rare variants have been identified in the interferon type I pathway that are responsible for inborn errors of immunity in a small percentage of patients and auto-antibodies against type I interferon genes in up to 10% of severe COVID-19 cases4,5. Toll-like receptors (TLRs) are crucial components in the initiation of innate immune response - the non specific-immune response-to a variety of pathogens, causing the production of pro-inflammatory cytokines (TNF-α, IL-1, and IL-6) and type I and II IFNs, that are responsible for innate antiviral responses. In particular the innate immune response is very sensitive in detecting potentially pathogenic antigens, activating downstream signaling to induce transcription factors in the nucleus, promoting synthesis and release of types I and type III interferons (IFNs) in addition to a number of other important proinflammatory cytokines. However, when the immune response is dysregulated, it can result in excessive inflammation, or in extreme cases, death. Among them, TLR7 recognises several single-stranded RNA viruses including SARS-CoV-26. We previously showed that another RNA virus, hepatitis C virus (HCV), is able to inhibit CD4 T cell function via Toll-like receptor 7 (TLR7)7. Recently, van der Made *et al*. have reported two independent families in which COVID-19 segregates like an X-linked recessive monogenic disorder conditioned by SARS-CoV-2, as an environmental factor8. Here, we performed a nested case-control study (NCC) within our prospectively recruited GEN-COVID cohort with the aim to determine whether the 2 families described by van der Made *et al*. represent an ultra-rare situation or the tip of the iceberg of a larger subset of young male patients. ## RESULTS AND DISCUSSION We applied LASSO logistic regression analysis, after correcting for Principal Components, to a synthetic boolean representation of the entire set of genes of the X chromosome on the extreme phenotypic ends of the young male subset of the Italian GEN-COVID cohort ([https://sites.google.com/dbm.unisi.it/gen-covid](https://sites.google.com/dbm.unisi.it/gen-covid))9. Cases were selected according to the following inclusion criteria: *i*. male gender; *ii*. young age (<60 years); *iii* endotracheal intubation or CPAP/biPAP ventilation (79 subjects). As controls, 77 subjects were selected using the sole criterion of being oligo-asymptomatic not requiring hospitalization. Cases and controls represented the extreme phenotypic presentations of the GEN-COVID cohort. Exclusion criteria for both cases and controls were: *i*. SARS-CoV-2 infection not confirmed by PCR; *ii*. non-caucasian ethnicity. The materials and methods details are listed in the Online Repository section. The study (GEN-COVID) was consistent with Institutional guidelines and approved by the University Hospital (Azienda Ospedaliero-Universitaria Senese) Ethical Review Board, Siena, Italy (Prot n. 16929, dated March 16, 2020). Only rare variants (≤1% in European Non-Finnish population) were considered in the boolean representation: the gene was set to 1 if it included at least a missense, splicing, or loss of function rare variant, and 0 otherwise. Fisher Exact test was then used for the specific data validation. Toll-like receptor 7 (*TLR7*) was picked up as one of the most important susceptibility genes by LASSO Logistic Regression analysis (**Figure 1**). We then queried the COVID-19 section of the Network of Italian Genome NIG database ([http://www.nig.cineca.it/](http://www.nig.cineca.it/), specifically, [http://nigdb.cineca.it](http://nigdb.cineca.it)) that houses the entire GEN-COVID cohort represented by more than 1000 WES data of COVID-19 patients and SARS-CoV-2 infected asymptomatic subjects7. By selecting for young (<60 year-old) males, we obtained rare (MAF≤1%) *TLR7* missense variants predicted to impact on protein function (CADD>12.28) in 5 out of 79 male patients (6,32%) with life-threatening COVID-19 (hospitalized intubated and hospitalized CPAP/BiPAP) and in none of the 77 SARS-CoV2 infected oligo-asymptomatic male subjects. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/27/2020.11.19.20234237/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/F1) Figure 1. Rare *TLR7* variants and association with COVID-19. LASSO logistic regression on boolean representation of rare variants of all genes of the X chromosome (X chr). TLR7 is picked up by LASSO logistic regression as one of the most important genes on the X chr (**Panel A**). The LASSO logistic regression model provides an embedded feature selection method within the binary classification tasks (male patients with life-threatening COVID-19 vs infected asymptomatic male subjects). The upward histograms (positive weights) reflect a susceptible behaviour of the features to the target COVID-19, whereas the downward histograms (negative weights) a protective action. In addition to the *TLR7* gene, other genes are picked up, including XPNPEP2 dysregulation bradykinine system. **Panel B** represents the cross-validation accuracy score for the grid of LASSO regularization parameters; the error bar is given by the standard deviation of the score within the 10 folds; the red point (1.26) corresponds to the parameter chosen for the fitting procedure. Performances are evaluated through the confusion matrix of the aggregated predictions in the 10 folds of the cross-validation (**Panel C**) and with the boxplot (**Panel D**) of accuracy (60% average value), precision (59%), sensitivity (75%), specificity (43%), and ROC-AUC score (68%). The box extends from the Q1 to Q3 quartile, with a line at the median (Q2) and a triangle for the average. A similar Spanish cohort, composed of male patients under 60 years of age without comorbidities (77 cases and 45 controls) was used to expand the cohort to another representative European population highly impacted by COVID-19. All subjects were white European. The Spanish Covid HGE cohort is under IRB approval PR127/20 from Bellvitge University Hospital, Barcelona, Spain. We found a *TLR7* variant in one of 77 cases (1,29%) and in none of 45 controls. Overall, the association between the presence of *TLR7* rare mutations and severe COVID-19 was significant (p= 0.04 by Fisher Exact test, **Table 1**). View this table: [Table 1.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/T1) Table 1. Fisher exact test of the overall combined cohorts in young males (<60 years). We then investigated the presence of *TLR7* mutations in the entire male cohort of COVID-19 (cases and controls) regardless of age. We found *TLR7* rare missense variants in 3 additional patients over 60 years of age, including 2 among cases and 1 among controls, the last bearing the mutation p.Val222Asp predicted to have a low impact on protein function (CADD of 5.36) (**Table 2**). View this table: [Table 2A.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/T2) Table 2A. *TLR7* mutations in severely affected Italian and Spanish males -all ages-(cases). View this table: [Table 2B.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/T3) Table 2B. *TLR7* mutations in asymptomatic males (controls). In order to functionally link the presence of the identified *TLR7* missense mutations and the effect on the downstream type I IFN-signaling, we performed a gene expression profile analysis in peripheral blood mononuclear cells (PBMCs) isolated from patients after stimulation with the TLR7 agonist imiquimod (as reported by van der Made *et al*.8). PBMCs from 5 out of 8 cases and from the control were available. All *TLR7* variants were tested except one (Ala288Val) as some different patients share the same. This analysis showed a statistically significant decrease of *IRF7* (member of the interferon regulatory transcription factor) and *IFN-*γ mRNA levels in all tested mutations (Val219Ile, Ser301Pro, Ala1032Thr, His630Tyr) identified in cases (P1, P3, P7, P8 and P6) compared with healthy (Ctl) demonstrating a complete impairment in IFN-□and IFN-γ production in response to TLR7 stimulation in all patients (**Figure 2**). A similarly significant reduction was shown for ISG15 (Interferon-stimulated gene 15) and *IFN-*□ mRNA level for 4 out of 5 patients (P3, P8, P6, P7). An analogous trend was observed for TLR7 in P8 and P6 cases. A significant reduction was also shown for all TLR7-related mRNA transcripts when cases were compared with the asymptomatic controls (C1) (**Supplementary Figure 1**). Interestingly, the p.Val222Asp variant proved to be functionally neutral in keeping with its discovery in control and not in cases (**Figure 2**). TLR7 expression was evaluated in monocytes and B cells from patients and healthy controls by flow cytometry. As shown in **Supplementary Figure 2** patients and controls expressed the TLR7 protein at intracellular level. The functional capacity of PBMC was evaluated after stimulation with the TLR4 agonist lipopolysaccharide (LPS). The analysis showed that LPS-induced production of IL6 by CD3-CD14+ cells was similar in patients and controls (**Supplementary Figure 3**). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/27/2020.11.19.20234237/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/F2) Figure 2. Gene expression profile analysis in peripheral blood mononuclear cells (PBMCs) after stimulation with a TLR7 agonist for 4 hours. 5×105 PBMCs from COVID-19 patients and four unaffected male and female controls were stimulated for 4 hours with the TLR7 agonist imiquimod at 5 μg/mL or cell culture medium. Quantitative PCR assay was performed and the 2- ΔΔ Ct calculated using *HPRT1* as housekeeping gene. Fold change in mRNA expression of *TLR7* and type 1 IFN-related genes *ISG15, IRF7, IFN-*□ and *IFN-*γ induced by TLR7 agonist imiquimod was compared with cell culture medium. Error bars show standard deviations. *P values* were calculated using an unpaired *t* test: *P < .05; **P < .01; \***|P < .001; \**\*|\*P < .0001. Ctl indicates healthy controls; C1, the asymptomatic mutated control; P1, P3, P8, P6, P7 severely affected mutated cases (as in **Table 2A**). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/27/2020.11.19.20234237/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2020/11/27/2020.11.19.20234237/F3) Figure 3. Segregation analysis in Italian (P3) and Spanish family (P6). Fold change in mRNA expression of the main *TLR7*-effector, *IRF7*, is shown in **Panel A**. Pedigree (**Panel B**) and respective segregation of *TLR7* variant and COVID-19 status (**Panel C**). Squares represent male family members; circles, females. Segregation analysis was available for two of 8 cases, P3 and P6 (**Figure 3**). In both pedigree, the disease nicely segregates as an X-linked disorder conditioned by environmental factors, i.e. SARS-Cov-2 (**Figure 3, panel B**). This was also supported by functional analysis on all *TLR7*-related genes (**Supplementary Figure 4**). As an example, we showed gene expression profile analysis for *IRF7* gene: in male patients *IRF7* mRNA levels are statistically significantly reduced compared to heterozygous females (**Figure 3, panel A**). Of note, only the infected mutated male subject had severe COVID-19, whereas the infected not mutated female (sister) was oligosymptomatic with symptoms lasting 7-10 days; the infected mutated female (one of the daughters) was oligosymptomatic with symptoms lasting one month (**Figure 3, panel C**). Both females did not require any hospital treatment. Our results showed that the two families with loss of function mutations reported by van der Made *et a*l.8 in males with severe COVID-19 with a mean age of 26 years are indeed the tip of the iceberg of a broader host genome scenario. Specifically, missense mutations in the X-linked recessive TLR7 disorder may represent the cause of disease susceptibility to COVID-19 in up to 4% of severely affected young male cases (6/156, 3,84%). Since not all identified mutations were functionally tested, the true percentage could be slightly lower. Overall, males with missense mutations shown here developed COVID-19 at a mean age of 58 years, considerably later than 26 years, in agreement with a predicted smaller impact on the protein than the loss of function mutations reported by van der Made *et al*.8. Similarly, the identified rare missense *TLR7* variants impaired the mRNA expression of *TLR7* as well as the downstream pathway. The observation reported here may lead to consider *TLR7* screening in severely affected male patients in order to start personalized interferon treatment for those with this specific genetic disorder. ## Supporting information Supplementary Figure 1 [[supplements/234237_file05.pdf]](pending:yes) Supplementary Figure 2 [[supplements/234237_file06.pdf]](pending:yes) Supplementary Figure 3 [[supplements/234237_file07.pdf]](pending:yes) Supplementary Figure 4 [[supplements/234237_file08.pdf]](pending:yes) Online Repository [[supplements/234237_file09.docx]](pending:yes) ## Data Availability The data and samples referenced here are housed in the GEN-COVID Patient Registry and the GEN-COVID Biobank and are available for consultation. For further information, you may contact the corresponding author, Prof. Alessandra Renieri (e-mail: alessandra.renieri{at}unisi.it). [http://www.nig.cineca.it](http://www.nig.cineca.it) [https://www.covid19hg.org/](https://www.covid19hg.org/) [http://nigdb.cineca.it](http://nigdb.cineca.it) ## ADDITIONAL INFORMATION ### GEN-COVID Multicenter Study ([https://sites.google.com/dbm.unisi.it/gen-covid](https://sites.google.com/dbm.unisi.it/gen-covid)) Floriana Valentino1,2, Gabriella Doddato1,2, Annarita Giliberti1,2, Rossella Tita9, Sara Amitrano9, Mirella Bruttini1,2,9, Susanna Croci1,2, Ilaria Meloni1,2, Maria Antonietta Mencarelli9, Caterina Lo Rizzo9, Anna Maria Pinto9, Laura Di Sarno1,2, Giada Beligni1,2, Andrea Tommasi1,2,9, Maria Palmieri1,2, Francesca Montagnani2,17, Arianna Emiliozzi2,17, Massimiliano Fabbiani17, Barbara Rossetti17, Giacomo Zanelli2,17, Elena Bargagli18, Laura Bergantini18, Miriana D’Alessandro18, Paolo Cameli18, David Bennet18, Federico Anedda19, Simona Marcantonio19, Sabino Scolletta19, Federico Franchi19, Maria Antonietta Mazzei20, Susanna Guerrini20, Edoardo Conticini21, Luca Cantarini21, Bruno Frediani21, Danilo Tacconi22, Chiara Spertilli22, Marco Feri23, Alice Donati23, Raffaele Scala24, Luca Guidelli24, Genni Spargi25, Marta Corridi25, Cesira Nencioni26, Leonardo Croci26, Gian Piero Caldarelli27, Maurizio Spagnesi16, Paolo Piacentini16, Elena Desanctis16, Silvia Cappelli16, Anna Canaccini28, Agnese Verzuri28, Valentina Anemoli28, Agostino Ognibene29, Antonella D’Arminio Monforte30, Esther Merlini30, Federica Gaia Miraglia30, Massimo Girardis31, Sophie Venturelli31, Marco Sita31, Andrea Cossarizza32, Andrea Antinori33, Alessandra Vergori33, Arianna Gabrieli15, Agostino Riva14,15, Daniela Francisci34,35, Elisabetta Schiaroli34, Pier Giorgio Scotton36, Francesca Andretta36, Sandro Panese37, Renzo Scaggiante38, Francesca Gatti38, Saverio Giuseppe Parisi39, Stefano Baratti39, Melania Degli Antoni12, Isabella Zanella12, Matteo Della Monica40, Carmelo Piscopo40, Mario Capasso41,42,43, Roberta Russo41,42, Immacolata Andolfo41,42, Achille Iolascon41,42, Giuseppe Fiorentino44, Massimo Carella45, Marco Castori45, Giuseppe Merla45, Filippo Aucella46, Pamela Raggi47, Carmen Marciano47, Rita Perna47, Matteo Bassetti48,49, Antonio Di Biagio49, Maurizio Sanguinetti50,51, Luca Masucci50,51, Serafina Valente52, Marco Mandalà53, Alessia Giorli53, Lorenzo Salerni53, Patrizia Zucchi54, Pierpaolo Parravicini54, Elisabetta Menatti55, Tullio Trotta56, Ferdinando Giannattasio56, Gabriella Coiro56, Fabio Lena57, Domenico A. Coviello58, Cristina Mussini59, Giancarlo Bosio60, Enrico Martinelli60, Sandro Mancarella61, Luisa Tavecchia61, Marco Gori8,6, Lia Crotti63,64,65,66, Agatha Schluter4,5,67, Laura Planas-Serra4,5,67, Marta Gut68, Chiara Gabbi69. 17) Dept of Specialized and Internal Medicine, Tropical and Infectious Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy 18) Unit of Respiratory Diseases and Lung Transplantation, Department of Internal and Specialist Medicine, University of Siena, Italy 19) Dept of Emergency and Urgency, Medicine, Surgery and Neurosciences, Unit of Intensive Care Medicine, Siena University Hospital, Italy 20) Department of Medical, Surgical and Neuro Sciences and Radiological Sciences, Unit of Diagnostic Imaging, University of Siena, Italy 21) Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, Italy 22) Department of Specialized and Internal Medicine, Infectious Diseases Unit, San Donato Hospital Arezzo, Italy 23) Dept of Emergency, Anesthesia Unit, San Donato Hospital, Arezzo, Italy 24) Department of Specialized and Internal Medicine, Pneumology Unit and UTIP, San Donato Hospital, Arezzo, Italy 25) Department of Emergency, Anesthesia Unit, Misericordia Hospital, Grosseto, Italy 26) Department of Specialized and Internal Medicine, Infectious Diseases Unit, Misericordia Hospital, Grosseto, Italy 27) Clinical Chemical Analysis Laboratory, Misericordia Hospital, Grosseto, Italy 28) Territorial Scientific Technician Department, Azienda USL Toscana Sud Est, Italy 29) Clinical Chemical Analysis Laboratory, San Donato Hospital, Arezzo, Italy 30) Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Italy 31) Department of Anesthesia and Intensive Care, University of Modena and Reggio Emilia, Modena, Italy 32) Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy 33) HIV/AIDS Department, National Institute for Infectious Diseases, IRCCS, Lazzaro Spallanzani, Rome, Italy 34) Infectious Diseases Clinic, Department of Medicine 2, Azienda Ospedaliera di Perugia and University of Perugia, Santa Maria Hospital, Perugia, Italy 35) Infectious Diseases Clinic, “Santa Maria” Hospital, University of Perugia, Perugia, Italy 36) Department of Infectious Diseases, Treviso Hospital, Local Health Unit 2 Marca Trevigiana, Treviso, Italy 37) Clinical Infectious Diseases, Mestre Hospital, Venezia, Italy 38) Infectious Diseases Clinic, ULSS1, Belluno, Italy 39) Department of Molecular Medicine, University of Padova, Italy 40) Medical Genetics and Laboratory of Medical Genetics Unit, A.O.R.N. “Antonio Cardarelli”, Naples, Italy 41) Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy 42) CEINGE Biotecnologie Avanzate, Naples, Italy 43) IRCCS SDN, Naples, Italy 44) Unit of Respiratory Physiopathology, AORN dei Colli, Monaldi Hospital, Naples, Italy 45) Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy 46) Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy 47) Clinical Trial Office, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy 48) Department of Health Sciences, University of Genova, Genova, Italy 49) Infectious Diseases Clinic, Policlinico San Martino Hospital, IRCCS for Cancer Research Genova, Italy 50) Microbiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Medicine, Rome, Italy 51) Department of Laboratory Sciences and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy 52) Department of Cardiovascular Diseases, University of Siena, Siena, Italy 53) Otolaryngology Unit, University of Siena, Italy 54) Department of Internal Medicine, ASST Valtellina e Alto Lario, Sondrio, Italy 55) Study Coordinator Oncologia Medica e Ufficio Flussi Sondrio, Italy 56) First Aid Department, Luigi Curto Hospital, Polla, Salerno, Italy 57) Local Health Unit-Pharmaceutical Department of Grosseto, Toscana Sud Est Local Health Unit, Grosseto, Italy 58) U.O.C. Laboratorio di Genetica Umana, IRCCS Istituto G. Gaslini, Genova, Italy 59) Infectious Diseases Clinics, University of Modena and Reggio Emilia, Modena, Italy 60) Department of Respiratory Diseases, Azienda Ospedaliera di Cremona, Cremona, Italy 61) U.O.C. Medicina, ASST Nord Milano, Ospedale Bassini, Cinisello Balsamo (MI), Italy 62) Université Côte d’Azur, Inria, CNRS, I3S, Maasai 63) Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Milan, Italy 64) Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy 65) Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin, Milan, Italy 66) Istituto Auxologico Italiano, IRCCS, Laboratory of Cardiovascular Genetics, Milan, Italy 67) Spanish Covid HGE 68) CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Carrer Baldiri i Reixac 4, 08028, Barcelona, Spain. 69) Independent Scientist, Milan, Italy ## ACKNOWLEDGEMENTS This study is part of the GEN-COVID Multicenter Study, [https://sites.google.com/dbm.unisi.it/gen-covid](https://sites.google.com/dbm.unisi.it/gen-covid), the Italian multicenter study aimed at identifying the COVID-19 host genetic bases. Specimens were provided by the COVID-19 Biobank of Siena, which is part of the Genetic Biobank of Siena, member of BBMRI-IT, of Telethon Network of Genetic Biobanks (project no. GTB18001), of EuroBioBank, and of D-Connect. We thank the CINECA consortium for providing computational resources and the Network for Italian Genomes (NIG) [http://www.nig.cineca.it](http://www.nig.cineca.it) for its support. We thank private donors for the support provided to A.R. (Department of Medical Biotechnologies, University of Siena) for the COVID-19 host genetics research project (D.L n.18 of March 17, 2020). We also thank the COVID-19 Host Genetics Initiative ([https://www.covid19hg.org/](https://www.covid19hg.org/)). MIUR project “Dipartimenti di Eccellenza 2018-2020” to the Department of Medical Biotechnologies University of Siena, Italy. Research on the Spanish cohort received funding from the European Union’s Horizon 2020 research and innovation programme under grant agreement No 824110 – EASI-Genomics, to AP. ## Footnotes * * Co-first author * $ Co-last author * **Funding Statement:** We thank the CINECA consortium for providing computational resources and the Network for Italian Genomes (NIG) [http://www.nig.cineca.it](http://www.nig.cineca.it) for its support. We thank private donors for the support provided to A.R. (Department of Medical Biotechnologies, University of Siena) for the COVID-19 host genetics research project (D.L n.18 of March 17, 2020). We also thank the COVID-19 Host Genetics Initiative ([https://www.covid19hg.org/](https://www.covid19hg.org/)); MIUR project “Dipartimenti di Eccellenza 2018-2020” to the Department of Medical Biotechnologies University of Siena, Italy. We also thank Intesa San Paolo for the 2020 charity fund dedicated to the project N. B/2020/0119 “Identificazione delle basi genetiche determinanti la variabilità clinica della risposta a COVID-19 nella popolazione italiana”. * **Disclosure Statement:** The authors declare no competing interests. ## Abbreviations Ala : Alanine amino acid Asp : Asparagine amino acid CADD : Combined Annotation Dependent Depletion COVID-19 : COronaVIrus Disease 19 His : Histidine amino acid HPRT1 : hypoxanthine phosphoribosyltransferase 1 INFs : Interferons IFN-α : Interferon Alpha IFN-β1 : Interferon Beta1 IFN-γ : Interferon Gamma IL-1 : Interleukin 1 IL-6 : Interleukin 6 Ile : Isoleucine amino acid IRF7 : Interferon Regulatory Factor 7 ISG15 : Interferon-Stimulated Gene 15 LASSO : Least Absolute Shrinkage and Selection Operator LBP : LipoPolySaccharide (LPS) : Binding Protein NIG : Network of Italian Genome PBMCs : Peripheral Blood Mononuclear Cells Pro : Proline amino acid RNA : RiboNucleic Acid Ser : Serine amino acid SARS-CoV-2 : Severe Acute Respiratory Syndrome caused by CoronaVirus Thr : Threonine amino acid TLRs : Toll-like Receptors TLR4 : Toll-like Receptor 4 TLR7 : Toll-like Receptor 7 TNF-α : Tumor Necrosis Factor α Tyr : Tyrosine amino acid Val : Valine amino acid WES : Whole Exome Sequencing * Received November 19, 2020. * Revision received November 19, 2020. * Accepted November 27, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## REFERENCES 1. 1.Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020 February 24. doi: 10.1001/jama.2020.2648. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2020.2648&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32091533&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F27%2F2020.11.19.20234237.atom) 2. 2.Hägg S, Jylhävä J, Wang Y, Xu H, Metzner C, Annetorp M, et al. Age, Frailty, and Comorbidity as Prognostic Factors for Short-Term Outcomes in Patients With Coronavirus Disease 2019 in Geriatric Care. J Am Med Dir Assoc. 2020 Aug 14:S1525-8610(20)30704-0. doi: 10.1016/j.jamda.2020.08.014. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jamda.2020.08.014&link_type=DOI) 3. 3.Stokes EK, Zambrano LD, Anderson KN, Marder EP, Raz KM, El Burai Felix S, et al. Coronavirus Disease 2019 Case Surveillance - United States, January 22-May 30, 2020. MMWR Morb Mortal Wkly Rep 2020; 69:759–65. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.15585/mmwr.mm6924e2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F27%2F2020.11.19.20234237.atom) 4. 4.Zhang Q, Bastard P, Liu Z, Le Pen J, Moncada-Velez M, Chen J, et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020 Sep 24:eabd4570. doi: 10.1126/science.abd4570. Epub ahead of print. PMID: 32972995. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjE3OiIzNzAvNjUxNS9lYWJkNDU3MCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzExLzI3LzIwMjAuMTEuMTkuMjAyMzQyMzcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 5. 5.Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann H-H, Zhang Y, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020 Oct 23; 370(6515):eabd4585. doi: 10.1126/science.abd4585. Epub 2020 Sep 24. PMID: 32972996. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjE3OiIzNzAvNjUxNS9lYWJkNDU4NSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzExLzI3LzIwMjAuMTEuMTkuMjAyMzQyMzcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 6. 6.Poulas K, Farsalinos K, Zanidis C. Activation of TLR7 and Innate Immunity as an Efficient Method Against COVID-19 Pandemic: Imiquimod as a Potential Therapy. Front Immunol. 2020 Jun 11;11:1373. doi: 10.3389/fimmu.2020.01373. PMID: 32612613; PMCID: PMC7307572. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2020.01373&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32612613&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F27%2F2020.11.19.20234237.atom) 7. 7.Mele D, Mantovani S, Oliviero B, Grossi G, Ludovisi S, Mondelli MU, et al. Hepatitis C virus inhibits CD4 T cell function via binding to Toll-like receptor 7. Antiviral Res. 137, 108–111 (2017). 8. 8.van der Made CI, Simons A, Schuurs-Hoeijmakers J, van den Heuvel G, Mantere T, Kersten S, et al. Presence of Genetic Variants Among Young Men With Severe COVID-19 [published online ahead of print, 2020 Jul 24]. JAMA. 324, 1–11 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2020.13719&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32706371&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F27%2F2020.11.19.20234237.atom) 9. 9.Daga S, Fallerini C, Baldassarri M, Fava F, Valentino F, Doddato G, et al. Employing a Systematic Approach to Biobanking and Analyzing Genetic and Clinical Data for Advancing COVID-19 Research. medRxiv 2020.07.24.20161307; doi: [https://doi.org/10.1101/2020.07.24.20161307](https://doi.org/10.1101/2020.07.24.20161307) (2020).