Inborn errors of type I IFN immunity in patients with life-threatening COVID-19

Qian Zhang, Paul Bastard, Zhiyong Liu, Jérémie Le Pen, Marcela Moncada-Velez, Jie Chen, Masato Ogishi, Ira K D Sabli, Stephanie Hodeib, Cecilia Korol, Jérémie Rosain, Kaya Bilguvar, Junqiang Ye, Alexandre Bolze, Benedetta Bigio, Rui Yang, Andrés Augusto Arias, Qinhua Zhou, Yu Zhang, Fanny Onodi, Sarantis Korniotis, Léa Karpf, Quentin Philippot, Marwa Chbihi, Lucie Bonnet-Madin, Karim Dorgham, Nikaïa Smith, William M Schneider, Brandon S Razooky, Hans-Heinrich Hoffmann, Eleftherios Michailidis, Leen Moens, Ji Eun Han, Lazaro Lorenzo, Lucy Bizien, Philip Meade, Anna-Lena Neehus, Aileen Camille Ugurbil, Aurélien Corneau, Gaspard Kerner, Peng Zhang, Franck Rapaport, Yoann Seeleuthner, Jeremy Manry, Cecile Masson, Yohann Schmitt, Agatha Schlüter, Tom Le Voyer, Taushif Khan, Juan Li, Jacques Fellay, Lucie Roussel, Mohammad Shahrooei, Mohammed F Alosaimi, Davood Mansouri, Haya Al-Saud, Fahd Al-Mulla, Feras Almourfi, Saleh Zaid Al-Muhsen, Fahad Alsohime, Saeed Al Turki, Rana Hasanato, Diederik van de Beek, Andrea Biondi, Laura Rachele Bettini, Mariella D'Angio', Paolo Bonfanti, Luisa Imberti, Alessandra Sottini, Simone Paghera, Eugenia Quiros-Roldan, Camillo Rossi, Andrew J Oler, Miranda F Tompkins, Camille Alba, Isabelle Vandernoot, Jean-Christophe Goffard, Guillaume Smits, Isabelle Migeotte, Filomeen Haerynck, Pere Soler-Palacin, Andrea Martin-Nalda, Roger Colobran, Pierre-Emmanuel Morange, Sevgi Keles, Fatma Çölkesen, Tayfun Ozcelik, Kadriye Kart Yasar, Sevtap Senoglu, Şemsi Nur Karabela, Carlos Rodríguez-Gallego, Giuseppe Novelli, Sami Hraiech, Yacine Tandjaoui-Lambiotte, Xavier Duval, Cédric Laouénan, COVID-STORM Clinicians, COVID Clinicians, Imagine COVID Group, French COVID Cohort Study Group, CoV-Contact Cohort, Amsterdam UMC Covid-19 Biobank, COVID Human Genetic Effort, NIAID-USUHS/TAGC COVID Immunity Group, Andrew L Snow, Clifton L Dalgard, Joshua D Milner, Donald C Vinh, Trine H Mogensen, Nico Marr, András N Spaan, Bertrand Boisson, Stéphanie Boisson-Dupuis, Jacinta Bustamante, Anne Puel, Michael J Ciancanelli, Isabelle Meyts, Tom Maniatis, Vassili Soumelis, Ali Amara, Michel Nussenzweig, Adolfo García-Sastre, Florian Krammer, Aurora Pujol, Darragh Duffy, Richard P Lifton, Shen-Ying Zhang, Guy Gorochov, Vivien Béziat, Emmanuelle Jouanguy, Vanessa Sancho-Shimizu, Charles M Rice, Laurent Abel, Luigi D Notarangelo, Aurélie Cobat, Helen C Su, Jean-Laurent Casanova

Abstract

Clinical outcome upon infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ranges from silent infection to lethal coronavirus disease 2019 (COVID-19). We have found an enrichment in rare variants predicted to be loss-of-function (LOF) at the 13 human loci known to govern Toll-like receptor 3 (TLR3)- and interferon regulatory factor 7 (IRF7)-dependent type I interferon (IFN) immunity to influenza virus in 659 patients with life-threatening COVID-19 pneumonia relative to 534 subjects with asymptomatic or benign infection. By testing these and other rare variants at these 13 loci, we experimentally defined LOF variants underlying autosomal-recessive or autosomal-dominant deficiencies in 23 patients (3.5%) 17 to 77 years of age. We show that human fibroblasts with mutations affecting this circuit are vulnerable to SARS-CoV-2. Inborn errors of TLR3- and IRF7-dependent type I IFN immunity can underlie life-threatening COVID-19 pneumonia in patients with no prior severe infection.

Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.

Figures

Inborn errors of TLR3- and IRF7-dependent type…
Inborn errors of TLR3- and IRF7-dependent type I IFN production and amplification underlie life-threatening COVID-19 pneumonia.
Molecules in red are encoded by core genes, deleterious variants of which underlie critical influenza pneumonia with incomplete penetrance, and deleterious variants of genes encoding biochemically related molecules in blue underlie other viral illnesses. Molecules represented in bold are encoded by genes with variants that also underlie critical COVID-19 pneumonia.
Fig. 1. Demographic and genetic data for…
Fig. 1. Demographic and genetic data for the COVID-19 cohort.
(A) Age and sex distribution of patients with life-threatening COVID-19. (B) PCA of patient (with or without LOF variants in the 13 candidate genes) and control cohorts (patients with mild or asymptomatic disease and individuals from the 1000 Genomes Project).
Fig. 2. Illustration of TLR3- and IRF7-dependent…
Fig. 2. Illustration of TLR3- and IRF7-dependent type I IFN production and amplification circuit.
Molecules in red are encoded by core genes, deleterious variants of which underlie critical influenza pneumonia with incomplete penetrance; deleterious variants of genes encoding biochemically related molecules in blue underlie other viral illnesses. Type I IFNs also induce themselves. ISGs, interferon-stimulated genes.
Fig. 3. Impact of TLR3 , TICAM1…
Fig. 3. Impact of TLR3, TICAM1, TBK1, IRF3, IRF7, IFNAR1, and IFNAR2 variants on type I IFN signaling.
(A) TLR3-deficient P2.1 fibrosarcoma cells were stably transfected with plasmids expressing WT or mutant forms of TLR3, and IFNL1 mRNA levels were determined by reverse transcription quantitative PCR. IFNL1 mRNA levels were expressed relative to the housekeeping gene GUS and then normalized. IFNL1 was undetectable in unstimulated cells. The differences between variants and WT were tested using one-way ANOVA (*P < 0.05). (B) TICAM1-deficient SV40-Fib cells were transiently transfected with WT or mutant forms of TICAM1, together with an IFN-β luciferase reporter and a constitutively expressed reporter. Normalized luciferase induction was measured 24 hours after transfection. The differences between variants and WT were tested using one-way ANOVA (*P < 0.05). (C) HEK293T cells were transiently transfected with WT and mutant forms of TBK1, together with an IFN-β luciferase reporter and a constitutively expressed reporter. Normalized luciferase activity was measured 24 hours after transfection. The differences between variants and WT were tested using one-way ANOVA (*P < 0.05). (D) IRF3-deficient HEK293T cells were transiently transfected with WT and mutant forms of IRF3, together with an IFN-β luciferase reporter and a constitutively expressed reporter. Cells were either left untreated or infected with Sendai virus for 24 hours before the normalized measurement of luciferase activity. The differences between variants and WT were evaluated using two-way ANOVA (*P < 0.05). (E) HEK293T cells were transiently transfected with WT and mutant forms of IRF7, together with an IFN-β luciferase reporter and a constitutively expressed reporter. Cells were either left untreated or infected with Sendai virus for 24 hours before the normalized measurement of luciferase activity. The differences between variants and WT were tested using two-way ANOVA (*P < 0.05). (F and G) IFNAR1- or IFNAR2-deficient SV40-Fib cells were transiently transfected with WT or mutant forms of IFNAR1 for 36 hours, and either left untreated or stimulated with IFN-α2 or IFN-γ. Fluorescence-activated cell sorting (FACS) staining with anti-p-STAT1 antibody and the z-score of the MFI were assessed. Asterisks indicate variants with MFI <50% of WT. Variants in red were identified in COVID-19 patients. Variants in blue are known deleterious variants and served as negative controls. EV, empty vector; LT, lipofectamine. Three technical repeats were performed for (A) to (E). Means and SD are shown in the columns and horizontal bars when appropriate.
Fig. 4. Type I IFN responses in…
Fig. 4. Type I IFN responses in patient cells defective for IRF7.
(A) Levels of the IRF7 protein in PHA-T cells from two patients with AR IRF7 deficiency (P1 and P3), one patient with AD IRF7 deficiency (P2), and four healthy donors (C1 to C4). Cells were either left untreated or stimulated with IFN-α2 for 24 hours, and protein levels were measured by Western blotting. MX1 was used as a positive control for IFN-α2 treatment. (B) pDCs isolated from an AR IRF7-deficient patient (P1) and a healthy donor (C1) were either left untreated or infected with influenza A virus (IAV) or SARS-CoV-2, and RNA-seq was performed. Genes with expression >2.5-fold higher or lower in C1 after infection are plotted as the fold change in expression. Red dots are type I IFN genes; blue dots are type III IFN genes. (C) pDCs isolated from healthy donor C and IRF7-deficient patient (P1) were either left untreated (Medium) or infected with IAV or SARS-CoV-2, and the production of IFN-α2 and IFN-λ1 was measured by CBA and ELISA, respectively, on the supernatant. ND, not detected.
Fig. 5. Type I IFN responses in…
Fig. 5. Type I IFN responses in patient cells defective for IFNAR1.
(A) FACS staining of IFNAR1 on the surface of PHA-T cells from a patient with AR IFNAR1 deficiency (P5) and healthy donors (C1 and C2). (B) PHA-T cells and SV40-Fib from a patient with AR IFNAR1 deficiency (P5) and a healthy donor (C3) were stimulated with IFN-α2 or IFN-β, and p-STAT1 levels were determined by FACS. Interleukin-27 stimulation served as a positive control on PHA-T cells, whereas IFN-γ stimulation served as a positive control on SV40-Fib cells.
Fig. 6. Cell-intrinsic type I IFN response…
Fig. 6. Cell-intrinsic type I IFN response to SARS-CoV-2.
SV40-Fib cells of TLR3−/−, TLR3+/−, IRF7−/−, and IRF7−/− SV40-Fib cells rescued with WT IRF7; IFNAR1−/− SV40-Fib cells, and IFNAR1−/− SV40-Fib cells rescued with WT IFNAR1 were transduced with ACE2 and TMPRSS2 and then either left untreated or treated with IFN-β for 4 hours. Cells were then infected with SARS-CoV-2 (MOI = 0.5). After staining, ACE2 and viral S-protein levels were measured by high-content microscopy with gating on ACE2+ cells. IRF7-deficient SV40-Fib cells were previously transduced with either WT IRF7 or negative control (Luc). IFNAR1-deficient cells were previously transduced with either WT IFNAR1 or empty vector (EV).
Fig. 7. In vivo type I IFN…
Fig. 7. In vivo type I IFN responses to SARS-CoV-2 infections.
Plasma levels of 13 IFN-α were measured by Simoa. Auto-Ab(+) without LOF variants indicates COVID-19 patients with neutralizing anti-IFN-α auto-Abs in our accompanying report (29). P values indicated were evaluated using one-way ANOVA.

References

    1. Morens D. M., Fauci A. S., Emerging pandemic diseases: How we got to COVID-19. Cell 182, 1077–1092 (2020). 10.1016/j.cell.2020.08.021
    1. Casanova J. L., Abel L., Lethal Infectious Diseases as Inborn Errors of Immunity: Toward a Synthesis of the Germ and Genetic Theories. Annu. Rev. Pathol. (2020).
    1. Casanova J. L., Abel L., The human genetic determinism of life-threatening infectious diseases: Genetic heterogeneity and physiological homogeneity? Hum. Genet. 139, 681–694 (2020). 10.1007/s00439-020-02184-w
    1. Casanova J. L., Su H. C.; COVID Human Genetic Effort , A global effort to define the human genetics of protective immunity to SARS-CoV-2 infection. Cell 181, 1194–1199 (2020). 10.1016/j.cell.2020.05.016
    1. Zhang Q., Human genetics of life-threatening influenza pneumonitis. Hum. Genet. 139, 941–948 (2020). 10.1007/s00439-019-02108-3
    1. Lim H. K., Huang S. X. L., Chen J., Kerner G., Gilliaux O., Bastard P., Dobbs K., Hernandez N., Goudin N., Hasek M. L., García Reino E. J., Lafaille F. G., Lorenzo L., Luthra P., Kochetkov T., Bigio B., Boucherit S., Rozenberg F., Vedrinne C., Keller M. D., Itan Y., García-Sastre A., Celard M., Orange J. S., Ciancanelli M. J., Meyts I., Zhang Q., Abel L., Notarangelo L. D., Snoeck H.-W., Casanova J.-L., Zhang S.-Y., Severe influenza pneumonitis in children with inherited TLR3 deficiency. J. Exp. Med. 216, 2038–2056 (2019). 10.1084/jem.20181621
    1. Ciancanelli M. J., Huang S. X. L., Luthra P., Garner H., Itan Y., Volpi S., Lafaille F. G., Trouillet C., Schmolke M., Albrecht R. A., Israelsson E., Lim H. K., Casadio M., Hermesh T., Lorenzo L., Leung L. W., Pedergnana V., Boisson B., Okada S., Picard C., Ringuier B., Troussier F., Chaussabel D., Abel L., Pellier I., Notarangelo L. D., García-Sastre A., Basler C. F., Geissmann F., Zhang S.-Y., Snoeck H.-W., Casanova J.-L., Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science 348, 448–453 (2015). 10.1126/science.aaa1578
    1. Hernandez N., Melki I., Jing H., Habib T., Huang S. S. Y., Danielson J., Kula T., Drutman S., Belkaya S., Rattina V., Lorenzo-Diaz L., Boulai A., Rose Y., Kitabayashi N., Rodero M. P., Dumaine C., Blanche S., Lebras M.-N., Leung M. C., Mathew L. S., Boisson B., Zhang S.-Y., Boisson-Dupuis S., Giliani S., Chaussabel D., Notarangelo L. D., Elledge S. J., Ciancanelli M. J., Abel L., Zhang Q., Marr N., Crow Y. J., Su H. C., Casanova J.-L., Life-threatening influenza pneumonitis in a child with inherited IRF9 deficiency. J. Exp. Med. 215, 2567–2585 (2018). 10.1084/jem.20180628
    1. Sancho-Shimizu V., Pérez de Diego R., Lorenzo L., Halwani R., Alangari A., Israelsson E., Fabrega S., Cardon A., Maluenda J., Tatematsu M., Mahvelati F., Herman M., Ciancanelli M., Guo Y., AlSum Z., Alkhamis N., Al-Makadma A. S., Ghadiri A., Boucherit S., Plancoulaine S., Picard C., Rozenberg F., Tardieu M., Lebon P., Jouanguy E., Rezaei N., Seya T., Matsumoto M., Chaussabel D., Puel A., Zhang S.-Y., Abel L., Al-Muhsen S., Casanova J.-L., Herpes simplex encephalitis in children with autosomal recessive and dominant TRIF deficiency. J. Clin. Invest. 121, 4889–4902 (2011). 10.1172/JCI59259
    1. Casrouge A., Zhang S.-Y., Eidenschenk C., Jouanguy E., Puel A., Yang K., Alcais A., Picard C., Mahfoufi N., Nicolas N., Lorenzo L., Plancoulaine S., Sénéchal B., Geissmann F., Tabeta K., Hoebe K., Du X., Miller R. L., Héron B., Mignot C., de Villemeur T. B., Lebon P., Dulac O., Rozenberg F., Beutler B., Tardieu M., Abel L., Casanova J.-L., Herpes simplex virus encephalitis in human UNC-93B deficiency. Science 314, 308–312 (2006). 10.1126/science.1128346
    1. Pérez de Diego R., Sancho-Shimizu V., Lorenzo L., Puel A., Plancoulaine S., Picard C., Herman M., Cardon A., Durandy A., Bustamante J., Vallabhapurapu S., Bravo J., Warnatz K., Chaix Y., Cascarrigny F., Lebon P., Rozenberg F., Karin M., Tardieu M., Al-Muhsen S., Jouanguy E., Zhang S.-Y., Abel L., Casanova J.-L., Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis. Immunity 33, 400–411 (2010). 10.1016/j.immuni.2010.08.014
    1. Herman M., Ciancanelli M., Ou Y.-H., Lorenzo L., Klaudel-Dreszler M., Pauwels E., Sancho-Shimizu V., Pérez de Diego R., Abhyankar A., Israelsson E., Guo Y., Cardon A., Rozenberg F., Lebon P., Tardieu M., Heropolitańska-Pliszka E., Chaussabel D., White M. A., Abel L., Zhang S.-Y., Casanova J.-L., Heterozygous TBK1 mutations impair TLR3 immunity and underlie herpes simplex encephalitis of childhood. J. Exp. Med. 209, 1567–1582 (2012). 10.1084/jem.20111316
    1. Andersen L. L., Mørk N., Reinert L. S., Kofod-Olsen E., Narita R., Jørgensen S. E., Skipper K. A., Höning K., Gad H. H., Østergaard L., Ørntoft T. F., Hornung V., Paludan S. R., Mikkelsen J. G., Fujita T., Christiansen M., Hartmann R., Mogensen T. H., Functional IRF3 deficiency in a patient with herpes simplex encephalitis. J. Exp. Med. 212, 1371–1379 (2015). 10.1084/jem.20142274
    1. Audry M., Ciancanelli M., Yang K., Cobat A., Chang H.-H., Sancho-Shimizu V., Lorenzo L., Niehues T., Reichenbach J., Li X.-X., Israel A., Abel L., Casanova J.-L., Zhang S.-Y., Jouanguy E., Puel A., NEMO is a key component of NF-κB- and IRF-3-dependent TLR3-mediated immunity to herpes simplex virus. J. Allergy Clin. Immunol. 128, 610–617.e4 (2011). 10.1016/j.jaci.2011.04.059
    1. Hernandez N., Bucciol G., Moens L., Le Pen J., Shahrooei M., Goudouris E., Shirkani A., Changi-Ashtiani M., Rokni-Zadeh H., Sayar E. H., Reisli I., Lefevre-Utile A., Zijlmans D., Jurado A., Pholien R., Drutman S., Belkaya S., Cobat A., Boudewijns R., Jochmans D., Neyts J., Seeleuthner Y., Lorenzo-Diaz L., Enemchukwu C., Tietjen I., Hoffmann H.-H., Momenilandi M., Pöyhönen L., Siqueira M. M., de Lima S. M. B., de Souza Matos D. C., Homma A., Maia M. L. S., da Costa Barros T. A., de Oliveira P. M. N., Mesquita E. C., Gijsbers R., Zhang S.-Y., Seligman S. J., Abel L., Hertzog P., Marr N., Martins R. M., Meyts I., Zhang Q., MacDonald M. R., Rice C. M., Casanova J.-L., Jouanguy E., Bossuyt X., Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J. Exp. Med. 216, 2057–2070 (2019). 10.1084/jem.20182295
    1. Duncan C. J., Mohamad S. M. B., Young D. F., Skelton A. J., Leahy T. R., Munday D. C., Butler K. M., Morfopoulou S., Brown J. R., Hubank M., Connell J., Gavin P. J., McMahon C., Dempsey E., Lynch N. E., Jacques T. S., Valappil M., Cant A. J., Breuer J., Engelhardt K. R., Randall R. E., Hambleton S., Human IFNAR2 deficiency: Lessons for antiviral immunity. Sci. Transl. Med. 7, 307ra154 (2015). 10.1126/scitranslmed.aac4227
    1. Dupuis S., Jouanguy E., Al-Hajjar S., Fieschi C., Al-Mohsen I. Z., Al-Jumaah S., Yang K., Chapgier A., Eidenschenk C., Eid P., Al Ghonaium A., Tufenkeji H., Frayha H., Al-Gazlan S., Al-Rayes H., Schreiber R. D., Gresser I., Casanova J.-L., Impaired response to interferon-alpha/beta and lethal viral disease in human STAT1 deficiency. Nat. Genet. 33, 388–391 (2003). 10.1038/ng1097
    1. Hambleton S., Goodbourn S., Young D. F., Dickinson P., Mohamad S. M. B., Valappil M., McGovern N., Cant A. J., Hackett S. J., Ghazal P., Morgan N. V., Randall R. E., STAT2 deficiency and susceptibility to viral illness in humans. Proc. Natl. Acad. Sci. U.S.A. 110, 3053–3058 (2013). 10.1073/pnas.1220098110
    1. Döffinger R., Smahi A., Bessia C., Geissmann F., Feinberg J., Durandy A., Bodemer C., Kenwrick S., Dupuis-Girod S., Blanche S., Wood P., Rabia S. H., Headon D. J., Overbeek P. A., Le Deist F., Holland S. M., Belani K., Kumararatne D. S., Fischer A., Shapiro R., Conley M. E., Reimund E., Kalhoff H., Abinun M., Munnich A., Israël A., Courtois G., Casanova J.-L., X-linked anhidrotic ectodermal dysplasia with immunodeficiency is caused by impaired NF-kappaB signaling. Nat. Genet. 27, 277–285 (2001). 10.1038/85837
    1. Zhang S. Y., Jouanguy E., Ugolini S., Smahi A., Elain G., Romero P., Segal D., Sancho-Shimizu V., Lorenzo L., Puel A., Picard C., Chapgier A., Plancoulaine S., Titeux M., Cognet C., von Bernuth H., Ku C.-L., Casrouge A., Zhang X.-X., Barreiro L., Leonard J., Hamilton C., Lebon P., Héron B., Vallée L., Quintana-Murci L., Hovnanian A., Rozenberg F., Vivier E., Geissmann F., Tardieu M., Abel L., Casanova J.-L., TLR3 deficiency in patients with herpes simplex encephalitis. Science 317, 1522–1527 (2007). 10.1126/science.1139522
    1. Zhang G., deWeerd N. A., Stifter S. A., Liu L., Zhou B., Wang W., Zhou Y., Ying B., Hu X., Matthews A. Y., Ellis M., Triccas J. A., Hertzog P. J., Britton W. J., Chen X., Feng C. G., A proline deletion in IFNAR1 impairs IFN-signaling and underlies increased resistance to tuberculosis in humans. Nat. Commun. 9, 85 (2018). 10.1038/s41467-017-02611-z
    1. Thomsen M. M., Jørgensen S. E., Storgaard M., Kristensen L. S., Gjedsted J., Christiansen M., Gad H. H., Hartmann R., Mogensen T. H., Identification of an IRF3 variant and defective antiviral interferon responses in a patient with severe influenza. Eur. J. Immunol. 49, 2111–2114 (2019). 10.1002/eji.201848083
    1. Thomsen M. M., Jørgensen S. E., Gad H. H., Storgaard M., Gjedsted J., Christiansen M., Hartmann R., Mogensen T. H., Defective interferon priming and impaired antiviral responses in a patient with an IRF7 variant and severe influenza. Med. Microbiol. Immunol. (Berl.) 208, 869–876 (2019). 10.1007/s00430-019-00623-8
    1. Tangye S. G., Al-Herz W., Bousfiha A., Chatila T., Cunningham-Rundles C., Etzioni A., Franco J. L., Holland S. M., Klein C., Morio T., Ochs H. D., Oksenhendler E., Picard C., Puck J., Torgerson T. R., Casanova J.-L., Sullivan K. E., Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J. Clin. Immunol. 40, 24–64 (2020). 10.1007/s10875-019-00737-x
    1. Bousfiha A., Jeddane L., Picard C., Al-Herz W., Ailal F., Chatila T., Cunningham-Rundles C., Etzioni A., Franco J. L., Holland S. M., Klein C., Morio T., Ochs H. D., Oksenhendler E., Puck J., Torgerson T. R., Casanova J.-L., Sullivan K. E., Tangye S. G., Human Inborn Errors of Immunity: 2019 Update of the IUIS Phenotypical Classification. J. Clin. Immunol. 40, 66–81 (2020). 10.1007/s10875-020-00758-x
    1. Notarangelo L. D., Bacchetta R., Casanova J.-L., Su H. C., Human inborn errors of immunity: An expanding universe. Sci. Immunol. 5, eabb1662 (2020). 10.1126/sciimmunol.abb1662
    1. Hadjadj J., Yatim N., Barnabei L., Corneau A., Boussier J., Smith N., Péré H., Charbit B., Bondet V., Chenevier-Gobeaux C., Breillat P., Carlier N., Gauzit R., Morbieu C., Pène F., Marin N., Roche N., Szwebel T.-A., Merkling S. H., Treluyer J.-M., Veyer D., Mouthon L., Blanc C., Tharaux P.-L., Rozenberg F., Fischer A., Duffy D., Rieux-Laucat F., Kernéis S., Terrier B., Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724 (2020). 10.1126/science.abc6027
    1. Trouillet-Assant S., Viel S., Gaymard A., Pons S., Richard J.-C., Perret M., Villard M., Brengel-Pesce K., Lina B., Mezidi M., Bitker L., Belot A.; COVID HCL Study Group , Type I IFN immunoprofiling in COVID-19 patients. J. Allergy Clin. Immunol. 146, 206–208.e2 (2020). 10.1016/j.jaci.2020.04.029
    1. Bastard P., Rosen L. B., Zhang Q., Michailidis E., Hoffmann H.-H., Zhang Y., Dorgham K., Philippot Q., Rosain J., Béziat V., Manry J., Shaw E., Haljasmägi L., Peterson P., Lorenzo L., Bizien L., Trouillet-Assant S., Dobbs K., Almeida de Jesus A., Belot A., Kallaste A., Catherinot E., Tandjaoui-Lambiotte Y., Le Pen J., Kerner G., Bigio B., Seeleuthner Y., Yang R., Bolze A., Spaan A. N., Delmonte O. M., Abers M. S., Aiuti A., Casari G., Lampasona V., Piemonti L., Ciceri F., Bilguvar K., Lifton R. P., Vasse M., Smadja D. M., Migaud M., Hadjadj J., Terrier B., Duffy D., Quintana-Murci L., van de Beek D., Roussel L., Vinh D. C., Tangye S. G., Haerynck F., Dalmau D., Martinez-Picado J., Brodin P., Nussenzweig M. C., Boisson-Dupuis S., Rodríguez-Gallego C., Vogt G., Mogensen T. H., Oler A. J., Gu J., Burbelo P. D., Cohen J., Biondi A., Bettini L. R., D'Angio M., Bonfanti P., Rossignol P., Mayaux J., Rieux-Laucat F., Husebye E. S., Fusco F., Ursini M. V., Imberti L., Sottini A., Paghera S., Quiros-Roldan E., Rossi C., Castagnoli R., Montagna D., Licari A., Marseglia G. L., Duval X., Ghosn J., HGID Lab, NIAID-USUHS Immune Response to COVID Group, COVID Clinicians, COVID-STORM Clinicians, Imagine COVID Group, French COVID Cohort Study Group, The Milieu Intérieur Consortium, CoV-Contact Cohort, Amsterdam UMC Covid-19 Biobank, COVID Human Genetic Effort, Tsang J. S., Goldbach-Mansky R., Kisand K., Lionakis M. S., Puel A., Zhang S.-Y., Holland S. M., Gorochov G., Jouanguy E., Rice C. M., Cobat A., Notarangelo L. D., Abel L., Su H. C., Casanova J.-L., Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science 10.1126/science.abd4585 (2020).
    1. DePristo M. A., Banks E., Poplin R., Garimella K. V., Maguire J. R., Hartl C., Philippakis A. A., del Angel G., Rivas M. A., Hanna M., McKenna A., Fennell T. J., Kernytsky A. M., Sivachenko A. Y., Cibulskis K., Gabriel S. B., Altshuler D., Daly M. J., A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011). 10.1038/ng.806
    1. Li H., Durbin R., Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009). 10.1093/bioinformatics/btp324
    1. Boisson B., Honda Y., Ajiro M., Bustamante J., Bendavid M., Gennery A. R., Kawasaki Y., Ichishima J., Osawa M., Nihira H., Shiba T., Tanaka T., Chrabieh M., Bigio B., Hur H., Itan Y., Liang Y., Okada S., Izawa K., Nishikomori R., Ohara O., Heike T., Abel L., Puel A., Saito M. K., Casanova J.-L., Hagiwara M., Yasumi T., Rescue of recurrent deep intronic mutation underlying cell type-dependent quantitative NEMO deficiency. J. Clin. Invest. 129, 583–597 (2019). 10.1172/JCI124011
    1. Gambin T., Akdemir Z. C., Yuan B., Gu S., Chiang T., Carvalho C. M. B., Shaw C., Jhangiani S., Boone P. M., Eldomery M. K., Karaca E., Bayram Y., Stray-Pedersen A., Muzny D., Charng W. L., Bahrambeigi V., Belmont J. W., Boerwinkle E., Beaudet A. L., Gibbs R. A., Lupski J. R., Homozygous and hemizygous CNV detection from exome sequencing data in a Mendelian disease cohort. Nucleic Acids Res. 45, 1633–1648 (2017).
    1. Backenroth D., Homsy J., Murillo L. R., Glessner J., Lin E., Brueckner M., Lifton R., Goldmuntz E., Chung W. K., Shen Y., CANOES: Detecting rare copy number variants from whole exome sequencing data. Nucleic Acids Res. 42, e97 (2014). 10.1093/nar/gku345
    1. M. Bouaziz, J. Mullaert, B. Bigio, Y. Seeleuthner, J.-L. Casanova, A. Alcais, L. Abel, A. Cobat, Controlling for human population stratification in rare variant association studies. bioRxiv 969477 [Preprint]. 28 February 2020. .10.1101/2020.02.28.969477
    1. Persyn E., Redon R., Bellanger L., Dina C., The impact of a fine-scale population stratification on rare variant association test results. PLOS ONE 13, e0207677 (2018). 10.1371/journal.pone.0207677
    1. Zhang Y., Shen X., Pan W., Adjusting for population stratification in a fine scale with principal components and sequencing data. Genet. Epidemiol. 37, 787–801 (2013). 10.1002/gepi.21764
    1. Boisson-Dupuis S., Ramirez-Alejo N., Li Z., Patin E., Rao G., Kerner G., Lim C. K., Krementsov D. N., Hernandez N., Ma C. S., Zhang Q., Markle J., Martinez-Barricarte R., Payne K., Fisch R., Deswarte C., Halpern J., Bouaziz M., Mulwa J., Sivanesan D., Lazarov T., Naves R., Garcia P., Itan Y., Boisson B., Checchi A., Jabot-Hanin F., Cobat A., Guennoun A., Jackson C. C., Pekcan S., Caliskaner Z., Inostroza J., Costa-Carvalho B. T., de Albuquerque J. A. T., Garcia-Ortiz H., Orozco L., Ozcelik T., Abid A., Rhorfi I. A., Souhi H., Amrani H. N., Zegmout A., Geissmann F., Michnick S. W., Muller-Fleckenstein I., Fleckenstein B., Puel A., Ciancanelli M. J., Marr N., Abolhassani H., Balcells M. E., Condino-Neto A., Strickler A., Abarca K., Teuscher C., Ochs H. D., Reisli I., Sayar E. H., El-Baghdadi J., Bustamante J., Hammarström L., Tangye S. G., Pellegrini S., Quintana-Murci L., Abel L., Casanova J.-L., Tuberculosis and impaired IL-23-dependent IFN-γ immunity in humans homozygous for a common TYK2 missense variant. Sci. Immunol. 3, eaau8714 (2018). 10.1126/sciimmunol.aau8714
    1. Fochi S., Bergamo E., Serena M., Mutascio S., Journo C., Mahieux R., Ciminale V., Bertazzoni U., Zipeto D., Romanelli M. G., TRAF3 Is Required for NF-κB Pathway Activation Mediated by HTLV Tax Proteins. Front. Microbiol. 10, 1302 (2019). 10.3389/fmicb.2019.01302
    1. Robbiani D. F., Gaebler C., Muecksch F., Lorenzi J. C. C., Wang Z., Cho A., Agudelo M., Barnes C. O., Gazumyan A., Finkin S., Hägglöf T., Oliveira T. Y., Viant C., Hurley A., Hoffmann H.-H., Millard K. G., Kost R. G., Cipolla M., Gordon K., Bianchini F., Chen S. T., Ramos V., Patel R., Dizon J., Shimeliovich I., Mendoza P., Hartweger H., Nogueira L., Pack M., Horowitz J., Schmidt F., Weisblum Y., Michailidis E., Ashbrook A. W., Waltari E., Pak J. E., Huey-Tubman K. E., Koranda N., Hoffman P. R., West A. P. Jr.., Rice C. M., Hatziioannou T., Bjorkman P. J., Bieniasz P. D., Caskey M., Nussenzweig M. C., Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature 584, 437–442 (2020). 10.1038/s41586-020-2456-9
    1. Q. Zhang, P. Bastard, Z. Liu, J. Le Pen, M. Moncada-Velez, J. Chen, M. Ogishi, I. K. D. Sabli, S. Hodeib, C. Korol, J. Rosain, K. Bilguvar, J. Ye, A. Bolze, B. Bigio, R. Yang, A. Augusto Arias Sierra, Q. Zhou, Y. Zhang, F. Onodi, S. Korniotis, L. Karpf, Q. Philippot, M. Chbihi, L. Bonnet-Madin, K. Dorgham, N. Smith, W. M. Schneider, B. S. Razooky, H.-H. Hoffmann, E. Michailidis, L. Moens, J. E. Han, L. Lorenzo, L. Bizien, P. Meade, A.-L. Neehus, A. C. Ugurbil, A. Corneau, G. Kerner, P. Zhang, F. Rapaport, Y. Seeleuthner, J. Manry, C. Masson, Y. Schmitt, A. Schlüter, T. Le Voyer, T. Khan, J. Li, J. Fellay, L. Roussel, M. Shahrooei, M. F. Alosaimi, D. Mansouri, H. Al-Saud, F. Al-Mulla, F. Almourfi, S. Z. Al-Muhsen, F. Alsohime, S. Al Turki, R. Hasanato, D. van de Beek, A. Biondi, L. R. Bettini, M. D’Angio, P. Bonfanti, L. Imberti, A. Sottini, S. Paghera, E. Quiros-Roldan, C. Rossi, A. J. Oler, M. F. Tompkins, C. Alba, I. Vandernoot, J.-C. Goffard, G. Smits, I. Migeotte, F. Haerynck, P. Soler-Palacin, A. Martin-Nalda, R. Colobran, P.-E. Morange, S. Keles, F. Çölkesen, T. Ozcelik, K. K. Yasar, S. Senoglu, Ş. N. Karabela, C. Rodríguez-Gallego, G. Novelli, S. Hraiech, Y. Tandjaoui-Lambiotte, X. Duval, C. Laouenan, COVID-STORM Clinicians, COVID Clinicians, Imagine COVID Group, French COVID Cohort Study Group, CoV-Contact Cohort, Amsterdam UMC Covid-19 Biobank, COVID Human Genetic Effort, NIAID-USUHS/TAGC COVID Immunity Group, A. L. Snow, C. L. Dalgard, J. Milner, D. C. Vinh, T. H. Mogensen, N. Marr, A. N. Spaan, B. Boisson, S. Boisson-Dupuis, J. Bustamante, A. Puel, M. Ciancanelli, I. Meyts, T. Maniatis, V. Soumelis, A. Amara, M. Nussenzweig, A. García-Sastre, F. Krammer, A. Pujol, D. Duffy, R. Lifton, S.-Y. Zhang, G. Gorochov, V. Béziat, E. Jouanguy, V. Sancho-Shimizu, C. M. Rice, L. Abel, L. D. Notarangelo, A. Cobat, H. C. Su, J.-L. Casanova, Detailed genotype counts for all coding variants for: Inborn errors of type I IFN immunity in patients with life-threatening COVID-19, Dryad (2020); . 10.5061/dryad.8pk0p2nkk
    1. M. Ogishi, R. Yang, C. Gruber, S. Pelham, A. N. Spaan, J. Rosain, M. Chbihi, J. E. Han, V. K. Rao, L. Kainulainen, J. Bustamante, B. Boisson, D. Bogunovic, S. Boisson-Dupuis, J.-L. Casanova, Multi-batch cytometry data integration for optimal immunophenotyping. bioRxiv 202432 [Preprint]. 15 July 2020. 10.1101/2020.07.14.202432
    1. Meade P., Kuan G., Strohmeier S., Maier H. E., Amanat F., Balmaseda A., Ito K., Kirkpatrick E., Javier A., Gresh L., Nachbagauer R., Gordon A., Krammer F., Influenza virus infection induces a narrow antibody response in children but a broad recall response in adults. mBio 11, e03243-19 (2020). 10.1128/mBio.03243-19

Source: PubMed

3
Subscribe