Try a new search

Format these results:

Searched for:

person:bigiob02

in-biosketch:yes

Total Results:

43


Inherited deficiency of stress granule ZNFX1 in patients with monocytosis and mycobacterial disease [Case Report]

Le Voyer, Tom; Neehus, Anna-Lena; Yang, Rui; Ogishi, Masato; Rosain, Jérémie; Alroqi, Fayhan; Alshalan, Maha; Blumental, Sophie; Al Ali, Fatima; Khan, Taushif; Ata, Manar; Rozen, Laurence; Demulder, Anne; Bastard, Paul; Gruber, Conor; Roynard, Manon; Seeleuthener, Yoann; Rapaport, Franck; Bigio, Benedetta; Chrabieh, Maya; Sng, Danielle; Berteloot, Laureline; Boddaert, Nathalie; Rozenberg, Flore; Al-Muhsen, Saleh; Bertoli-Avella, Aida; Abel, Laurent; Bogunovic, Dusan; Marr, Nico; Mansouri, Davood; Al Mutairi, Fuad; Béziat, Vivien; Weil, Dominique; Mahdaviani, Seyed Alireza; Ferster, Alina; Zhang, Shen-Ying; Reversade, Bruno; Boisson-Dupuis, Stéphanie; Casanova, Jean-Laurent; Bustamante, Jacinta
Human inborn errors of IFN-γ underlie mycobacterial disease, due to insufficient IFN-γ production by lymphoid cells, impaired myeloid cell responses to this cytokine, or both. We report four patients from two unrelated kindreds with intermittent monocytosis and mycobacterial disease, including bacillus Calmette-Guérin-osis and disseminated tuberculosis, and without any known inborn error of IFN-γ. The patients are homozygous for ZNFX1 variants (p.S959* and p.E1606Rfs*10) predicted to be loss of function (pLOF). There are no subjects homozygous for pLOF variants in public databases. ZNFX1 is a conserved and broadly expressed helicase, but its biology remains largely unknown. It is thought to act as a viral double-stranded RNA sensor in mice, but these patients do not suffer from severe viral illnesses. We analyze its subcellular localization upon overexpression in A549 and HeLa cell lines and upon stimulation of THP1 and fibroblastic cell lines. We find that this cytoplasmic protein can be recruited to or even induce stress granules. The endogenous ZNFX1 protein in cell lines of the patient homozygous for the p.E1606Rfs*10 variant is truncated, whereas ZNFX1 expression is abolished in cell lines from the patients with the p.S959* variant. Lymphocyte subsets are present at normal frequencies in these patients and produce IFN-γ normally. The hematopoietic and nonhematopoietic cells of the patients tested respond normally to IFN-γ. Our results indicate that human ZNFX1 is associated with stress granules and essential for both monocyte homeostasis and protective immunity to mycobacteria.
PMCID:8053974
PMID: 33876776
ISSN: 1091-6490
CID: 5065102

Inhibition of HECT E3 ligases as potential therapy for COVID-19

Novelli, Giuseppe; Liu, Jing; Biancolella, Michela; Alonzi, Tonino; Novelli, Antonio; Patten, J J; Cocciadiferro, Dario; Agolini, Emanuele; Colona, Vito Luigi; Rizzacasa, Barbara; Giannini, Rosalinda; Bigio, Benedetta; Goletti, Delia; Capobianchi, Maria Rosaria; Grelli, Sandro; Mann, Justin; McKee, Trevor D; Cheng, Ke; Amanat, Fatima; Krammer, Florian; Guarracino, Andrea; Pepe, Gerardo; Tomino, Carlo; Tandjaoui-Lambiotte, Yacine; Uzunhan, Yurdagul; Tubiana, Sarah; Ghosn, Jade; Notarangelo, Luigi D; Su, Helen C; Abel, Laurent; Cobat, Aurélie; Elhanan, Gai; Grzymski, Joseph J; Latini, Andrea; Sidhu, Sachdev S; Jain, Suresh; Davey, Robert A; Casanova, Jean-Laurent; Wei, Wenyi; Pandolfi, Pier Paolo
SARS-CoV-2 is responsible for the ongoing world-wide pandemic which has already taken more than two million lives. Effective treatments are urgently needed. The enzymatic activity of the HECT-E3 ligase family members has been implicated in the cell egression phase of deadly RNA viruses such as Ebola through direct interaction of its VP40 Protein. Here we report that HECT-E3 ligase family members such as NEDD4 and WWP1 interact with and ubiquitylate the SARS-CoV-2 Spike protein. Furthermore, we find that HECT family members are overexpressed in primary samples derived from COVID-19 infected patients and COVID-19 mouse models. Importantly, rare germline activating variants in the NEDD4 and WWP1 genes are associated with severe COVID-19 cases. Critically, I3C, a natural NEDD4 and WWP1 inhibitor from Brassicaceae, displays potent antiviral effects and inhibits viral egression. In conclusion, we identify the HECT family members of E3 ligases as likely novel biomarkers for COVID-19, as well as new potential targets of therapeutic strategy easily testable in clinical trials in view of the established well-tolerated nature of the Brassicaceae natural compounds.
PMCID:7987752
PMID: 33762578
ISSN: 2041-4889
CID: 5065092

TLR3 controls constitutive IFN-β antiviral immunity in human fibroblasts and cortical neurons

Gao, Daxing; Ciancanelli, Michael J; Zhang, Peng; Harschnitz, Oliver; Bondet, Vincent; Hasek, Mary; Chen, Jie; Mu, Xin; Itan, Yuval; Cobat, Aurélie; Sancho-Shimizu, Vanessa; Bigio, Benedetta; Lorenzo, Lazaro; Ciceri, Gabriele; McAlpine, Jessica; Anguiano, Esperanza; Jouanguy, Emmanuelle; Chaussabel, Damien; Meyts, Isabelle; Diamond, Michael S; Abel, Laurent; Hur, Sun; Smith, Gregory A; Notarangelo, Luigi; Duffy, Darragh; Studer, Lorenz; Casanova, Jean-Laurent; Zhang, Shen-Ying
Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/β induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-β protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-β secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-β immunity.
PMCID:7773389
PMID: 33393505
ISSN: 1558-8238
CID: 5065082

Herpes simplex encephalitis in a patient with a distinctive form of inherited IFNAR1 deficiency [Case Report]

Bastard, Paul; Manry, Jeremy; Chen, Jie; Rosain, Jérémie; Seeleuthner, Yoann; AbuZaitun, Omar; Lorenzo, Lazaro; Khan, Taushif; Hasek, Mary; Hernandez, Nicholas; Bigio, Benedetta; Zhang, Peng; Lévy, Romain; Shrot, Shai; Reino, Eduardo J Garcia; Lee, Yoon-Seung; Boucherit, Soraya; Aubart, Mélodie; Gijsbers, Rik; Béziat, Vivien; Li, Zhi; Pellegrini, Sandra; Rozenberg, Flore; Marr, Nico; Meyts, Isabelle; Boisson, Bertrand; Cobat, Aurélie; Bustamante, Jacinta; Zhang, Qian; Jouangy, Emmanuelle; Abel, Laurent; Somech, Raz; Casanova, Jean-Laurent; Zhang, Shen-Ying
Inborn errors of TLR3-dependent IFN-α/β- and IFN-λ-mediated immunity in the CNS can underlie herpes simplex virus 1 (HSV-1) encephalitis (HSE). The respective contributions of IFN-α/β and IFN-λ are unknown. We report a child homozygous for a genomic deletion of the entire coding sequence and part of the 3'-UTR of the last exon of IFNAR1, who died of HSE at the age of 2 years. An older cousin died following vaccination against measles, mumps, and rubella at 12 months of age, and another 17-year-old cousin homozygous for the same variant has had other, less severe, viral illnesses. The encoded IFNAR1 protein is expressed on the cell surface but is truncated and cannot interact with the tyrosine kinase TYK2. The patient's fibroblasts and EBV-B cells did not respond to IFN-α2b or IFN-β, in terms of STAT1, STAT2, and STAT3 phosphorylation or the genome-wide induction of IFN-stimulated genes. The patient's fibroblasts were susceptible to viruses, including HSV-1, even in the presence of exogenous IFN-α2b or IFN-β. HSE is therefore a consequence of inherited complete IFNAR1 deficiency. This viral disease occurred in natural conditions, unlike those previously reported in other patients with IFNAR1 or IFNAR2 deficiency. This experiment of nature indicates that IFN-α/β are essential for anti-HSV-1 immunity in the CNS.
PMCID:7773360
PMID: 32960813
ISSN: 1558-8238
CID: 5065052

Autoantibodies against type I IFNs in patients with life-threatening COVID-19

Bastard, Paul; Rosen, Lindsey B; Zhang, Qian; Michailidis, Eleftherios; Hoffmann, Hans-Heinrich; Zhang, Yu; Dorgham, Karim; Philippot, Quentin; Rosain, Jérémie; Béziat, Vivien; Manry, Jérémy; Shaw, Elana; Haljasmägi, Liis; Peterson, Pärt; Lorenzo, Lazaro; Bizien, Lucy; Trouillet-Assant, Sophie; Dobbs, Kerry; de Jesus, Adriana Almeida; Belot, Alexandre; Kallaste, Anne; Catherinot, Emilie; Tandjaoui-Lambiotte, Yacine; Le Pen, Jeremie; Kerner, Gaspard; Bigio, Benedetta; Seeleuthner, Yoann; Yang, Rui; Bolze, Alexandre; Spaan, András N; Delmonte, Ottavia M; Abers, Michael S; Aiuti, Alessandro; Casari, Giorgio; Lampasona, Vito; Piemonti, Lorenzo; Ciceri, Fabio; Bilguvar, Kaya; Lifton, Richard P; Vasse, Marc; Smadja, David M; Migaud, Mélanie; Hadjadj, Jérome; Terrier, Benjamin; Duffy, Darragh; Quintana-Murci, Lluis; van de Beek, Diederik; Roussel, Lucie; Vinh, Donald C; Tangye, Stuart G; Haerynck, Filomeen; Dalmau, David; Martinez-Picado, Javier; Brodin, Petter; Nussenzweig, Michel C; Boisson-Dupuis, Stéphanie; Rodríguez-Gallego, Carlos; Vogt, Guillaume; Mogensen, Trine H; Oler, Andrew J; Gu, Jingwen; Burbelo, Peter D; Cohen, Jeffrey I; Biondi, Andrea; Bettini, Laura Rachele; D'Angio, Mariella; Bonfanti, Paolo; Rossignol, Patrick; Mayaux, Julien; Rieux-Laucat, Frédéric; Husebye, Eystein S; Fusco, Francesca; Ursini, Matilde Valeria; Imberti, Luisa; Sottini, Alessandra; Paghera, Simone; Quiros-Roldan, Eugenia; Rossi, Camillo; Castagnoli, Riccardo; Montagna, Daniela; Licari, Amelia; Marseglia, Gian Luigi; Duval, Xavier; Ghosn, Jade; Tsang, John S; Goldbach-Mansky, Raphaela; Kisand, Kai; Lionakis, Michail S; Puel, Anne; Zhang, Shen-Ying; Holland, Steven M; Gorochov, Guy; Jouanguy, Emmanuelle; Rice, Charles M; Cobat, Aurélie; Notarangelo, Luigi D; Abel, Laurent; Su, Helen C; Casanova, Jean-Laurent
Interindividual clinical variability in the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is vast. We report that at least 101 of 987 patients with life-threatening coronavirus disease 2019 (COVID-19) pneumonia had neutralizing immunoglobulin G (IgG) autoantibodies (auto-Abs) against interferon-ω (IFN-ω) (13 patients), against the 13 types of IFN-α (36), or against both (52) at the onset of critical disease; a few also had auto-Abs against the other three type I IFNs. The auto-Abs neutralize the ability of the corresponding type I IFNs to block SARS-CoV-2 infection in vitro. These auto-Abs were not found in 663 individuals with asymptomatic or mild SARS-CoV-2 infection and were present in only 4 of 1227 healthy individuals. Patients with auto-Abs were aged 25 to 87 years and 95 of the 101 were men. A B cell autoimmune phenocopy of inborn errors of type I IFN immunity accounts for life-threatening COVID-19 pneumonia in at least 2.6% of women and 12.5% of men.
PMID: 32972996
ISSN: 1095-9203
CID: 5065072

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

Zhang, Qian; Bastard, Paul; Liu, Zhiyong; Le Pen, Jérémie; Moncada-Velez, Marcela; Chen, Jie; Ogishi, Masato; Sabli, Ira K D; Hodeib, Stephanie; Korol, Cecilia; Rosain, Jérémie; Bilguvar, Kaya; Ye, Junqiang; Bolze, Alexandre; Bigio, Benedetta; Yang, Rui; Arias, Andrés Augusto; Zhou, Qinhua; Zhang, Yu; Onodi, Fanny; Korniotis, Sarantis; Karpf, Léa; Philippot, Quentin; Chbihi, Marwa; Bonnet-Madin, Lucie; Dorgham, Karim; Smith, Nikaïa; Schneider, William M; Razooky, Brandon S; Hoffmann, Hans-Heinrich; Michailidis, Eleftherios; Moens, Leen; Han, Ji Eun; Lorenzo, Lazaro; Bizien, Lucy; Meade, Philip; Neehus, Anna-Lena; Ugurbil, Aileen Camille; Corneau, Aurélien; Kerner, Gaspard; Zhang, Peng; Rapaport, Franck; Seeleuthner, Yoann; Manry, Jeremy; Masson, Cecile; Schmitt, Yohann; Schlüter, Agatha; Le Voyer, Tom; Khan, Taushif; Li, Juan; Fellay, Jacques; Roussel, Lucie; Shahrooei, Mohammad; Alosaimi, Mohammed F; Mansouri, Davood; Al-Saud, Haya; Al-Mulla, Fahd; Almourfi, Feras; Al-Muhsen, Saleh Zaid; Alsohime, Fahad; Al Turki, Saeed; Hasanato, Rana; van de Beek, Diederik; Biondi, Andrea; Bettini, Laura Rachele; D'Angio', Mariella; Bonfanti, Paolo; Imberti, Luisa; Sottini, Alessandra; Paghera, Simone; Quiros-Roldan, Eugenia; Rossi, Camillo; Oler, Andrew J; Tompkins, Miranda F; Alba, Camille; Vandernoot, Isabelle; Goffard, Jean-Christophe; Smits, Guillaume; Migeotte, Isabelle; Haerynck, Filomeen; Soler-Palacin, Pere; Martin-Nalda, Andrea; Colobran, Roger; Morange, Pierre-Emmanuel; Keles, Sevgi; Çölkesen, Fatma; Ozcelik, Tayfun; Yasar, Kadriye Kart; Senoglu, Sevtap; Karabela, Åžemsi Nur; Rodríguez-Gallego, Carlos; Novelli, Giuseppe; Hraiech, Sami; Tandjaoui-Lambiotte, Yacine; Duval, Xavier; Laouénan, Cédric; Snow, Andrew L; Dalgard, Clifton L; Milner, Joshua D; Vinh, Donald C; Mogensen, Trine H; Marr, Nico; Spaan, András N; Boisson, Bertrand; Boisson-Dupuis, Stéphanie; Bustamante, Jacinta; Puel, Anne; Ciancanelli, Michael J; Meyts, Isabelle; Maniatis, Tom; Soumelis, Vassili; Amara, Ali; Nussenzweig, Michel; García-Sastre, Adolfo; Krammer, Florian; Pujol, Aurora; Duffy, Darragh; Lifton, Richard P; Zhang, Shen-Ying; Gorochov, Guy; Béziat, Vivien; Jouanguy, Emmanuelle; Sancho-Shimizu, Vanessa; Rice, Charles M; Abel, Laurent; Notarangelo, Luigi D; Cobat, Aurélie; Su, Helen C; Casanova, Jean-Laurent
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.
PMID: 32972995
ISSN: 1095-9203
CID: 5065062

A genome-wide case-only test for the detection of digenic inheritance in human exomes

Kerner, Gaspard; Bouaziz, Matthieu; Cobat, Aurélie; Bigio, Benedetta; Timberlake, Andrew T; Bustamante, Jacinta; Lifton, Richard P; Casanova, Jean-Laurent; Abel, Laurent
Whole-exome sequencing (WES) has facilitated the discovery of genetic lesions underlying monogenic disorders. Incomplete penetrance and variable expressivity suggest a contribution of additional genetic lesions to clinical manifestations and outcome. Some monogenic disorders may therefore actually be digenic. However, only a few digenic disorders have been reported, all discovered by candidate gene approaches applied to at least one locus. We propose here a two-locus genome-wide test for detecting digenic inheritance in WES data. This approach uses the gene as the unit of analysis and tests all pairs of genes to detect pairwise gene × gene interactions underlying disease. It is a case-only method, which has several advantages over classic case-control tests, in particular by avoiding recruitment of controls. Our simulation studies based on real WES data identified two major sources of type I error inflation in this case-only test: linkage disequilibrium and population stratification. Both were corrected by specific procedures. Moreover, our case-only approach is more powerful than the corresponding case-control test for detecting digenic interactions in various population stratification scenarios. Finally, we confirmed the potential of our unbiased, genome-wide approach by successfully identifying a previously reported digenic lesion in patients with craniosynostosis. Our case-only test is a powerful and timely tool for detecting digenic inheritance in WES data from patients.
PMCID:7430978
PMID: 32719112
ISSN: 1091-6490
CID: 5065042

Fatal Cytomegalovirus Infection in an Adult with Inherited NOS2 Deficiency [Case Report]

Drutman, Scott B; Mansouri, Davood; Mahdaviani, Seyed Alireza; Neehus, Anna-Lena; Hum, David; Bryk, Ruslana; Hernandez, Nicholas; Belkaya, Serkan; Rapaport, Franck; Bigio, Benedetta; Fisch, Robert; Rahman, Mahbuba; Khan, Taushif; Al Ali, Fatima; Marjani, Majid; Mansouri, Nahal; Lorenzo-Diaz, Lazaro; Emile, Jean-François; Marr, Nico; Jouanguy, Emmanuelle; Bustamante, Jacinta; Abel, Laurent; Boisson-Dupuis, Stéphanie; Béziat, Vivien; Nathan, Carl; Casanova, Jean-Laurent
BACKGROUND:) are susceptible to the related murine CMV infection. METHODS:We studied a previously healthy 51-year-old man from Iran who after acute CMV infection had an onset of progressive CMV disease that led to his death 29 months later. We hypothesized that the patient may have had a novel type of inborn error of immunity. Thus, we performed whole-exome sequencing and tested candidate mutant alleles experimentally. RESULTS:variants that we found in homozygosity in public databases encoded functional proteins, as did all other variants with an allele frequency greater than 0.001. CONCLUSIONS:These findings suggest that inherited NOS2 deficiency was clinically silent in this patient until lethal infection with CMV. Moreover, NOS2 appeared to be redundant for control of other pathogens in this patient. (Funded by the National Center for Advancing Translational Sciences and others.).
PMCID:7063989
PMID: 31995689
ISSN: 1533-4406
CID: 5065032

Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-β

Li, Juan; Ritelli, Marco; Ma, Cindy S; Rao, Geetha; Habib, Tanwir; Corvilain, Emilie; Bougarn, Salim; Cypowyj, Sophie; Grodecká, Lucie; Lévy, Romain; Béziat, Vivien; Shang, Lei; Payne, Kathryn; Avery, Danielle T; Migaud, Mélanie; Boucherit, Soraya; Boughorbel, Sabri; Guennoun, Andrea; Chrabieh, Maya; Rapaport, Franck; Bigio, Benedetta; Itan, Yuval; Boisson, Bertrand; Cormier-Daire, Valérie; Syx, Delfien; Malfait, Fransiska; Zoppi, Nicoletta; Abel, Laurent; Freiberger, Tomáš; Dietz, Harry C; Marr, Nico; Tangye, Stuart G; Colombi, Marina; Casanova, Jean-Laurent; Puel, Anne
Genetic etiologies of chronic mucocutaneous candidiasis (CMC) disrupt human IL-17A/F-dependent immunity at mucosal surfaces, whereas those of connective tissue disorders (CTDs) often impair the TGF-β-dependent homeostasis of connective tissues. The signaling pathways involved are incompletely understood. We report a three-generation family with an autosomal dominant (AD) combination of CMC and a previously undescribed form of CTD that clinically overlaps with Ehlers-Danlos syndrome (EDS). The patients are heterozygous for a private splice-site variant of MAPK8, the gene encoding c-Jun N-terminal kinase 1 (JNK1), a component of the MAPK signaling pathway. This variant is loss-of-expression and loss-of-function in the patients' fibroblasts, which display AD JNK1 deficiency by haploinsufficiency. These cells have impaired, but not abolished, responses to IL-17A and IL-17F. Moreover, the development of the patients' TH17 cells was impaired ex vivo and in vitro, probably due to the involvement of JNK1 in the TGF-β-responsive pathway and further accounting for the patients' CMC. Consistently, the patients' fibroblasts displayed impaired JNK1- and c-Jun/ATF-2-dependent induction of key extracellular matrix (ECM) components and regulators, but not of EDS-causing gene products, in response to TGF-β. Furthermore, they displayed a transcriptional pattern in response to TGF-β different from that of fibroblasts from patients with Loeys-Dietz syndrome caused by mutations of TGFBR2 or SMAD3, further accounting for the patients' complex and unusual CTD phenotype. This experiment of nature indicates that the integrity of the human JNK1-dependent MAPK signaling pathway is essential for IL-17A- and IL-17F-dependent mucocutaneous immunity to Candida and for the TGF-β-dependent homeostasis of connective tissues.
PMID: 31784499
ISSN: 2470-9468
CID: 5065022

Severe influenza pneumonitis in children with inherited TLR3 deficiency [Case Report]

Lim, Hye Kyung; Huang, Sarah X L; Chen, Jie; Kerner, Gaspard; Gilliaux, Olivier; Bastard, Paul; Dobbs, Kerry; Hernandez, Nicholas; Goudin, Nicolas; Hasek, Mary L; García Reino, Eduardo Javier; Lafaille, Fabien G; Lorenzo, Lazaro; Luthra, Priya; Kochetkov, Tatiana; Bigio, Benedetta; Boucherit, Soraya; Rozenberg, Flore; Vedrinne, Catherine; Keller, Michael D; Itan, Yuval; García-Sastre, Adolfo; Celard, Marie; Orange, Jordan S; Ciancanelli, Michael J; Meyts, Isabelle; Zhang, Qian; Abel, Laurent; Notarangelo, Luigi D; Snoeck, Hans-Willem; Casanova, Jean-Laurent; Zhang, Shen-Ying
Autosomal recessive IRF7 and IRF9 deficiencies impair type I and III IFN immunity and underlie severe influenza pneumonitis. We report three unrelated children with influenza A virus (IAV) infection manifesting as acute respiratory distress syndrome (IAV-ARDS), heterozygous for rare TLR3 variants (P554S in two patients and P680L in the third) causing autosomal dominant (AD) TLR3 deficiency. AD TLR3 deficiency can underlie herpes simplex virus-1 (HSV-1) encephalitis (HSE) by impairing cortical neuron-intrinsic type I IFN immunity to HSV-1. TLR3-mutated leukocytes produce normal levels of IFNs in response to IAV. In contrast, TLR3-mutated fibroblasts produce lower levels of IFN-β and -λ, and display enhanced viral susceptibility, upon IAV infection. Moreover, the patients' iPSC-derived pulmonary epithelial cells (PECs) are susceptible to IAV. Treatment with IFN-α2b or IFN-λ1 rescues this phenotype. AD TLR3 deficiency may thus underlie IAV-ARDS by impairing TLR3-dependent, type I and/or III IFN-mediated, PEC-intrinsic immunity. Its clinical penetrance is incomplete for both IAV-ARDS and HSE, consistent with their typically sporadic nature.
PMCID:6719423
PMID: 31217193
ISSN: 1540-9538
CID: 5065012