scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Tuberculosis and impaired IL-23–dependent IFN-γ immunity in humans homozygous for a common TYK2 missense variant

Stéphanie Boisson-Dupuis1, Stéphanie Boisson-Dupuis2, Stéphanie Boisson-Dupuis3, Noé Ramírez-Alejo1, Zhi Li2, Zhi Li4, Etienne Patin5, Etienne Patin4, Geetha Rao6, Gaspard Kerner3, Gaspard Kerner2, Che Kang Lim7, Che Kang Lim8, Dimitry N. Krementsov9, Nicholas Hernandez1, Cindy S. Ma10, Cindy S. Ma6, Qian Zhang11, Qian Zhang1, Janet Markle1, Rubén Martínez-Barricarte1, Kathryn Payne6, Robert Fisch1, Caroline Deswarte2, Caroline Deswarte3, Joshua Halpern1, Matthieu Bouaziz3, Matthieu Bouaziz2, Jeanette Mulwa1, Durga Sivanesan12, Durga Sivanesan13, Tomi Lazarov14, Rodrigo Naves15, Patricia García16, Yuval Itan1, Yuval Itan17, Bertrand Boisson2, Bertrand Boisson1, Bertrand Boisson3, Alix Checchi3, Alix Checchi2, Fabienne Jabot-Hanin2, Fabienne Jabot-Hanin3, Aurélie Cobat3, Aurélie Cobat2, Andrea Guennoun11, Carolyn C. Jackson14, Carolyn C. Jackson1, Sevgi Pekcan, Zafer Caliskaner, Jaime Inostroza18, Beatriz Tavares Costa-Carvalho19, Jose Antonio Tavares de Albuquerque20, Humberto García-Ortiz, Lorena Orozco, Tayfun Ozcelik21, Ahmed Abid, Ismail Abderahmani Rhorfi22, Hicham Souhi, Hicham Naji Amrani, Adil Zegmout, Frederic Geissmann14, Stephen W. Michnick13, Ingrid Müller-Fleckenstein22, Bernhard Fleckenstein22, Anne Puel1, Anne Puel2, Anne Puel3, Michael J. Ciancanelli1, Nico Marr11, Hassan Abolhassani8, Hassan Abolhassani23, María Elvira Balcells16, Antonio Condino-Neto20, Alexis Strickler24, Katia Abarca16, Cory Teuscher9, Hans D. Ochs25, Ismail Reisli, Esra Hazar Sayar, Jamila El-Baghdadi, Jacinta Bustamante, Lennart Hammarström26, Lennart Hammarström8, Lennart Hammarström7, Stuart G. Tangye6, Stuart G. Tangye10, Sandra Pellegrini4, Sandra Pellegrini2, Lluis Quintana-Murci5, Lluis Quintana-Murci4, Laurent Abel1, Laurent Abel2, Laurent Abel3, Jean-Laurent Casanova 
21 Dec 2018-Science immunology (American Association for the Advancement of Science)-Vol. 3, Iss: 30
TL;DR: Homozygosity for the catalytically inactive P1104A missense variant of the TYK2 Janus kinase selectively disrupts the induction of IFN-γ by IL-23 and is a common monogenic etiology of tuberculosis.
Abstract: Inherited IL-12Rβ1 and TYK2 deficiencies impair both IL-12- and IL-23-dependent IFN-γ immunity and are rare monogenic causes of tuberculosis, each found in less than 1/600,000 individuals. We show that homozygosity for the common TYK2 P1104A allele, which is found in about 1/600 Europeans and between 1/1000 and 1/10,000 individuals in regions other than East Asia, is more frequent in a cohort of patients with tuberculosis from endemic areas than in ethnicity-adjusted controls (P = 8.37 × 10-8; odds ratio, 89.31; 95% CI, 14.7 to 1725). Moreover, the frequency of P1104A in Europeans has decreased, from about 9% to 4.2%, over the past 4000 years, consistent with purging of this variant by endemic tuberculosis. Surprisingly, we also show that TYK2 P1104A impairs cellular responses to IL-23, but not to IFN-α, IL-10, or even IL-12, which, like IL-23, induces IFN-γ via activation of TYK2 and JAK2. Moreover, TYK2 P1104A is properly docked on cytokine receptors and can be phosphorylated by the proximal JAK, but lacks catalytic activity. Last, we show that the catalytic activity of TYK2 is essential for IL-23, but not IL-12, responses in cells expressing wild-type JAK2. In contrast, the catalytic activity of JAK2 is redundant for both IL-12 and IL-23 responses, because the catalytically inactive P1057A JAK2, which is also docked and phosphorylated, rescues signaling in cells expressing wild-type TYK2. In conclusion, homozygosity for the catalytically inactive P1104A missense variant of TYK2 selectively disrupts the induction of IFN-γ by IL-23 and is a common monogenic etiology of tuberculosis.

Summary (3 min read)

INTRODUCTION

  • About a quarter of the world’s population is infected with Mycobacterium tuberculosis, but this bacterium causes tuberculosis in less than 10% of infected individuals, generally within 2 years of infection (a situation referred to here as primary tuberculosis) (1–3).
  • These results suggest that homozygosity for P1104A, which is still present in about 1/600 Europeans and between 1/10,000 and 1/1000 individuals in other regions of the world, with the exception of East Asia, where the allele is almost absent, has been a major human genetic determinant of primary tuberculosis during the course of human history.
  • Like patients with complete TYK2 and IL-12R1 deficiencies, P1104A homozygotes did not respond to IL-23, in terms of IFN- production, as shown by comparison with healthy controls.

Whole blood ***

  • TYK2 P1104A patients thus displayed impaired IL-23–mediated IFN- immunity.
  • The authors then studied the capacity of naïve CD4+.
  • These cells were unable to produce IL-17A/ IL-17F, consistent with the impairment of IL-23 signaling, as observed in IL-12R1– and TYK2–deficient patients (Fig. 6D).
  • T cells from P1104A homozygotes were able to differentiate into IFN-–producing TH1 cells in an IL-12– dependent manner (TAE beads and IL-12), like control cells and cells from IL-23R–deficient patients, but unlike cells from TYK2-, IL-12R1–, and IL-12R2–deficient patients (Fig. 6D and companion paper).

DISCUSSION

  • In conclusion, homozygosity for TYK2 P1104A confers a predisposition to severe mycobacterial diseases, including MSMD and, more frequently, primary tuberculosis.
  • Several genome-wide association studies have shown that homozygosity for TYK2 P1104A has a strong protective effect (ORs ranging from 0.1 to 0.3) against various autoinflammatory or autoimmune conditions (41).
  • The gradual decline of the P1104A allele in the European continent, which requires further investigation using more ancient DNA samples from different geographic locations and epochs, suggests that tuberculosis has been continuously endemic from the Neolithic until the middle of the 20th century (56).
  • Genetic testing before travels into endemic areas may, however, be warranted.
  • This observation has important clinical implications, because injections of recombinant IFN- would probably be beneficial in these patients, as it is in patients with IL-12R1 deficiency (12, 19, 88).

MATERIALS AND METHODS Study design

  • The authors studied the contributions of two common TYK2 missense variants, I684S and P1104A, to predisposition to mycobacterial diseases.
  • The authors screened their WES database including 463 patients with MSMD, 454 with tuberculosis, and 2835 with non-mycobacterial infections used as controls, as well as the WES data of the 2504 from the 1000 Genomes Project (a total of 5339 controls).
  • The authors tested the association of the two TYK2 variants with MSMD and tuberculosis using a logistic regression model including the first three principal components of the PCA to account for the ethnic heterogeneity of the cohorts.
  • The authors analyzed the occurrence of negative selection acting on the two TYK2 variants by testing whether their frequency in Europeans has decreased more than other variants that were in the same frequency range 4000 years ago.
  • The authors also tested the patient’s primary leukocytes.

Ethics statement

  • This study was conducted in accordance with the Helsinki Declaration, with written informed consent obtained from the patients’ families.
  • Approval for this study was obtained from the French Ethics Committee “Comité de Protection des Personnes,” The French National Agency for Medicine and Health Product Safety (ANSM) and the Institut National de la Santé et de la Recherche Médicale in France, and the Rockefeller University Institutional Review Board (IRB), New York, USA.

Whole-blood activation experiments

  • Venous blood samples from controls and patients were collected into heparin-containing tubes and processed according to a modified version of the protocol described by Feinberg et al. (89).
  • ELISA was then performed on the collected supernatants, with the human IFN- ELISA Kit (Ready-SET-Go! from eBioscience or PeliPair from Sanquin), in accordance with the manufacturer’s instructions.
  • The plasmids were used to transfect Phoenix-A packing cells to generate retroviral particles carrying each allelic variant.
  • Positively transfected cells were selected with puromycin at a concentration of 2 g/ml until all the cells were positive for the surface expression of NGFR, as assessed by fluorescence-activated cell sorting with Alexa Fluor 647 anti-NGFR staining (BD Pharmingen).

Cell culture and stimulation

  • EBV-B cells were cultured in RPMI supplemented with 10% fetal bovine serum (FBS) .
  • The cells were then starved for 2 hours by incubation in serum-free RPMI.
  • A dose-response experiment was performed on EBV-B cells, with different concentrations of rhIL-23 ranging from Boisson-Dupuis et al., Sci. Immunol.

Western blotting

  • Protein extracts were separated by SDS–polyacrylamide gel electrophoresis, and the resulting bands were electroblotted onto polyvinylidene difluoride membranes.
  • Last, the blots were washed with washing buffer, and antibody binding was detected with the SuperSignal West Femto System (Thermo Fisher Scientific).
  • S3D, immunoblots were revealed using enhanced chemiluminescence detection reagent (Western Lightning, PerkinElmer) and signals were acquired with Fuji ImageQuant LAS 4000.
  • The level of phosphorylated band in control cells was set as 100.
  • Statistical analysis was performed considering technical replicates for the transduced cells with the TYK2 alleles, and technical and biological replicates for EBV-B and HVS-T cell lines derived from controls and patients.

Population genetic analysis

  • The authors investigated the occurrence of strong negative selection acting on the candidate TYK2 missense variants over the last few hundreds of generations, by comparing the current allele frequency of these variants in Europeans with that estimated from the low-coverage sequenced genomes of 22 individuals from Late Stone Age (LSA) Central Europe (52).
  • Only variants that were still segregating in the current European population (CEU, TSI, or FIN from the 1000 Genomes Project) were considered in this analysis.
  • The authors checked that these empirical observations were not biased due to sampling or sequencing errors, by performing 100,000 forward simulations under the Wright-Fisher neutral model (R code available upon request).
  • The lower quantiles of the simulated and observed distributions were largely similar (Fig. 1E), suggesting that their analyses were unbiased.

WES and RNA-seq analyses

  • WES was performed as previously described (47).
  • Gene expression levels were estimated with TPM (transcripts per kilobase million).
  • Gene expression profiles are expressed as the fold change in expression between the values obtained before and after stimulation.

Variant enrichment analysis

  • The authors performed an enrichment analysis of TYK2 variants in their two cohorts of 463 MSMD patients and 454 tuberculosis patients.
  • To account for the ethnic heterogeneity of the cohorts, the first three principal components of the PCA were systematically included in the logistic regression model, as previously described (48).
  • Table S1. Summary of the TYK2 genotypes among the different cohorts of patients and healthy individuals.
  • Raw data used to generate dot plots and bar graphs.

REFERENCES AND NOTES

  • The global burden of latent tuberculosis infection: A re-estimation using mathematical modelling.
  • Two rare disease-associated Tyk2 variants are catalytically impaired but signaling competent.
  • The Laboratory of Human Genetics of Infectious Diseases was supported, in part, by grants from the French National Agency for Research (ANR) under the “Investissement d’avenir” program (grant no. ANR-10-IAHU-01), the TBPATHGEN project (grant no, also known as Funding.
  • The Yale Center for Mendelian Genomics (UM1HG006504) is funded by the National Human Genome Research Institute.

Did you find this useful? Give us your feedback

Figures (7)

Content maybe subject to copyright    Report

Boisson-Dupuis et al., Sci. Immunol. 3, eaau8714 (2018) 21 December 2018
SCIENCE IMMUNOLOGY
|
RESEARCH ARTICLE
1 of 19
TUBERCULOSIS
Tuberculosis and impaired IL-23–dependent IFN-
immunity in humans homozygous for a common TYK2
missense variant
Stéphanie Boisson-Dupuis
1,2,3
*
, Noe Ramirez-Alejo
1†
, Zhi Li
4,5‡
, Etienne Patin
6,7,8‡
, Geetha Rao
9‡
,
Gaspard Kerner
2,3‡
, Che Kang Lim
10,11
, Dimitry N. Krementsov
12
, Nicholas Hernandez
1
, Cindy S. Ma
9,13
,
Qian Zhang
1,14
, Janet Markle
1
, Ruben Martinez-Barricarte
1
, Kathryn Payne
9
, Robert Fisch
1
,
Caroline Deswarte
2,3
, Joshua Halpern
1
, Matthieu Bouaziz
2,3
, Jeanette Mulwa
1
, Durga Sivanesan
15,16
,
Tomi Lazarov
17
, Rodrigo Naves
18
, Patricia Garcia
19
, Yuval Itan
1,20,21
, Bertrand Boisson
1,2,3
, Alix Checchi
2,3
,
Fabienne Jabot-Hanin
2,3
, Aurélie Cobat
2,3
, Andrea Guennoun
14
, Carolyn C. Jackson
1,22
,
Sevgi Pekcan
23
, Zafer Caliskaner
24
, Jaime Inostroza
25
, Beatriz Tavares Costa-Carvalho
26
,
Jose Antonio Tavares de Albuquerque
27
, Humberto Garcia-Ortiz
28
, Lorena Orozco
28
,
Tayfun Ozcelik
29
, Ahmed Abid
30
, Ismail Abderahmani Rhorfi
30,31
, Hicham Souhi
30
,
Hicham Naji Amrani
30
, Adil Zegmout
30
, Frédéric Geissmann
17
, Stephen W. Michnick
15
,
Ingrid Muller-Fleckenstein
31
, Bernhard Fleckenstein
31
, Anne Puel
1,2,3
, Michael J. Ciancanelli
1
,
Nico Marr
14
, Hassan Abolhassani
10,32
, María Elvira Balcells
33
, Antonio Condino-Neto
27
,
Alexis Strickler
34
, Katia Abarca
35
, Cory Teuscher
36
, Hans D. Ochs
37
, Ismail Reisli
38
, Esra H. Sayar
38
,
Jamila El-Baghdadi
39
, Jacinta Bustamante
1,2,3,40§
, Lennart Hammarström
10,11,41§
, Stuart G. Tangye
9,13§
,
Sandra Pellegrini
4,5§
, Lluis Quintana-Murci
6,7,
, Laurent Abel
1,2,3||
, Jean-Laurent Casanova
1,2,3,42,43
*
||
Inherited IL-12R1 and TYK2 deficiencies impair both IL-12– and IL-23–dependent IFN- immunity and are rare
monogenic causes of tuberculosis, each found in less than 1/600,000 individuals. We show that homozygosity for
the common TYK2 P1104A allele, which is found in about 1/600 Europeans and between 1/1000 and 1/10,000 indi-
viduals in regions other than East Asia, is more frequent in a cohort of patients with tuberculosis from endemic
areas than in ethnicity-adjusted controls (P = 8.37 × 10
−8
; odds ratio, 89.31; 95% CI, 14.7 to 1725). Moreover, the
frequency of P1104A in Europeans has decreased, from about 9% to 4.2%, over the past 4000 years, consistent
with purging of this variant by endemic tuberculosis. Surprisingly, we also show that TYK2 P1104A impairs cellular
responses to IL-23, but not to IFN-, IL-10, or even IL-12, which, like IL-23, induces IFN- via activation of TYK2 and
JAK2. Moreover, TYK2 P1104A is properly docked on cytokine receptors and can be phosphorylated by the proximal
JAK, but lacks catalytic activity. Last, we show that the catalytic activity of TYK2 is essential for IL-23, but not IL-12,
responses in cells expressing wild-type JAK2. In contrast, the catalytic activity of JAK2 is redundant for both IL-12
and IL-23 responses, because the catalytically inactive P1057A JAK2, which is also docked and phosphorylated,
rescues signaling in cells expressing wild-type TYK2. In conclusion, homozygosity for the catalytically inactive
P1104A missense variant of TYK2 selectively disrupts the induction of IFN- by IL-23 and is a common monogenic
etiology of tuberculosis.
INTRODUCTION
About a quarter of the world’s population is infected with Mycobacterium
tuberculosis, but this bacterium causes tuberculosis in less than 10% of
infected individuals, generally within 2 years of infection (a situa-
tion referred to here as primary tuberculosis) (1–3). In the countries
in which tuberculosis is highly endemic, primary tuberculosis is
particularly common in children, who often develop life-threatening
disease (4–6). Clinical and epidemiological studies have long suggested
that tuberculosis in humans has a strong genetic basis (7–9). Auto-
somal recessive (AR) complete interleukin-12 receptor 1 (IL-12R1)
and tyrosine kinase 2 (TYK2) deficiencies are the only two inborn
errors of immunity reported to date to underlie primary tuberculosis
in otherwise healthy patients in two or more kindreds (10–17). Cells
from patients with IL-12R1 deficiency do not respond to IL-12 or
IL-23 (12, 18–24). These patients are susceptible to weakly virulent
mycobacteria, such as the Bacille Calmette-Guérin (BCG) vaccine and
environmental species [Mendelian susceptibility to mycobacterial
disease (MSMD)], to the more virulent species M. tuberculosis, and
more rarely to Candida albicans (20, 25). They are prone to MSMD
and tuberculosis because they produce too little interferon- (IFN-)
(7, 12, 26, 27) and, in some cases, to chronic mucocutaneous candi-
diasis (CMC) because they produce too little IL-17A/F (28–32).
In patients with TYK2 deficiency, cellular responses to IL-12 and
IL-23 are severely impaired, but not abolished (10, 3335). These
patients are, thus, also prone to MSMD and tuberculosis, although
probably with a lower penetrance than for IL-12R1 deficiency, be-
cause they display residual responses to IL-12 and IL-23. They do
not seem to be susceptible to C. albicans, which may merely reflect
the lower penetrance of candidiasis and smaller number of patients,
when compared with IL-12R1 deficiency. However, unlike patients
with IL-12R1 deficiency, they are susceptible to viral diseases due
to the impairment of their responses to IFN-
/ (10, 36). In vitro,
their cells respond poorly to IL-10, but this defect, which is not
observed in patients with IL-12R1 deficiency, is clinically silent
(10, 37, 38). Both IL-12R1 and TYK2 deficiencies are caused by
rare or private alleles, accounting for each deficiency being found in
Copyright © 2018
The
Authors, some
rights reserved;
exclusive licensee
American Association
for the Advancement
of Science. No claim
to original U.S.
Government Works

Boisson-Dupuis et al., Sci. Immunol. 3, eaau8714 (2018) 21 December 2018
SCIENCE IMMUNOLOGY
|
RESEARCH ARTICLE
2 of 19
no more than 1/600,000 individuals worldwide. Here, we tested the
hypothesis that two common and catalytically inactive missense
TYK2 variants, P1104A and I684S (39), might underlie MSMD,
tuberculosis, or both.
RESULTS
Ten homozygotes for TYK2 P1104A suffered from
mycobacterial diseases
The common TYK2 variants P1104A (rs34536443) and I684S
(rs12720356) are both catalytically impaired, as shown by in vitro
kinase assays in reconstituted TYK2-deficient fibrosarcoma cells
(U1A cells) (39). Other studies with selective small-molecule kinase
inhibitors suggested that the catalytic activity of TYK2 was required
for T cell responses to IL-12 and IL-23, but not IFN- and IL-10
(40). Consistently, the P1104A variant has been reported to im-
pair cellular responses to both IL-12 and IL-23 in human memory
T cells, whereas discordant results were obtained for IFN- (39, 41).
The response to IL-10 was normal in human leukocytes (41). On
the basis of the gnomAD database (42) (gnomAD: http://gnomad.
broadinstitute.org), these two missense variants are rare (<0.02%)
in East Asian populations, but otherwise common (>0.8%) in the
other four main gnomAD populations, reaching their highest fre-
quencies in Europeans (4.2% for P1104A and 9% for I684S) (fig. S1,
A and B) (43, 44). On the basis of the 1000 Genomes Project data-
base (45), these two variants are not in linkage disequilibrium.
We investigated the possibility that these variants might confer a
predisposition to MSMD, tuberculosis, or both. We screened our
whole- exome sequencing (WES) data for 463 patients with MSMD
and 291 children with tuberculosis, from different geographic loca-
tions and ancestries, and for 163 adults of North African ancestry
with early-onset pulmonary tuberculosis (table S1). None of these
patients carried pathogenic mutations in known MSMD- and
tuberculosis-
causing genes (12, 46). Our WES data for 2835 other
patients, from various ethnic origins (fig. S1C) and with various
genetically unexplained non-mycobacterial infections, were used as
a control. Among the 3752 exomes available in total, we identified
366 I684S heterozygotes, 168 P1104A heterozygotes, 18 I684S
homozygotes, and 6 I684S/P1104A compound heterozygotes, with
no clustering of any of these genotypes within any of the patient
cohorts (table S1). By contrast, we identified 11 unrelated P1104A
homozygotes, which were confirmed by Sanger sequencing: 7 with
tuberculosis (3 children under the age of 15 years and 4 adults
under the age of 40 years), 3 with MSMD (all under 3 years of age),
and 1 with CMC (aged 1 year) (Fig.1, A to C; fig. S1D; and Supple-
mentary Materials and Methods). We further Sanger sequenced
TYK2 in parents and siblings of these 11 patients. We found that, in
kindred K with the CMC patient, homozygosity for P1104A did
not segregate with CMC, because one sibling with CMC was het-
erozygous for P1104A, implying that there is another genetic cause
for CMC in this kindred (fig. S1D). We also found only one as-
ymptomatic P1104A homozygote among the relatives of the other
10 patients (kindred G, I.1). In total, we identified 10 unrelated P1104A
TYK2 homozygotes with MSMD (3 patients) or primary tuberculosis
(7 patients).
P1104A homozygosity is strongly enriched in patients
with tuberculosis
Principal components analysis (PCA) based on the WES data
(fig. S1C) (47) confirmed the diverse ancestries of the 10 patients.
Eight were living in their countries of origin (Fig.1C and fig. S1B).
The Mexican patient was living in the United States, and the 10th
patient, who was living in Brazil, had mixed European and African
ancestry. We compared the proportions of individuals with P1104A
in each cohort and estimated odds ratios (ORs) by logistic regres-
sion, with adjustment for the first three principal components of the
PCA to account for ethnic heterogeneity (48). In addition to the
2835 exomes already used as controls, we used all 2504 available
1
St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
2
Laboratory of Human Genetics of Infectious
Diseases, Necker Branch, INSERM U1163, Paris, France.
3
Paris Descartes University, Imagine Institute, Paris, France.
4
Cytokine Signaling Unit, Pasteur Institute, Paris, France.
5
INSERM U1221, Paris, France.
6
Human Evolutionary Genetics Unit, Pasteur Institute, Paris, France.
7
CNRS UMR2000, Paris, France.
8
Center of Bioinformatics, Biostatistics
and Integrative Biology, Pasteur Institute, Paris, France.
9
Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
10
Division
of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.
11
Department of Clinical
Translational Research, Singapore General Hospital, Singapore, Singapore.
12
Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, USA.
13
St. Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales, Australia.
14
Sidra Medicine, Doha, Qatar.
15
Department of Biochemistry,
University of Montreal, Montreal, Quebec, Canada.
16
Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
17
Im-
munology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
18
Institute of Biochemical Sciences, Faculty of Medicine, Uni-
versity of Chile, Santiago, Chile.
19
Laboratory of Microbiology, Clinical Laboratory Department School of Medicine, Pontifical Catholic University of Chile, Santiago, Chile.
20
The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
21
Department of Genetics and Genomic Sciences,
Icahn School of Medicine at Mount Sinai, New York, NY, USA.
22
Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
23
Department of
Pediatric Pulmonology, Necmettin Erbakan University, Meram Medical Faculty, Konya, Turkey.
24
Meram Faculty of Medicine, Department of Internal Medicine, Division of
Allergy and Immunology, Necmettin Erbakan University, Konya, Turkey.
25
Jeffrey Modell Center for Diagnosis and Research in Primary Immunodeficiencies, Faculty of
Medicine University of La Frontera, Temuco, Chile.
26
Department of Pediatrics, Federal University of São Paulo Medical School, São Paulo, Brazil.
27
Department of Immu-
nology, Institute of Biomedical Sciences, and Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil.
28
National Institute of Genomic Medicine, Mexico
City, Mexico.
29
Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.
30
Department of Pneumology, Military Hospital Mohammed V, Rabat,
Morocco.
31
Institute of Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany.
32
Research Center for Immunodeficiencies, Pediatrics
Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
33
Department of Infectious Diseases, Medical School, Pontifical Catholic
University of Chile, Santiago, Chile.
34
Department of Pediatrics, San Sebastián University, Santiago, Chile.
35
Department of Infectious Diseases and Pediatric Immunology,
School of Medicine, Pontifical Catholic University of Chile, Santiago, Chile.
36
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT,
USA.
37
Seattle Children's Research Institute and Department of Pediatrics, University of Washington, Seattle, WA, USA.
38
Department of Pediatric Immunology and
Allergy, Necmettin Erbakan University, Meram Medical Faculty, Konya, Turkey.
39
Genetics Unit, Military Hospital Mohamed V, Hay Riad, Rabat, Morocco.
40
Center for the
Study of Primary Immunodeficiencies, AP-HP, Necker Hospital for Sick Children, Paris, France.
41
Beijing Genomics Institute BGI-Shenzhen, Shenzhen, China.
42
Pediatric
Hematology-Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France.
43
Howard Hughes Medical Institute, New York, NY, USA.
*Corresponding author. Email: stbo603@rockefeller.edu (S.B.-D.); jean-laurent.casanova@rockefeller.edu (J.-L.C.)
†These authors contributed equally to this work.
‡These authors contributed equally to this work.
§These authors contributed equally to this work.
||These authors contributed equally to this work.

Boisson-Dupuis et al., Sci. Immunol. 3, eaau8714 (2018) 21 December 2018
SCIENCE IMMUNOLOGY
|
RESEARCH ARTICLE
3 of 19
Kindred A Kindred D
P2
1 2
12
Kindred E
m/m
1
WT/m
2
1
WT/m
Kindred I
m/m
P1
1
E?
2
1
E?
Kindred F
m/m
P4
1
E?
2
1
E?
Kindred G
m/m
P5
1
WT/m
2
12
m/m
Kindred C
E? m/m
P7
1
WT/m
2
12
Kindred H
E? m/m
P8
E?
E? E?
1
E?
2
1
E?
m/m
P3
WT/WT
WT/m WT/m
1
E?
2
1
E?
m/m
P6
Kindred B
3 4
1
E?
2
1
E?
m/m
P9
WT/m
3
FERM
SH2 Pseudokinase
Kinase
NH2 COOH
C70HfsX21
L767X
T1106HfsX4
E154XS50HfsX1 R638X
P1104A
P216HfsX14
I684S
I
II
3
I
II
MSM
D
TB
MSMD
TB
A
B
C
D
E
WT/m
451584 8891187
28
1567 8751173
0.1 0.2 0.3 0.4 0.50.0 0.1 0.2 0.3 0.4 0.50.0
0.1 0.2 0.3 0.4 0.50.0
0 500 1000 1500
0 500 1000 1500
0 500 1000 1500
I684S TYK2 M694V MEFV C282Y HFE
Controls
(n = 5339)
Homoz. Homoz. OR Homoz. OR Homoz. OR
carriers carriers (95% CI) carriers (95%CI)carriers(95% CI)
89.3123.53 53.72
(14.7–1725) (2.9–483)(10.1–993)
0.46 0.76 0.61
(0.03–2.2)(0.12–2.6)(0.15–1.8)
10 4.87 × 10
−8
I684S221 0.38 20.713 0.4
Variant P value P value P value
P1104A 17 8.37 × 10
−8
33.27 × 10
−3
Tuberculosis
(n = 454)
MSMD
(n = 463)
TB + MSMD
(n = 917)
Current frequency in western Europe
0
0.1 0.2 0.3 0.4 0.5
500 1000 1500 2000
0.0
Number of variants
P1104A TYK2
Kindred J
1
E?
2
1
E?
m/m
P10
CountryMAF TB incidence* BCG Age of onset of
Patients Disease
of residence
Origin by PCA
gnomAD#(/100,000) vaccination symptoms (years)
P1 BCG osteomyelitis Sweden European 0.0429.2 Yes1
P2 MAC osteomyelitis USA American/Mexican 0.0123.2 No 1
P3 BCG disseminated IranMiddle Eastern0.031 15 Yes2
P4 PulmonaryBrazilMixed European/African 0.01841Yes 6
P5 PulmonaryAlgeria North African 0.01875Yes 40
P6 Pulmonary Morocco North African 0.018107 Yes27
P7 Miliary Turkey Turkish0.021 18 Yes15
P8 Pulmonary Chile American/Chilean 0.01216Yes 13
P9 Pulmonary Morocco North African 0.018107 Yes35
P10Pulmonary Chile American/Chilean 0.01216Yes 33
#: Allele frequency in the country of origin in the gnomAD database
*TB incidence in the country of residence from WHO 2015
%
Homoz@
0.41
1.58
3.27
2.85
1.48
4.16
5.53
0.6
0.82
0.27
@: Percentage of homozygosity
TYK2
Fig. 1. Familial segregation and
clinical information for patients
homozygous for TYK2 P1104A.
(A) Schematic diagram of the TYK2
protein with its various domains
(FERM, SH2, pseudokinase, and
tyrosine kinase). The positions of
the previously reported TYK2 mu-
tations resulting in premature STOP
codons are indicated in red. The
positions of the I684S and P1104A
polymorphisms are indicated in blue
and green, respectively. (B) Pedi-
grees of the 10 TYK2-deficient
families. Each generation is desig-
nated by a Roman numeral (I–II),
and each individual by an Arabic
numeral. The double lines connect-
ing the parents indicate consan-
guinity based on interview and/
or a homozygosity rate of >4%
estimated from the exome data.
Solid shapes indicate disease status.
Individuals whose genetic status
could not be determined are in-
dicated by “E?”, and “m” indicates
a TYK2 P1104A allele. (C) Sum-
mary table of clinical details and
origin of the patients associated
with the MAF in the country of ori-
gin. The incidence of tuberculosis
(TB) in the country of residence is
also mentioned. MAC indicates
Mycobacterium avium complex.
(D) Summary of WES, indicating
the numbers of individuals with
tuberculosis or MSMD and of con-
trols carrying the I684S or P1104A
variant of TYK2 in the homozygous
state, and the associated P value
and OR. (E) Distributions of the cur-
rent allele frequencies of variants
that segregated 4000 years ago at
frequencies similar to those of the
P1104A and I684S TYK2, M694V
MEFV, and C282Y HFE variants.
The red vertical lines indicate the
current frequency of the four
variants of interest. Colored bars
indicate the distribution of cur-
rent allele frequency, in the 1000
Genomes Project, for variants with
frequencies in ancient European
human DNA similar to those of
the four candidate variants (52).
Black lines indicate the distribu-
tion of simulated frequencies, in
the present generation, for alleles
with a past frequency similar to
that of the four candidate variants,
with propagation over 160 gen-
erations (corresponding to a pe-
riod of ~4000 years) under the Wright-Fisher neutral model. For instance, for the P1104A allele, which had a frequency of ~9% in ancient Europeans, colored bars
indicate the observed distribution of current frequencies for the 31,276 variants with a frequency of 8 to 10% 4000 years ago. The black lines indicate the distribution
of frequencies for 100,000 simulated alleles obtained after 160 generations under the Wright-Fisher neutral model.

Boisson-Dupuis et al., Sci. Immunol. 3, eaau8714 (2018) 21 December 2018
SCIENCE IMMUNOLOGY
|
RESEARCH ARTICLE
4 of 19
individuals from the 1000 Genomes Project (45), giving a total of
5339 controls for whom we have complete WES data (Fig.1D).
P1104A homozygosity was more enriched among patients with
MSMD than among controls [P = 3.27 × 10
−3
; OR, 23.53; 95%
confidence interval (CI), 2.9 to 483], and an even higher level of
enrichment was observed among patients with tuberculosis (P =
8.37 × 10
−8
; OR, 89.31; 95% CI, 14.7 to 1725). The level of enrich-
ment in homozygosity for this variant was intermediate but more
significant when both groups were analyzed together (OR, 53.72;
95% CI, 10.1 to 993; P = 4.87 × 10
−8
). By contrast, no enrichment
in homozygosity for this variant was observed among the patients
with other infections studied in the laboratory (table S1) (49, 50).
Aside from the 10 MSMD and tuberculosis patients, we identi-
fied only one other P1104A homozygote by WES: a CMC patient
whose P1104A homozygosity was not CMC-causing, living in the
United States, where infants are not inoculated with BCG and
M. tuberculosis is not endemic (fig. S1D). No homozygotes were
observed among the 2504 individuals of the 1000 Genomes Project.
No significant enrichment in P1104A heterozygosity was observed
in any of the cohorts studied, including patients with MSMD (P =
0.57) or tuberculosis (P = 0.49), demonstrating the recessive na-
ture of P1104A inheritance for both mycobacterial conditions.
Moreover, no significant enrichment in I684S heterozygotes or
homozygotes or in P1104A/I684S compound heterozygotes was
observed in any of the cohorts studied (table S1). Last, the TYK2
P1104A allele yielded the highest OR at genome-wide level in an
independent enrichment analysis performed under the assump-
tion of a recessive mode of inheritance and considering all com-
mon missense or potential loss-of-function (LOF) alleles in our
entire cohort of 3752 patients (fig. S1E). These results strongly sug-
gest that homozygosity for P1104A is a genetic etiology of primary
tuberculosis and MSMD.
TYK2 P1104A allele frequency has decreased in Europe over
the past 4000 years
The higher risk of life-threatening tuberculosis in P1104A homo-
zygotes suggests that this variant has been subject to negative
selection in areas in which this disease has long been endemic, such
as Europe (51). We analyzed changes in the frequencies of the
P1104A and I684S TYK2 variants in the European population,
from ancient to modern times (52). Only three nonsynonymous
TYK2 variants—P1104A, I684S, and V362F—were found in an
available sample of central European individuals who lived during
the late Neolithic age ~4000 years ago (52). Over this period, the
frequency of TYK2 P1104A has significantly decreased in Europeans,
from about 9% to 4.2% (Fig.1E). Of the 31,276 variants with fre-
quencies in the 8 to 10% range 4000 years ago, P1104A is among
the 5% displaying the largest decrease in frequency (empirical P =
0.048; Fig.1E). Furthermore, the neutral model of evolution was
significantly rejected for P1104A in Wright-Fisher simulations
(simulation P = 0.050; Fig.1E and Supplementary Materials and
Methods), suggesting an absence of bias in the empirical analyses.
As a negative control, the frequency of V362F remained stable
(from 25% to 26.2%) and that of I684S did not decrease signifi-
cantly over this period (empirical P = 0.181). The frequency of
I684S was about 14% 4000 years ago and is now 9%, placing this
variant among the 80% of the 36,469 polymorphisms considered
with a frequency that was in the 13 to 15% range 4000 years ago and
has remained relatively stable.
TYK2 P1104A allele was possibly purged in Europe
by tuberculosis
We subsequently analyzed, as positive controls, two relatively com-
mon mutations known to cause life-threatening AR disorders and
present in ancient Europeans: the MEFV M694V variant underly-
ing Mediterranean fever (MF) (53) and the HFE C282Y underlying
hemochromatosis (which also decreases male fertility) (54). Both
these variants decreased significantly in frequency over the same
period, from about 11% to 0.4% for MEFV M694V and from 16% to
5.7% for HFE C282Y (empirical P = 0.016 for both variants; Fig.1E).
Therefore, our preliminary assessments suggest that TYK2 P1104A,
MEFV M694V, and HFE C282Y have been subject to negative
selection in Europeans, whereas TYK2 I684S has not. The stronger
selection operating on MEFV M694V, and to a lesser extent HFE
C282Y, than on TYK2 P1104A is consistent with the inevitability of
MF and hemochromatosis in patients with these mutations, whereas
tuberculosis development also requires exposure to M. tuberculosis.
These results suggest that, unlike I684S, P1104A has been undergoing
a purge in Europe since the Neolithic period due to the continued
endemic nature of life-threatening tuberculosis (51). No other in-
tramacrophagic infection, whose control depends on IFN-, has
been endemic for so long in Europe (55, 56). The purging of delete-
rious mutations is expected to be much less effective in the absence
of continued exposure (57, 58), which has been the case for other
infections that killed a sizeable proportion of Europeans, albeit for
no more than several decades or a few centuries, such as plague
(59). The observed decline in P1104A allele frequency is consistent
with the purging of a recessive trait that kills in childhood or when
the individual is of reproductive age. This decrease would be much
steeper for a dominant trait with a similar fitness effect. These re-
sults suggest that homozygosity for P1104A, which is still present in
about 1/600 Europeans and between 1/10,000 and 1/1000 individuals
in other regions of the world, with the exception of East Asia, where the
allele is almost absent, has been a major human genetic determinant
of primary tuberculosis during the course of human history.
TYK2 P1104A impairs IL-23 but not IFN-, IL-12,
and IL-10 signaling
We performed a functional characterization of the I684S and
P1104A TYK2 alleles, focusing on the four known human TYK2-
dependent signaling pathways (10). In reconstituted U1A cells
stimulated with IFN- in vitro, both mutant proteins were previ-
ously shown to be catalytically inactive, i.e., unable to autophos-
phorylate or phosphorylate a substrate such as signal transducer
and activator of transcription 3 (STAT3) (39). However, both could
be phosphorylated by Janus kinase 1 (JAK1), unlike the prototypical
kinase-dead adenosine 5 triphosphate (ATP)–binding mutant K930R
(39). Epstein-Barr virus (EBV)–transformed B (EBV-B) cells and her-
pesvirus saimiri (HVS)–transformed T (HVS-T) cells derived from
a TYK2-deficient patient without TYK2 protein expression (10) were
stably transduced with a retrovirus generated with an empty vector
or a vector containing the wild-type (WT), P1104A, I684S, or
K930R TYK2 complementary DNA (cDNA) (60). Transduction
with the WT or any mutant TYK2 restored both TYK2 expres-
sion, as shown by Western blotting, and the corresponding TYK2
scaffolding-dependent surface expression of IFN-R1, IL-10R2,
and IL-12R1, as shown by flow cytometry (Fig.2, A and B, and
fig. S2, A and B). In P1104A-expressing cells, the IFN-– and IL-12–
dependent signaling pathways were normal, as shown by the levels

Boisson-Dupuis et al., Sci. Immunol. 3, eaau8714 (2018) 21 December 2018
SCIENCE IMMUNOLOGY
|
RESEARCH ARTICLE
5 of 19
AB
CD
E
TYK2
GAPDH
EV
EV
I684S
P1104A
WT
K930R
I684S
P1004A
WT
K930R
TYK2
−/−
EBV-B cells TYK2
−/−
HVS-T cells
EV
WT
P1104A
I684S
K930R
0
500
1000
1500
2000
2500
MFI
***
EV
WT
P1104A
I684S
K930R
0
1000
2000
3000
4000
***
EV
WT
P1104A
I684S
K930R
0
2000
4000
6000
TYK2
−/−
EBV-B cells TYK2
−/−
HVS-T cells
EV
I684S
P1104A
WT
K930R
+−+−+−+
−+
IL-23
pTYK2
pJAK2
TYK2
JAK2
GAPDH
TYK2
−/−
EBV-B cells
EV
I684S
P1104A
WT
K930R
+−+−+−+−+
pTYK2
pJAK1
TYK2
JAK1
GAPDH
TYK2
−/−
EBV-B cells
EV
I684S
P1104A
WT
K930R
+−+−+−+−+
IL-12
pTYK2
pJAK2
TYK2
JAK2
GAPDH
TYK2
−/−
HVS-T cells
pSTAT1
STAT1
TYK2
GAPDH
pSTAT4
STAT4
TYK2
G
F
pSTAT3
150
150
38
150
150
150
38
150
150
38
150
150
150
150
38
150
150
MW
MW MW
MW
102
102
38
150
102
102
76
76
150
TYK2
Tubulin
52
pSTAT1
STAT1
STAT3
TYK2
−/−
HVS-T cells
102
102
150
MFI pSTAT4
0
200
400
600
800
1000
−−+ + +
EV
WT
P1104
A
I684
S
K930R
IL-12
+−− + +
+
+
+
+
**
***
ns
**
***
***
***
ns
ns
ns
102
STAT1
102
pSTAT1
GAPDH
38
EV
WT
P1101A
I681S
E779K
0.00
0.05
0.10
0.15
0.20
Mouse TYK2
−/−
MEF cells and VSV
Human TYK2
−/−
U1A cells and VSV
*
ns
*
*
*
ns
ns
EV
WT
P1104A
I684S
0.00
0.02
0.04
0.06
0.08
0.10
***
Fig. 2. Cellular responses to IFN-, IL-12 and IL-23 in transduced EBV-B and HVS-T cells. TYK2-deficient EBV-B and HVS-T cells were transduced with a retrovirus gener-
ated with an empty vector (EV), or vectors encoding WT TYK2, or the P1104A, I684S, or K930R TYK2 alleles. (A) Levels of TYK2 in transduced EBV-B (left) and HVS-T (right)
cells, as determined by Western blotting. (B) Levels of IL-12R1 and IFN-R1 in transduced EBV-B (left) and HVS-T (right) cells, as determined by flow cytometry. ***P <
0.001, two-tailed Student’s t test. Error bars indicate SEM. (C, D, and F) Phosphorylation of JAKs and STATs in unstimulated (−) transduced EBV-B or HVS-T cells or in these cells
after stimulation (+) with IFN- (C) (pTYK2, pJAK1, and pSTAT1), IL-12 (D) (pTYK2, pJAK2, pSTAT1, and pSTAT4), and IL-23 (F) (pTYK2, pJAK2, pSTAT3, and pSTAT1), as assessed
by Western blotting with specific antibodies recognizing phospho-TYK2, phospho-JAK1, phospho-JAK2, phospho-STAT1, phospho-STAT4, and phospho-STAT3. MW,
molecular weight. (E) Phosphorylation of STAT4 in response to IFN- and IL-12, as determined by flow cytometry in HVS-transduced T cells and expression as mean fluorescence
intensity (MFI). **P < 0.01, ***P < 0.001, two-tailed Student’s t test. ns, not significant. (G) IFN- response of U1A (left) and MEF (right) cells, both lacking TYK2, after transduc-
tion with the indicated human and mouse TYK2 alleles, respectively, or with empty vector control, as measured in an IFN-–induced antiviral activity assay (see Materials
and Methods). A unique dose is shown: an IFN- dose of 0.01 ng/ml for human cells and 1 IU/ml for mouse cells.

Citations
More filters
Journal ArticleDOI
Qian Zhang1, Paul Bastard2, Paul Bastard3, Zhiyong Liu1  +169 moreInstitutions (34)
23 Oct 2020-Science
TL;DR: The COVID Human Genetic Effort established to test the general hypothesis that life-threatening COVID-19 in some or most patients may be caused by monogenic inborn errors of immunity to SARS-CoV-2 with incomplete or complete penetrance finds an enrichment in variants predicted to be loss-of-function (pLOF), with a minor allele frequency <0.001.
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.

1,659 citations

Journal ArticleDOI
16 Apr 2019-Immunity
TL;DR: Significant knowledge gaps are presented, including how similar signals from these cytokines can mediate distinct outcomes, and how a better understanding of the biology of the IL-12 family provides new therapeutic opportunities.

254 citations

Journal ArticleDOI
TL;DR: These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-γ–dependent immunity to mycobacteria, both individually and much more so cooperatively.
Abstract: Hundreds of patients with autosomal recessive, complete IL-12p40 or IL-12Rβ1 deficiency have been diagnosed over the last 20 years. They typically suffer from invasive mycobacteriosis and, occasionally, from mucocutaneous candidiasis. Susceptibility to these infections is thought to be due to impairments of IL-12-dependent IFN-γ immunity and IL-23-dependent IL-17A/IL-17F immunity, respectively. We report here patients with autosomal recessive, complete IL-12Rβ2 or IL-23R deficiency, lacking responses to IL-12 or IL-23 only, all of whom, unexpectedly, display mycobacteriosis without candidiasis. We show that αβ T, γδ T, B, NK, ILC1, and ILC2 cells from healthy donors preferentially produce IFN-γ in response to IL-12, whereas NKT cells and MAIT cells preferentially produce IFN-γ in response to IL-23. We also show that the development of IFN-γ-producing CD4+ T cells, including, in particular, mycobacterium-specific TH1* cells (CD45RA-CCR6+), is dependent on both IL-12 and IL-23. Last, we show that IL12RB1, IL12RB2, and IL23R have similar frequencies of deleterious variants in the general population. The comparative rarity of symptomatic patients with IL-12Rβ2 or IL-23R deficiency, relative to IL-12Rβ1 deficiency, is, therefore, due to lower clinical penetrance. There are fewer symptomatic IL-23R- and IL-12Rβ2-deficient than IL-12Rβ1-deficient patients, not because these genetic disorders are rarer, but because the isolated absence of IL-12 or IL-23 is, in part, compensated by the other cytokine for the production of IFN-γ, thereby providing some protection against mycobacteria. These experiments of nature show that human IL-12 and IL-23 are both required for optimal IFN-γ-dependent immunity to mycobacteria, both individually and much more so cooperatively.

139 citations

Journal ArticleDOI
TL;DR: BMS-986165 is a potent oral agent that similarly blocks receptor-stimulated activation of TYK2 allosterically and with high selectivity and potency afforded through optimized binding to a regulatory domain of the protein.
Abstract: TYK2 is a nonreceptor tyrosine kinase involved in adaptive and innate immune responses. A deactivating coding variant has previously been shown to prevent receptor-stimulated activation of this kinase and provides high protection from several common autoimmune diseases but without immunodeficiency. An agent that recapitulates the phenotype of this deactivating coding variant may therefore represent an important advancement in the treatment of autoimmunity. BMS-986165 is a potent oral agent that similarly blocks receptor-stimulated activation of TYK2 allosterically and with high selectivity and potency afforded through optimized binding to a regulatory domain of the protein. Signaling and functional responses in human TH17, TH1, B cells, and myeloid cells integral to autoimmunity were blocked by BMS-986165, both in vitro and in vivo in a phase 1 clinical trial. BMS-986165 demonstrated robust efficacy, consistent with blockade of multiple autoimmune pathways, in murine models of lupus nephritis and inflammatory bowel disease, supporting its therapeutic potential for multiple immune-mediated diseases.

122 citations

Journal ArticleDOI
TL;DR: This Review summarizes recent advances in the molecular, cellular, and clinical characterization of human inborn errors of immunity and challenges a number of immunological notions initially established in inbred mice.
Abstract: Molecular, cellular, and clinical studies of human inborn errors of immunity have revolutionized our understanding of their pathogenesis, considerably broadened their spectrum of immunological and clinical phenotypes, and enabled successful targeted therapeutic interventions. These studies have also been of great scientific merit, challenging a number of immunological notions initially established in inbred mice while revealing previously unrecognized mechanisms of host defense by leukocytes and other cells and of both innate and adaptive tolerance to self.

121 citations

References
More filters
Journal Article
TL;DR: Copyright (©) 1999–2012 R Foundation for Statistical Computing; permission is granted to make and distribute verbatim copies of this manual provided the copyright notice and permission notice are preserved on all copies.
Abstract: Copyright (©) 1999–2012 R Foundation for Statistical Computing. Permission is granted to make and distribute verbatim copies of this manual provided the copyright notice and this permission notice are preserved on all copies. Permission is granted to copy and distribute modified versions of this manual under the conditions for verbatim copying, provided that the entire resulting derived work is distributed under the terms of a permission notice identical to this one. Permission is granted to copy and distribute translations of this manual into another language, under the above conditions for modified versions, except that this permission notice may be stated in a translation approved by the R Core Team.

272,030 citations

Journal ArticleDOI
TL;DR: The Spliced Transcripts Alignment to a Reference (STAR) software based on a previously undescribed RNA-seq alignment algorithm that uses sequential maximum mappable seed search in uncompressed suffix arrays followed by seed clustering and stitching procedure outperforms other aligners by a factor of >50 in mapping speed.
Abstract: Motivation Accurate alignment of high-throughput RNA-seq data is a challenging and yet unsolved problem because of the non-contiguous transcript structure, relatively short read lengths and constantly increasing throughput of the sequencing technologies. Currently available RNA-seq aligners suffer from high mapping error rates, low mapping speed, read length limitation and mapping biases. Results To align our large (>80 billon reads) ENCODE Transcriptome RNA-seq dataset, we developed the Spliced Transcripts Alignment to a Reference (STAR) software based on a previously undescribed RNA-seq alignment algorithm that uses sequential maximum mappable seed search in uncompressed suffix arrays followed by seed clustering and stitching procedure. STAR outperforms other aligners by a factor of >50 in mapping speed, aligning to the human genome 550 million 2 × 76 bp paired-end reads per hour on a modest 12-core server, while at the same time improving alignment sensitivity and precision. In addition to unbiased de novo detection of canonical junctions, STAR can discover non-canonical splices and chimeric (fusion) transcripts, and is also capable of mapping full-length RNA sequences. Using Roche 454 sequencing of reverse transcription polymerase chain reaction amplicons, we experimentally validated 1960 novel intergenic splice junctions with an 80-90% success rate, corroborating the high precision of the STAR mapping strategy. Availability and implementation STAR is implemented as a standalone C++ code. STAR is free open source software distributed under GPLv3 license and can be downloaded from http://code.google.com/p/rna-star/.

30,684 citations

Journal ArticleDOI
TL;DR: This work introduces PLINK, an open-source C/C++ WGAS tool set, and describes the five main domains of function: data management, summary statistics, population stratification, association analysis, and identity-by-descent estimation, which focuses on the estimation and use of identity- by-state and identity/descent information in the context of population-based whole-genome studies.
Abstract: Whole-genome association studies (WGAS) bring new computational, as well as analytic, challenges to researchers. Many existing genetic-analysis tools are not designed to handle such large data sets in a convenient manner and do not necessarily exploit the new opportunities that whole-genome data bring. To address these issues, we developed PLINK, an open-source C/C++ WGAS tool set. With PLINK, large data sets comprising hundreds of thousands of markers genotyped for thousands of individuals can be rapidly manipulated and analyzed in their entirety. As well as providing tools to make the basic analytic steps computationally efficient, PLINK also supports some novel approaches to whole-genome data that take advantage of whole-genome coverage. We introduce PLINK and describe the five main domains of function: data management, summary statistics, population stratification, association analysis, and identity-by-descent estimation. In particular, we focus on the estimation and use of identity-by-state and identity-by-descent information in the context of population-based whole-genome studies. This information can be used to detect and correct for population stratification and to identify extended chromosomal segments that are shared identical by descent between very distantly related individuals. Analysis of the patterns of segmental sharing has the potential to map disease loci that contain multiple rare variants in a population-based linkage analysis.

26,280 citations

Journal ArticleDOI
TL;DR: This work presents HTSeq, a Python library to facilitate the rapid development of custom scripts for high-throughput sequencing data analysis, and presents htseq-count, a tool developed with HTSequ that preprocesses RNA-Seq data for differential expression analysis by counting the overlap of reads with genes.
Abstract: Motivation: A large choice of tools exists for many standard tasks in the analysis of high-throughput sequencing (HTS) data. However, once a project deviates from standard workflows, custom scripts are needed. Results: We present HTSeq, a Python library to facilitate the rapid development of such scripts. HTSeq offers parsers for many common data formats in HTS projects, as well as classes to represent data, such as genomic coordinates, sequences, sequencing reads, alignments, gene model information and variant calls, and provides data structures that allow for querying via genomic coordinates. We also present htseq-count, a tool developed with HTSeq that preprocesses RNA-Seq data for differential expression analysis by counting the overlap of reads with genes. Availability and implementation: HTSeq is released as an opensource software under the GNU General Public Licence and available from http://www-huber.embl.de/HTSeq or from the Python Package Index at https://pypi.python.org/pypi/HTSeq. Contact: sanders@fs.tum.de

15,744 citations

Journal ArticleDOI
Adam Auton1, Gonçalo R. Abecasis2, David Altshuler3, Richard Durbin4  +514 moreInstitutions (90)
01 Oct 2015-Nature
TL;DR: The 1000 Genomes Project set out to provide a comprehensive description of common human genetic variation by applying whole-genome sequencing to a diverse set of individuals from multiple populations, and has reconstructed the genomes of 2,504 individuals from 26 populations using a combination of low-coverage whole-generation sequencing, deep exome sequencing, and dense microarray genotyping.
Abstract: The 1000 Genomes Project set out to provide a comprehensive description of common human genetic variation by applying whole-genome sequencing to a diverse set of individuals from multiple populations. Here we report completion of the project, having reconstructed the genomes of 2,504 individuals from 26 populations using a combination of low-coverage whole-genome sequencing, deep exome sequencing, and dense microarray genotyping. We characterized a broad spectrum of genetic variation, in total over 88 million variants (84.7 million single nucleotide polymorphisms (SNPs), 3.6 million short insertions/deletions (indels), and 60,000 structural variants), all phased onto high-quality haplotypes. This resource includes >99% of SNP variants with a frequency of >1% for a variety of ancestries. We describe the distribution of genetic variation across the global sample, and discuss the implications for common disease studies.

12,661 citations

Related Papers (5)
Frequently Asked Questions (11)
Q1. What are the contributions in "Tuberculosis and impaired il-23–dependent ifn- immunity in humans homozygous for a common tyk2 missense variant" ?

For example, Boisson-Dupuis et al. this paper showed that the IL-12R1 deficiency in primary tuberculosis patients can be linked to weakly virulent mycobacteria, such as the Bacille Calmette-Guérin vaccine and environmental species. 

T cells of P1104A homozygotes have impaired IFN- production, due to their very weak response to IL-23, accounting for the susceptibility to MSMD or primary tuberculosis. 

the TYK2-dependent response to IL-23 that is disrupted by P1104A is essential for antimycobacterial IFN- immunity, but seems to be redundant for anti-fungal IL-17 immunity, given the absence of Candida infection in the patients described here. 

The impact of TYK2 variants on cellular responses to IL-12 and IL-23 is irrelevant in nonhematopoietic cells, because the receptors for these cytokines are expressed only on leukocytes. 

Several genome-wide association studies have shown that homozygosity for TYK2 P1104A has a strong protective effect (ORs ranging from 0.1 to 0.3) against various autoinflammatory or autoimmune conditions (41). 

Funding: The Laboratory of Human Genetics of Infectious Diseases was supported, in part, by grants from the French National Agency for Research (ANR) under the “Investissement d’avenir” program (grant no. ANR-10-IAHU-01), the TBPATHGEN project (grant no. 

The stronger selection operating on MEFV M694V, and to a lesser extent HFE C282Y, than on TYK2 P1104A is consistent with the inevitability of MF and hemochromatosis in patients with these mutations, whereas tuberculosis development also requires exposure to M. tuberculosis. 

The authors analyzed the occurrence of negative selection acting on the two TYK2 variants by testing whether their frequency in Europeans has decreased more than other variants that were in the same frequency range 4000 years ago. 

Between 1/10,000 and 1/1000 individuals are homozygous in endemic regions of the world (other than East Asia), where P1104A TYK2 is likely to define a strictly recessive but relatively common etiology of severe primary tuberculosis (about 0.5% of cases). 

In P1104A homozygous cells, the response to IFN- was modestly reduced in terms of JAK1, TYK2, STAT3, and STAT1 phosphorylation (Fig. 3C and fig. 

P1104A homozygosity was more enriched among patients with MSMD than among controls [P = 3.27 × 10−3; OR, 23.53; 95% confidence interval (CI), 2.9 to 483], and an even higher level of enrichment was observed among patients with tuberculosis (P = 8.37 × 10−8; OR, 89.31; 95% CI, 14.7 to 1725).