T cell dynamics and response of the microbiota after gene therapy to treat X-linked severe combined immunodeficiency

Erik L Clarke, A Jesse Connell, Emmanuelle Six, Nadia A Kadry, Arwa A Abbas, Young Hwang, John K Everett, Casey E Hofstaedter, Rebecca Marsh, Myriam Armant, Judith Kelsen, Luigi D Notarangelo, Ronald G Collman, Salima Hacein-Bey-Abina, Donald B Kohn, Marina Cavazzana, Alain Fischer, David A Williams, Sung-Yun Pai, Frederic D Bushman, Erik L Clarke, A Jesse Connell, Emmanuelle Six, Nadia A Kadry, Arwa A Abbas, Young Hwang, John K Everett, Casey E Hofstaedter, Rebecca Marsh, Myriam Armant, Judith Kelsen, Luigi D Notarangelo, Ronald G Collman, Salima Hacein-Bey-Abina, Donald B Kohn, Marina Cavazzana, Alain Fischer, David A Williams, Sung-Yun Pai, Frederic D Bushman

Abstract

Background: Mutation of the IL2RG gene results in a form of severe combined immune deficiency (SCID-X1), which has been treated successfully with hematopoietic stem cell gene therapy. SCID-X1 gene therapy results in reconstitution of the previously lacking T cell compartment, allowing analysis of the roles of T cell immunity in humans by comparing before and after gene correction.

Methods: Here we interrogate T cell reconstitution using four forms of high throughput analysis. (1) Estimation of the numbers of transduced progenitor cells by monitoring unique positions of integration of the therapeutic gene transfer vector. (2) Estimation of T cell population structure by sequencing of the recombined T cell receptor (TCR) beta locus. (3) Metagenomic analysis of microbial populations in oropharyngeal, nasopharyngeal, and gut samples. (4) Metagenomic analysis of viral populations in gut samples.

Results: Comparison of progenitor and mature T cell populations allowed estimation of a minimum number of cell divisions needed to generate the observed populations. Analysis of microbial populations showed the effects of immune reconstitution, including normalization of gut microbiota and clearance of viral infections. Metagenomic analysis revealed enrichment of genes for antibiotic resistance in gene-corrected subjects relative to healthy controls, likely a result of higher healthcare exposure.

Conclusions: This multi-omic approach enables the characterization of multiple effects of SCID-X1 gene therapy, including T cell repertoire reconstitution, estimation of numbers of cell divisions between progenitors and daughter T cells, normalization of the microbiome, clearance of microbial pathogens, and modulations in antibiotic resistance gene levels. Together, these results quantify several aspects of the long-term efficacy of gene therapy for SCID-X1. This study includes data from ClinicalTrials.gov numbers NCT01410019, NCT01175239, and NCT01129544.

Conflict of interest statement

Ethics approval and consent to participate

Written informed consent for all subjects described in this study was obtained as previously described [19, 33] and under IRB 12-010072. For the previous studies, local institutional review boards at the participating sites (Paris, Boston, Cincinnati, and Los Angeles) approved enrollment and study protocols. All human subject research was performed in accordance with the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Sampling schedule for 14 SCID-X1 gene-corrected subjects studied for integration site distributions and TCRB CDR3 sequence composition. a Times of sample acquisition after cell infusion. b Population sizes of inferred progenitor cells deduced from marking with unique sites of vector integration. Integration site data is in Additional file 1: Table S2, and summaries of genes near sites of vector integration are in Additional file 2: Figure S1. The x-axis shows the time since cell infusion. The y-axis shows the population size reconstructed using Chao1 from the numbers of unique integration sites and replicate sampling. Samples are named for the site of gene correction (U indicates USA, F indicates France; the next digit indicates the trial 1 = SCIDn1, 2 = SCIDn2, and the next two digits indicate the patient number within that trial). Patients F107 and F110 suffered severe adverse events at months 68 and 33, respectively. Both recovered by the final timepoints listed
Fig. 2
Fig. 2
Analysis of TCRB CDR3 sequences. a The unique numbers of rearranged genes detected are shown. The colors indicate in frame rearrangements (blue), frameshifts (tan), and stop codons (red). b Richness and evenness of the inferred TCRB CDR3 populations. Patients are color coded as indicated on the right, and all replicates for each patient timepoint displayed. The ranges of healthy adults and children are shown by the light and dark gray diamonds, respectively. c Clustering of the samples sequenced using Bray-Curtis similarity and t-SNE. The association of patients with samples is shown by the key at the right. d V gene usage. The patient of origin is marked at the top of each panel. The V genes shown were the top most commonly found V genes in healthy children, ordered by prevalence; not all V genes used by each patient are shown. All panels show the same x-axis of V genes. Chi-squared test used to assess differences between gene distribution in patients compared to healthy children; red label indicates significant difference from healthy (p < 0.05). e J gene usage, with J genes determined by the same manner as in D. f Heat map summarizing the frequencies of utilization of the most common V and J pairs. Subjects studied are marked at the top. Time of sampling is shown on the right
Fig. 3
Fig. 3
Minimum numbers of cell divisions between progenitors and daughter T cells. The x-axis shows time after corrected cell infusion. The y-axis shows the estimated number of cell divisions calculated as described in the text. The subjects studied are indicated beneath the figure as indicated by the color code. Circles indicate subjects from the second trial, and squares indicate subjects from the first. Stars next to the points indicate PBMCs were sequenced rather than sorted CDR3+ cells. Replicates were not available for U205b and so this subject was not analyzed
Fig. 4
Fig. 4
Longitudinal analysis of the microbiome during SCID-X1 gene correction. a Timing of sample acquisition. b Longitudinal analysis of the nasopharyngeal microbiome. Each column indicates a sample. Samples are grouped by subject as indicated at the top. Each row summarizes the proportions of a specific microbial taxa inferred using Kraken. Abundance is color coded as indicated to the right and reflects the number of reads assigned to that taxa as a proportion of all non-human reads. c As in B, but oropharyngeal samples. d As in B, but stool samples
Fig. 5
Fig. 5
Microbiome community analysis. a Sample clustering using Bray-Curtis dissimilarity. Different sample types are marked by the colors, healthy versus SCID are shown by the shapes. b Comparison of stool samples for each patient queried to healthy controls. Samples were clustered using Bray-Curtis dissimilarity. Each panel compares one SCID subject samples (indicated at the top) to healthy control samples (shown in gray). Elapsed time is shown using the color code (bottom). c The longitudinal species richness of each patient’s oral, nasal, and stool samples is shown in separate panels. The 95% CI of the healthy child richness for that sample type is shown in tan. d Representation of selected antibiotic resistance genes in the stool samples studied. Each column indicates a metagenomic data set from the subject listed at the top. Each row summarizes the abundance of an antibiotic resistance gene class. The tiles are colored by reads per kilobase of target per million sequence reads (RPKM); the color code is to the right of the panel
Fig. 6
Fig. 6
Virome analysis. a Heat map summarizing RNA viruses detected. Each column indicates a sample from the patient indicated at the top of the heatmap, each row indicates a type of virus. The tiles are colored according to abundance. b Heat map summarizing DNA viruses detected. Markings as in A

References

    1. Aiuti A, Slavin S, Aker M, Ficara F, Deola S, Mortellaro A, Morecki S, Andolfi G, Tabucchi A, Carlucci F, et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002;296(5577):2410–2413. doi: 10.1126/science.1070104.
    1. Aiuti A, Cassani B, Andolfi G, Mirolo M, Biasco L, Recchia A, Urbinati F, Valacca C, Scaramuzza S, Aker M, et al. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J Clin Invest. 2007;117(8):2233–2240. doi: 10.1172/JCI31666.
    1. Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, Dionisio F, Calabria A, Giannelli S, Castiello MC, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 2013;341(6148):1233151. doi: 10.1126/science.1233151.
    1. Biffi A, Montini E, Lorioli L, Cesani M, Fumagalli F, Plati T, Baldoli C, Martino S, Calabria A, Canale S, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013;341(6148):1233158. doi: 10.1126/science.1233158.
    1. Cavazzana-Calvo M, Andre-Schmutz I, Fischer A. Haematopoietic stem cell transplantation for SCID patients: where do we stand? Br J Haematol. 2013;160(2):146–152. doi: 10.1111/bjh.12119.
    1. Hacein-Bey-Abina S, Le Deist F, Carlier F, Bouneaud C, Hue C, De Villartay JP, Thrasher AJ, Wulffraat N, Sorensen R, Dupuis-Girod S, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med. 2002;346(16):1185–1193. doi: 10.1056/NEJMoa012616.
    1. Gaspar HB, Parsley KL, Howe S, King D, Gilmour KC, Sinclair J, Brouns G, Schmidt M, Von Kalle C, Barington T, et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet. 2004;364(9452):2181–2187. doi: 10.1016/S0140-6736(04)17590-9.
    1. Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, Martinache C, Rieux-Laucat F, Latour S, Belohradsky BH, et al. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2010;363(4):355–364. doi: 10.1056/NEJMoa1000164.
    1. Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Adams S, Howe SJ, Al Ghonaium A, Bayford J, Brown L, Davies EG, et al. Long-term persistence of a polyclonal T cell repertoire after gene therapy for X-linked severe combined immunodeficiency. Sci Transl Med. 2011;3(97):97ra79.
    1. Noguchi M, Yi H, Rosenblatt HM, Filipovich AH, Adelstein S, Modi WS, McBride OW, Leonard WJ. Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. Cell. 1993;73(1):147–157. doi: 10.1016/0092-8674(93)90167-O.
    1. Puck JM, Deschênes SM, Porter JC, Dutra AS, Brown CJ, Willard HF, Henthorn PS. The interleukin-2 receptor gamma chain maps to Xq13.1 and is mutated in X-linked severe combined immunodeficiency, SCIDX1. Hum Mol Genet. 1993;2(8):1099–1104. doi: 10.1093/hmg/2.8.1099.
    1. Kovanen PE, Leonard WJ. Cytokines and immunodeficiency diseases: critical roles of the gamma(c)-dependent cytokines interleukins 2, 4, 7, 9, 15, and 21, and their signaling pathways. Immunol Rev. 2004;202:67–83. doi: 10.1111/j.0105-2896.2004.00203.x.
    1. Puel A, Ziegler SF, Buckley RH, Leonard WJ. Defective IL7R expression in T(−)B(+)NK(+) severe combined immunodeficiency. Nat Genet. 1998;20(4):394–397. doi: 10.1038/3877.
    1. Kennedy MK, Glaccum M, Brown SN, Butz EA, Viney JL, Embers M, Matsuki N, Charrier K, Sedger L, Willis CR, et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J Exp Med. 2000;191(5):771–780. doi: 10.1084/jem.191.5.771.
    1. Lodolce JP, Boone DL, Chai S, Swain RE, Dassopoulos T, Trettin S, Ma A. IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation. Immunity. 1998;9(5):669–676. doi: 10.1016/S1074-7613(00)80664-0.
    1. Hacein-Bey-Abina S, von Kalle C, Schmidt M, Le Deist F, Wulffraat N, McIntyre E, Radford I, Villeval JL, Fraser CC, Cavazzana-Calvo M, et al. A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2003;348(3):255–256. doi: 10.1056/NEJM200301163480314.
    1. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, Lim A, Osborne CS, Pawliuk R, Morillon E, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302(5644):415–419. doi: 10.1126/science.1088547.
    1. Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, Brugman MH, Pike-Overzet K, Chatters SJ, de Ridder D, et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest. 2008;118(9):3143–3150. doi: 10.1172/JCI35798.
    1. Hacein-Bey-Abina S, Pai SY, Gaspar HB, Armant M, Berry CC, Blanche S, Bleesing J, Blondeau J, de Boer H, Buckland KF, et al. A modified gamma-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med. 2014;371(15):1407–1417. doi: 10.1056/NEJMoa1404588.
    1. Berry C, Hannenhalli S, Leipzig J, Bushman FD. Selection of target sites for mobile DNA integration in the human genome. PLoS Comput Biol. 2006;2(11):e157. doi: 10.1371/journal.pcbi.0020157.
    1. Berry CC, Gillet NA, Melamed A, Gormley N, Bangham CR, Bushman FD. Estimating abundances of retroviral insertion sites from DNA fragment length data. Bioinformatics. 2012;28(6):755–762. doi: 10.1093/bioinformatics/bts004.
    1. Schroder ARW, Shinn P, Chen HM, Berry C, Ecker JR, Bushman F. HIV-1 integration in the human genome favors active genes and local hotspots. Cell. 2002;110(4):521–529. doi: 10.1016/S0092-8674(02)00864-4.
    1. Mitchell RS, Beitzel BF, Schroder AR, Shinn P, Chen H, Berry CC, Ecker JR, Bushman FD. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2004;2(8):E234. doi: 10.1371/journal.pbio.0020234.
    1. Wang GP, Ciuffi A, Leipzig J, Berry CC, Bushman FD. HIV integration site selection: analysis by massively parallel pyrosequencing reveals association with epigenetic modifications. Genome Res. 2007;17(8):1186–1194. doi: 10.1101/gr.6286907.
    1. Wang GP, Berry CC, Malani N, Leboulch P, Fischer A, Hacein-Bey-Abina S, Cavazzana-Calvo M, Bushman FD. Dynamics of gene-modified progenitor cells analyzed by tracking retroviral integration sites in a human SCID-X1 gene therapy trial. Blood. 2010;115(22):4356–4366. doi: 10.1182/blood-2009-12-257352.
    1. Sherman E, Nobles C, Berry CC, Six E, Wu Y, Dryga A, Malani N, Male F, Reddy S, Bailey A, et al. INSPIIRED: a pipeline for quantitative analysis of sites of new DNA integration in cellular genomes. Mol Ther Methods Clin Dev. 2017;4:39–49. doi: 10.1016/j.omtm.2016.11.002.
    1. Berry CC, Nobles C, Six E, Wu Y, Malani N, Sherman E, Dryga A, Everett JK, Male F, Bailey A, et al. INSPIIRED: quantification and visualization tools for analyzing integration site distributions. Mol Ther Methods Clin Dev. 2017;4:17–26. doi: 10.1016/j.omtm.2016.11.003.
    1. Boyd SD, Marshall EL, Merker JD, Maniar JM, Zhang LN, Sahaf B, Jones CD, Simen BB, Hanczaruk B, Nguyen KD, et al. Measurement and clinical monitoring of human lymphocyte clonality by massively parallel VDJ pyrosequencing. Sci Transl Med. 2009;1(12):12ra23. doi: 10.1126/scitranslmed.3000540.
    1. Weinstein JA, Jiang N, White RA, 3rd, Fisher DS, Quake SR. High-throughput sequencing of the zebrafish antibody repertoire. Science. 2009;324(5928):807–810. doi: 10.1126/science.1170020.
    1. Robins HS, Srivastava SK, Campregher PV, Turtle CJ, Andriesen J, Riddell SR, Carlson CS, Warren EH. Overlap and effective size of the human CD8+ T cell receptor repertoire. Sci Transl Med. 2010;2(47):47ra64. doi: 10.1126/scitranslmed.3001442.
    1. Campregher PV, Srivastava SK, Deeg HJ, Robins HS, Warren EH. Abnormalities of the alphabeta T-cell receptor repertoire in advanced myelodysplastic syndrome. Exp Hematol. 2010;38(3):202–212. doi: 10.1016/j.exphem.2009.12.004.
    1. Robins HS, Campregher PV, Srivastava SK, Wacher A, Turtle CJ, Kahsai O, Riddell SR, Warren EH, Carlson CS. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood. 2009;114(19):4099–4107. doi: 10.1182/blood-2009-04-217604.
    1. Cavazzana-Calvo M, Hacein-Bey S, de Saint BG, Gross F, Yvon E, Nusbaum P, Selz F, Hue C, Certain S, Casanova JL, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science. 2000;288(5466):669–672002E. doi: 10.1126/science.288.5466.669.
    1. Hacein-Bey Abina S, Gaspar HB, Blondeau J, Caccavelli L, Charrier S, Buckland K, Picard C, Six E, Himoudi N, Gilmour K, et al. Outcomes following gene therapy in patients with severe Wiskott-Aldrich syndrome. JAMA. 2015;313(15):1550–1563. doi: 10.1001/jama.2015.3253.
    1. Robins H, Desmarais C, Matthis J, Livingston R, Andriesen J, Reijonen H, Carlson C, Nepom G, Yee C, Cerosaletti K. Ultra-sensitive detection of rare T cell clones. J Immunol Methods. 2012;375(1–2):14–19. doi: 10.1016/j.jim.2011.09.001.
    1. Minot S, Bryson A, Chehoud C, Wu GD, Lewis JD, Bushman FD. Rapid evolution of the human gut virome. Proc Natl Acad Sci U S A. 2013;110(30):12450–12455. doi: 10.1073/pnas.1300833110.
    1. Clarke Erik L., Lauder Abigail P., Hofstaedter Casey E., Hwang Young, Fitzgerald Ayannah S., Imai Ize, Biernat Wojciech, Rękawiecki Bartłomiej, Majewska Hanna, Dubaniewicz Anna, Litzky Leslie A., Feldman Michael D., Bittinger Kyle, Rossman Milton D., Patterson Karen C., Bushman Frederic D., Collman Ronald G. Microbial Lineages in Sarcoidosis. A Metagenomic Analysis Tailored for Low–Microbial Content Samples. American Journal of Respiratory and Critical Care Medicine. 2018;197(2):225–234. doi: 10.1164/rccm.201705-0891OC.
    1. Abbas A. A., Diamond J. M., Chehoud C., Chang B., Kotzin J. J., Young J. C., Imai I., Haas A. R., Cantu E., Lederer D. J., Meyer K. C., Milewski R. K., Olthoff K. M., Shaked A., Christie J. D., Bushman F. D., Collman R. G. The Perioperative Lung Transplant Virome: Torque Teno Viruses Are Elevated in Donor Lungs and Show Divergent Dynamics in Primary Graft Dysfunction. American Journal of Transplantation. 2016;17(5):1313–1324. doi: 10.1111/ajt.14076.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26(5):589–595. doi: 10.1093/bioinformatics/btp698.
    1. Camacho C, Coulouris G, Avagyan V, Ma N, Papadopoulos J, Bealer K, Madden TL. BLAST+: architecture and applications. BMC Bioinformatics. 2009;10:421. doi: 10.1186/1471-2105-10-421.
    1. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014;15(3):R46. doi: 10.1186/gb-2014-15-3-r46.
    1. O'Leary NA, Wright MW, Brister JR, Ciufo S, Haddad D, McVeigh R, Rajput B, Robbertse B, Smith-White B, Ako-Adjei D, et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 2016;44(D1):D733–D745. doi: 10.1093/nar/gkv1189.
    1. Kaminski J, Gibson MK, Franzosa EA, Segata N, Dantas G, Huttenhower C. High-specificity targeted functional profiling in microbial communities with ShortBRED. PLoS Comput Biol. 2015;11(12):e1004557. doi: 10.1371/journal.pcbi.1004557.
    1. Jia B, Raphenya AR, Alcock B, Waglechner N, Guo P, Tsang KK, Lago BA, Dave BM, Pereira S, Sharma AN, et al. CARD 2017: expansion and model-centric curation of the comprehensive antibiotic resistance database. Nucleic Acids Res. 2017;45(D1):D566–D573. doi: 10.1093/nar/gkw1004.
    1. Shugay Mikhail, Bagaev Dmitriy V, Zvyagin Ivan V, Vroomans Renske M, Crawford Jeremy Chase, Dolton Garry, Komech Ekaterina A, Sycheva Anastasiya L, Koneva Anna E, Egorov Evgeniy S, Eliseev Alexey V, Van Dyk Ewald, Dash Pradyot, Attaf Meriem, Rius Cristina, Ladell Kristin, McLaren James E, Matthews Katherine K, Clemens E Bridie, Douek Daniel C, Luciani Fabio, van Baarle Debbie, Kedzierska Katherine, Kesmir Can, Thomas Paul G, Price David A, Sewell Andrew K, Chudakov Dmitriy M. VDJdb: a curated database of T-cell receptor sequences with known antigen specificity. Nucleic Acids Research. 2017;46(D1):D419–D427. doi: 10.1093/nar/gkx760.
    1. Team RC . R: a language and environment for statistical computing. 2017.
    1. Dixon P. VEGAN, a package of R functions for community ecology. J Veg Sci. 2003;14(6):927–930. doi: 10.1111/j.1654-1103.2003.tb02228.x.
    1. Wang GP, Garrigue A, Ciuffi A, Ronen K, Leipzig J, Berry C, Lagresle-Peyrou C, Benjelloun F, Hacein-Bey-Abina S, Fischer A, et al. DNA bar coding and pyrosequencing to analyze adverse events in therapeutic gene transfer. Nucleic Acids Res. 2008;36(9):e49. doi: 10.1093/nar/gkn125.
    1. Wang GP, Levine BL, Binder GK, Berry CC, Malani N, McGarrity G, Tebas P, June CH, Bushman FD. Analysis of lentiviral vector integration in HIV+ study subjects receiving autologous infusions of gene modified CD4+ T cells. Mol Ther. 2009;17(5):844–850. doi: 10.1038/mt.2009.16.
    1. Thrasher AJ, Gaspar HB, Baum C, Modlich U, Schambach A, Candotti F, Otsu M, Sorrentino B, Scobie L, Cameron E, et al. Gene therapy: X-SCID transgene leukaemogenicity. Nature. 2006;443(7109):E5–E6. doi: 10.1038/nature05219.
    1. Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, Adisetiyo H, Zabih S, Lincez PJ, Bittinger K, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171(7):647–654. doi: 10.1001/jamapediatrics.2017.0378.
    1. Bousso P, Wahn V, Douagi I, Horneff G, Pannetier C, Le Deist F, Zepp F, Niehues T, Kourilsky P, Fischer A, et al. Diversity, functionality, and stability of the T cell repertoire derived in vivo from a single human T cell precursor. Proc Natl Acad Sci U S A. 2000;97(1):274–278. doi: 10.1073/pnas.97.1.274.
    1. Vely F, Barlogis V, Vallentin B, Neven B, Piperoglou C, Ebbo M, Perchet T, Petit M, Yessaad N, Touzot F, et al. Evidence of innate lymphoid cell redundancy in humans. Nat Immunol. 2016;17(11):1291–1299. doi: 10.1038/ni.3553.
    1. Lane JP, Stewart CJ, Cummings SP, Gennery AR. Gut microbiome variations during hematopoietic stem cell transplant in severe combined immunodeficiency. J Allergy Clin Immunol. 2015;135(6):1654–1656. doi: 10.1016/j.jaci.2015.01.024.

Source: PubMed

3
S'abonner