Skip to main content
Top
Published in: Journal of Clinical Immunology 5/2021

01-07-2021 | Graft-Versus-Host Disease | Commentary

The “Editors” Take to RAG: Promise of CRISPR/Cas9/rAAV6-Based Gene Therapy for RAG2 Deficiency

Authors: Vivien Béziat, Isabelle Meyts

Published in: Journal of Clinical Immunology | Issue 5/2021

Login to get access

Excerpt

Severe combined immunodeficiencies (SCID) are inborn errors of immunity characterized by an absence of T lymphocytes [1]. The concomitant presence or absence of B and/or natural killer (NK) cells usually points to specific genetic defects. The presence of NK cells coupled to an absence of T and B cells (TBNK+ SCID) is the hallmark of T cell receptor (TCR) and B cell receptor (BCR) rearrangement defects, due to mutations affecting the V(D)J recombination machinery. V(D)J recombination allows the generation of highly diverse TCR and BCR repertoires. TCR and BCR rearrangements are initiated by the recombination-activating gene (RAG) complex, a tetramer composed of two RAG1 and RAG2 heterodimers [2]. Upon binding to the recombination signal sequences (RSS) flanking the V, D, and J gene segments, the RAG complex introduces double-strand breaks into the DNA that are processed and joined by the non-homologous end-joining (NHEJ) pathway. These two genes, and their role in V(D)J recombination and T and B cell differentiation, were characterized in a series of landmark studies in mice in the late 1980s and early 1990s [36]. Shortly afterwards, the role of RAG mutations in T and B cell lymphopenia in humans was confirmed [7]. Biallelic RAG mutations abolishing RAG activity cause SCID; biallelic hypomorphic RAG mutations that markedly reduce, but do not completely abolish, recombination activity are the most common cause of Omenn syndrome. Clinically, this syndrome is characterized by early-onset generalized erythroderma, lymphadenopathy, hepatosplenomegaly, and typical laboratory features, including eosinophilia, severe hypogammaglobulinemia with high IgE levels, and autologous, oligoclonal, activated T cells infiltrating multiple organs. Hypomorphic mutations of RAG leading to even milder deficiencies of RAG activity cause a phenotype of combined immunodeficiency (CID), characterized by granulomatous inflammation and various autoimmune manifestations. Since the first report in 1996, more than 400 patients with RAG deficiency have been described [8]. In a recent US report, RAG1/RAG2 deficiency accounted for 20% of SCID cases [9]. …
Literature
1.
go back to reference Fischer A, Notarangelo LD, Neven B, Cavazzana M, Puck JM. Severe combined immunodeficiencies and related disorders. Nat Rev Dis Primers. 2015;1:15061.CrossRef Fischer A, Notarangelo LD, Neven B, Cavazzana M, Puck JM. Severe combined immunodeficiencies and related disorders. Nat Rev Dis Primers. 2015;1:15061.CrossRef
2.
go back to reference Notarangelo LD, Kim M-S, Walter JE, Lee YN. Human RAG mutations: biochemistry and clinical implications. Nat Rev Immunol. 2016;16:234–46. Notarangelo LD, Kim M-S, Walter JE, Lee YN. Human RAG mutations: biochemistry and clinical implications. Nat Rev Immunol. 2016;16:234–46.
3.
go back to reference Schatz DG, Oettinger MA, Baltimore D. The V(D)J recombination activating gene, RAG-1. Cell. 1989;59:1035–48.CrossRef Schatz DG, Oettinger MA, Baltimore D. The V(D)J recombination activating gene, RAG-1. Cell. 1989;59:1035–48.CrossRef
4.
go back to reference Oettinger MA, Schatz DG, Gorka C, Baltimore D. RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science. 1990;248:1517–23. Oettinger MA, Schatz DG, Gorka C, Baltimore D. RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination. Science. 1990;248:1517–23.
5.
go back to reference Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–77.CrossRef Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–77.CrossRef
6.
go back to reference Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell. 1992;68:855–67. Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell. 1992;68:855–67.
7.
go back to reference Schwarz K, Gauss GH, Ludwig L, Pannicke U, Li Z, Lindner D, et al. RAG mutations in human B cell-negative SCID. Science. 1996;274:97–9. Schwarz K, Gauss GH, Ludwig L, Pannicke U, Li Z, Lindner D, et al. RAG mutations in human B cell-negative SCID. Science. 1996;274:97–9.
8.
go back to reference Delmonte OM, Schuetz C, Notarangelo LD. RAG deficiency: two genes, many diseases. J Clin Immunol. 2018;38:646–55.CrossRef Delmonte OM, Schuetz C, Notarangelo LD. RAG deficiency: two genes, many diseases. J Clin Immunol. 2018;38:646–55.CrossRef
9.
go back to reference Dvorak CC, Haddad E, Buckley RH, Cowan MJ, Logan B, Griffith LM, et al. The genetic landscape of severe combined immunodeficiency in the United States and Canada in the current era (2010-2018). J Allergy Clin Immunol. 2019;143:405–7. Dvorak CC, Haddad E, Buckley RH, Cowan MJ, Logan B, Griffith LM, et al. The genetic landscape of severe combined immunodeficiency in the United States and Canada in the current era (2010-2018). J Allergy Clin Immunol. 2019;143:405–7.
10.
go back to reference Ciurea SO, Borges Bittencourt MC, Milton DR, Cao K, Kongtim P, Rondon G, et al. Is a matched unrelated donor search needed for all allogeneic transplant candidates? Blood. 2018;132:3450–0. Ciurea SO, Borges Bittencourt MC, Milton DR, Cao K, Kongtim P, Rondon G, et al. Is a matched unrelated donor search needed for all allogeneic transplant candidates? Blood. 2018;132:3450–0.
11.
go back to reference Neven B, Leroy S, Decaluwe H, Le Deist F, Picard C, Moshous D, et al. Long-term outcome after hematopoietic stem cell transplantation of a single-center cohort of 90 patients with severe combined immunodeficiency. Blood. 2009;113:4114–24.CrossRef Neven B, Leroy S, Decaluwe H, Le Deist F, Picard C, Moshous D, et al. Long-term outcome after hematopoietic stem cell transplantation of a single-center cohort of 90 patients with severe combined immunodeficiency. Blood. 2009;113:4114–24.CrossRef
13.
go back to reference Haddad E, Logan BR, Griffith LM, Buckley RH, Parrott RE, Prockop SE, et al. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery. Blood. 2018;132:1737–49. Haddad E, Logan BR, Griffith LM, Buckley RH, Parrott RE, Prockop SE, et al. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery. Blood. 2018;132:1737–49.
14.
go back to reference Heimall J, Buckley RH, Puck J, Fleisher TA, Gennery AR, Haddad E, et al. Recommendations for screening and management of late effects in patients with severe combined immunodeficiency after allogenic hematopoietic cell transplantation: a consensus statement from the second Pediatric blood and marrow transplant consortium international conference on late effects after pediatric HCT. Biol Blood Marrow Transplant. 2017;23:1229–40. Heimall J, Buckley RH, Puck J, Fleisher TA, Gennery AR, Haddad E, et al. Recommendations for screening and management of late effects in patients with severe combined immunodeficiency after allogenic hematopoietic cell transplantation: a consensus statement from the second Pediatric blood and marrow transplant consortium international conference on late effects after pediatric HCT. Biol Blood Marrow Transplant. 2017;23:1229–40.
15.
go back to reference Puck JM. Newborn screening for severe combined immunodeficiency and T-cell lymphopenia. Immunol Rev. 2019;287:241–52.CrossRef Puck JM. Newborn screening for severe combined immunodeficiency and T-cell lymphopenia. Immunol Rev. 2019;287:241–52.CrossRef
16.
go back to reference Kuo CY, Garabedian E, Puck J, Cowan MJ, Sullivan KE, Buckley RH, et al. Adenosine deaminase (ADA)–deficient severe combined immune deficiency (SCID) in the US Immunodeficiency Network (USIDNet) Registry. J Clin Immunol. 2020;40:1124–31. Kuo CY, Garabedian E, Puck J, Cowan MJ, Sullivan KE, Buckley RH, et al. Adenosine deaminase (ADA)–deficient severe combined immune deficiency (SCID) in the US Immunodeficiency Network (USIDNet) Registry. J Clin Immunol. 2020;40:1124–31.
17.
go back to reference Touzot F, Moshous D, Creidy R, Neven B, Frange P, Cros G, et al. Faster T-cell development following gene therapy compared with haploidentical HSCT in the treatment of SCID-X1. Blood. 2015;125:3563–9. Touzot F, Moshous D, Creidy R, Neven B, Frange P, Cros G, et al. Faster T-cell development following gene therapy compared with haploidentical HSCT in the treatment of SCID-X1. Blood. 2015;125:3563–9.
18.
go back to reference van Til NP, Sarwari R, Visser TP, Hauer J, Lagresle-Peyrou C, van der Velden G, et al. Recombination-activating gene 1 (Rag1)–deficient mice with severe combined immunodeficiency treated with lentiviral gene therapy demonstrate autoimmune Omenn-like syndrome. J Allergy Clin Immunol. 2014;133:1116–23. van Til NP, Sarwari R, Visser TP, Hauer J, Lagresle-Peyrou C, van der Velden G, et al. Recombination-activating gene 1 (Rag1)–deficient mice with severe combined immunodeficiency treated with lentiviral gene therapy demonstrate autoimmune Omenn-like syndrome. J Allergy Clin Immunol. 2014;133:1116–23.
19.
go back to reference Capo V, Castiello MC, Fontana E, Penna S, Bosticardo M, Draghici E, et al. Efficacy of lentivirus-mediated gene therapy in an Omenn syndrome recombination-activating gene 2 mouse model is not hindered by inflammation and immune dysregulation. J Allergy Clin Immunol. 2018;142:928–941.e8.CrossRef Capo V, Castiello MC, Fontana E, Penna S, Bosticardo M, Draghici E, et al. Efficacy of lentivirus-mediated gene therapy in an Omenn syndrome recombination-activating gene 2 mouse model is not hindered by inflammation and immune dysregulation. J Allergy Clin Immunol. 2018;142:928–941.e8.CrossRef
20.
go back to reference Pike-Overzet K, Rodijk M, Ng Y-Y, Baert MRM, Lagresle-Peyrou C, Schambach A, et al. Correction of murine Rag1 deficiency by self-inactivating lentiviral vector-mediated gene transfer. Leukemia. 2011;25:1471–83. Pike-Overzet K, Rodijk M, Ng Y-Y, Baert MRM, Lagresle-Peyrou C, Schambach A, et al. Correction of murine Rag1 deficiency by self-inactivating lentiviral vector-mediated gene transfer. Leukemia. 2011;25:1471–83.
21.
go back to reference Lagresle-Peyrou C, Benjelloun F, Hue C, Andre-Schmutz I, Bonhomme D, Forveille M, et al. Restoration of human B-cell differentiation into NOD-SCID mice engrafted with gene-corrected CD34+ cells isolated from Artemis or RAG1-deficient patients. Mol Ther. 2008;16:396–403. Lagresle-Peyrou C, Benjelloun F, Hue C, Andre-Schmutz I, Bonhomme D, Forveille M, et al. Restoration of human B-cell differentiation into NOD-SCID mice engrafted with gene-corrected CD34+ cells isolated from Artemis or RAG1-deficient patients. Mol Ther. 2008;16:396–403.
22.
go back to reference Mostoslavsky G, Fabian AJ, Rooney S, Alt FW, Mulligan RC. Complete correction of murine Artemis immunodeficiency by lentiviral vector-mediated gene transfer. Proc Natl Acad Sci U S A. 2006;103:16406–11.CrossRef Mostoslavsky G, Fabian AJ, Rooney S, Alt FW, Mulligan RC. Complete correction of murine Artemis immunodeficiency by lentiviral vector-mediated gene transfer. Proc Natl Acad Sci U S A. 2006;103:16406–11.CrossRef
23.
go back to reference Yates F, Malassis-Séris M, Stockholm D, Bouneaud C, Larousserie F, Noguiez-Hellin P, et al. Gene therapy of RAG-2−/− mice: sustained correction of the immunodeficiency. Blood. 2002;100:3942–9.CrossRef Yates F, Malassis-Séris M, Stockholm D, Bouneaud C, Larousserie F, Noguiez-Hellin P, et al. Gene therapy of RAG-2−/− mice: sustained correction of the immunodeficiency. Blood. 2002;100:3942–9.CrossRef
24.
go back to reference Lagresle-Peyrou C, Yates F, Malassis-Séris M, Hue C, Morillon E, Garrigue A, et al. Long-term immune reconstitution in RAG-1-deficient mice treated by retroviral gene therapy: a balance between efficiency and toxicity. Blood. 2006;107:63–72. Lagresle-Peyrou C, Yates F, Malassis-Séris M, Hue C, Morillon E, Garrigue A, et al. Long-term immune reconstitution in RAG-1-deficient mice treated by retroviral gene therapy: a balance between efficiency and toxicity. Blood. 2006;107:63–72.
25.
go back to reference Bak RO, Dever DP, Porteus MH. CRISPR/Cas9 genome editing in human hematopoietic stem cells. Nat Protoc. 2018;13:358–76. Bak RO, Dever DP, Porteus MH. CRISPR/Cas9 genome editing in human hematopoietic stem cells. Nat Protoc. 2018;13:358–76.
26.
go back to reference Pavel-Dinu M, Wiebking V, Dejene BT, Srifa W, Mantri S, Nicolas CE, et al. Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nat Commun Nature Publishing Group. 2019;10:1634.CrossRef Pavel-Dinu M, Wiebking V, Dejene BT, Srifa W, Mantri S, Nicolas CE, et al. Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nat Commun Nature Publishing Group. 2019;10:1634.CrossRef
27.
go back to reference Themeli M, Chhatta A, Boersma H, Prins HJ, Cordes M, de Wilt E, et al. iPSC-based modeling of RAG2 severe combined immunodeficiency reveals multiple T cell developmental arrests. Stem Cell Reports. 2020;14:300–11. Themeli M, Chhatta A, Boersma H, Prins HJ, Cordes M, de Wilt E, et al. iPSC-based modeling of RAG2 severe combined immunodeficiency reveals multiple T cell developmental arrests. Stem Cell Reports. 2020;14:300–11.
Metadata
Title
The “Editors” Take to RAG: Promise of CRISPR/Cas9/rAAV6-Based Gene Therapy for RAG2 Deficiency
Authors
Vivien Béziat
Isabelle Meyts
Publication date
01-07-2021
Publisher
Springer US
Published in
Journal of Clinical Immunology / Issue 5/2021
Print ISSN: 0271-9142
Electronic ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-021-01024-4

Other articles of this Issue 5/2021

Journal of Clinical Immunology 5/2021 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.