Severe combined immune deficiency (SCID) includes a variety of genetic defects characterized by severe numerical and functional defects of T lymphocytes, variably associated with defects of B and NK lymphocytes. Differences exist between mice and humans with mutations in orthologue SCID- associated genes. Moreover, for some forms of SCID no animal models are currently available. Finally, SCID have offered proof of principle that gene therapy can cure human diseases; however, leukemic proliferation due to insertional mutagenesis has been reported in 5 SCID patients treated by gene therapy. Altogether, these notions emphasize the importance of studying human models to gain insights into pathological human T cell development and to develop novel and safer approaches to gene therapy. Co-culture of human hematopoietic stem cells (HSCs) with mouse OP9 stromal cells engineered to express the Notch1 ligands Delta-like 1 (Dll1) or Delta-like 4 (Dll4) has been used to study T cell differentiation. Moreover, severely immunodeficient mice can serve as an in vivo system to study T cell development from human HSCs. However, these approaches can be hardly applied to the study of human SCID, given the rarity of this condition that limits access to freshly isolated HSCs from the patients. Induced pluripotent stem cells (iPSCs) represent a novel tool for disease modeling and correction and may be used to overcome technical and feasibility barriers that have so far hampered the study of T cell development in human SCID. We have assembled a team of investigators with specific and complementary expertise in immunodeficiency, iPSC biology, T cell development and integration site analysis upon gene transfer. We have created a large repository of fibroblast cell lines from patients with various forms of SCID. We now intend to generate iPSCs from these patients. We will characterize the stemness and pluripotency profile and verify genetic integrity of patient-derived iPSCs. To model SCID, we will differentiate iPSCs into T lymphocytes both in vitro and in vivo. We also intend to use patient-derived iPSCs as a platform to explore the efficacy and safety of self-inactivating lentiviral vectors in the correction of various forms of SCID both in vitro and in viv in immunodeficient mice. We anticipate that SCID patients-derived iPSCs will represent a novel and robust platform to study human T cell development and for preclinical assessment of efficacy and safety of gene therapy for these diseases.

Public Health Relevance

Severe combined immune deficiency (SCID) includes several genetic defects of the immune system. SCID has offered proof of principle that gene therapy can cure human disease; however, leukemia has been observed in some patients as a result of this treatment. Availability of a limitless source of stem cells from patients with SCID could help improve our knowledge of the disease and assist in the C. We will differentiate skin cells (fibroblasts) from SCID patients into induced pluripotent stem cells and analyze their ability to give rise to functional lymphocytes. We will also introduce a normal copy of the gene into patient iPSCs and analyze reconstitution of T cell development in vitro and in animal models.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI100887-04
Application #
8874092
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Voulgaropoulou, Frosso
Project Start
2012-07-01
Project End
2016-06-30
Budget Start
2015-07-01
Budget End
2016-06-30
Support Year
4
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Children's Hospital Boston
Department
Type
DUNS #
076593722
City
Boston
State
MA
Country
United States
Zip Code
Ott de Bruin, L M; Bosticardo, M; Barbieri, A et al. (2018) Hypomorphic Rag1 mutations alter the preimmune repertoire at early stages of lymphoid development. Blood 132:281-292
Rowe, Jared H; Stadinski, Brian D; Henderson, Lauren A et al. (2017) Abnormalities of T-cell receptor repertoire in CD4+ regulatory and conventional T cells in patients with RAG mutations: Implications for autoimmunity. J Allergy Clin Immunol 140:1739-1743.e7
Dobbs, Kerry; Tabellini, Giovanna; Calzoni, Enrica et al. (2017) Natural Killer Cells from Patients with Recombinase-Activating Gene and Non-Homologous End Joining Gene Defects Comprise a Higher Frequency of CD56bright NKG2A+++ Cells, and Yet Display Increased Degranulation and Higher Perforin Content. Front Immunol 8:798
Kumánovics, Attila; Lee, Yu Nee; Close, Devin W et al. (2017) Estimated disease incidence of RAG1/2 mutations: A case report and querying the Exome Aggregation Consortium. J Allergy Clin Immunol 139:690-692.e3
Volpi, Stefano; Yamazaki, Yasuhiro; Brauer, Patrick M et al. (2017) EXTL3 mutations cause skeletal dysplasia, immune deficiency, and developmental delay. J Exp Med 214:623-637
Dobbs, Kerry; Tabellini, Giovanna; Calzoni, Enrica et al. (2017) Corrigendum: Natural Killer Cells from Patients with Recombinase-Activating Gene and Non-Homologous End Joining Gene Defects Comprise a Higher Frequency of CD56bright NKG2A+++ Cells, and Yet Display Increased Degranulation and Higher Perforin Content. Front Immunol 8:1244
Walter, Jolan E; Rosen, Lindsey B; Csomos, Krisztian et al. (2016) Broad-spectrum antibodies against self-antigens and cytokines in RAG deficiency. J Clin Invest 126:4389
Lee, Yu Nee; Frugoni, Francesco; Dobbs, Kerry et al. (2016) Characterization of T and B cell repertoire diversity in patients with RAG deficiency. Sci Immunol 1:
Felgentreff, Kerstin; Baxi, Sachin N; Lee, Yu Nee et al. (2016) Ligase-4 Deficiency Causes Distinctive Immune Abnormalities in Asymptomatic Individuals. J Clin Immunol 36:341-53
Brauer, Patrick M; Singh, Jastaranpreet; Xhiku, Sintia et al. (2016) T Cell Genesis: In Vitro Veritas Est? Trends Immunol 37:889-901

Showing the most recent 10 out of 31 publications