Your browser doesn't support javascript.
loading
Modeling altered T-cell development with induced pluripotent stem cells from patients with RAG1-dependent immune deficiencies.
Brauer, Patrick M; Pessach, Itai M; Clarke, Erik; Rowe, Jared H; Ott de Bruin, Lisa; Lee, Yu Nee; Dominguez-Brauer, Carmen; Comeau, Anne M; Awong, Geneve; Felgentreff, Kerstin; Zhang, Yuhang H; Bredemeyer, Andrea; Al-Herz, Waleed; Du, Likun; Ververs, Francesca; Kennedy, Marion; Giliani, Silvia; Keller, Gordon; Sleckman, Barry P; Schatz, David G; Bushman, Frederic D; Notarangelo, Luigi D; Zúñiga-Pflücker, Juan Carlos.
Afiliación
  • Brauer PM; Sunnybrook Research Institute, and Department of Immunology, University of Toronto, Toronto, ON, Canada;
  • Pessach IM; Division of Immunology, Boston Children's Hospital, Boston, MA; Department of Pediatric Critical Care, Sheba Medical Center, Tel-Aviv University, Tel-Aviv, Israel;
  • Clarke E; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA;
  • Rowe JH; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Ott de Bruin L; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Lee YN; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Dominguez-Brauer C; Ontario Cancer Institute, University Health Network, Toronto, ON, Canada;
  • Comeau AM; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA;
  • Awong G; Sunnybrook Research Institute, and Department of Immunology, University of Toronto, Toronto, ON, Canada; McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada;
  • Felgentreff K; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Zhang YH; Departments of Immunobiology and Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT;
  • Bredemeyer A; Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO;
  • Al-Herz W; Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait;
  • Du L; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Ververs F; Division of Immunology, Boston Children's Hospital, Boston, MA;
  • Kennedy M; McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada;
  • Giliani S; "Angelo Nocivelli" Institute for Molecular Medicine, University of Brescia, Brescia, Italy;
  • Keller G; McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, Canada;
  • Sleckman BP; Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO;
  • Schatz DG; Departments of Immunobiology and Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT; Howard Hughes Medical Institute, New Haven, CT; and.
  • Bushman FD; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA;
  • Notarangelo LD; Division of Immunology, Boston Children's Hospital, Boston, MA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA.
  • Zúñiga-Pflücker JC; Sunnybrook Research Institute, and Department of Immunology, University of Toronto, Toronto, ON, Canada;
Blood ; 128(6): 783-93, 2016 08 11.
Article en En | MEDLINE | ID: mdl-27301863
ABSTRACT
Primary immunodeficiency diseases comprise a group of heterogeneous genetic defects that affect immune system development and/or function. Here we use in vitro differentiation of human induced pluripotent stem cells (iPSCs) generated from patients with different recombination-activating gene 1 (RAG1) mutations to assess T-cell development and T-cell receptor (TCR) V(D)J recombination. RAG1-mutants from severe combined immunodeficient (SCID) patient cells showed a failure to sustain progression beyond the CD3(--)CD4(-)CD8(-)CD7(+)CD5(+)CD38(-)CD31(-/lo)CD45RA(+) stage of T-cell development to reach the CD3(-/+)CD4(+)CD8(+)CD7(+)CD5(+)CD38(+)CD31(+)CD45RA(-) stage. Despite residual mutant RAG1 recombination activity from an Omenn syndrome (OS) patient, similar impaired T-cell differentiation was observed, due to increased single-strand DNA breaks that likely occur due to heterodimers consisting of both an N-terminal truncated and a catalytically dead RAG1. Furthermore, deep-sequencing analysis of TCR-ß (TRB) and TCR-α (TRA) rearrangements of CD3(-)CD4(+)CD8(-) immature single-positive and CD3(+)CD4(+)CD8(+) double-positive cells showed severe restriction of repertoire diversity with preferential usage of few Variable, Diversity, and Joining genes, and skewed length distribution of the TRB and TRA complementary determining region 3 sequences from SCID and OS iPSC-derived cells, whereas control iPSCs yielded T-cell progenitors with a broadly diversified repertoire. Finally, no TRA/δ excision circles (TRECs), a marker of TRA/δ locus rearrangements, were detected in SCID and OS-derived T-lineage cells, consistent with a pre-TCR block in T-cell development. This study compares human T-cell development of SCID vs OS patients, and elucidates important differences that help to explain the wide range of immunologic phenotypes that result from different mutations within the same gene of various patients.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Linfocitos T / Inmunodeficiencia Combinada Grave / Proteínas de Homeodominio / Células Madre Pluripotentes Inducidas Tipo de estudio: Prognostic_studies Límite: Humans / Infant Idioma: En Revista: Blood Año: 2016 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Linfocitos T / Inmunodeficiencia Combinada Grave / Proteínas de Homeodominio / Células Madre Pluripotentes Inducidas Tipo de estudio: Prognostic_studies Límite: Humans / Infant Idioma: En Revista: Blood Año: 2016 Tipo del documento: Article