Your browser doesn't support javascript.
loading
Dominant-negative IKZF1 mutations cause a T, B, and myeloid cell combined immunodeficiency.
Boutboul, David; Kuehn, Hye Sun; Van de Wyngaert, Zoé; Niemela, Julie E; Callebaut, Isabelle; Stoddard, Jennifer; Lenoir, Christelle; Barlogis, Vincent; Farnarier, Catherine; Vely, Frédéric; Yoshida, Nao; Kojima, Seiji; Kanegane, Hirokazu; Hoshino, Akihiro; Hauck, Fabian; Lhermitte, Ludovic; Asnafi, Vahid; Roehrs, Philip; Chen, Shaoying; Verbsky, James W; Calvo, Katherine R; Husami, Ammar; Zhang, Kejian; Roberts, Joseph; Amrol, David; Sleaseman, John; Hsu, Amy P; Holland, Steven M; Marsh, Rebecca; Fischer, Alain; Fleisher, Thomas A; Picard, Capucine; Latour, Sylvain; Rosenzweig, Sergio D.
Afiliação
  • Boutboul D; Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Inserm UMR 1163, Paris, France.
  • Kuehn HS; Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland, USA.
  • Van de Wyngaert Z; Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Inserm UMR 1163, Paris, France.
  • Niemela JE; University Paris Descartes Sorbonne Paris Cité, Imagine Institute, Paris, France.
  • Callebaut I; Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland, USA.
  • Stoddard J; Centre National de la Recherche Scientifique UMR 7590, Sorbonne Universities, University Pierre et Marie Curie-Paris 6-MNHN-IRD-IUC, Paris, France.
  • Lenoir C; Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland, USA.
  • Barlogis V; Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Inserm UMR 1163, Paris, France.
  • Farnarier C; Department of Paediatric Haematology-Oncology, La Timone Hospital, Marseille, France.
  • Vely F; Assistance Publique - Hôpitaux de Marseille (APHM) Hôpital Timone Enfants, Service d'Immunologie - Marseille Immunopôle, Marseille, France.
  • Yoshida N; Aix Marseille University, APHM, CNRS, Inserm, Centre d'Immunologie de Marseille-Luminy (CIML), Hôpital Timone Enfants, Service d'Immunologie - Marseille Immunopôle, Marseille, France.
  • Kojima S; Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Japan.
  • Kanegane H; Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
  • Hoshino A; Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
  • Hauck F; Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
  • Lhermitte L; Department of Pediatric Immunology and Rheumatology, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
  • Asnafi V; University Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Inserm 1151 and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (APHP), Necker-Enfants Malades Hospital, Paris, France.
  • Roehrs P; University Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Inserm 1151 and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (APHP), Necker-Enfants Malades Hospital, Paris, France.
  • Chen S; Levine Children's Hospital, Carolinas Healthcare System, Charlotte, North Carolina, USA.
  • Verbsky JW; Department of Pediatrics, Division of Rheumatology, Medical College of Wisconsin, Madison, Wisconsin, USA.
  • Calvo KR; Department of Pediatrics, Division of Rheumatology, Medical College of Wisconsin, Madison, Wisconsin, USA.
  • Husami A; Hematology section, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland, USA.
  • Zhang K; Division of Human Genetics and Division of Immune Deficiency and Bone Marrow Transplant, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.
  • Roberts J; Division of Human Genetics and Division of Immune Deficiency and Bone Marrow Transplant, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.
  • Amrol D; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA.
  • Sleaseman J; University of South Carolina School of Medicine, Columbia, South Carolina, USA.
  • Hsu AP; Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA.
  • Holland SM; Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, Maryland, USA.
  • Marsh R; Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, Maryland, USA.
  • Fischer A; Division of Human Genetics and Division of Immune Deficiency and Bone Marrow Transplant, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.
  • Fleisher TA; University Paris Descartes Sorbonne Paris Cité, Imagine Institute, Paris, France.
  • Picard C; Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, APHP, Paris, France.
  • Latour S; Collège de France, Paris, France.
  • Rosenzweig SD; Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, Maryland, USA.
J Clin Invest ; 128(7): 3071-3087, 2018 07 02.
Article em En | MEDLINE | ID: mdl-29889099
ABSTRACT
Ikaros/IKZF1 is an essential transcription factor expressed throughout hematopoiesis. IKZF1 is implicated in lymphocyte and myeloid differentiation and negative regulation of cell proliferation. In humans, somatic mutations in IKZF1 have been linked to the development of B cell acute lymphoblastic leukemia (ALL) in children and adults. Recently, heterozygous germline IKZF1 mutations have been identified in patients with a B cell immune deficiency mimicking common variable immunodeficiency. These mutations demonstrated incomplete penetrance and led to haploinsufficiency. Herein, we report 7 unrelated patients with a novel early-onset combined immunodeficiency associated with de novo germline IKZF1 heterozygous mutations affecting amino acid N159 located in the DNA-binding domain of IKZF1. Different bacterial and viral infections were diagnosed, but Pneumocystis jirovecii pneumonia was reported in all patients. One patient developed a T cell ALL. This immunodeficiency was characterized by innate and adaptive immune defects, including low numbers of B cells, neutrophils, eosinophils, and myeloid dendritic cells, as well as T cell and monocyte dysfunctions. Notably, most T cells exhibited a naive phenotype and were unable to evolve into effector memory cells. Functional studies indicated these mutations act as dominant negative. This defect expands the clinical spectrum of human IKZF1-associated diseases from somatic to germline, from haploinsufficient to dominant negative.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Mutação em Linhagem Germinativa / Fator de Transcrição Ikaros / Mutação com Perda de Função / Síndromes de Imunodeficiência Tipo de estudo: Prognostic_studies Limite: Adolescent / Adult / Child / Child, preschool / Female / Humans / Infant / Male Idioma: En Revista: J Clin Invest Ano de publicação: 2018 Tipo de documento: Article País de afiliação: França

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Mutação em Linhagem Germinativa / Fator de Transcrição Ikaros / Mutação com Perda de Função / Síndromes de Imunodeficiência Tipo de estudo: Prognostic_studies Limite: Adolescent / Adult / Child / Child, preschool / Female / Humans / Infant / Male Idioma: En Revista: J Clin Invest Ano de publicação: 2018 Tipo de documento: Article País de afiliação: França