Your browser doesn't support javascript.
loading
Oncogenetic landscape of lymphomagenesis in coeliac disease.
Cording, Sascha; Lhermitte, Ludovic; Malamut, Georgia; Berrabah, Sofia; Trinquand, Amélie; Guegan, Nicolas; Villarese, Patrick; Kaltenbach, Sophie; Meresse, Bertrand; Khater, Sherine; Dussiot, Michael; Bras, Marc; Cheminant, Morgane; Tesson, Bruno; Bole-Feysot, Christine; Bruneau, Julie; Molina, Thierry Jo; Sibon, David; Macintyre, Elizabeth; Hermine, Olivier; Cellier, Christophe; Asnafi, Vahid; Cerf-Bensussan, Nadine.
Afiliação
  • Cording S; Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France.
  • Lhermitte L; Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France.
  • Malamut G; Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
  • Berrabah S; Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France.
  • Trinquand A; Department of Gastroenterology, AP-HP, Hôpital Cochin, Paris, France.
  • Guegan N; Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France.
  • Villarese P; Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France.
  • Kaltenbach S; Haematology Department, National Children's Research Centre, Children's Health Ireland at Crumlin, Dublin, Ireland.
  • Meresse B; Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, INSERM UMR 1163, Paris, France.
  • Khater S; Université de Paris, Institut Necker-Enfants Malades, INSERM UMR 1151, Paris, France.
  • Dussiot M; Laboratory of Onco-Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
  • Bras M; Department of Cytogenetics, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
  • Cheminant M; Université de Lille, CHU Lille, INSERM UMR 1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France.
  • Tesson B; Department of Gastroenterology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France.
  • Bole-Feysot C; Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France.
  • Bruneau J; Université de Paris, Imagine Institute, Bioinformatics Platform, Paris, France.
  • Molina TJ; Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France.
  • Sibon D; Clinical Haematology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
  • Macintyre E; Institut Carnot CALYM, Paris, France.
  • Hermine O; Université de Paris, Imagine Institute, Genomics Platform, Paris, France.
  • Cellier C; Department of Pathology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
  • Asnafi V; Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France.
  • Cerf-Bensussan N; Department of Pathology, AP-HP, Hôpital Necker Enfants-Malades, Paris, France.
Gut ; 71(3): 497-508, 2022 03.
Article em En | MEDLINE | ID: mdl-33579790
ABSTRACT

OBJECTIVE:

Enteropathy-associated T-cell lymphoma (EATL) is a rare but severe complication of coeliac disease (CeD), often preceded by low-grade clonal intraepithelial lymphoproliferation, referred to as type II refractory CeD (RCDII). Knowledge on underlying oncogenic mechanisms remains scarce. Here, we analysed and compared the mutational landscape of RCDII and EATL in order to identify genetic drivers of CeD-associated lymphomagenesis.

DESIGN:

Pure populations of RCDII-cells derived from intestinal biopsies (n=9) or sorted from blood (n=2) were analysed by whole exome sequencing, comparative genomic hybridisation and RNA sequencing. Biopsies from RCDII (n=50), EATL (n=19), type I refractory CeD (n=7) and uncomplicated CeD (n=18) were analysed by targeted next-generation sequencing. Moreover, functional in vitro studies and drug testing were performed in RCDII-derived cell lines.

RESULTS:

80% of RCDII and 90% of EATL displayed somatic gain-of-functions mutations in the JAK1-STAT3 pathway, including a remarkable p.G1097 hotspot mutation in the JAK1 kinase domain in approximately 50% of cases. Other recurrent somatic events were deleterious mutations in nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) regulators TNFAIP3 and TNIP3 and potentially oncogenic mutations in TET2, KMT2D and DDX3X. JAK1 inhibitors, and the proteasome inhibitor bortezomib could block survival and proliferation of malignant RCDII-cell lines.

CONCLUSION:

Mutations activating the JAK1-STAT3 pathway appear to be the main drivers of CeD-associated lymphomagenesis. In concert with mutations in negative regulators of NF-κB, they may favour the clonal emergence of malignant lymphocytes in the cytokine-rich coeliac intestine. The identified mutations are attractive therapeutic targets to treat RCDII and block progression towards EATL.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos / Doença Celíaca / Linfoma de Células T Associado a Enteropatia / Mutação com Ganho de Função Tipo de estudo: Etiology_studies / Incidence_studies / Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Adult / Aged / Aged80 / Female / Humans / Male / Middle aged País/Região como assunto: Europa Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos / Doença Celíaca / Linfoma de Células T Associado a Enteropatia / Mutação com Ganho de Função Tipo de estudo: Etiology_studies / Incidence_studies / Observational_studies / Prognostic_studies / Risk_factors_studies Limite: Adult / Aged / Aged80 / Female / Humans / Male / Middle aged País/Região como assunto: Europa Idioma: En Ano de publicação: 2022 Tipo de documento: Article