Your browser doesn't support javascript.
loading
Reprogramming identifies functionally distinct stages of clonal evolution in myelodysplastic syndromes.
Hsu, Jasper; Reilly, Andreea; Hayes, Brian J; Clough, Courtnee A; Konnick, Eric Q; Torok-Storb, Beverly; Gulsuner, Suleyman; Wu, David; Becker, Pamela S; Keel, Siobán B; Abkowitz, Janis L; Doulatov, Sergei.
Afiliação
  • Hsu J; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA.
  • Reilly A; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA.
  • Hayes BJ; Fred Hutchinson Cancer Research Center, Seattle, WA; and.
  • Clough CA; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA.
  • Konnick EQ; Department of Laboratory Medicine.
  • Torok-Storb B; Fred Hutchinson Cancer Research Center, Seattle, WA; and.
  • Gulsuner S; Division of Medical Genetics, Department of Medicine.
  • Wu D; Department of Laboratory Medicine.
  • Becker PS; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA.
  • Keel SB; Fred Hutchinson Cancer Research Center, Seattle, WA; and.
  • Abkowitz JL; Institute for Stem Cell and Regenerative Medicine, and.
  • Doulatov S; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA.
Blood ; 134(2): 186-198, 2019 07 11.
Article em En | MEDLINE | ID: mdl-31010849
ABSTRACT
Myeloid neoplasms, including myelodysplastic syndromes (MDS), are genetically heterogeneous disorders driven by clonal acquisition of somatic mutations in hematopoietic stem and progenitor cells (HPCs). The order of premalignant mutations and their impact on HPC self-renewal and differentiation remain poorly understood. We show that episomal reprogramming of MDS patient samples generates induced pluripotent stem cells from single premalignant cells with a partial complement of mutations, directly informing the temporal order of mutations in the individual patient. Reprogramming preferentially captured early subclones with fewer mutations, which were rare among single patient cells. To evaluate the functional impact of clonal evolution in individual patients, we differentiated isogenic MDS induced pluripotent stem cells harboring up to 4 successive clonal abnormalities recapitulating a progressive decrease in hematopoietic differentiation potential. SF3B1, in concert with epigenetic mutations, perturbed mitochondrial function leading to accumulation of damaged mitochondria during disease progression, resulting in apoptosis and ineffective erythropoiesis. Reprogramming also informed the order of premalignant mutations in patients with complex karyotype and identified 5q deletion as an early cytogenetic anomaly. The loss of chromosome 5q cooperated with TP53 mutations to perturb genome stability, promoting acquisition of structural and karyotypic abnormalities. Reprogramming thus enables molecular and functional interrogation of preleukemic clonal evolution, identifying mitochondrial function and chromosome stability as key pathways affected by acquisition of somatic mutations in MDS.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Síndromes Mielodisplásicas / Células-Tronco Hematopoéticas / Células-Tronco Pluripotentes / Reprogramação Celular / Evolução Clonal Limite: Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Síndromes Mielodisplásicas / Células-Tronco Hematopoéticas / Células-Tronco Pluripotentes / Reprogramação Celular / Evolução Clonal Limite: Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article