4.7 Article

SF3B1-initiating mutations in MDS-RSs target lymphomyeloid hematopoietic stem cells

期刊

BLOOD
卷 130, 期 7, 页码 881-890

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood-2017-03-776070

关键词

-

资金

  1. Swedish Cancer Society (Cancerfonden) [150269]
  2. Cancer Research Foundations of Radiumhemmets (Radiumhemmets Forskningsfonder) [151103]
  3. Swedish Research Council (Vetenskapsradet) [521-2013-3577]
  4. international recruitment grant from the Swedish Medical Research Council
  5. Tobias Foundation
  6. Center for Innovative Medicine at KI
  7. Versus Arthritis
  8. Cancer Research UK [21019] Funding Source: researchfish

向作者/读者索取更多资源

Mutations in the RNA splicing gene SF3B1 are found in >80% of patients with myelodysplastic syndrome with ring sideroblasts (MDS-RS). We investigated the origin of SF3B1 mutations within the bone marrow hematopoietic stem and progenitor cell compartments in patients with MDS-RS. Screening for recurrently mutated genes in the mononuclear cell fraction revealed mutations in SF3B1 in 39 of 40 cases (97.5%), combined with TET2 and DNMT3A in 11 (28%) and 6 (15%) patients, respectively. All recurrent mutations identified in mononuclear cells could be tracked back to the phenotypically defined hematopoietic stem cell (HSC) compartment in all investigated patients and were also present in downstream myeloid and erythroid progenitor cells. While in agreement with previous studies, little or no evidence for clonal (SF3B1 mutation) involvement could be found in mature B cells, consistent involvement at the pro-B-cell progenitor stage was established, providing definitive evidence for SF3B1 mutations targeting lymphomyeloid HSCs and compatible with mutated SF3B1 negatively affecting lymphoid development. Assessment of stem cell function in vitro as well as in vivo established that only HSCs and not investigated progenitor populations could propagate the SF3B1 mutated clone. Upon transplantation into immune-deficient mice, SF3B1 mutated MDS-RS HSCs differentiated into characteristic ring sideroblasts, the hallmark of MDS-RS. Our findings provide evidence of a multipotent lymphomyeloid HSC origin of SF3B1 mutations in MDS-RS patients and provide a novel in vivo platform for mechanistically and therapeutically exploring SF3B1 mutated MDS-RS.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据