Your browser doesn't support javascript.
loading
Heterozygous mutation SLFN14 K208N in mice mediates species-specific differences in platelet and erythroid lineage commitment.
Stapley, Rachel J; Smith, Christopher W; Haining, Elizabeth J; Bacon, Andrea; Lax, Sian; Pisareva, Vera P; Pisarev, Andrey V; Watson, Steve P; Khan, Abdullah O; Morgan, Neil V.
Afiliación
  • Stapley RJ; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences.
  • Smith CW; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences.
  • Haining EJ; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences.
  • Bacon A; MRC Centre for Immune Regulation, Transgenics Facility, and.
  • Lax S; Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, United Kingdom.
  • Pisareva VP; Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY; and.
  • Pisarev AV; Department of Cell Biology, SUNY Downstate Health Sciences University, Brooklyn, NY; and.
  • Watson SP; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences.
  • Khan AO; Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, United Kingdom.
  • Morgan NV; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences.
Blood Adv ; 5(2): 377-390, 2021 01 26.
Article en En | MEDLINE | ID: mdl-33496736
ABSTRACT
Schlafen 14 (SLFN14) has recently been identified as an endoribonuclease responsible for cleaving RNA to regulate and inhibit protein synthesis. Early studies revealed that members of the SLFN family are capable of altering lineage commitment during T-cell differentiation by using cell-cycle arrest as a means of translational control by RNase activity. SLFN14 has been reported as a novel gene causing an inherited macrothrombocytopenia and bleeding in human patients; however, the role of this endoribonuclease in megakaryopoiesis and thrombopoiesis remains unknown. To investigate this, we report a CRISPR knock-in mouse model of SLFN14 K208N homologous to the K219N mutation observed in our previous patient studies. We used hematological analysis, in vitro and in vivo studies of platelet and erythrocyte function, and analysis of spleen and bone marrow progenitors. Mice homozygous for this mutation do not survive to weaning age, whereas heterozygotes exhibit microcytic erythrocytosis, hemolytic anemia, splenomegaly, and abnormal thrombus formation, as revealed by intravital microscopy, although platelet function and morphology remain unchanged. We also show that there are differences in erythroid progenitors in the spleens and bone marrow of these mice, indicative of an upregulation of erythropoiesis. This SLFN14 mutation presents distinct species-specific phenotypes, with a platelet defect reported in humans and a severe microcytic erythrocytosis in mice. Thus, we conclude that SLFN14 is a key regulator in mammalian hematopoiesis and a species-specific mediator of platelet and erythroid lineage commitment.
Asunto(s)

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Plaquetas / Endorribonucleasas / Eritropoyesis Límite: Animals / Humans Idioma: En Revista: Blood Adv Año: 2021 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Plaquetas / Endorribonucleasas / Eritropoyesis Límite: Animals / Humans Idioma: En Revista: Blood Adv Año: 2021 Tipo del documento: Article