4.8 Article

A variant erythroferrone disrupts iron homeostasis in SF3B1-mutated myelodysplastic syndrome

Journal

SCIENCE TRANSLATIONAL MEDICINE
Volume 11, Issue 500, Pages -

Publisher

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/scitranslmed.aav5467

Keywords

-

Funding

  1. INSERM
  2. Institut National du Cancer INCa PLBio [INCa_9290]
  3. INCa
  4. Direction Generale de l'Offre de Soins (DGOS) of the French Ministry of Social Affairs and Health through the Programme Hospitalier de Recherche Clinique [PHRC MDS-04, INCa-DGOS_5480]
  5. Site de Recherche Integree sur le Cancer (SIRIC) CAncer Research for PErsonalized Medicine (CARPEM) - FEDER
  6. Canceropole Ile de France [ANR-16-ACHN-0002-01]
  7. European Research Council (ERC) under the European Union [715491]
  8. NIH [U54HL119893]
  9. UCLA Center for Accelerated Innovation
  10. European Research Council (ERC) [715491] Funding Source: European Research Council (ERC)
  11. Agence Nationale de la Recherche (ANR) [ANR-16-ACHN-0002] Funding Source: Agence Nationale de la Recherche (ANR)

Ask authors/readers for more resources

Myelodysplastic syndromes (MDS) with ring sideroblasts are hematopoietic stem cell disorders with erythroid dysplasia and mutations in the SF3B1 splicing factor gene. Patients with MDS with SF3B1 mutations often accumulate excessive tissue iron, even in the absence of transfusions, but the mechanisms that are responsible for their parenchymal iron overload are unknown. Body iron content, tissue distribution, and the supply of iron for erythropoiesis are controlled by the hormone hepcidin, which is regulated by erythroblasts through secretion of the erythroid hormone erythroferrone (ERFE). Here, we identified an alternative ERFE transcript in patients with MDS with the SF3B1 mutation. Induction of this ERFE transcript in primary SF3B1-mutated bone marrow erythroblasts generated a variant protein that maintained the capacity to suppress hepcidin transcription. Plasma concentrations of ERFE were higher in patients with MDS with an SF3B1 gene mutation than in patients with SF3B1 wild-type MDS. Thus, hepcidin suppression by a variant ERFE is likely responsible for the increased iron loading in patients with SF3B1-mutated MDS, suggesting that ERFE could be targeted to prevent iron-mediated toxicity. The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available