4.7 Article

miR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2

Journal

JCI INSIGHT
Volume 5, Issue 1, Pages -

Publisher

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/jci.insight.121781

Keywords

-

Funding

  1. NIH/National Center for Advancing Translational Sciences [UH3TR00943-01]
  2. NIH/National Cancer Institute (NCI) [1 R01 CA182905-01]
  3. Team Department of Defense grant [CA160445P1]
  4. Ladies Leukemia League grant
  5. Chronic Lymphocytic Leukemia Moonshot Flagship project
  6. Sister Institution Network Fund 2017 grant
  7. Estate of C. G. Johnson, Jr
  8. NCI [P50 CA140388]
  9. NIH Clinical Research Loan Repayment Program
  10. Fundacao para a Ciencia e a Tecnologia [SFRH/BD/85968/2012]
  11. MPN Foundation
  12. Oklahoma Center for the Advancement of Science and Technology
  13. Programul Operational Competitivitate grant [35/01.09.2016, 37_796]
  14. Fundacao de Amparo a Pesquisa do Estado de Sao Paulo [BEPE 2016/09349-4]
  15. U54 grant UPR/MDACC Partnership for Excellence in Cancer Research 2016 Pilot Project
  16. Fundação para a Ciência e a Tecnologia [SFRH/BD/85968/2012] Funding Source: FCT

Ask authors/readers for more resources

Myelofibros is (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAK(V617F) mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK(V617F) inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-S43 was significantly upregulated in non responders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2(V617F) mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available