Abstract

Background: The alteration of hematopoiesis in MDS patients is deeply associated with microenvironment alterations, in particular in MSCs. Stromal cells are epigenetically reprogrammed to function in cooperation with leukemic cells and propagate the disease as a “tumor unit”. Notably, dysfunctions of MDS-MSCs persist following expansion ex vivo suggesting a hereditable epigenetic dysregulation which endures despite removal of disease-associated microenvironment factors. Aims: Here, we investigated the role of histone H2A variant MacroH2A1 (mH2A1) in promoting pro-tumorigenic inflammation and metabolic reprogramming of MSCs. Methods: MSCs were collected from BM of MDS patients (n=18) and matched healthy controls (n=8; HC). HS-5 cells were used as model of healthy MSCs. Real time, western blot, immunohistochemistry and immunofluorescence were used. Proteomic and metabolic analysis were performed. Results: MDS patients had significant higher signature of mH2A1 in bone marrow slides compared to age-matched control. Investigating mH2A1 expression in stromal cells, MDS-MSCs had increased levels of expression in respect of HC-MSCs. In accordance with the aberrant inflammation described in MDS microenvironment, MDS-MSCs showed higher levels of the sum of NO2+ and NO3+ associated to increased GSH and NADP+/NAPH. TLR4 also was upregulated and positively correlated to mH2A1 expression. To better investigate the relationship between mH2A1 and TLR4 in MDS-MSCs, we induced mH2A1 overexpression in HS-5 cells (mH2A1-OE) by mH2A1-CT-MYC plasmid. Our data showed that mH2A1-OE upregulated TLR4 and increased NFkB nuclear translocation compared to cells transfected with empty vector (CTL). Proteomic analysis confirmed upregulation of intracellular serine protease inhibitors (SerpinB2, B8, B6) strongly induced during inflammation and important for the maintenance of TLR4 activation. Moreover, proteomic approach identified upregulation of several proteins associated to hypermethylation of DNA and histones in mH2A1-OE. In particular, S-adenosylhomocysteine hydrolase which regulates the concentration of S-adenosylhomocysteine (SAH), a strong inhibitor of methyltransferase reactions and of the methyl donor S-adenosyl-methionine (SAM), resulted overexpressed. HPLC analysis showed higher SAM/SAH ratio associated to a significant reduction of SAH in mH2A1-OE, confirming the increase of the methylation index. In addition, the higher levels of CBX3, a suppressive epigenetic mark which recognizes histone H3K9me3, contributes to the maintenance of the heterochromatin. The higher levels of H3K9me3 in mh2A1-OE were confirmed by western blot analysis. Overexpression of mH2A1 also induced metabolic reprogramming with the acquisition of a more glycolytic metabolism characterized by decreased levels of NAD+/NADH, upregulation of LDHA and MCT4. As LDHA could translocate into the nucleus, we evaluated if it used a noncanonical enzymatic activity in mh2A1-OE. Data showed increased nuclear localization of LDHA producing histone H3K79 hypermethylation. Nuclear LDHA was also observed in MDS-MSC. Finally, we treated ex-vivo HC- and MDS-MSCs with azacytidine founding a significant reduction both of mH2A1 and TLR4 associated to lower levels of nuclear NFKB. Summary/Conclusion: Our data provide a key role of mH2A1in driving the crosstalk between inflammation, metabolism and epigenetic signatures in MDS-MSC. Future experiments will define the contribution of mH2A to ineffective hematopoiesis and leukemic evolution.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call