Epigenetic silencing of tumor-suppressor genes has potential therapeutic relevance if the diagnostic and prognostic utility of particular DNA methylation patterns could be established. SHP1 is a tumor-suppressor gene that can be downregulated in various neoplasms. The aims of this study are (i) whether SHP1 was lost/ suppressed in diffuse large B cell lymphoma (DLBCL) (ii) Understanding the role of various epigenetic mechanisms including CpG hypermethylation and histone modifications in SHP1 loss, and (iii) whether SHP1 in DLBCL can be re-expressed with epigenetic therapy.

We initially analyzed SHP1 mRNA expression in DLBCL (n=9) patient specimens and normal B-cells by QRT-PCR. Decreased expression of SHP1 mRNA was observed in all the DLBCL patient samples as compared to normal B cells. DLBCL tumor samples (n=37) along with normal tonsils (n=10) were stained for SHP1 protein by immunohistochemistry (IHC). SHP1 expression was lost or suppressed in 53% of DLBCL tumors compared to normal controls. These data, when taken together, confirm that SHP1 is strongly expressed in normal B-cells and can be lost or suppressed in DLBCL tumors. We performed epigenetic studies of SHP1 CpG promoter 2 (named CPG1 P2) in 37 DLBCL tumors and 6 DLBCL cell lines by methylation specific (MSP1) PCR and pyrosequencing methods. Surprisingly, SHP1 CpG1 P2 hypermethylation (0/37) was not detected in patient samples or cell lines by both methods. Our data conclusively demonstrate that SHP1 CpG1 P2 hypermethylation is absent in DLBCL tumors. The finding that SHP1 was lost or silenced in many of the clinical samples without finding any CpG1 P2 hypermethylation led us to extend our methylation studies to a novel, more proximal CpG island within P2 (named CpG2) that has not been previously studied in any hematological malignancies. We designed the primers specific for these CpG2 sites and ran the pyrosequencing on the same 37 DLBCL tumors as used above for CpG1 methylation. Using a cut off of >25% (intermediate to high methylation) 57% (21/37) of patient samples were methylated. The Ly3, Ly10, and DHL2 DLBCL cell lines were also hypermethylated at CpG2. To confirm the role of CpG2 methylation in SHP1 expression, we treated the Ly3 and DHL2 cells with the DNA methyltransferase inhibitor, 5-Azacytidine (Aza). We observed increased SHP1 expression compared to untreated control cells in Ly3 and DHL2 cells followed by a progressive demethylation of SHP1 CpG2, as shown by positive U-MSP with increasing amplification intensity. This resulted in a corresponding down-regulation of activated STAT3 in a similar time dependent manner. Treatment of Ly3 and DHL2 cells with 5-Aza for 0-6 days significantly (p=0.0008 for Ly3 and p=0.03 for DHL2) reduced the cell survival (40% decrease at day 6). To further investigate epigenetic control of SHP1 in DLBCL, we proceeded to study whether histone modifications in the SHP1 promoter were involved in the regulation of SHP1 expression. Chromatin immunoprecipitation assays were performed in the Ly3, Raji cell line and normal B cells, by using antibodies to repressing marks on histones. We observed that the SHP1 promoter in the Ly3 and Raji cells was enriched for both H3K9me3 and H3K27me3 repressing marks as compared to normal B cells. To confirm the role of histone modifications in SHP1 expression, we treated Ly3 cells with the histone methyltransferase inhibitor (HMT) DZNEP and the HDAC inhibitor LBH589 (Novartis Pharmaceuticals). DZNEP has been shown to be very specific for histone suppressive marks H3K27me3 that are mediated through EZH2. Treatment with DZNEP partially reverted the H3K27me3 histone marks in the SHP1 promoter followed by increased the SHP1 expression in Ly3 cells. LBH589 treatment was not able to inhibit H3K27me3 marks, but was able to inhibit the CpG2 methylation within the SHP1 promoter 2 alone or in combination with 5- Aza.

In conclusion, these data provide a comprehensive characterization of the epigenetic mechanisms leading to SHP1 deregulation in the DLBCL tumors. These findings support the notion that control of SHP1 P2 in DLBCL tumors is associated with both DNA promoter methylation and histone methylation, which explains our results that treatment with 5-Aza, DZNEP and LBH589 could reactivate SHP1 expression and inhibit DLBCL cell survival. Therefore, SHP1 may prove useful diagnostic and prognostic marker and provide a rationale to use epigenetic therapy for patients with DLBCL.

Disclosures:

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution