scholarly journals Combined inhibition of PI3K-related DNA damage response kinases and mTORC1 induces apoptosis in MYC-driven B-cell lymphomas

Blood ◽  
2013 ◽  
Vol 121 (15) ◽  
pp. 2964-2974 ◽  
Author(s):  
Jake Shortt ◽  
Benjamin P. Martin ◽  
Andrea Newbold ◽  
Katherine M. Hannan ◽  
Jennifer R. Devlin ◽  
...  

Key Points MYC-driven lymphomas demonstrate activation of mTORC1 and an endogenous DNA damage response. BEZ235 inhibits PI3K-related DNA damage response kinases and mTORC1, inducing p53-independent upregulation of proapoptotic BMF.

2019 ◽  
Vol 18 (7) ◽  
pp. 1255-1264 ◽  
Author(s):  
Valentina Restelli ◽  
Monica Lupi ◽  
Rosaria Chilà ◽  
Micaela Vagni ◽  
Chiara Tarantelli ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 224-224 ◽  
Author(s):  
Stella M. Ranuncolo ◽  
Jose M. Polo ◽  
Jamil Dierov ◽  
Martin Carroll ◽  
Ari M. Melnick

Abstract The BCL6 (B-Cell-Lymphoma-6) transcriptional repressor is a critical oncogene in B-cell lymphomas and is required for establishment of germinal centers (GCs) by normal B-cells. However, the mechanisms by which BCL6 licenses GC formation and lymphomagenesis remain unknown. To characterize this mechanism we identified BCL6 target genes through several methods. Most notable among these was ATR, a master DNA damage response mediator. We showed that primary BCL6-expressing GC centroblasts purified from human tonsils do not express ATR, do not activate downstream targets of ATR (Chk1) and exhibit an overall attenuated DNA damage checkpoint response (as shown in COMET assays, H2AX phosphorylation assays, and other methods). ATR expression, the activation of ChK1, and the DNA damage phenotype were fully rescued by BCL6 loss of function (induced by shRNA or by a specific BCL6 inhibitor that we designed). BCL6 expressing DLBCL cell lines exhibited the same BCL6-dependent DNA damage response phenotype. This could be attributed almost entirely to ATR since ectopic expression of ATR could restore DNA damage sensing even in the presence of BCL6. Reciprocally, loss of ATR in BCL6 depleted B-cells could completely prevent them from restoring their damage response pathways. These effects are independent of p53, which in contrast to ATR is expressed in primary centroblasts and in many primary DLBCL cases. Gain of function experiments with BCL6 in isolated human tonsilar naïve B-cells could induce this same damage phenotype in an ATR dependent/p53 independent manner. Most remarkably, we discovered that CD40 signaling, which occurs once centroblasts mature into centrocytes in the GC light zone, rescues ATR from BCL6 mediated repression by signaling to the BCL6 repression complex through NFKB. This causes the SMRT and N-CoR corepressors to be released from BCL6 repressor complexes associated with the ATR promoter, induces ATR expression, its downstream target ChK1 activation and restores DNA damage sensing. As a consequence, B-cells that have undergone extensive damage (as a by-product of antibody affinity maturation and intense proliferation) can now undergo apoptosis, while those with lower level damage undergo cell cycle arrest, DNA repair, and further differentiation. We thus described a molecular feedback loop through which BCL6 mediates GC formation by directly repressing ATR and inducing a DNA damage checkpoint deficient phenotype, and through which subsequent CD40 signaling rescues this phenotype by disrupting the BCL6 repression complex. We showed that sustained BCL6 expression leads to DNA misrepair and genomic instability in primary B cells consistent with its role in lymphomagenesis. Reciprocally, therapeutic targeting of BCL6 synergized with chemotherapy and radiation to kill DLBCL cells (by restoring DNA damage checkpoints) and would thus be expected to improve therapeutic outcomes of DLBCL patients.


Blood ◽  
2017 ◽  
Vol 130 (24) ◽  
pp. 2631-2641 ◽  
Author(s):  
Brenton G. Mar ◽  
S. Haihua Chu ◽  
Josephine D. Kahn ◽  
Andrei V. Krivtsov ◽  
Richard Koche ◽  
...  

Key Points Alterations of SETD2, a histone 3 lysine 36 trimethyl (H3K36me3) transferase leads to resistance to DNA damaging-chemotherapy in leukemia. Low H3K36me3 levels impair DNA damage response and increase mutation rate, which may be targeted by H3K36me3 demethylase inhibition.


2013 ◽  
Vol 110 (4) ◽  
pp. 1404-1409 ◽  
Author(s):  
Roy L. Maute ◽  
Christof Schneider ◽  
Pavel Sumazin ◽  
Antony Holmes ◽  
Andrea Califano ◽  
...  

Author(s):  
Theodora Manolakou ◽  
Aggelos Banos ◽  
Anastasia Filia ◽  
Antigone Pieta ◽  
Panayotis Verginis ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (12) ◽  
pp. 1473-1482 ◽  
Author(s):  
Diamantis G. Konstantinidis ◽  
Katie M. Giger ◽  
Mary Risinger ◽  
Suvarnamala Pushkaran ◽  
Ping Zhou ◽  
...  

Key Points RhoA GTPase activates pMRLC and localizes to the site of midbody formation to regulate erythroblast cytokinesis. Cytokinesis failure in erythroblasts caused by RhoA deficiency triggers p53-mediated DNA-damage response, cell-cycle arrest, and apoptosis.


2017 ◽  
Vol 108 (8) ◽  
pp. 1556-1564 ◽  
Author(s):  
Kenichi Tahara ◽  
Makiko Takizawa ◽  
Arito Yamane ◽  
Yohei Osaki ◽  
Takuma Ishizaki ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3015-3015
Author(s):  
Enrico Derenzini ◽  
Claudio Agostinelli ◽  
Ilaria Iacobucci ◽  
Enrica Imbrogno ◽  
Beatrice Casadei ◽  
...  

Abstract Introduction Genomic instability and constitutive activation of the DNA damage response (DDR) pathway has been recently described in models of aggressive myc-driven lymphoid malignancies. The MYC oncogene has been reported to induce genomic instability by a mechanism involving replication stress. On the other hand, MYC is overexpressed in a fraction of diffuse large B-cell lymphomas (DLBCLs), and its overexpression has been reported to be associated with poor prognosis. The checkpoint kinases 1 (CHK1) and 2 (CHK2), are serine-threonine kinase involved in the DDR pathway. DDR activation triggers the phosphorylation of the histone H2AX at ser 139, a known marker of DNA damage and genomic instability. The correlation between genomic instability, MYC expression, and prognosis has not been investigated yet in DLBCL. Methods Immunohistochemistry (IHC) for phospho (γ) H2AX, pCHK1, pCHK2 was performed in tissue microarrays (TMAs) from 97 consecutive patients treated at our Institution between 2004 and 2011 with R-CHOP/CHOP-like regimens, with available paraffin embedded tissue from initial diagnosis. Moreover, to evaluate the therapeutic potential of DDR pathway inhibition in DLBCL, the DLBCL cell lines HBL-1, U2932, TMD8, SUDHL-6, BJAB, SUDHL-4 and primary DLBCL cells were incubated with the CHK inhibitor PF-0477736 (Pfizer). Results In the TMA study 57% of patients (n=55) displayed high levels of basal γH2AX (>30% of positive cells), 55% (n=53) displayed pCHK1/pCHK2 activation and of note all DLBCL cell lines showed detectable baseline activation of CHK1/CHK2 and/or H2AX phosphorylation, by western immunoblotting. γH2AX positive cases distributed equally in germinal center (GC) and in non GC DLBCLs, and were significantly associated with MYC expression (p<0.01). Five-year survival rate was 70% vs 41% for γH2AX-low and γH2AX-high patients respectively (p=0.01). Factors significantly related to the outcome in multivariate analysis were International Prognostic Index (IPI) score and γH2AX expression. Remarkably the prognostic significance of γH2AX was particularly evident in the low risk IPI group (0-2 risk factors), identifying a subgroup characterized by worse outcome (54% 5-year OS). In the in vitro study a significant growth inhibition (WST-1 assay), was evident after 48 hrs in all cell lines (IC50 10-230 nM). PF-0477736 25-500 nM induced cell death by apoptosis (annexin V- propidium iodide staining) in a time and dose dependent manner. Notably PF-0477736 demonstrated activity also in primary DLBCL cells (IC 50 of 50-500 nM, 24 hrs). We observed inhibition of phosphorylation of the downstream target CDC25c (ser 216), coupled with a marked increase in γH2AX ser 139 and CHK1 phosphorylation (ser317 and 345) following treatment. Conclusions A significant fraction of DLBCLs shows high levels of inherent genomic instability; the DDR activation marker γH2AX is a poor prognostic predictor in DLBCL and interestingly is significantly associated with MYC expression. DDR inhibition resulted to be highly effective in DLBCL cell lines and primary DLBCL cells; on treatment modifications of CHK1 and H2AX phosphorylation could be useful biomarkers of CHK inhibitors activity. These data provide strong rationale for targeting the DDR pathway and for clinical investigation of CHK inhibitors in DLBCL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2279-2279
Author(s):  
Hakim Bouamar ◽  
Long Wang ◽  
Manoela Ortega ◽  
An-Ping Lin ◽  
Daifeng Jiang ◽  
...  

Abstract The germinal center (GC) reaction includes two critical events: somatic hypermutation (SHM) and class switch DNA recombination (CSR). These processes are tightly controlled, thus preventing excessive DNA injury which could lead to loss of normal B lymphocytes as well as the survival of cells with oncogenic DNA lesions. MicroRNA-155 (miR-155) plays an important role in immune cell biology; miR-155 knock-out (KO) mice have a defective mature B cell development characterized by a decreased number of GC B cells, whereas the Eμ-miR-155 transgenic mouse model develops and oligoclonal proliferation which evolves to B cell lymphoma. These observations suggest that miR-155 may regulate B cell sensitivity and response to DNA damage, which could mechanistically explain the phenotypes observed in the gain and loss-of-function animal models. To examine this concept, we purified mature B cells from multiple pairs of miR-155 KO and WT littermates, and stimulated them with LPS and IL4 thus recapitulating the GC reaction in vitro. Next, we used immunohistochemistry to quantify γH2AX at the DNA double-strand breaks (DSBs) foci, western blot to evaluate the phospho-p53 (Ser15) levels, and real-time RT-PCR to quantify the expression of p53 target genes (p21, GADD45a, cdc25c, PCNA). In these assays, we found a significantly higher γH2AX staining in B cells null for miR-155 than in WT controls (number of foci per nucleus: 4.1±0.9 vs. 1.4±0.2, p<0.01), and a markedly elevated p53 activity, defined by its phospho-level and target genes expression. To confirm these observations in a more physiologic setting, we immunized miR-155 WT and KO mice with NP-CGG, collected spleens and purified mature B-cells. Quantification of subpopulation by FACS confirmed that miR-155 mice developed fewer GC B cells, and the examination of DSBs foci and p53 target genes expression supported our in vitro observation of a heightened sensitivity to DNA damage and p53 engagement in miR-155 null cells. We concluded that miR-155 deficiency in B cells associates with excessive DNA damage and p53 activity. To further isolate define the role of miR-155 in these events, we used a retrovirus system to rescue miR-155 expression in B cells derived from the KO mouse, and defined the pattern of DNA damage response. In these assays, cells transduced with a MSCV-miR-155 construct formed fewer DSBs foci than their control counterparts transduced with an empty vector (9.0±2.1 vs. 4.9±1.8, p<0.05) and showed significantly lower p53 activity, defined by target gene expression. To test if miR-155 controls the DNA damage induced by broad genotoxic stresses, we exposed miR-155 KO or WT B cells, thymocytes and bone marrow cells to 5Gy of ionizing radiation (IR) or etoposide (4µM). In these assays, albeit a robust induction of γH2AX foci formation and p53 activation were detected, there was no significant difference between WT and miR-155 KO mice, in any of the cell types analyzed. We concluded that the heightened sensitivity to genotoxic stress in miR-155 KO mice is specific to B cells undergoing the GC reaction. This observation suggests a potential role in this process for activation-induced cytidine deaminase (AID), a key regulator of the DNA damage inducing SHM and CSR, which is also a miR-155 target gene. We confirmed that AID expression is higher in miR-155 KO than in WT B cells, and to test its contribution to the excessive DSB and p53 activity found miR-155 null cells we used an RNAi approach. We found that the inhibition of AID levels in miR-155 KO B cells significantly reduced γH2AX foci formation and expression of p53 target genes. Together, our data highlight a hitherto unappreciated interaction between miR-155 and the p53 pathway, involving DNA lesions that are germane to the GC reaction and the control of AID expression/activity. The excessive engagement of p53 is this setting may explain, at least in part, the loss of normal GC B cells found in the miR-155 KO mice. Conversely, it is possible that cells expressing abnormally high miR-155 levels show a subpar activation of the DNA damage response thus leading to the accumulation of oncogenic mutations. This paradigm would provide a mechanistic explanation for the initial poly/oligoclonal proliferation reported in the Eμ-miR-155 mice, which eventually evolves into a B-cell lymphoma Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document