Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent …

O Gilad, BY Nabet, RL Ragland, DW Schoppy… - Cancer research, 2010 - AACR
O Gilad, BY Nabet, RL Ragland, DW Schoppy, KD Smith, AC Durham, EJ Brown
Cancer research, 2010AACR
Previous studies indicate that oncogenic stress activates the ATR-Chk1 pathway. Here, we
show that ATR-Chk1 pathway engagement is essential for limiting genomic instability
following oncogenic Ras transformation. ATR pathway inhibition in combination with
oncogenic Ras expression synergistically increased genomic instability, as quantified by
chromatid breaks, sister chromatid exchanges, and H2AX phosphorylation. This level of
instability was significantly greater than that observed following ATR suppression in …
Abstract
Previous studies indicate that oncogenic stress activates the ATR-Chk1 pathway. Here, we show that ATR-Chk1 pathway engagement is essential for limiting genomic instability following oncogenic Ras transformation. ATR pathway inhibition in combination with oncogenic Ras expression synergistically increased genomic instability, as quantified by chromatid breaks, sister chromatid exchanges, and H2AX phosphorylation. This level of instability was significantly greater than that observed following ATR suppression in untransformed control cells. In addition, consistent with a deficiency in long-term genome maintenance, hypomorphic ATR pathway reduction to 16% of normal levels was synthetic lethal with oncogenic Ras expression in cultured cells. Notably, elevated genomic instability and synthetic lethality following suppression of ATR were not due to accelerated cycling rates in Ras-transformed cells, indicating that these synergistic effects were generated on a per-cell-cycle basis. In contrast to the synthetic lethal effects of hypomorphic ATR suppression, subtle reduction of ATR expression (haploinsufficiency) in combination with endogenous levels of K-rasG12D expression elevated the incidence of lung adenocarcinoma, spindle cell sarcoma, and thymic lymphoma in p53 heterozygous mice. K-rasG12D-induced tumorigenesis in ATR+/−p53+/− mice was associated with intrachromosomal deletions and loss of wild-type p53. These findings indicate that synergistic increases in genomic instability following ATR reduction in oncogenic Ras-transformed cells can produce 2 distinct biological outcomes: synthetic lethality upon significant suppression of ATR expression and tumor promotion in the context of ATR haploinsufficiency. These results highlight the importance of the ATR pathway both as a barrier to malignant progression and as a potential target for cancer treatment. Cancer Res; 70(23); 9693–702. ©2010 AACR.
AACR