首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Evidence for DNA Damage Checkpoint Activation in Barrett Esophagus
Authors:Urs von Holzen  Tina Chen  Amelie Boquoi  Joel E Richter  Gary W Falk  Andres J Klein-Szanto  Harry Cooper  Sam Litwin  David S Weinberg  Greg H Enders
Institution:2. Epigenetics and Progenitor Cell Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;3. Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;4. Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA;5. Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH 44195, USA
Abstract:Barrett esophagus is an epithelial metaplasia that predisposes to adenocarcinoma. Better markers of cancer risk are urgently needed to identify those patients who are likely to benefit most from emerging methods of endoscopic ablation. Disease progression is associated with genomic DNA changes (segmental gains, losses, or loss of heterozygosity). Although these changes are not easily assayed directly, we hypothesized that the underlying DNA damage should activate a DNA damage response (DDR), detectable by immunohistochemical (IHC) assays of checkpoint proteins and the resulting replicative phase cell cycle delays. Surgical specimens and endoscopic biopsies (N = 28) were subjected to IHC for the cell cycle markers cyclin A and phosphorylated histone H3 (P-H3), the DDR markers γH2AX and phosphorylated ATM/ATR substrates (P-ATM/ATRsub), and the DNA damage-responsive tumor suppressors p16 and p53. Correlations were made with histologic diagnoses. The fractions of cells that stained for cyclin A, P-H3, and γH2AX increased in parallel in dysplastic tissue, consistent with checkpoint-mediated cell cycle delays. Foci of nuclear γH2AX and P-ATM/ATRsub were demonstrated by standard and confocal immunofluorescence. Staining for p16 was more prevalent in early-stage disease with lower staining for γH2AX and P-H3. Staining for p53 was moderately increased in some early-stage disease and strongly increased in some advanced disease, consistent with checkpoint-mediated induction and mutational inactivation of p53, respectively. We suggest that IHC for DDR-associated markers may help stratify risk of disease progression in Barrett.
Keywords:
本文献已被 ScienceDirect 等数据库收录!
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号