Oxidative DNA Damage as a Potential Early Biomarker of Helicobacter pylori Associated Carcinogenesis

2014 ◽  
Vol 20 (4) ◽  
pp. 839-846 ◽  
Author(s):  
Yasir Raza ◽  
Adnan Khan ◽  
Amber Farooqui ◽  
Muhammad Mubarak ◽  
Alex Facista ◽  
...  
2005 ◽  
Vol 58 (1) ◽  
pp. 166-176 ◽  
Author(s):  
Ge Wang ◽  
Praveen Alamuri ◽  
M. Zafri Humayun ◽  
Diane E. Taylor ◽  
Robert J. Maier

2015 ◽  
Vol 83 (7) ◽  
pp. 2935-2943 ◽  
Author(s):  
Lisa G. Kuhns ◽  
Ge Wang ◽  
Robert J. Maier

Thioredoxins are highly conserved throughout a wide range of organisms, and they are essential for the isurvival of oxygen-sensitive cells. The gastric pathogenHelicobacter pyloriuses the thioredoxin system to maintain its thiol/disulfide balance. There are two thioredoxins present inH. pylori, Trx1 and Trx2 (herein referred to as TrxA and TrxC). TrxA has been shown to be important as an electron donor for some antioxidant enzymes, but the function of TrxC remains unknown (L. M. Baker, A. Raudonikiene, P. S. Hoffman, and L. B. Poole, J Bacteriol 183:1961–1973, 2001; P. Alamuri and R. J. Maier, J Bacteriol 188:5839–5850, 2006). We demonstrate that both TrxA and TrxC are important in protectingH. pylorifrom oxidative stress. Individual ΔtrxAand ΔtrxCdeletion mutant strains each show a greater abundance of lipid peroxides and suffer more DNA damage and more protein carbonylation than the parent. Both deletion mutants were much more sensitive to O2-mediated viability loss than the parent. Unexpectedly, the oxidative DNA damage and protein carbonylation was more severe in the ΔtrxCmutant than in the ΔtrxAmutant; it had 20-fold- and 4-fold-more carbonylated protein content than the wild type and the ΔtrxAstrain, respectively, after 4 h of atmospheric O2stress.trxtranscript abundance was altered by the deletion of the heterologoustrxgene. The ΔtrxCmutant lacked mouse colonization ability, while the ability to colonize mouse stomachs was significantly reduced in the ΔtrxAmutant.


2001 ◽  
Vol 120 (5) ◽  
pp. A656-A657
Author(s):  
Alfredo Papa ◽  
Silvio Danese ◽  
Alessandro Sgambato ◽  
Raffaele Ardito ◽  
Antonino Grillo ◽  
...  

2017 ◽  
Vol 06 (02) ◽  
Author(s):  
Doaa Waseem Nada ◽  
Amal Mohamad El Barbary ◽  
Sally Abd El monem El Nawasany ◽  
Rasha Ahmed Gaber ◽  
Radwa Mahmoud El sharaby ◽  
...  

DNA Repair ◽  
2007 ◽  
Vol 6 (1) ◽  
pp. 19-26 ◽  
Author(s):  
Rory Eutsey ◽  
Ge Wang ◽  
Robert J. Maier

2007 ◽  
Vol 53 (1) ◽  
pp. 248-255 ◽  
Author(s):  
Marcelo S. P. Ladeira ◽  
Roberta C. A Bueno ◽  
Bruna Fornazari dos Santos ◽  
Carla L. S. Pinto ◽  
Renato P. Prado ◽  
...  

2020 ◽  
Vol 295 (32) ◽  
pp. 11082-11098 ◽  
Author(s):  
Ibrahim M. Sayed ◽  
Ayse Z. Sahan ◽  
Tatiana Venkova ◽  
Anirban Chakraborty ◽  
Dibyabrata Mukhopadhyay ◽  
...  

Infection with the Gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and oxidative DNA damage in gastric epithelial cells that can lead to gastric cancer (GC). However, the underlying pathogenic mechanism is largely unclear. Here, we report that the suppression of Nei-like DNA glycosylase 2 (NEIL2), a mammalian DNA glycosylase that specifically removes oxidized bases, is one mechanism through which H. pylori infection may fuel the accumulation of DNA damage leading to GC. Using cultured cell lines, gastric biopsy specimens, primary cells, and human enteroid-derived monolayers from healthy human stomach, we show that H. pylori infection greatly reduces NEIL2 expression. The H. pylori infection-induced downregulation of NEIL2 was specific, as Campylobacter jejuni had no such effect. Using gastric organoids isolated from the murine stomach in coculture experiments with live bacteria mimicking the infected stomach lining, we found that H. pylori infection is associated with the production of various inflammatory cytokines. This response was more pronounced in Neil2 knockout (KO) mouse cells than in WT cells, suggesting that NEIL2 suppresses inflammation under physiological conditions. Notably, the H. pylori-infected Neil2-KO murine stomach exhibited more DNA damage than the WT. Furthermore, H. pylori-infected Neil2-KO mice had greater inflammation and more epithelial cell damage. Computational analysis of gene expression profiles of DNA glycosylases in gastric specimens linked the reduced Neil2 level to GC progression. Our results suggest that NEIL2 downregulation is a plausible mechanism by which H. pylori infection impairs DNA damage repair, amplifies the inflammatory response, and initiates GC.


2000 ◽  
Vol 118 (4) ◽  
pp. A182
Author(s):  
Romilda Cardin ◽  
Valentina Russo ◽  
Graziella Busatto ◽  
Massimo Rugge ◽  
Fabio Farinati

Sign in / Sign up

Export Citation Format

Share Document