Skip to main content

Advertisement

Log in

Barrett’s esophagus: A molecular perspective

  • Published:
Current Gastroenterology Reports Aims and scope Submit manuscript

Abstract

Carcinogenesis in Barrett’s esophagus involves the accumulation of DNA abnormalities that enable cells to 1) provide their own growth signals; 2) ignore growthinhibitory signals; 3) avoid apoptosis; 4) replicate without limit; 5) sustain angiogenesis; and 6) invade and proliferate in unnatural locations. This report reviews recent publications describing molecular abnormalities in Barrett’s esophagus that could lead to the acquisition of these key physiologic hallmarks of malignancy. Some recent reports suggest that the gastroesophageal reflux of acid and bile can activate molecular pathways that promote proliferation and interfere with apoptosis in Barrett’s metaplastic cells. Inactivation of the p16 and p53 tumor suppressor genes through promoter methylation, gene mutation, or loss of heterozygosity appears to be important for carcinogenesis in Barrett’s esophagus. Finally, this report discusses recent data regarding the role of the Cdx2 gene in the development of esophageal intestinal metaplasia.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References and Recommended Reading

  1. Spechler SJ: Barrett’s esophagus. N Engl J Med 2002, 346:836–842.

    Article  PubMed  Google Scholar 

  2. Blot WJ, McLaughlin JK: The changing epidemiology of esophageal cancer. Semin Oncol 1999, 26:2–8.

    PubMed  CAS  Google Scholar 

  3. Hanahan D, Weinberg RA: The hallmarks of cancer. Cell 2000, 100:57–70.

    Article  PubMed  CAS  Google Scholar 

  4. Morales CP, Souza RF, Spechler SJ: Hallmarks of cancer progression in Barrett’s oesophagus. Lancet 2002, 360:1587–1589.

    Article  PubMed  Google Scholar 

  5. Campbell SL, Khosravi-Far R, Rossman KL, et al.: Increasing complexity of Ras signaling. Oncogene 1998, 17:1395–1413.

    Article  PubMed  CAS  Google Scholar 

  6. Karin M: The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem 1995, 270:16483–16486.

    PubMed  CAS  Google Scholar 

  7. Souza RF, Shewmake K, Terada LS, Spechler SJ: Acid exposure activates the mitogen activated protein kinase pathways in Barrett’s esophagus. Gastroenterology 2002, 122:299–307.

    Article  PubMed  CAS  Google Scholar 

  8. Souza RF, Shewmake K, Beer DG, et al.: Selective inhibition of cyclooxygenase-2 suppresses growth and induces apoptosis in human esophageal adenocarcinoma cells. Cancer Res 2000, 60:5767–5772.

    PubMed  CAS  Google Scholar 

  9. Souza RF, Shewmake K, Pearson S, et al.: Acid increases proliferation via ERK and p38 MAPK-mediated increases in cyclooxygenase-2 in Barrett’s adenocarcinoma cells. Am J Physiol 2004, 287:G743-G748. In a Barrett’s adenocarcinoma cell line, brief acid exposure caused a 2.8-fold increase in COX-2 mRNA levels, an effect that could be attenuated by treatment with specific MAPK inhibitors. These observations indicate that transient acidification increases COX-2 expression in Barrett’s adenocarcinoma cells through activation of the MAPK pathways.

    CAS  Google Scholar 

  10. Sommerer F, Vieth M, Markwarth A, et al.: Mutations of BRAF and KRAS2 in the development of Barrett’s adenocarcinoma. Oncogene 2004, 23:554–558. K-Ras mutations were found in four of 19 (21%) Barrett’s adenocarcinomas and in three of 27 (11%) specimens with high-grade dysplasia, whereas activating B-Raf mutations were found in two of 19 (11%) Barrett’s adenocarcinomas and in one of 27 specimens (4%) with high-grade dysplasia. No specimen exhibited mutations in both K-Ras and B-Raf. It appears that mutations in these molecules, which are activators of the MAPK pathways, occur commonly during carcinogenesis in Barrett’s esophagus.

    Article  PubMed  CAS  Google Scholar 

  11. Haigh CR, Attwood SE, Thompson DG, et al.: Gastrin induces proliferation in Barrett’s metaplasia through activation of the CCK2 receptor. Gastroenterology 2003, 124:615–625.

    Article  PubMed  CAS  Google Scholar 

  12. Todisco A, Ramamoorthy S, Witham T, et al.: Molecular mechanisms for the antiapoptotic action of gastrin. Am J Physiol Gastrointest Liver Physiol 2001, 280:G298-G307.

    PubMed  CAS  Google Scholar 

  13. Harris JC, Clarke PA, Awan A, et al.: An antiapoptotic role for gastrin and the gastrin/CCK-2 receptor in Barrett’s esophagus. Cancer Res 2004, 64:1915–1919.

    Article  PubMed  CAS  Google Scholar 

  14. El-Serag H, Aguirre TV, Davis S, et al.: Proton pump inhibitors are associated with reduced incidence of dysplasia in Barrett’s esophagus. Am J Gastroenterol 2004, 99:1877–1883.

    Article  PubMed  CAS  Google Scholar 

  15. Jaiswal K, Tello V, Lopez-Guzman C, et al.: Bile salt exposure causes phosphatidylinositol-3-kinase-mediated proliferation in a Barrett’s adenocarcinoma cell line. Surgery 2004, 136:160–168. Barrett’s adenocarcinoma cells exposed to a conjugated bile salt exhibited a dose-dependent increase in cell number, an effect that could be blunted by treatment with a PI3 kinase inhibitor. These findings suggest that bile reflux might activate the PI3 kinase/Akt pathway in Barrett’s esophagus to increase proliferation.

    Article  PubMed  Google Scholar 

  16. Stoler DL, Chen N, Basik M, et al.: The onset and extent of genomic instability in sporadic colorectal tumor progression. Proc Natl Acad Sci U S A 1999, 96:15121–15126.

    Article  PubMed  CAS  Google Scholar 

  17. Maley CC, Galipeau PC, Li X, et al.: advantageous mutations and hitchhikers in neoplasms: p16 lesions are selected in Barrett’s esophagus. Cancer Res 2004, 64:3414–3427. Esophageal biopsy specimens from 211 patients with Barrett’s esophagus were assayed for a number of DNA abnormalities, and the investigators used statistical techniques to separate advantageous mutations from neutral mutations ("hitchhikers"). Inactivation of the p16 tumor suppressor gene was found to be the most likely advantageous lesion and, after at least one p16 gene had been inactivated, a lesion in the p53 tumor suppressor gene also appeared to be advantageous. All other genetic lesions evaluated appeared to be hitchhikers carried along by the p16 and p53 abnormalities.

    Article  PubMed  CAS  Google Scholar 

  18. Wong DJ, Paulson TG, Prevo LJ, et al.: p16(INK4a) lesions are common, early abnormalities that undergo clonal expansion in Barrett’s metaplastic epithelium. Cancer Res 2001, 61:8284–8289.

    PubMed  CAS  Google Scholar 

  19. Maley CC, Galipeau PC, Li X, et al.: The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res 2004, 64:7629–7633.

    Article  PubMed  CAS  Google Scholar 

  20. Shay JW, Bacchetti S: A survey of telomerase activity in human cancer. Eur J Cancer 1997, 33:787–791.

    Article  PubMed  CAS  Google Scholar 

  21. Morales CP, Lee EL, Shay JW: In situ hybridization for the detection of telomerase RNA in the progression fryom Barrett’s esophagus to esophageal adenocarcinoma. Cancer 1998, 83:652–659.

    Article  PubMed  CAS  Google Scholar 

  22. Shammas MA, Koley H, Beer DG, et al.: Growth arrest, apoptosis, and telomere shortening of Barrett’s-associated adenocarcinoma cells by a telomerase inhibitor. Gastroenterology 2004, 126:1337–1346. The investigators explored the effects of a telomerase inhibitor (PPA) on Barrett’s adenocarcinoma cells. Treatment with PPA resulted in the loss of telomerase activity, telomere shortening, and replicative arrest. These findings suggest a potential therapeutic role for telomerase inhibitors in patients who have cancer in Barrett’s esophagus.

    Article  PubMed  CAS  Google Scholar 

  23. Couvelard A, Paraf F, Gratio V, et al.: Angiogenesis in the neoplastic sequence of Barrett’s esophagus: correlation with VEGF expression. J Pathol 2000, 192:14–18.

    Article  PubMed  CAS  Google Scholar 

  24. Auvinen MI, Sihvo EIT, Ruohtula T, et al.: Incipient angiogenesis in Barrett’s epithelium and lymphangiogenesis in Barrett’s adenocarcinoma. J Clin Oncol 2002, 20:2971–2979.

    Article  PubMed  Google Scholar 

  25. Möbius C, Stein HJ, Becker I, et al.: Vascular endothelial growth factor expression and neovascularization in Barrett’s carcinoma. World J Surg 2004, 28:675–679.

    Article  PubMed  Google Scholar 

  26. Beck F: The role of Cdx genes in the mammalian gut. Gut 2004, 53:1394–1396.

    Article  PubMed  CAS  Google Scholar 

  27. Phillips RW, Frierson HF, Moskaluk CA: Cdx2 as a marker of epithelial intestinal differentiation in the esophagus. Am J Surg Pathol 2003, 27:1442–1447. Immunostaining for Cdx2 was found in all of 34 specimens of specialized intestinal metaplasia without dysplasia, in all of 32 specimens of specialized intestinal metaplasia with dysplasia, and in all of six specimens with adenocarcinoma. In contrast, Cdx2 was detected in only 20 of 62 (30%) specimens of junctional-type epithelium (which contains no goblet cells). These findings suggest that Cdx2 immunostaining is a sensitive marker for specialized intestinal metaplasia in Barrett’s esophagus, and that Cdx2 immunostaining may be helpful in establishing the diagnosis of the condition in histologically equivocal cases.

    Article  PubMed  Google Scholar 

  28. Moons LMG, Bax DA, Kuipers EJ, et al.: The homeodomain protein CDX2 is an early marker of Barrett’s oesophagus. J Clin Pathol 2004, 57:1063–1068.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Spechler, S.J. Barrett’s esophagus: A molecular perspective. Curr Gastroenterol Rep 7, 177–181 (2005). https://doi.org/10.1007/s11894-005-0031-z

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11894-005-0031-z

Keywords

Navigation