Skip to main content
Log in

Zelluläre Seneszenz im kolorektalen Karzinom

Cellular senescence and colorectal cancer

  • Nachwuchsakademie
  • Published:
Der Pathologe Aims and scope Submit manuscript

Zusammenfassung

Zelluläre Seneszenz ist neben Proliferation und Zelltod eine wichtige Reaktion auf stressassoziierte Stimuli. Ursprünglich als In-vitro-Phänomen beschrieben, ist sie an vielen physiologischen und pathophysiologischen Prozessen beteiligt. Im Rahmen der Entstehung solider und generalisierter Tumorerkrankungen stellt Seneszenzinduktion beispielsweise eine wichtige Barriere gegen die Progression der Krankheit dar. Dies konnte für maligne Lymphome, Melanome und verschiedene Karzinome im Tiermodell demonstriert werden. Doch seneszente Zellen bleiben hochgradig sekretorisch aktiv und haben einen wichtigen Effekt auf ihre Nachbarzellen sowie den gesamten Gewebeverband. Im vorliegenden Artikel soll ein Einblick in die aktuelle Studienlage gegeben und es sollen klinische Implikationen und zukünftige Anwendungsmöglichkeiten diskutiert werden.

Abstract

Apart from proliferation and cell death, cellular senescence is an important response to numerous stress-associated stimuli. Originally described as an in vitro phenomenon, it is involved in various physiological and pathophysiological processes. For example, during the development of solid and generalized tumors, senescence induction poses an important barrier against disease progression. This could be demonstrated for malignant lymphomas, melanomas and various carcinomas using sophisticated animal models. However, senescent cells remain highly secretorically active and have a profound effect on neighboring cells as well as the entire tissue network. This article tries to provide insight into the current literature and discusses clinical implications and future applications.

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.

Abb. 1
Abb. 2
Abb. 3

Literatur

  1. Baker DJ, Childs BG, Durik M et al (2016) Naturally occurring p16 Ink4a-positive cells shorten healthy lifespan. Nature. https://doi.org/10.1038/nature16932

    Google Scholar 

  2. Baker DJ, Wijshake T, Tchkonia T et al (2011) Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479:232–236. https://doi.org/10.1038/nature10600

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bartkova J, Rezaei N, Liontos M et al (2006) Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444:633–637. https://doi.org/10.1038/nature05268

    Article  CAS  PubMed  Google Scholar 

  4. Bennecke M, Kriegl L, Bajbouj M et al (2010) Ink4a/Arf and oncogene-induced senescence prevent tumor progression during alternative colorectal tumorigenesis. Cancer Cell 18:135–146

    Article  CAS  PubMed  Google Scholar 

  5. Campisi J, d’Adda di Fagagna F (2007) Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol 8:729–740. https://doi.org/10.1038/nrm2233

    Article  CAS  PubMed  Google Scholar 

  6. Childs BG, Baker DJ, Wijshake T et al (2016) Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354:472–477. https://doi.org/10.1126/science.aaf6659

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cho KR, Vogelstein B (1992) Genetic alterations in the adenoma – carcinoma sequence. Cancer 70:1727–1731

    Article  CAS  PubMed  Google Scholar 

  8. Cipriano R, Kan CE, Graham J et al (2011) TGF-beta signaling engages an ATM-CHK2-p53-independent RAS-induced senescence and prevents malignant transformation in human mammary epithelial cells. Proc Natl Acad Sci U S A 108:8668–8673. https://doi.org/10.1073/pnas.1015022108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Collado M, Serrano M (2006) The power and the promise of oncogene-induced senescence markers. Nat Rev Cancer 6:472–476. https://doi.org/10.1038/nrc1884

    Article  CAS  PubMed  Google Scholar 

  10. Collado M, Serrano M (2010) Senescence in tumours: evidence from mice and humans. Nat Rev Cancer 10:51–57. https://doi.org/10.1038/nrc2772

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Coppé J‑P, Desprez P‑Y, Krtolica A, Campisi J (2010) The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5:99–118. https://doi.org/10.1146/annurev-pathol-121808-102144

    Article  PubMed  PubMed Central  Google Scholar 

  12. Demaria M, O’Leary MN, Chang J et al (2017) Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov 7:165–176. https://doi.org/10.1158/2159-8290.CD-16-0241

    Article  CAS  PubMed  Google Scholar 

  13. Evan GI, d’Adda di Fagagna F (2009) Cellular senescence: hot or what? Curr Opin Genet Dev 19:25–31

    Article  CAS  PubMed  Google Scholar 

  14. Evan GI, Vousden KH (2001) Proliferation, cell cycle and apoptosis in cancer. Nature 411:342–348

    Article  CAS  PubMed  Google Scholar 

  15. Evangelou K, Lougiakis N, Rizou SV et al (2017) Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell 16:192–197. https://doi.org/10.1111/acel.12545

    Article  CAS  PubMed  Google Scholar 

  16. Fearon ER, Vogelstein B (1990) A genetic model for colorectal tumorigenesis. Cell 61:759–767

    Article  CAS  PubMed  Google Scholar 

  17. Foersch S, Sperka T, Lindner C et al (2015) VEGFR2 signaling prevents Colorectal cancer cell senescence to promote Tumorigenesis in mice with colitis. Gastroenterology 149:177–189.e10. https://doi.org/10.1053/j.gastro.2015.03.016

    Article  CAS  PubMed  Google Scholar 

  18. Gil J, Peters G (2006) Regulation of the INK4b-ARF-INK4a tumour suppressor locus: all for one or one for all. Nat Rev Mol Cell Biol 7:667–677. https://doi.org/10.1038/nrm1987

    Article  CAS  PubMed  Google Scholar 

  19. Guinney J, Dienstmann R, Wang X et al (2016) The consensus molecular subtypes of colorectal cancer. Nat Med 21:1350–1356. https://doi.org/10.7303/syn2623706

    Article  Google Scholar 

  20. Haugstetter a M, Loddenkemper C, Lenze D et al (2010) Cellular senescence predicts treatment outcome in metastasised colorectal cancer. Br J Cancer 103:505–509. https://doi.org/10.1038/sj.bjc.6605784

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hayflick L, Moorhead PS (1961) The serial cultivation of human diploid cell strains. Exp Cell Res 25:585–621

    Article  CAS  PubMed  Google Scholar 

  22. Kang T‑W, Yevsa T, Woller N et al (2011) Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479:547–551. https://doi.org/10.1038/nature10599

    Article  CAS  PubMed  Google Scholar 

  23. Krizhanovsky V, Yon M, Dickins RA et al (2008) Senescence of activated stellate cells limits liver fibrosis. Cell 134:657–667. https://doi.org/10.1016/j.cell.2008.06.049

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Michaloglou C, Vredeveld LCW, Soengas MS et al (2005) BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436:720–724. https://doi.org/10.1038/nature03890

    Article  CAS  PubMed  Google Scholar 

  25. Pistritto G, Trisciuoglio D, Ceci C et al (2016) Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8:603–619. https://doi.org/10.18632/aging.100934

    Article  Google Scholar 

  26. Powell SM, Zilz N, Beazer-Barclay Y et al (1992) APC mutations occur early during colorectal tumorigenesis. Nature 359:235–237. https://doi.org/10.1038/359235a0

    Article  CAS  PubMed  Google Scholar 

  27. Prieur A, Besnard E, Babled A, Lemaitre J‑M (2011) p53 and p16(INK4A) independent induction of senescence by chromatin-dependent alteration of S‑phase progression. Nat Commun. https://doi.org/10.1038/ncomms1473

    PubMed  Google Scholar 

  28. Roxburgh CS, Richards CH, Macdonald AI et al (2013) The in situ local immune response, tumour senescence and proliferation in colorectal cancer. Br J Cancer 109:2207–2216. https://doi.org/10.1038/bjc.2013.556

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Schmitt C, Fridman J, Yang M, Lee S (2002) A senescence program controlled by p53 and p16 INK4a contributes to the outcome of cancer therapy. Cell 109:335–346

    Article  CAS  PubMed  Google Scholar 

  30. Serrano M, Lin AW, McCurrach ME et al (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88:593–602

    Article  CAS  PubMed  Google Scholar 

  31. Severino J, Allen RG, Balin S et al (2000) Is beta-galactosidase staining a marker of senescence in vitro and in vivo? Exp Cell Res 257:162–171

    Article  CAS  PubMed  Google Scholar 

  32. Shima K, Nosho K, Baba Y et al (2011) Prognostic significance of CDKN2A (p16) promoter methylation and loss of expression in 902 colorectal cancers: Cohort study and literature review. Int J Cancer 128:1080–1094. https://doi.org/10.1002/ijc.25432

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tchkonia T, Zhu Y, Van Deursen J et al (2013) Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest 123:966–972. https://doi.org/10.1172/JCI64098

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ventura A, Kirsch DG, McLaughlin ME et al (2007) Restoration of p53 function leads to tumour regression in vivo. Nature 445:661–665. https://doi.org/10.1038/nature05541

    Article  CAS  PubMed  Google Scholar 

  35. Waldner MJ, Wirtz S, Jefremow A et al (2010) VEGF receptor signaling links inflammation and tumorigenesis in colitis-associated cancer. J Exp Med 207:2855–2868

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Xue W, Zender L, Miething C et al (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445:656–660. https://doi.org/10.1038/nature05529

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang NC, Hu ML (2005) The limitations and validities of senescence associated-??-galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells. Exp Gerontol 40:813–819. https://doi.org/10.1016/j.exger.2005.07.011

    Article  CAS  PubMed  Google Scholar 

  38. Robert Koch-Institut (2016) Bericht zum Krebsgeschehen in Deutschland 2016. Zentrum für Krebsregisterdaten im Robert Koch-Institut, Berlin

    Google Scholar 

Download references

Förderung

S. Foersch wird in seiner Arbeit durch das Stufe-1-Programm der Universitätsmedizin Mainz, durch die Manfred-Stolte-Stiftung und durch das UCT TransMed Fellowship unterstützt.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to S. Foersch.

Ethics declarations

Interessenkonflikt

S. Foersch gibt an, dass kein Interessenkonflikt besteht.

Dieser Beitrag beinhaltet keine vom Autor durchgeführten Studien an Menschen oder Tieren, welche noch nicht publiziert wurden. Hinsichtlich der Einhaltung ethischer Richtlinien beachten Sie die jeweiligen Originalarbeiten.

The supplement containing this article is not sponsored by industry.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Foersch, S. Zelluläre Seneszenz im kolorektalen Karzinom. Pathologe 38 (Suppl 2), 205–210 (2017). https://doi.org/10.1007/s00292-017-0357-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00292-017-0357-y

Schlüsselwörter

Keywords

Navigation