An overview of apoptosis and the prevention of colorectal cancer

https://doi.org/10.1016/j.critrevonc.2005.06.005Get rights and content

Abstract

Colorectal cancer arises as a result of the accumulation of genetic errors many of which affect the control of apoptosis. Effective chemoprevention strategies for colorectal cancer must rectify these genetic defects. Mutation of apc is often the initiating genetic lesion in colorectal cancers that develop along the chromosomal instability pathway. Depending on the cellular context, loss of apc activates the Wnt signalling pathway causing immediate widespread apoptosis of colorectal epithelial cells and defects in differentiation and cell migration. Only cells that are inherently resistant to apoptosis survive this initial wave of apoptosis. These surviving cells constitute the epithelial population that develop into adenomas. Two gene targets of the Wnt signalling pathway are of particular relevance to apoptosis. Although controversial, survivin may function to inhibit apoptosis. MYC has two outputs in normal cells, the induction of apoptosis and proliferation. These opposing functions work so that MYC can only induce cell proliferation in cells if apoptosis is disabled. p53 couples apoptosis to mitogenic signals and survival pathways. Under some circumstances, NF-κB can act as an inhibitor of apoptosis possibly through increased expression of bcl-xL. Tumours that evolve by the microsatellite instability pathway often have mutations in the proapoptotic gene bax. Colonic adenomas express cyclo-oxygenase-2 (COX-2) and may be targets of chemoprevention before the development of malignancy. However, the recent discovery that coxibs increase the risk of serious cardiovascular events limits their use as chemopreventive agents. Nevertheless, aspirin remains a drug of great interest as it is already known to reduce the risk of colorectal cancer by up to 50%. The balance of evidence shows that high vegetable fibre diets can prevent colorectal cancer, probably via the fermentation of butyrate enhancing the apoptotic response to DNA damage.

Introduction

On a global scale, colorectal cancer is the fourth commonest malignant neoplasm after lung, breast and prostate [1]. As will be discussed below, approximately 90% of colorectal cancers are derived from benign adenomatous lesions which are estimated to take 5–15 years to evolve into invasive cancer (for review, see [2], [3]). A wealth of epidemiological and clinical trial data suggests that if these premalignant lesions are identified and removed, the subsequent development of colorectal cancer is aborted [4], [5], [6]. Unfortunately, adenomas are largely asymptomatic and so require population-based screening programmes for their detection. Colorectal cancer develops through a number of well characterized stages based on the degree of invasion. When the cancer is confined to the wall of the bowel (Stage 1, T1N0M0), resection is essentially curative with 5 year survival rates of 90% or more. However, in Stage IV disease where distant metastases are present, the 5 year survival drops to 15% [7]. Overall, the 5 year survival rate of colorectal cancer is 50%.

These diagnostic challenges have led investigators to attempt to prevent adenoma formation with the minimum of risk. The long natural history of adenomas, their accessibility to biopsy and the existence of two inherited forms of colorectal cancer whose genetic changes mirror those of sporadic cancer have enabled investigation of molecular pathogenesis of sporadic colorectal cancer in great detail, perhaps more than any other neoplasm [8]. These investigations have shown that numerous errors in pathways controlling apoptosis (programmed cell death) occur during the development of colorectal cancer. These abnormalities render cells with clinically significant mutations resistant to elimination through apoptosis (programmed cell death). Abnormalities in core apoptosis mechanisms go hand in hand with inherent resistance to chemotherapy and radiotherapy. Clinical evidence is emerging that defective apoptosis is of pathogenic significance [9]. Apoptosis rates in rectal mucosa are inversely related to the presence of adenomas, while the clinical response of adenomas to treatment with the COX-2 inhibitor celecoxib correlates positively with apoptosis [10], [11]. Thus, a full understanding of molecular abnormalities in apoptosis and its relationship to other molecular pathogenic mechanisms is essential for the rational development of prevention strategies against colorectal cancer.

Section snippets

Molecular pathogenesis of colorectal cancer

Hahn and Weinberg have argued that a normal cell has only to acquire six phenotypes in order to behave as a fully transformed malignant cell. These phenotypes are resistance to growth inhibition, immortalization, independence from mitogenic stimulation, the ability to acquire their own blood supply, the ability to invade and metastasize and the ability to suppress or evade apoptosis [12]. These characteristics are acquired through the mutation of key genes regulating these functions. The

Vegetable products

A number of dietary components have been reported to cause apoptosis with the implication that mutant cells will, in this way, be eliminated thereby preventing cancer. Such inferences are fraught with difficulty as modulation of cell death may not necessarily be responsible for cancer protection in the complex environment of the human gut [110]. Diets high in fibre (non-starch polysaccharides) have been recommended to prevent colorectal cancer [111], [112]. Epidemiological studies and

Conclusions

The majority of genes that are functionally important in the pathogenesis of colorectal cancer in some way inactivate or raise the threshold for induction of apoptosis, giving a growth advantage to neoplastic and malignant clones. Clinical data are emerging that apoptosis in human colonic epithelium is inversely related to cancer development. Studies of mouse models are starting to illustrate how apoptosis fits in and interacts with the other important phenotypes required by cancer cells as

Reviewers

Prof. Chris Paraskeva, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK.

Prof. E.G.E. De Vries, M.D., Ph.D., Groningen University Hospital, Department of Medical Oncology, P.O. Box 30.001, NL-9700 RB Groningen, The Netherlands.

Dr. Nadir Arber, Tel Aviv Sourasky Medical Center, Weizmann 6, Tel Aviv 64238, Israel.

Alastair J.M. Watson is professor of medicine at the University of Liverpool and head of the Division of Gastroenterology in the School of Clinical Sciences. He qualified in 1980 from Cambridge and London Universities. In 1985, he became a research fellow with Michael Farthing in St. Bartholomew's hospital from where he obtained in 1989 a Medical Doctorate on the absorption of short chain fatty acids. He became a post-doctoral fellow with Mark Donowitz and Chip Montrose at Johns Hopkins Medical

References (217)

  • A.M. Verhagen et al.

    Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins

    Cell

    (2000)
  • Z. Song et al.

    Direct interaction between survivin and Smac/DIABLO is essential for the anti-apoptotic activity of survivin during taxol-induced apoptosis

    J Biol Chem

    (2003)
  • I.A. McNeish et al.

    Survivin interacts with Smac/DIABLO in ovarian carcinoma cells but is redundant in Smac-mediated apoptosis

    Exp Cell Res

    (2005)
  • E. Beltrami et al.

    Acute ablation of survivin uncovers p53-dependent mitotic checkpoint functions and control of mitochondrial apoptosis

    J Biol Chem

    (2004)
  • A.O. Hueber et al.

    Traps to catch unwary oncogenes

    Trends Genet

    (1998)
  • W.S. el-Deiry et al.

    WAF1, a potential mediator of p53 tumor suppression

    Cell

    (1993)
  • C. de la Cova et al.

    Drosophila myc regulates organ size by inducing cell competition

    Cell

    (2004)
  • E. Moreno et al.

    dMyc transforms cells into super-competitors

    Cell

    (2004)
  • J. Secombe et al.

    Myc: a weapon of mass destruction

    Cell

    (2004)
  • T.D. Donaldson et al.

    Cancer cell biology: Myc wins the competition

    Curr Biol

    (2004)
  • N. Arber et al.

    Activation of c-K-ras mutations in human gastrointestinal tumors

    Gastroenterology

    (2000)
  • Y. Shi et al.

    Mechanisms of TGF-beta signaling from cell membrane to the nucleus

    Cell

    (2003)
  • Y. Yamamura et al.

    Critical role of Smads and AP-1 complex in transforming growth factor-beta-dependent apoptosis

    J Biol Chem

    (2000)
  • S.K. Mithani et al.

    Smad3 has a critical role in TGF-beta-mediated growth inhibition and apoptosis in colonic epithelial cells

    J Surg Res

    (2004)
  • Y. Zhu et al.

    Smad3 mutant mice develop metastatic colorectal cancer

    Cell

    (1998)
  • L.J. Hofseth et al.

    p53: 25 years after its discovery

    Trends Pharmacol Sci

    (2004)
  • C.A. Schmitt et al.

    Dissecting p53 tumor suppressor functions in vivo

    Cancer Cell

    (2002)
  • V. Stambolic et al.

    Regulation of PTEN transcription by p53

    Mol Cell

    (2001)
  • T. Serafini et al.

    The netrins define a family of axon outgrowth-promoting proteins homologous to C. elegans UNC-6

    Cell

    (1994)
  • F. Balkwill et al.

    Inflammation and cancer: back to Virchow?

    Lancet

    (2001)
  • K.V. Winther et al.

    Long-term risk of cancer in ulcerative colitis: a population-based cohort study from Copenhagen County

    Clin Gastroenterol Hepatol

    (2004)
  • G. Bonizzi et al.

    The two NF-kappaB activation pathways and their role in innate and adaptive immunity

    Trends Immunol

    (2004)
  • F.R. Greten et al.

    IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer

    Cell

    (2004)
  • S. Rakoff-Nahoum et al.

    Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis

    Cell

    (2004)
  • UK CR

    Cancer statistics

    (2005)
  • A. Leslie et al.

    The colorectal adenoma–carcinoma sequence

    Br J Surg

    (2002)
  • S.J. Winawer

    Natural history of colorectal cancer

    Am J Med

    (1999)
  • S.J. Winawer et al.

    Prevention of colorectal cancer by colonoscopic polypectomy. The National Polyp Study Workgroup

    N Engl J Med

    (1993)
  • W.S. Atkin et al.

    Surveillance guidelines after removal of colorectal adenomatous polyps

    Gut

    (2002)
  • A. Bedi et al.

    Inhibition of apoptosis during development of colorectal cancer

    Cancer Res

    (1995)
  • F.A. Sinicrope et al.

    Cell proliferation and apoptotic indices predict adenoma regression in a placebo-controlled trial of celecoxib in familial adenomatous polyposis patients

    Cancer Epidemiol Biomarkers Prev

    (2004)
  • W.C. Hahn et al.

    Rules for making human tumor cells

    N Engl J Med

    (2002)
  • B. Iacopetta

    Are there two sides to colorectal cancer?

    Int J Cancer

    (2002)
  • K. Konishi et al.

    Clinicopathological differences between colonic and rectal carcinomas: are they based on the same mechanism of carcinogenesis?

    Gut

    (1999)
  • K. Birkenkamp-Demtroder et al.

    Differential gene expression in colon cancer of the caecum versus the sigmoid and rectosigmoid

    Gut

    (2005)
  • P. Distler et al.

    Are right- and left-sided colon neoplasms distinct tumors?

    Dig Dis

    (1997)
  • D.L. Stoler et al.

    The onset and extent of genomic instability in sporadic colorectal tumor progression

    Proc Natl Acad Sci USA

    (1999)
  • I.M. Shih et al.

    Evidence that genetic instability occurs at an early stage of colorectal tumorigenesis

    Cancer Res

    (2001)
  • O.J. Sansom et al.

    Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration

    Genes Dev

    (2004)
  • L.K. Su et al.

    APC binds to the novel protein EB1

    Cancer Res

    (1995)
  • Cited by (105)

    • Immunological paradox for maintaining normal flora: it is all by design, not by chance

      2022, Microbial Crosstalk with Immune System: New Insights in Therapeutics
    • Review of short-chain fatty acids effects on the immune system and cancer

      2020, Food Bioscience
      Citation Excerpt :

      SCFA prevents colon cancer (Nilsson et al., 2003) through the cancer-specific induction of apoptosis (Hinnebusch et al., 2002) and autophagy (Donohoe et al., 2011). Butyrate arrests the cell cycle and induces apoptosis in cancer cells but does not affect the proliferation of the colon's normal epithelial cells (Davie, 2003; Watson, 2006). Normal colon epithelium cells use butyrate as an energy source, thus promoting epithelial growth.

    • Enoyl-coenzyme A hydratase in cancer

      2015, Clinica Chimica Acta
    View all citing articles on Scopus

    Alastair J.M. Watson is professor of medicine at the University of Liverpool and head of the Division of Gastroenterology in the School of Clinical Sciences. He qualified in 1980 from Cambridge and London Universities. In 1985, he became a research fellow with Michael Farthing in St. Bartholomew's hospital from where he obtained in 1989 a Medical Doctorate on the absorption of short chain fatty acids. He became a post-doctoral fellow with Mark Donowitz and Chip Montrose at Johns Hopkins Medical School (1988–1990). He returned to the University of Manchester to work with Lord Turnberg on apoptosis in the GI epithelium. His main research interests the regulation of apoptosis in intestinal epithelium and the role of apoptosis in colorectal cancer, pancreatic disease and inflammatory bowel disease. He is on the Council of the British Society of Gastroenterology and is Deputy Editor of Gut.

    View full text