Summary and Concluding Remarks

Similar documents
From crypt stem cell to colorectal cancer

CHAPTER 6 SUMMARIZING DISCUSSION

The Wnt target MASH-2 regulates expression of the putative intestinal specific stemcell markers Sox4 and Sfrp5.

CHAPTER 3. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 23: (2005)

icamp: Cancer biology tutorial II: recent developments in tumor biology, experimental methodology, and reference identification

Cancer and Oncogenes Bioscience in the 21 st Century. Linda Lowe-Krentz

Supplemental Figure 1: Lrig1-Apple expression in small intestine. Lrig1-Apple is observed at the crypt base and in insterstial cells of Cajal, but is

Genotype analysis by Southern blots of nine independent recombinated ES cell clones by

CANCER. Inherited Cancer Syndromes. Affects 25% of US population. Kills 19% of US population (2nd largest killer after heart disease)

Development of Carcinoma Pathways

Section D: The Molecular Biology of Cancer

Type of file: PDF Size of file: 0 KB Title of file for HTML: Supplementary Information Description: Supplementary Figures

Early Embryonic Development

Regulation of Gene Expression in Eukaryotes

Negative Regulation of c-myc Oncogenic Activity Through the Tumor Suppressor PP2A-B56α

Biochemistry of Carcinogenesis. Lecture # 35 Alexander N. Koval

INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 19: ,

mirna Dr. S Hosseini-Asl

Molecular Regulation of Intestinal Stem Cells

The Wnt/βcatenin signaling pathway

Karyotype analysis reveals transloction of chromosome 22 to 9 in CML chronic myelogenous leukemia has fusion protein Bcr-Abl

oncogenes-and- tumour-suppressor-genes)

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

Genetics and Cancer Ch 20

Targeting the cgmp Pathway to Treat Colorectal Cancer

SUPPLEMENTARY INFORMATION

Wnt signaling. Ramray Bhat.

Introduction to Cancer Biology

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Stem cells in homeostasis and cancer of the gut

Lactase, sucrase-isomaltase, and carbamoyl phosphate synthase I expression in human intestine van Beers, E.H.

ANAT3231: lectures overview

The Hallmarks of Cancer

A Genetic Program for Embryonic Development

Regulation of Intestinal Stem Cells

5-ASA and Carcinogenesis in IBD

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

Colonic polyps and colon cancer. Andrew Macpherson Director of Gastroentology University of Bern

Determination Differentiation. determinated precursor specialized cell

Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Characteristics of Cancer Stem Cells (CSCs)

The silence of the genes: clinical applications of (colorectal) cancer epigenetics

Profiles of gene expression & diagnosis/prognosis of cancer. MCs in Advanced Genetics Ainoa Planas Riverola

Cancer and Oncogenes Bioscience in the 21 st Century. Linda Lowe-Krentz October 11, 2013

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Colon Cancer and Hereditary Cancer Syndromes

Carcinogenesis in IBD

supplementary information

Agro/Ansc/Bio/Gene/Hort 305 Fall, 2017 MEDICAL GENETICS AND CANCER Chpt 24, Genetics by Brooker (lecture outline) #17

Epstein-Barr virus driven promoter hypermethylated genes in gastric cancer

Pancreatic intraepithelial

The Biology and Genetics of Cells and Organisms The Biology of Cancer

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Cancer Cells. It would take another 20 years and a revolution in the techniques of biological research to answer these questions.

number Done by Corrected by Doctor Maha Shomaf

7.012 Quiz 3 Answers

Enterprise Interest Nothing to declare

p53 cooperates with DNA methylation and a suicidal interferon response to maintain epigenetic silencing of repeats and noncoding RNAs

Activation of cellular proto-oncogenes to oncogenes. How was active Ras identified?

Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines

Modelling multiscale aspects of colorectal cancer

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes.

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation

Functional characterisation of hepatitis B viral X protein/microrna-21 interaction in HBVassociated hepatocellular carcinoma

Microarray Analysis and Liver Diseases

Multistep nature of cancer development. Cancer genes

A class of genes that normally suppress cell proliferation. p53 and Rb..ect. suppressor gene products can release cells. hyperproliferation.

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

ANAT3231: lectures overview

Problem Set 8 Key 1 of 8

LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway

Experimental Therapeutics I

Cancer genetics

Bi 8 Lecture 17. interference. Ellen Rothenberg 1 March 2016

mtorc1-mediated translational elongation limits intestinal tumour initiation and growth

Cancer and sornat ic evolution

MicroRNA in Cancer Karen Dybkær 2013

MicroRNA expression profiling and functional analysis in prostate cancer. Marco Folini s.c. Ricerca Traslazionale DOSL

Concomitant Gain of Function of Notch and Loss of p53 Signalling trigger an EMT Like Programme Driving Metastatic Intestinal Cancers

A calibrated agent-based computer model of stochastic cell dynamics in normal human colon crypts useful for in silico experiments

Journal Club. 03/04/2012 Lama Nazzal

Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation

Molecular Pathology of Ovarian Carcinoma with Morphological Correlation

VIII Curso Internacional del PIRRECV. Some molecular mechanisms of cancer

1. Basic principles 2. 6 hallmark features 3. Abnormal cell proliferation: mechanisms 4. Carcinogens: examples. Major Principles:

BCHM3972 Human Molecular Cell Biology (Advanced) 2013 Course University of Sydney

Dominic J Smiraglia, PhD Department of Cancer Genetics. DNA methylation in prostate cancer

Human Lung Cancer Pathology and Cellular Biology Mouse Lung Tumor Workshop

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CELL CYCLE REGULATION AND CANCER. Cellular Reproduction II

Colorectal adenocarcinoma leading cancer in developed countries In US, annual deaths due to colorectal adenocarcinoma 57,000.

The Effect of Inflammation on Stem Cell Mutagenesis and Carcinogenesis Induced by Azoxymethane in Wild type and Immune Compromised Mice

Signaling Vascular Morphogenesis and Maintenance

INTRODUCTION: The intestinal epithelium is one of the most rapidly dividing tissues in the

Mathematics and Physics of Cancer: Questions. Robijn Bruinsma, UCLA KITP Colloquium May 6, ) Cancer statistics and the multi-stage model.

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Generating Mouse Models of Pancreatic Cancer

Computer Science, Biology, and Biomedical Informatics (CoSBBI) Outline. Molecular Biology of Cancer AND. Goals/Expectations. David Boone 7/1/2015

CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION. Androgen deprivation therapy is the most used treatment of de novo or recurrent

Histone methyltransferase SETD2 modulates alternative splicing to inhibit intestinal tumorigenesis

Transcription:

Summary and Concluding Remarks

Chapter 6 The intestinal epithelium provides an excellent model system for investigating molecular mechanisms regulating cell lineage establishment, stem cell proliferation, morphogenesis, the specialization of cell function during terminal differentiation and the deregulation of these processes during cancer initiation and development. In vertebrates, the intestinal lumen is lined by an endoderm-derived epithelial sheet, a monolayer consisting of four principal cell types: Paneth cells, Enterocytes, Goblet cells and Enteroendocrine cells. Lineage tracing experiments demonstrated that these four cell types are all derived from a small population of multipotential stem cells present near the villus base (the crypt) 1,2. The differentiating cells migrate from the crypt towards the villus tip, where they eventually die and are extruded into the lumen. Within the epithelium, the absorptive enterocytes are the predominant cell type and they form a highly organized apical brush border. Goblet cells secrete mucus through exported granules and Enteroendocrine cells secrete various hormones and growth factors. The fourth cell type, the anti-microbial secreting Paneth cell, differentiates during migration toward the base of the crypt [reviewed in 3 ]. The intestine constitutes the most rapidly self-renewing tissue in adult mammals that produces an impressive number of descendant epithelial cells each day: 70 billion in humans ( 0.25 kg) and 200 million in mice. This massive rate of cell production is compensated by apoptosis at the tip of the villus. In the mouse, the intestinal epithelium turns over entirely within 3 5 days. This enormous cell renewal, which takes places throughout life, makes the intestine sensitive for additional mutations and, as a consequence, increases the risk of developing of cancer. The cessation of proliferation, the induction of differentiation, migration, adhesion and cell death in the intestine are highly coordinated processes. This width variety of cellular responses is coordinated by a relatively small number of highly evolutionarily conserved signalling pathways. These pathways include the BMP, Sonic Hedgehog, Notch and Wnt signalling pathway [reviewed in 4 ]. This is particularly well illustrated by the role of the Wnt pathway in the development and maintenance of intestinal epithelium and by the fact that deregulation of this pathway causes intestinal cancer. Current evidence indicates that the Wnt cascade is the dominant force in controlling cell fate along the crypt-villus axis. In neonatal mice lacking Tcf4, the differentiated villus epithelium appears unaffected, but the crypt progenitor compartment is entirely absent 5. This implies that physiological Wnt signaling is required for the establishment of this progenitor compartment. Inhibition of Wnt signaling by transgenic or viral expression of a secreted Wnt inhibitor (Dkk-1) in adult mice induces the complete loss of crypts, identifying Wnt as the dominant mitogen for crypt progenitor cells throughout life 6,7. This notion has been confirmed by transgenic expression of the Wnt agonist R-spondin-1, which results in a massive hyperproliferation of intestinal crypts 8. Wnt proteins are physiologically expressed by the crypt epithelial cells rather than by 88

Summary and concluding remarks the surrounding mesenchyme 9. These Wnt proteins not only stimulate the proliferation of crypt progenitors but also promote the terminal differentiation of Paneth cells, residing at the bottoms of the crypts 10. Three regulatory genes in the Wnt pathway encoding APC, β-catenin and Axin, are known to be mutated in human cancers [reviewed in 11 ]. All of these mutations result in constitutively, uncontrolled activation of gene transcription and thereby promoting proliferation and carcinogenesis. Moreover, recent evidence indicates that loss of functional APC may also affect other cellular processes besides transcription that can promote carcinogenesis, such as interference with normal chromosome segregation 12,13. The aim of the study described in this thesis was to address a number of questions related to the role of the Wnt signaling pathway in normal development and maintenance of the intestine in relation to the development of intestinal cancer. These questions were: what is the contribution of aberrant activation of Wnt signalling to the colorectal transformation process, how can genes that are overexpressed in cancer and/or contribute to the survival and progression of tumor cells be silenced, what is the overlap between the Wnt responsive genes with the gene profile of adenomas, adenocarcinomas versus normal colonic epithelium and what is the contribution of individual Wnt target genes to these processes? Answering these questions is of paramount importance since unraveling the growth regulatory pathways and the process of tumorgenesis may lead to new diagnostic and prognostic markers and potential targets for therapeutic intervention. The transition of an intestinal epithelial cell into a fully transformed, metastatic cancer cell requires mutations in multiple proto-oncogenes and key tumor suppressor genes, including those of the Wnt pathway 14. To further delineate the contribution of aberrant activation of Wnt signalling to this colorectal transformation process, we described in chapter 2 a largescale analysis of the downstream genetic program activated by β-catenin/tcf4 in colorectal cancer cells (CRC s). Inhibition of Wnt signalling in these cells leads to downregulation of genes expressed in the proliferating region of the crypt, e.g. c-myc, BMP-4, EphB3 and NRP/B. Simultaneously, upregulated genes represented differentiation markers of mucosecretory and/ or absorptive intestinal cells, for example MUC-2, FABPL, CAII. Physiological evidence by immunohistochemical analyses of adult colonic crypts, adenomas and early colorectal lesions confirmed the array results. In cells that exhibit nuclear β-catenin, proliferation markers are highly expressed and differentiation markers are absent. Identically, inducing cell cycle arrest by inhibition of β-catenin/tcf-4 activity forces the CRC cells into a differentiation program. From this study, we concluded that constitutive Wnt signalling in CRC cells activates the genetic program that is physiologically expressed in the crypt progenitor cells, in order to maintain 89

Chapter 6 progenitor cell proliferation. As a consequence, they continue to behave as crypt progenitor cells in the surface epithelium, persisting for many years and given ample opportunity for the acquisition of further mutations. This study validated the disruption of the β-catenin/tcf complex as a therapeutic strategy to revert the transformed phenotype in colorectal cancer. So far, a number of different approaches have successfully been used to disrupt Tcf/β-catenin signalling in preclinical studies, including the inhibition of β-catenin expression, the induction of β-catenin degradation, the modulation of the subcellular localisation of β-catenin, the activation of oncolytic viruses or the induction of apoptosis inducing genes upon aberrant β-catenin signalling activity [reviewed in 15 ]. Moreover, small molecules have been identified that disrupt Tcf-4/β-catenin complexes using a high throughput in vitro screen of natural products 16. However, further challenges will be to improve on the selectivity and/or in vivo efficacy. Silencing genes that contribute to the survival and progression of tumor cells is a major research objective. However, mammalian genetic approaches to study gene function have been hampered by the lack of efficient tools to generate stable loss-of-function phenotypes. Recently RNAi technology for gene knockdown in mammalian cells has been proven to be effective. Using polymerase III H1-RNA gene promoters like H1 and U7, vectors for stable expression of small interfering RNAs (sirnas) have been generated (for example psuper.) 17. These vectors drive expression of shrnas, allowing efficient and specific inactivation of targeted genes, generating a loss-of-function phenotype. In chapter 3, we described the generation of a doxycycline-inducible version of a Polymerase III H1-RNA gene promoter driven shrna vector (pter). As a proof of principle, we inactivated the key regulator of the Wnt pathway: β-catenin. Down-regulation of β-catenin in stable pter LS174T CRC cells transfectants resulted in a G1 arrest and differentiation. The knockdown of β-catenin/tcf activity by a loss-of-function approach reproduced the phenotypic changes that occur in CRC cells in which dominant-negative versions of TCF proteins (dntcf) are overexpressed. The results of the study described in chapter 2 are thus confirmed by inducible knock-down of β-catenin gene expression by sirna. Moreover, the successful knockdown of two other independent genes (Pygopus-2, c-myc) showed that this vector system is widely applicable for the inducible knockdown of gene expression. This is particularly useful for the analysis of genes which functions are difficult to evaluate due to effects on growth or differentiation of cells, like cell-cycle regulators, oncogenes, tumor suppressor genes and genes that control apoptosis. Moreover, RNAi may also lead to promising novel therapeutic strategies. However, one critical factor is the ability to deliver intact sirnas into target cells/organs in vivo. Recently, several studies based on the systemic or local application of naked sirnas or the uses of various viral and non-viral sirna delivery systems weredescribed [reviewed in 18 ]. 90

Summary and concluding remarks DNA array techniques are rapidly evolving and genome-wide oligonucleotide array platforms are now common use, containing up to 55000 probes. DNA arrays are extensively used to get a gene profiling set in a whole array of human carcinomas, relating it to growth characteristics, prognosis and the expected response to therapy. In chapter 4 we analyzed the expression profile of LS174 CRC cells stably transfected with inducible dntcf1 and dntcf4. The 300-400 selected Wnt responsive genes were subsequently compared to the gene profile of adenomas and adenocarcinomas versus normal colonic epithelium. This analysis provided us with a list of genes that were expressed both in adenomas and carcinomas and controlled by TCF. We believe these genes represent the Wnt signature. In situ hybridizations were performed on intestinal tissue of APC min mice for 80 of the Wnt signature genes. These genes could be divided into 3 groups based on differential expression in distinct parts of the crypts. One group, 80% of the selected genes, is expressed in the proliferative compartment of the crypts. A second category consists of Paneth cell maturation markers. A third category, expressed near the crypt bottom, distinct from the Paneth cells, correlating with the supposed stem cell compartment in the crypts. These data demonstrated that different transcriptional outputs of Wnt signalling in one organ system, the gut, correspond with the proposed Wnt functions of maintaining the crypt progenitor phenotype, driving the terminal differentiation of Paneth cells and stem cell renewal, respectively. Moreover, the identification of the Tcf/β-catenin target gene program activated in Wnt driven cancers should facilitate the selection of novel targets for therapeutic antibodies or small-molecule inhibition [reviewed in 19 ]. However, it will be a challenge to target colon cancer by abrogating Wnt signalling while not affecting normal gut homeostasis, since ample experimental evidence indicates that Wnt signalling is crucial for tissue renewal in the intestine. As noted above Wnt signalling regulates both proliferation and differentiation of intestinal crypt cells. By now numerous TCF target genes have been identified but little is known about their individual role in these processes. In a study described in chapter 5 we focused on Mash-2, one of the identified target genes of the Wnt signalling pathway. Mash-2 is a basic helix-loop-helix transcription factor, so far only known to be required for the development of the spongiotrophoblast in order to form a functional placenta. In the normal intestine, Mash-2 is expressed at the position of the presumptive crypt stem cell. No Mash-2 expression was found in the TCF4 KO mice and in adenomas of APC min mice Mash-2 is highly expressed. This confirmed the observation that Mash-2 is indeed a target of the Wnt pathway. To further define the contribution of Mash-2 to the Wnt dependent phenotype in the gut, we generated a Villin-Mash-2-transgenic mouse. In the small bowel of these mice Mash-2 is highly expressed in both crypt and villus. This over- and misexpression leads to architectural 91

Chapter 6 abnormalities, i.e. enlarged crypts, disorganization of the villi and the formation of aberrant crypt-like structures in the villi. A few cells in the aberrant crypt foci stained positive for the proliferation marker Ki67 and/or the Wnt target gene c-myc. c-myc, normally expressed at the bottom of the crypts, is implicated in the control of cellular proliferation, differentiation and apoptosis. Mash-2 expression in the crypts and villi of transgenic mice induced upregulation of the Wnt targets Sox-4 and Ets-2, as well as sfrp5, a Wnt inhibitor. These Wnt target genes, whose expression is normally restricted to the intestinal crypts, seemed to be regulated by Mash- 2. Ets-2 is being implicated in tumour progression. Sox-4 and sfrp5 are both expressed by the cells at position plus 4, the position were the potential stem cells reside. These data implicate a role for Mash-2 in the Wnt dependent proliferation and stem/progenitor cell maintenance in the crypts as well as in unrestricted cell growth seen in colorectal cancer. This makes Mash-2, a possible stem cell regulator which might confer selective advantage to the cancer cell, a promising and more selective target for anti-cancer therapy. The Wnt signalling cascade plays a central role in embryonic patterning, stem cell renewal, maintenance of the crypt progenitor phenotype, tissue repair and regeneration. Aberrant activation drives the formation of various human cancers. The results described in this thesis, using a width variety of sophisticated techniques, has further increased our knowledge of the role of the Wnt pathway in these processes. This may be a basis for the ongoing efforts for the development of drugs targeting the inhibition of the Wnt pathway. References 1 Marshman E., Booth C., Potten C.S. (2002) The intestinal epithelial stem cell. Bioessays. 24:91-98. 2 Bjerknes M., Cheng H. (1999) Clonal analysis of mouse intestinal epithelial progenitors. Gastroenterology 116:7-14. 3 Roberts, D.J. (2000) Molecular mechanisms of development of the gastrointestinal tract. Dev. Dyn. 219, 109. 4 Batlle, E. et al (2004) Signaling pathways in intestinal development and cancer. Annual Review and Cell and Developmental Biology 20, 695. 5 Korinek, V. et al (1998) Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet. 19, 379. 6 Pinto, D. et al (2003) Canonical Wnt signals are essential for homeostasis of the intestinal epithelium. Genes Dev. 17, 1709. 7 Kuhnert, F. et al (2004) Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1. Proc. Natl. Acad. Sci. 101, 266. 92

Summary and concluding remarks 8 Kim, K et al (2005) Mitogenic Influence of Human R-Spondin1 on the Intestinal Epithelium Science 309, 1256. 9 Gregorieff A. et al (2005) Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 129, 626. 10 van Es, J.H. et al (2005) Wnt signalling induces maturation of Paneth cells in intestinal crypts. Nat Cell Biol. 7, 381. 11 Clevers H. (2006) Wnt/beta-catenin signaling in development and disease. Cell 127, 469. 12 Fodde R, Kuipers J, Rosenberg C, Smits R, Kielman M, Gaspar C et al. Mutations in the APC tumor suppressor gene cause chromosomal instability. Nat Cell Biol 2001; 3:433 8. 13 Pellman D. Cancer: a CINtillating new job for the APC tumor suppressor. Science 2001; 291:2555 6. 14 Fearon, E.R. and Vogelstein, B. (1990) A genetic model for colorectal tumorigenesis. Cell 61, 759. 15 Dihlmann, S. and von Knebel Doeberitz, M. (2005) Wnt β-catenin-pathway as a molecular target for future anti-cancer therapeutics. Int. J. Cancer 113, 515. 16 Lepourcelet, M. et al (2004) Small-molecule antagonists of the oncogenic Tcf/β-catenin protein complex. Cancer Cell 5, 91. 17 Brummelkamp T.R. et al (2002) A system for stable expression of short interfering RNAs in mammalian cells. Science 296,550. 18 Aigner, A. (2006) Delivery Systems for the Direct Application of sirnas to Induce RNA Interference (RNAi) In Vivo. J Biomed Biotechnol. 2006, 71659. 19 Barker, N. and Clevers H. (2006) Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov. 5, 997 93

Chapter 6 94