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

Similar documents
Summary and Concluding Remarks

CHAPTER 6 SUMMARIZING DISCUSSION

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

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

Immature organoids appear after hours.

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

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

From crypt stem cell to colorectal cancer

SUPPLEMENTARY INFORMATION

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

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

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

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

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

Supplemental Experimental Procedures

SUPPLEMENTARY INFORMATION

Snai1 regulates cell lineage allocation and stem cell maintenance in the mouse intestinal epithelium

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

Supplementary Figure 1: Hsp60 / IEC mice are embryonically lethal (A) Light microscopic pictures show mouse embryos at developmental stage E12.

General Structure of Digestive Tract

Wnt signaling. Ramray Bhat.

Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/-

Nature Immunology: doi: /ni Supplementary Figure 1. Huwe1 has high expression in HSCs and is necessary for quiescence.

Biochemistry of Carcinogenesis. Lecture # 35 Alexander N. Koval

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

Ex vivo Human Antigen-specific T Cell Proliferation and Degranulation Willemijn Hobo 1, Wieger Norde 1 and Harry Dolstra 2*

SSM signature genes are highly expressed in residual scar tissues after preoperative radiotherapy of rectal cancer.

ANAT3231: lectures overview

LECTURE 12: MUCOSAL IMMUNITY GUT STRUCTURE

Meeting Report. From December 8 to 11, 2012 at Atlanta, GA, U.S.A

Anatomy & Histology of The Small intestine

ANAT3231: lectures overview

DIGESTIVE TRACT ESOPHAGUS

Supplemental Figure S1. RANK expression on human lung cancer cells.

Supplementary Information and Figure legends

Enterprise Interest Nothing to declare

Alan G. Casson FRCSC Professor of Surgery & VP Integrated Health Services University of Saskatchewan & Saskatoon Health Region

Imaging of glycolytic metabolism in primary glioblastoma cells with

SUPPLEMENTARY INFORMATION

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human)

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

Fig. S1. Upregulation of K18 and K14 mrna levels during ectoderm specification of hescs. Quantitative real-time PCR analysis of mrna levels of OCT4

SUPPLEMENTARY FIGURES

Aminimal definition of stemness states that a stem

GFP/Iba1/GFAP. Brain. Liver. Kidney. Lung. Hoechst/Iba1/TLR9!

The Hallmarks of Cancer

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

Stem cells in homeostasis and cancer of the gut

Extended Mammosphere Culture of Human Breast Cancer Cells

Supplementary Figure 1. Genotyping strategies for Mcm3 +/+, Mcm3 +/Lox and Mcm3 +/- mice and luciferase activity in Mcm3 +/Lox mice. A.

Development of Carcinoma Pathways

PBS Class #2 Introduction to the Immune System part II Suggested reading: Abbas, pgs , 27-30

Supplementary Figure 1 Lymphocytes can be tracked for at least 4 weeks after

SUPPLEMENTARY INFORMATION

Ahtiainen et al., http :// /cgi /content /full /jcb /DC1

SUPPLEMENTARY INFORMATION

Supplementary Figure 1. H-PGDS deficiency does not affect GI tract functions and anaphylactic reaction. (a) Representative pictures of H&E-stained

Aberrant cell Growth. Younas Masih New Life College of Nursing Karachi. 3/4/2016 Younas Masih ( NLCON)

CYTOMORPHOLOGY MODULE 28.1 INTRODUCTION OBJECTIVES 28.2 GENERAL GUIDELINES. Notes

A adipose cells. B capillary. C epithelium

Parenteral Nutrition. Outline. Potential Biomarkers for Use in Intestinal Adaptation. Jejunum is primary site of digestion and absorption

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

(b) Stomach s function 1. Dilution of food materials 2. Acidification of food (absorption of dietary Fe in small intestine) 3. Partial chemical digest

DYNAMICS OF CHANGES IN MORPHOLOGICAL STRUCTURES WITH PREPARATIONS OF NUCLEIC NATURE ADMINISTERED

Evaluation of directed and random motility in microslides Assessment of leukocyte adhesion in flow chambers

A classification of epithelial tissues

Dr. Abeer.c.Yousif. Histology -2 nd stage. What is histology?

SUPPLEMENTARY INFORMATION

Gut Microbiota and IBD. Vahedi. H M.D Associate Professor of Medicine DDRI

Dr. Heba Kalbouneh. Dr. Heba Kalbouneh. Dr. Heba Kalbouneh

Beuling _SupplFig. 1

Regulation of Gene Expression in Eukaryotes

VEGFR2-Mediated Vascular Dilation as a Mechanism of VEGF-Induced Anemia and Bone Marrow Cell Mobilization

Tissues and organs PART 1

Opposing Activities of Notch and Wnt Signaling Regulate Intestinal Stem Cells and Gut Homeostasis

Resident cardiac stem cells: how to find and use them

Supplemental Information. Otic Mesenchyme Cells Regulate. Spiral Ganglion Axon Fasciculation. through a Pou3f4/EphA4 Signaling Pathway

Epithelial tumors. Dr. F.F. Khuzin, PhD Dr. M.O. Mavlikeev

The autoimmune disease-associated PTPN22 variant promotes calpain-mediated Lyp/Pep

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

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

Nature Methods: doi: /nmeth Supplementary Figure 1

Supplementary Figure S1. Venn diagram analysis of mrna microarray data and mirna target analysis. (a) Western blot analysis of T lymphoblasts (CLS)

Cells and Tissues 3PART C. PowerPoint Lecture Slide Presentation by Patty Bostwick-Taylor, Florence-Darlington Technical College

Supplemental Information. Tissue Myeloid Progenitors Differentiate. into Pericytes through TGF-b Signaling. in Developing Skin Vasculature

LIST OF ORGANS FOR HISTOPATHOLOGICAL ANALYSIS:!! Neural!!!!!!Respiratory:! Brain : Cerebrum,!!! Lungs and trachea! Olfactory, Cerebellum!!!!Other:!

The toll-like receptor 4 ligands Mrp8 and Mrp14 play a critical role in the development of autoreactive CD8 + T cells

COPD lungs show an attached stratified mucus layer that separate. bacteria from the epithelial cells resembling the protective colonic

Haematopoietic stem cells

Supplemental Figure 1. Intracranial transduction of a modified ptomo lentiviral vector in the mouse

Histology = the study of tissues. Tissue = a complex of cells that have a common function

Determination of the temporal pattern and importance of BALF1 expression in Epstein-Barr viral infection

Phosphate buffered saline (PBS) for washing the cells TE buffer (nuclease-free) ph 7.5 for use with the PrimePCR Reverse Transcription Control Assay

SUPPLEMENTAL MATERIAL. Supplementary Methods

CRIPTO-1 A POSSIBLE NEW BIOMARKER IN GLIOBLASTOMA MULTIFORME PIA OLESEN, MD, PHD STUDENT

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2

Epithelium Characteristics cont. 2. Apical Surface

Multistep nature of cancer development. Cancer genes

Superior Fluorescent Labeling Dyes Spanning the Full Visible Spectrum...1. Trademarks: HiLyte Fluor (AnaSpec, Inc.)

Transcription:

INTRODUCTION: The intestinal epithelium is one of the most rapidly dividing tissues in the mammalian organism, with near complete replacement of the epithelial monolayer every 7-10 days (Wright et al., 1984). The start site of this impressive cellular renewal and division begins with a pool of intestinal epithelial stem cells (IESCs) that reside at the base of the crypts. The crypts are small depressions in the intestine that extend to the lamina propria and house both the IESCs and supporting niche cells at its base (Cheng et al., 1974). As the IESCs divide, their progeny migrate upward to the top of the crypt towards the lumen, into what is termed the transit-amplifying zone. Once in this zone, the transit amplifying cells mature into one of four well defined postmitotic cell lineages: absorptive enterocytes, goblet cells, enteroendocrine cells, and Paneth cells (Figure 1). With the exception of the Paneth cells, all of these cell types continue to migrate upward into the large finger-like villi, where they live for several days before dying and being sloughed off at the tip of the villus (Cheng et al., 1974). Each cell type in the villus has a distinct role; absorptive enterocytes are responsible for absorbing nutrients and transferring them to the bloodstream, goblet cells secrete a protective mucus layer over the epithelium, and enteroendocrine cells produce Figure 1. Diagram depicting crypt-villus axis, showing direction of cell migration from the crypt base to villus tip. important signaling hormones (Cheng et al., 1974). Paneth cells are unique in that they migrate downward from the transit-amplifying zone back to the crypt base where they secrete antimicrobial peptides and serve as niche cells to the stem cells (Sato et al., 2011). This rapid

cycle of tissue renewal, coupled with well-defined post-mitotic lineages, make the intestinal epithelium an ideal model for the study of factors that regulate the potency and self renewing capability of the IESCs, which to date remain poorly understood. Mutations or deficiencies in these properties of stem cells, collectively considered stemness, lie at the heart of many gastrointestinal disorders. Establishing a better understanding of the processes and transcription factors that regulate stemness is critical to developing novel cellular and gene-based therapies for those afflicted with GI disorders and cancers. The Sry-related HMG box (Sox) is a group of highly conserved transcription factors that is broadly expressed across tissue systems, particularly during embryogenesis. Interestingly, knockout experiments of many Sox factors demonstrate a failure to undergo organogenesis and implicate this group of transcription factors as potent regulators of stem cell maintenance and differentiation (Gracz et al., 2011). Sox4 causes lethality at embryonic day 10.5 in knockout mice due to malformations in the endocardial ridge (Schilham et al., 1996). In addition, Sox4 has been shown to play an important role in B-cell, T-cell, and neuronal proliferation and differentiation, providing further evidence that Sox4 has crucial regulatory functions in the maintenance and differentiation of stem cell populations (Schilham et al., 1996, 1997, Bergsland et al, 2006). Recently, in situ hybridization has been used to show that Sox4 is expressed at the base of intestinal crypts, in the known stem cell Figure 2. 10X and 40X in situ hybridization showing Sox4 mrna localizing to the base of the crypt (Magness, unpublished data). compartment (Figure 2). Given the role of Sox4 in other tissues and its localization to the stem cell zone, these preliminary data collectively suggest

that Sox4 may be playing a critical role in maintaining and regulating the differentiation of the IESCs. In addition to regulating proliferation and differentiation in a variety of tissues, Sox4 has long been implicated as playing an oncogenic role in a variety of cancer types. There exists a large body of literature showing Sox4 expression is upregulated in cancers of virtually every type (for a summary of this research see Table 1, adapted from Vervoort et al., 2013). Despite this sizable and growing body of literature, little is known about how Sox4 is contributing to tumorigenesis in these cancers. A landmark paper demonstrated Sox4 to be a master regulator of the biological process called Epithelial-Mesenchymal Transition (EMT) in breast tissue. In this process, cells begin to lose their epithelial traits such as cell-cell adhesion and polarity, and take on more mesenchymal characteristics such as the ability to migrate and invade other tissue types (Christofori et al., 2013). This process of EMT mediates epithelial tumor metastasis by generating cells capable of invading the basement membrane and entering the circulatory system. Recently, cancer cell lines have been used to show a role for Sox4 induced EMT contributing to tumor progression in breast, prostate, and liver cancers. These experiments demonstrated that overexpression of Sox4 was sufficient to cause cells to undergo EMT and become increasingly invasive and migratory in vitro. However, almost no research on the role of Sox4 in tumor metastasis via EMT induction has been examined in vivo. And while it has long been known that EMT plays an important process in human colorectal cancer metastasis, the mechanistic and regulatory details of this potentially Sox4-dependent EMT remains largely unknown (Loboda et al., 2011). I hypothesize that Sox4 is the driving force for EMT in colorectal cancer, by promoting tumor progression and metastasis.

Table 1. Summary of existing research showing Sox4 upregulation in many cancer types. Table adapted from Vervoort et al., 2013. METHODS: The Sox4 fl/fl :VilCre Mouse Model: A genome-wide knockout of Sox4 results in embryonic lethality. To study the role of Sox4 in the adult intestine, we generated conditional transgenic knockout mice using the Cre-lox recombination system (Marioue et al., 2004). In our model, Cre-recombinase expression is driven by a villin promoter, a gene that is expressed exclusively in the intestinal epithelium. This system allows for an intestinal specific knockout of Sox4 (Figure 3). To attempt to discern the role of Sox4 in the epithelium, this mouse model will be examined using a variety of techniques including RNA and protein analysis.

The APC min mouse model:. The APC gene is an component of the destruction complex which targets cytoplasmic catenin for degradation. This mouse model contains a truncation in one copy of the APC gene Following random inactivation of the remaining copy of APC, Wnt signaling is constitutively turned on and -catenin is localized to the nucleus where it can act as a transcription factor, promoting Figure 3. Schematic diagram showing excision of Sox4 by Cre recombinase protein specifically in the intestinal epithelium. genes for proliferation and cell growth. In this model, mice begin to display multiple intestinal neoplasias as early as 50-60 days, with more severe and damaging tumor growth coming at about 14 weeks (Moser et al. 1990, Su et al. 1992). Tissue Processing: Knockout mice with littermate controls for immunohistochemistry and quantification were sacrificed at 8-9 weeks after birth. Intestines were dissected out and divided into three equal length segments, with segment 1 representing the most proximal section and segment 3 the most distal. These sections were then flushed with ice cold Phosphate Buffered Saline (PBS) and fixed in 4% Paraformaldehyde (PFA). After fixing for 24 hours at 4 C, tissue was transferred to a 30% sucrose solution before it was filleted open to expose the lumen and give a flat intestinal segment. The flattened segments were then rolled up in a manner similar to a swiss-roll, and frozen in optimal cutting temperature embedding media (OCT). The swiss-rolls were then sectioned at a thickness of 6-8 µm, Immunohistochemistry and quantification: Staining was performed according to previously described protocols (Gracz et al. 2013). Primary antibodies used were raised against murine Ki67, lysozyme, Ep-CAM, Mucin-2, Chromogranin-A, and Sox4 and visualized with Cy3 conjugated

secondary antibodies. Nuclear staining was done using bisbenzimide, following primary and secondary antibody incubation. For quantification, crypts counted had to meet certain criteria; crypts had to be open to the luminal space and extend their base to the lamina propria or muscle layer. For goblet cells and enteroendocrine cells, which exist in both the crypt and the villus, images for quantification were taken and cell number was quantified separately in the crypt and in the villus. Imaging analysis software (ImageJ version 1.48a) was used for cell quantification and morphometric analysis. Quantitative Real-Time PCR (qrt-pcr): After sacrificing mice at 8-12 weeks of age, 0.5 cm segments of jejunal segment number 2 and proximal and distal colon were dissected out and placed into RNAlater (Qiagen) for 24 hours at 4 C. RNA was then extracted and purified, and cdna was made using the iscript cdna Synthesis Kit (Bio-Rad). qrt- PCR was performed with SsoFast Probes Supermix with ROX. qrt-pcr data was analyzed using StepOne Plus Software. Tumor dissociation and flow cytometry APC min mice between 14-16 weeks were sacrificed, and the intestines were removed and flushed as normal. The intestines were then opened longitudinally, and tumors were dissected out under a dissecting microscope. Figure 4. A) Confocal image of SOX4 protein expression, B) overlayed with nuclei. C) Quantification of SO4 high and low expressing cell position, n=3 animals, 10 crypts per animal.

These tumors were minced with a razor blade to a paste-like consistency and placed in an EDTA chelation buffer for 45 minutes at 4 C. These chunks of tissue were then spun down at 1500 rpm for 5 minutes and resuspended in pre-warmed Hank s Balanced Salt Solution (HBSS, Gibco) containing dispase and DNase. This suspension was placed at 37 C and shaken lightly every 1.5 minutes, with samples taken to observe dissociation progress after each step. After 6-10 minutes of this enzymatic digestion, the cells were put through a 40 µm filter directly into ice cold PBS containing 10% Fetal Bovine Serum (FBS). The single cells were then washed twice with cold PBS, and resuspended in IESC media (DMEM, 10% FBS, B27, N2, Pen/Strep, Y27632, NAC). Cells in IESC media were stained for 1 hours with EpCAM antibody (EpCAM FITC, Biolegend), and washed twice. Live/dead cell exclusion stains were added 10 minutes before flow cytometry. All flow cytometry was performed using the Beckman-Coulter Cyan machine in the Lineburger Cancer Center. RESULTS Characterizing Sox4 expression patterns While in situ hybridization data showed that Sox4 was expressed in the crypts, more specific localization of Sox4 expression was needed to associate Sox4 expression with cell position in the crypt. To investigate the expression pattern of SOX4 with greater accuracy, a Sox4 antibody was used to immunofluorescently label Sox4 expressing cells. In order for this stain to work, a protocol first had to be optimized. This was devised to include antigen retrieval, an extended primary antibody incubation time, and signal amplification using the biotin-streptavidin system. This protocol yielded highly specific binding with little background signal, and revealed a clear and distinct expression pattern.

SOX4 was expressed at both a high and low level in different cells of the crypt (Figure 4). The cell position of each subset was quantified and showed that Sox4 low expressing cells were primarily at the crypt base, around position +1 to +3 and becoming less frequent towards the top of the crypt. The Sox4 high expressing cells, however, were found most frequently at cell position +5, a position that is consistent with that of the reserve ISC population (Van Landeghem et al., 2012). Loss of Sox4 results in crypt hyperplasia and hyperproliferation Upon first examination of knockout mouse tissue, there was no overt phenotype, with knockout mice exhibiting no change in body mass and appearing to have normal crypt-villus architecture. However, closer examination revealed that crypts in knockout animals were hyperplastic, extending deeper than those in the control animals. To ensure any significant morphological changes were discovered, measurements of both crypt depth and width as well as villus height and width, were measured using ImageJ software. Morphometric analysis confirmed that crypts were on average 150% larger in knockout animals compared to controls. Due to the proliferative nature of cells in the crypt, it was hypothesized that hyperproliferation might be the source of the hyperplastic crypt morphology.

Figure 5. A, B, C, D) Confocal imagery of EpCAM with quantification of crypt and villus dimensions. E, F, G) Confocal images of KI67 staining with quantification, n=3 animals, 25 crypts per animal. H) qpcr on several direct Wnt target genes, n=3 animals, * indicates p<.05 To test whether or not an increase in cell proliferation was responsible for the oversized crypts, slides were stained with the proliferative marker KI67. The percentage of KI67 positive crypt cells were quantified and this showed that there was increased proliferation in the crypts of Sox4 knockout mice compared to controls (Figure 5). While this increase was obvious in many crypts, it appeared more subtly in others, but overall was statistically significant (n=3 animals, 25 crypts per animal, p<0.05). Additionally, it was qualitatively observed that there appeared to be more Ki67 positive cells lower in the crypt near the stem cell zone and higher towards the top of the crypt, although cell position has yet to be quantified. Sox4 regulates Wnt signaling

Due to the significant in KI67 positive proliferating cells, it was hypothesized that Sox4 may be involved in regulating Wnt signaling, a well studied pathway that controls cellular growth and proliferation 14. Quantitative Real-Time PCR on several direct Wnt target genes such as Myc, Cyclin D-1, and CD44 confirmed that there was increased Wnt signaling in the knockout animals. This result was also observed in tissue from both the proximal and distal colon sections. These data suggest that Sox4 is acting as an inhibitor of proliferation, at least partially by downregulating Wnt signaling. Sox4 regulates secretory lineage allocation in the intestinal epithelium Figure 6. A-F) Example images of goblet, enteroendocrine, and Paneth cells with quantification in crypt and villus. G) Quantification of Paneth cell numbers at each cell position. For Goblet cells and enteroendocrine cells n=3 animals, 50 crypts/villi per animal. For Paneth cells, n=3 animals, 25 crypts per animal. * indicates p<.05

Investigating the secretory lineages revealed an interesting pattern, marked by a significant decrease in enteroendocrine cells and a concomitant increase in the number of Paneth cells (Figure 6). No significant change in the number of goblet cells was observed and goblet cell numbers per crypt and per villus was highly variable across biological replicates. Sox4 high expressing cells are not label-retaining To directly interrogate whether or not Sox4 expressing cells were acting in the label-retaining cell described in Buczacki et al. 2013, cells were labeled with the thymidine analog EdU over a 28 day period using subcutaneously implanted osmotic mini-pumps. Over this 28 day period, every cell that enters S phase of the cell cycle is labeled with the thymidine analog which can later be visualized with the (Click-it EdU Kit, Invitrogen). The label is then allowed to wash out, such that all cells that divide lose their fluorescent label, and those that are long-lived and dividing at a slow rate remain labeled. Because the Buczacki paper considers a cell to be label-retaining at 7 to 13 days, we examined co-localization of Sox4 and label retaining cells at three wash out timepoints: 0 hours, 1 week, and 2 weeks. The results showed that Sox4 only co-localized with label-retaining cells at a frequency of about 8-9%, and many of these cells may have been mature Paneth cells, which are also known to retain labels due to their long-lived nature (Figure 7). Additionally, all of the 8-9% of cells which exhibited co-localization were expressing Sox4 at a low level, contrary to what was predicted.

Figure 7. SOX4 and EdU colocalization at three timpoints, with quantification n=100 SOX4 expressing cells. Sox4 is highly expressed in small intestinal adenomas and is correlated with decreased EpCAM expression To begin to investigate the role of Sox4 in tumorigenesis, tissue from APC min mice was stained for Sox4, showing that it is highly expressed in cells of small intestinal adenomas. Interestingly, when stained simultaneously with Epithelial Cell Adhesion Molecule (EpCAM), it can be seen that cells highly expressing Sox4 appear to have a severely decreased expression of EpCAM (Figure 8). These same cells also begin to collapse from a polarized epithelium into an unorganized tissue mass and blend with the mesenchyme previously in the deeper tissue layers. This process is suggestive of a potential EMT and the invasion that is necessary for tumor metastasis.

Flow cytometry can also be used to validate the transition of cells from a high to low EpCAM expression profile. Following dissociation of tumors to single cells, a histogram of cells expressing EpCAM at different levels confirms that many cells are showing decreased levels of EpCAM. This flow cytometric analysis will allow us to derive an appropriate gating strategy for using fluorescent activated cell-sorting (FACS) to sort out these cell populations for subsequent RNA analysis. Figure 8. A-D) Confocal image of a small intestinal adenoma, expressing SOX4 at a high level. E). Flow cytometry overlay graph showing the EpCAM profile of cells dissociated from a tumor (red) and from healthy adjacent tissue (green). CONCLUSIONS: The expression pattern of Sox4 protein suggests that it may be acting in a context-dependent manner, in two different cell types. First, it showed that Sox4 was being expressed at a low level in the stem cells, which occupy the base of the crypt at positions +1 +3. The identity of cells in which Sox4 is expressed at a high level is a more complicated question. Early label-retaining studies propose a quiescent, slowly-dividing stem cell at the +4 position,

and a recent study using a novel Cyp1a1-H2B-YFP mouse model to more precisely locate the label-retaining cell found it predominantly at position +3, but in a variety of positions from +1 to +6. It is also well known that the transit amplifying cells occupy positions +6 and up in the crypt. Given these facts, it remains possible that Sox4 is acting at a high level in either a long-lived, label-retaining, reserve stem cell, or in the rapidly-dividing transit amplifying progenitor cells. The increased levels of KI67 by immunofluorescent quantification and qrt-pcr, combined with Sox4 expression in the stem cells, strongly suggest that Sox4 is involved in regulating the proliferation of intestinal epithelial stem cells. More specifically, the data suggests that in normal tissue, Sox4 is limiting proliferation by down-regulating genes that regulate cell cycle entry and growth through the Wnt pathway. Quantification of secretory cells in knockout animals reveals a lineage allocation defect where cells are preferentially fated towards Paneth cells rather than enteroendocrine cells in the absence of SOX4. This result is striking in light of the existence of the label-retaining secretory precursor cell that gives rise to both enteroendocrine and Paneth cell lineages described in Buczacki et al. 2013. This result lends support to the hypothesis that Sox4 high cells may be acting in a label-retaining secretory precursor, pushing this cell towards an enteroendocrine fate rather than a Paneth cell fate. Given these two functions, a model of Sox4 s context dependent manner can be drawn out, where Sox4 is acting at a low level in the stem cells to regulate proliferation, and at a higher level in either a progenitor or label retaining cell to regulate secretory cell fate decision (Figure 9). While the label-retaining cell studies do not support the hypothesis that Sox4 is highly expressed in these cells to regulate their fate decision, it does not necessarily rule it out. Since only 100 SOX4 positive cells were counted, it remains possible that Sox4 is turned on very

Figure 9. Diagram with proposed model of Sox4 in normal intestinal epithelium transiently in the label-retaining secretory precursor and we have simply not caught these cells at the right time in their lifespan. However, given the data it seems more likely that Sox4 is regulating a short-lived transit amplifying progenitor cell. This joint function of regulating proliferation and differentiation is consistent with roles shown in other tissues (Schilham et al., 1996, 1997, Bergsland et al, 2006). In the cancer context, it appears as though Sox4 may have a role unique from its function in normal intestinal epithelium homeostasis. The correlation between increased Sox4 levels and decreased EpCAM levels suggests that a possible EMT may be occurring in our mouse model of colorectal cancer. While it is well established that EMT is an essential program in colorectal cancer metastasis, the driver of this program remains unknown. The highly aberrant expression

of Sox4 in the adenomas combined with previous work in breast tissue implicating Sox4 as a master regulator of EMT, provides tanatalizing clues that Sox4 may be responsible for promoting the EMT program in colorectal cancer. While further study in this area is certainly needed, confirmation of this finding would have profound impact on colorectal cancer research and would identify Sox4 as a therapeutic target to manage tumor progression and metastasis. References: 1. Wright N, Allison, M. The Biology of Epithelial Cell Populations. Oxford: Clarindon, 1984. 2. Cheng H. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. II. Mucous cells. Am J Anat 141: 481-501, 1974. 3. Cheng H. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. IV. Paneth cells. Am J Anat 141: 521-535, 1974. 4. Cheng H, and Leblond CP. Origin, differentiation and renewal of the four main epithelial cell types in mouse small intestine. I. Columnar cell. Am J Anat 141: 461-479, 1974. 5. Cheng H, and Leblond CP. Origin, differentiation and renewal of the four main epithelial cell types in mouse small intestine. III. Entero-endocrine cells. Am J Anat 141: 503-519, 1974. 6. Cheng H, and Leblond CP. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Unitarian Theory of the origin of the four epithelial cell types. Am J Anat 141: 537-561, 1974. 7. Sato T, van Es JH, Snippert HJ, Stange DE, Vries RG, van den Born M, Barker N, Shroyer NF, van de Wetering M, and Clevers H. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469: 415-418, 2011.

8. Gracz AD, and Magness ST. Sry-box (Sox) transcription factors in gastrointestinal physiology and disease. Am J Physiol Gastrointest Liver Physiol 300: G503-515, 2011. 9. Schilham MW, Oosterwegel MA, Moerer P, Ya J, de Boer PA, van de Wetering M, Verbeek S, Lamers WH, Kruisbeek AM, Cumano A, and Clevers H. Defects in cardiac outflow tract formation and pro- B-lymphocyte expansion in mice lacking Sox-4. Nature 380: 711-714, 1996. 10. Bergsland M, Werme M, Malewicz M, Perlmann T, and Muhr J. The establishment of neuronal properties is controlled by Sox4 and Sox11. Genes Dev 20: 3475-3486, 2006. 11. Schilham MW, Moerer P, Cumano A, and Clevers HC. Sox-4 facilitates thymocyte differentiation. Eur J Immunol 27: 1292-1295, 1997. 12. Marjou FE, Robine, S. Tissue-specific and inducible Cre-mediated recombination in the gut epithelium. Genesis 39: 186-193, 2004. 13. Gracz AD, Puthoff BJ, Magness ST. Identification, isolation, and culture of intestinal epithelial stem cells from murine intestine. Methods mol bio 879: 89-107, 2012. 14. Gregorieff A, Pinto D, Begthel H, Destree O, Kielman M, and Clevers H. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 129: 626-638, 2005. 15. Buczacki A, Zecchini H, Winton DJ. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 000; 0-5, 2013. 16. Christofori et al. Sox4 is a master regulator of Epithelial-Mesenchymal Transition by controlling Ezh2 expression and epigenetic reprogramming. Cancer Cell 23: 768-783, 2013. 17. Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 247: 322-324, 1990.

18. Su LK, Kinzler LK, Vogelstein B, Preisinger AC, Moser AR, Luongo C, Gould KA, Dove WF. Multiple intestinal neoplasia cause by a mutation in the murine homolog of the APC gene. Science 256: 668-670, 1992. 19. Vervoort SJ, Boxtel RV, Coffer PJ. The role of SRY-related HMG box transcription factor 4 (SOX4) in tumorigenesis and metastasis: friend or foe? Oncogene 32: 3397-3409, 2013. 20. Loboda, A, Nebozhyn, MV, Watters JW, Buser CA, Shaw PM, Huang PS, Veer LV, Tollenaar RA, Jackson DB, Agrawal D, Dai H, Yeatman TJ. EMT is the dominant program in human colon cancer. BMC Medical Genomics 4: 9-19, 2011. 21. Landeghem LV, Santoro MA, Krebs, AE, Dehmer JJ, Gracz AD, Scull BP, McNaughton K, Magness ST, Lund PK. Activation of two distinct Sox9-EGFP-expressing intestinal stem cell populations during crypt regeneration after irradiation. Am J Physiol Gastrointest Liver Physiol 10: 302-312, 2012.