ArticlePDF Available

LGR5 positivity defines stem-like cells in colorectal cancer

Authors:

Abstract and Figures

Like normal colorectal epithelium, colorectal carcinomas (CRC) are organized hierarchically and include populations of cells with stem-like properties. Leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5) is associated with these stem cells in normal colorectal epithelium, however the precise function of LGR5 in CRC remains largely unknown. Here, we analyzed the functional and molecular consequences of short hairpin RNA-mediated silencing of LGR5 in CRC cell lines SW480 and HT-29. Additionally, we exposed Lgr5-EGFP-IRES-CreERT2 mice to azoxymethane/dextrane sodium sulfate (AOM/DSS) which induces inflammation-driven colon tumors. Tumors were then flow-sorted into fractions of epithelial cells that expressed high or low levels of Lgr5 and were molecularly characterized using gene expression profiling and array comparative genomic hybridization. Silencing of LGR5 in SW480 CRC cells resulted in a depletion of spheres but did not affect adherently growing cells. Spheres expressed higher levels of several stem cell-associated genes than adherent cells, including LGR5. Silencing of LGR5 reduced proliferation, migration and colony formation in vitro, and tumorigenicity in vivo. In accordance with these results, NOTCH signaling was down-regulated upon LGR5 silencing. In AOM/DSS-induced colon tumors Lgr5 high cells showed higher levels of several stem cell-associated genes and higher Wnt signaling than Lgr5 low tumor cells and Lgr5 high normal colon cells. Array comparative genomic hybridization revealed no genomic imbalances in either tumor cell fraction. Our data elucidate mechanisms that define the role of LGR5 as a marker for stem-like cells in CRC.
Expression levels of LGR5 in CRC cell lines and shRNA-mediated silencing of LGR5 in SW480 and HT-29 CRC cells. (A) LGR5 mRNA levels in CRC cell lines as determined by real-time qRT?PCR. LGR5 mRNA levels were normalized to YWHAZ and are expressed as fold changes relative to normal colorectal epithelial cells. Columns and error bars represent means ? SEM of two independent experiments using triplicate measurements in each experiment. (B) LGR5 silencing efficiency in CRC cell lines SW480 and HT-29 upon lentiviral shRNA transduction as determined by real-time qRT?PCR. LGR5 mRNA levels were normalized to YWHAZ and are expressed as fold changes relative to control. Columns and error bars represent means ? SEM of three independent experiments using triplicate measurements in each experiment. (C) Representative phase-contrast images of SW480 and HT-29 cells upon LGR5 silencing. SW480 is composed of two morphologically distinct subpopulations (spheres and adherent cells), whereas HT-29 shows one phenotype. After LGR5 silencing, spheres in SW480 were completely lost with no apparent changes in HT-29. Scale bars, 100 ?m. (D) Expression of LGR5 in spheres versus adherent cells of SW480. Spheres express high levels of LGR5, whereas in adherent cells, virtually no LGR5 expression can be detected by real-time qRT?PCR. LGR5 mRNA levels were normalized to YWHAZ and are expressed as fold changes relative to control. Data represent means ? SEM of three independent experiments using triplicate measurements in each experiment. *P < 0.05, **P < 0.005 and ***P < 0.0005.
… 
Content may be subject to copyright.
Published by Oxford University Press 2013.
Carcinogenesis vol.35 no.4 pp.849–858, 2014
doi:10.1093/carcin/bgt377
Advance Access publication November 26, 2013
LGR5 positivity denes stem-like cells in colorectalcancer
DanielaHirsch1,2, NickBarker3, NicoleMcNeil1, YueHu1,
JordiCamps1, KatherineMcKinnon4, HansClevers5,
ThomasRied1,* and TimoGaiser6
1Section of Cancer Genomics, Genetics Branch, Center for Cancer Research,
National Cancer Institute, National Institutes of Health, Bethesda, MD
20892, USA, 2Experimental Medicine and Therapy Research, University of
Regensburg, 93053 Regensburg, Germany, 3Institute of Medical Biology,
Singapore 138648, Singapore, 4FACS Core Facility, Vaccine Branch, Center
for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, MD 20892, USA, 5Hubrecht Institute, 3584 CT Utrecht,
The Netherlands and 6Institute of Pathology, University Medical Center
Mannheim, 68167 Mannheim, Germany
*To whom correspondence should be addressed. Tel:+1 301 5943118;
Fax: +1 301 4021204;
Email: riedt@mail.nih.gov
Correspondence may also be addressed to Timo Gaiser. Tel:+49 621
3832556; Fax: +49 621 3832005;
Email: timo.gaiser@umm.de
Like normal colorectal epithelium, colorectal carcinomas (CRCs)
are organized hierarchically and include populations of cells with
stem-like properties. Leucine-rich-repeat-containing G-protein-
coupled receptor 5 (LGR5) is associated with these stem cells
in normal colorectal epithelium; however, the precise function
of LGR5 in CRC remains largely unknown. Here, we analyzed
the functional and molecular consequences of short hairpin
RNA-mediated silencing of LGR5 in CRC cell lines SW480 and
HT-29. Additionally, we exposed Lgr5-EGFP-IRES-CreERT2
mice to azoxymethane/dextrane sodium sulfate (AOM/DSS),
which induces inammation-driven colon tumors. Tumors were
then ow-sorted into fractions of epithelial cells that expressed
high or low levels of Lgr5 and were molecularly characterized
using gene expression proling and array comparative genomic
hybridization. Silencing of LGR5 in SW480 CRC cells resulted
in a depletion of spheres but did not affect adherently growing
cells. Spheres expressed higher levels of several stem cell-asso-
ciated genes than adherent cells, including LGR5. Silencing of
LGR5 reduced proliferation, migration and colony formation in
vitro and tumorigenicity in vivo. In accordance with these results,
NOTCH signaling was downregulated upon LGR5 silencing. In
AOM/DSS-induced colon tumors, Lgr5 high cells showed higher
levels of several stem cell-associated genes and higher Wnt sign-
aling than Lgr5 low tumor cells and Lgr5 high normal colon
cells. Array comparative genomic hybridization revealed no
genomic imbalances in either tumor cell fraction. Our data elu-
cidate mechanisms that dene the role of LGR5 as a marker for
stem-like cells in CRC.
Introduction
Colorectal tumorigenesis is associated with the accumulation of a
number of specic genetic changes, which drive the transition from
normal epithelium through adenomas to invasive carcinomas. These
genetic changes include mutations of specic genes, such as adeno-
matous polyposis coli (APC) and KRAS, and tumor-specic genomic
imbalances (1–3). Similar to normal colorectal epithelium, colorectal
tumors consist of heterogeneous cell populations at various levels
of differentiation. Although a few years ago all cells within a tumor
were considered to be tumorigenic, recent ndings suggested that
only a subpopulation of tumor cells can regenerate the tumor (4,5).
These cells, termed cancer stem cells (CSCs), may also be involved
in therapy resistance, tumor relapse and metastasis. Accordingly, the
identication and characterization of these cells was the subject of
considerable research efforts. With respect to colorectal carcinomas
(CRCs), putative CSCs can be identied by leucine-rich-repeat-con-
taining G-protein-coupled receptor 5 (Lgr5; also known as G-protein-
coupled receptor 49, Gpr49). Lgr5, a Wnt target gene that acts as
receptor for the Wnt agonist R-spondin, is a marker gene for adult
intestinal stem cells as revealed by in vivo lineage tracing (6–8).
Selective deletion of Apc in the mouse in either Lgr5 positive intes-
tinal stem cells or more differentiated cells revealed that mainly the
Lgr5 positive stem cell fraction is capable of forming tumors upon
Wnt pathway activation, suggesting Lgr5 positive stem cells as the
cells-of-origin of intestinal epithelial tumors (9). Although the cell-
of-origin for tumorigenesis and the CSC, which propagates the
tumor, need not necessarily be identical, in vivo lineage tracing pro-
vides direct evidence for a stem cell activity of Lgr5 positive cells
in mouse intestinal adenomas generated by deletion of Apc in Lgr5
positive stem cells (10,11). Resembling the situation in normal intes-
tinal epithelium, adenomas contain a small fraction of Lgr5 positive
cells (5–10%) that are able to generate all cell types present within
the adenomas, including additional Lgr5 positive cells (11). In human
CRC, LGR5 expression is highly enriched in EPHB2 positive cells,
which have similar expression proles to normal intestinal stem cells
and—in contrast to EPHB2 negative cells—display reproducible
tumorigenic capacity in immunodecient mice (12). Cataloging the
genetic idiosyncrasies of LGR5 positive and negative cells might help
to identify the mechanisms that cause these differences in tumorigenic
potential. We have therefore investigated the functional and molecular
consequences of short hairpin RNA (shRNA)-mediated LGR5 silenc-
ing in CRC cell lines SW480 and HT-29. To date, studies on LGR5 in
primary CRC samples have been constrained by the lack of a reliable
antibody against LGR5. We therefore induced inammation-driven
colon tumors in mice that were engineered to contain one enhanced
green uorescent protein (EGFP)-tagged Lgr5 allele (6). This allowed
ow cytometric separation of Lgr5 high and low cells based on GFP
expression and thus enabled a genome and transcriptome characteri-
zation of these two cell fractions. Our loss-of-function experiments
conclusively indicate that LGR5 acts as a marker for stem-like cells
in CRC.
Materials and methods
Cell lines and lentiviral transduction
The six human CRC cell lines (Caco-2, HCT 116, HT-29, SW480, SW620 and
T84) were purchased from the American Type Culture Collection (Manassas,
VA). All cell lines were cultured in media as recommended by the American
Type Culture Collection supplemented with fetal bovine serum (10% v/v),
-glutamine (2 mM), penicillin (100 U/ml) and streptomycin (100 µg/ml).
Lentiviral shRNA transduction of SW480 and HT-29 cells was done using
high-titer lentivirus (Clone ID: V3LHS_635055, Open Biosystems, Thermo
Fisher Scientic, Lafayette, CO) according to the manufacturer’s instructions.
Mice
Athymic nude mice (strain NCr-nu/nu) were obtained from Frederick National
Laboratory for Cancer Research (Frederick, MD). Heterozygous Lgr5-EGFP-
IRES-CreERT2 mice [strain B6.129P2-Lgr5tm1(cre/ERT2)Cle/J, henceforth
referred to as 'Lgr5-EGFP mice'] were ordered from Jackson Laboratory (Bar
Harbor, ME) (6). All mice were bred and housed in a pathogen-free environ-
ment and used in experiments in accordance with institutional guidelines at
the Center for Cancer Research, National Cancer Institute, National Institutes
of Health. All experimental procedures conducted in this study were approved
by the Animal Care and Use Committee of the National Institutes of Health.
Abbreviations: AOM, azoxymethane; APC, adenomatous polyposis coli; CRC,
colorectal carcinoma; CSC, cancer stem cell; DSS, dextrane sodium sulfate;
EGFP, enhanced green uorescent protein; LGR5, leucine-rich-repeat-containing
G-protein-coupled receptor 5; mRNA, messenger RNA; qRT–PCR, quantitative
reverse transcription–polymerase chain reaction; shRNA, short hairpin RNA.
849
D.Hirsch etal.
Tumorigenicityassay
Tumorigenicity assay was performed as described previously (13).
Microarray gene expression proling of celllines
Total RNA was isolated from SW480 shLGR5 and control cells and from
SW480 spheres and adherent cells using the RNeasy Mini Kit (Qiagen,
Hilden, Germany) including DNase Itreatment (RNase-Free DNase Set,
Qiagen). RNA integrity was assessed by 2100 Bioanalyzer (RNA 6000
Nano LabChip Kit, Agilent Technologies, Santa Clara, CA). Appropriate
LGR5 status was confirmed by real-time quantitative reverse transcrip-
tion–polymerase chain reaction (qRT–PCR). Total RNA (700 ng) was
labeled using the Quick Amp Labeling Kit, one-color (Agilent) and sub-
sequently hybridized on Human GE 4x44K v2 Microarrays (Agilent)
according to the manufacturer’s protocol version 6.5. Slides were scanned
with microarray scanner G2565BA (Agilent). Images were analyzed and
data were quality controlled using Feature Extraction software version
10.7.1.1 (Agilent).
Carcinogen-induced inammation-driven colon tumorigenesismodel
To induce colon tumors, Lgr5-EGFP mice aged 2–4 months were injected
with azoxymethane (AOM, 12.5µg/g body weight; A5486, Sigma, St Louis,
MO) twice and subjected to three cycles of dextrane sodium sulfate (DSS,
2.5%; molecular weight = 36 000–50 000, MP Biomedicals, Solon, OH) in
the drinking water (14,15). Tumor growth was monitored by colonoscopies.
About 100days after the rst AOM injection, mice were killed.
RNA amplication and microarray gene expression proling of ow-sorted
normal mouse colons and mouse colontumors
RNA isolated from ow-sorted normal mouse colons and mouse colon
tumors was amplied together with spike-ins (1µl of a 1:50 000 dilution
per reaction, One-Color RNA Spike-In Kit, Agilent) using the Ovation Pico
WTA System (NuGEN Technologies, San Carlos, CA). The amplied com-
plementary DNA was labeled using the BioPrime® Total Genomic Labeling
Module (Invitrogen, Life Technologies, Carlsbad, CA) and subsequently 4µg
of Alexa Fluor® 3 labeled target was hybridized on Whole Mouse Genome
Microarrays 4x44K (Agilent) according to NuGEN’s Agilent Solution
Application Note #1. Scanning, image analysis and data quality control were
done as for cell lines.
DNA amplication and array comparative genomic hybridization of ow-
sorted mouse colontumors
DNA amplication and array comparative genomic hybridization were per-
formed as described previously and are summarized in Supplementary
Materials and methods, available at Carcinogenesis Online (16). Array-
based comparative genomic hybridization data have been deposited in Gene
Expression Omnibus database (data accession number: GSE46711).
Statistical analysis
Differences between groups were estimated by Student’s t-test, Fisher’s exact
test or two-way repeated measures analysis of variance (one factor repetition).
P<0.05 was considered signicant.
Microarray gene expression analysis
Log2 intensities were normalized to the 75% percentile according to the manu-
facturer’s protocol (Agilent). Only probes with intensities higher than 50 were
used for the analysis. Unsupervised hierarchical clustering with Euclidean
distance and Ward method was performed with Genomics Suite™ software
(Partek Incorporated, St Louis, MO). The corresponding functional annota-
tion of differentially expressed genes and their afliation with specic genetic
pathways were interrogated using Ingenuity Pathway Analysis software
(Ingenuity Systems, Redwood City, CA). Microarray gene expression data
have been deposited in Gene Expression Omnibus database (data accession
number: GSE46200).
Results
LGR5 is overexpressed in human CRC celllines
LGR5 expression is upregulated in primary CRC samples compared
with normal colorectal epithelium (17). Using real-time qRT–PCR,
we could show that LGR5 expression was also upregulated in CRC
cell lines Caco-2, HT-29, SW480, SW620 and T84 (Figure 1A)
(18,19). The only cell line without overexpression of LGR5 was HCT
116. This is consistent with recent ndings, suggesting that HCT 116
may not be organized hierarchically and may therefore not contain a
stem-like cell fraction (13,20,21).
LGR5 silencing leads to depletion of a morphologically distinct
subpopulation of SW480 CRCcells
To investigate the function of LGR5 in CRC, we transduced two
CRC cell lines (SW480 and HT-29), which both showed a simi-
lar moderate overexpression of LGR5, with lentiviral shRNA con-
structs. This signicantly reduced LGR5 expression compared with
empty vector control (henceforth referred to as ‘control’) cells as
assessed by real-time qRT–PCR (Figure1B). In SW480, silencing
of LGR5 led to a remarkable morphologic change, whereas HT-29
did not display an apparent difference (Figure1C). Unlike HT-29,
SW480 typically comprises two morphologically distinct subpopu-
lations, i.e. spheres and adherent cells, when cultured in serum-con-
taining medium. Upon LGR5 silencing, spheres were completely
depleted and only an adherent cell layer remained. Spheres, along
with LGR5 expression, remained undetectable in SW480 shLGR5
cells over more than 12 months of continuous passage, demon-
strating long-lasting effects of the shRNA on LGR5 expression. To
exclude that the disappearance of spheres was induced by cell split-
ting, we next separately analyzed LGR5 messenger RNA (mRNA)
expression in spheres and adherent cells of SW480 control cells.
Although LGR5 was highly expressed in spheres, adherent cells
displayed virtually no LGR5 expression, supporting our conclu-
sion that the morphologic change was caused by silencing of LGR5
(Figure1D).
LGR5 silencing in SW480 and HT-29 CRC cells reduces prolifera-
tion, migration and colony formation in vitro
To evaluate the inuence of LGR5 on cell proliferation, we com-
pared the cleavage of the tetrazolium salt WST-1 between shLGR5
and control cells. Upon silencing of LGR5, cleavage of WST-1 was
signicantly decreased in HT-29 cells (Figure 2A). SW480 cells
showed the same tendency although not reaching statistical signi-
cance (P=0.096). This might be attributable to the loss of spheres
in SW480 upon silencing of LGR5; spheres usually grow slower than
isogenic adherent cells (22). In other words, the proliferating cells of
SW480 reside within the LGR5 low adherent cells and not within the
LGR5 high spheres. This explains the small effect of LGR5 silencing
on proliferation inSW480.
The effect of LGR5 on migration was assessed by wound healing
and transwell migration assays. In the wound healing assay, SW480
shLGR5 cells tended to cover a smaller area of the initial scratch than
respective control cells at all time points, reaching statistical signi-
cance after 72 h (Figure2B). To reduce the impact of proliferation
as a confounding variable, we additionally monitored migration for
each group through transwell membranes and observed a signi-
cantly smaller number of migrated shLGR5 cells compared with con-
trol cells after incubating for 24 h (Figure2C). For HT-29, none of
the assays allowed a proper assessment of migration dependent on
LGR5 status owing to too little migratory activity of HT-29 cells in
our experiments.
In the colony formation assay, both cell lines formed signicantly
fewer colonies when LGR5 was silenced compared with control cells
(Figure2D).
LGR5 silencing reduces tumorigenic capacity of SW480 CRC cells
after xenotransplantation
To corroborate our in vitro colony formation results in vivo, we
injected CRC cells with differential LGR5 expression levels subcu-
taneously into the anks of nude mice and followed the appearance
of tumors. SW480 shLGR5 cells were less tumorigenic than control
cells and the tumors grew slower (Table I and Supplementary Figure
S1A–C, available at Carcinogenesis Online). Reduced tumorigenic-
ity was not observed for HT-29 cells upon LGR5 silencing (Table I).
However, tumors derived from HT-29 shLGR5 cells also grew slower
than those from control cells (Supplementary Figure S1D, available
at Carcinogenesis Online). Sufcient silencing of LGR5 in xenografts
was conrmed by real-time qRT–PCR (Supplementary Figure S1E,
available at Carcinogenesis Online).
850
LGR5 in colorectalcancer
Lacking a reliable LGR5 antibody, we utilized the phenotypic
change of SW480 upon LGR5 silencing to separate LGR5 high cells
(spheres) and LGR5 low cells (adherent cells), which we then injected
subcutaneously into the anks of nude mice as a proof-of-principle
experiment. Conrming our hypothesis, only LGR5 high cells gener-
ated tumors; however, the small number of animals prevented reach-
ing statistical signicance (P=0.40) (Table I).
NOTCH signaling is downregulated upon LGR5 silencing
To examine whether differential LGR5 expression levels in CRC
cells would be reected in specic changes to the cellular transcrip-
tome, we performed microarray gene expression proling of SW480
shLGR5 and control cells and of SW480 spheres and adherent cells.
As LGR5 silencing in SW480 resulted in a profound morphologic
change, we reasoned that genes regulated by LGR5 might overlap
with genes differentially expressed between spheres and adherent
cells. Indeed, gene expression proling revealed a signicant overlap
of deregulated genes (false discovery rate < 1.0E-103; Figure3A).
These genes included known oncogenes (e.g. MYB, MYCN) and cer-
tain drug efux genes (e.g. ABCB1, ABCC2), whose expression was
positively correlated with LGR5 expression. The expression differ-
ences between spheres and adherent cells were more pronounced than
between shLGR5 and control cells, yet unsupervised hierarchical clus-
tering showed a clear separation of all four cell fractions (Figure3B).
Ingenuity Pathway Analysis revealed that the NOTCH signaling
pathway was downregulated when LGR5 was silenced. Conversely,
it was upregulated in LGR5 high spheres. Apart from LGR5, spheres
also expressed higher levels of several other stem cell-associated
genes including SOX2, ALDH1A1 and SMOC2 when compared
with adherent cells (12,23–25). Our gene expression results were
Fig.1. Expression levels of LGR5 in CRC cell lines and shRNA-mediated silencing of LGR5 in SW480 and HT-29 CRC cells. (A) LGR5 mRNA levels in CRC
cell lines as determined by real-time qRT–PCR. LGR5 mRNA levels were normalized to YWHAZ and are expressed as fold changes relative to normal colorectal
epithelial cells. Columns and error bars represent means ± SEM of two independent experiments using triplicate measurements in each experiment. (B) LGR5
silencing efciency in CRC cell lines SW480 and HT-29 upon lentiviral shRNA transduction as determined by real-time qRT–PCR. LGR5 mRNA levels were
normalized to YWHAZ and are expressed as fold changes relative to control. Columns and error bars represent means ± SEM of three independent experiments
using triplicate measurements in each experiment. (C) Representative phase-contrast images of SW480 and HT-29 cells upon LGR5 silencing. SW480 is
composed of two morphologically distinct subpopulations (spheres and adherent cells), whereas HT-29 shows one phenotype. After LGR5 silencing, spheres
in SW480 were completely lost with no apparent changes in HT-29. Scale bars, 100µm. (D) Expression of LGR5 in spheres versus adherent cells of SW480.
Spheres express high levels of LGR5, whereas in adherent cells, virtually no LGR5 expression can be detected by real-time qRT–PCR. LGR5 mRNA levels were
normalized to YWHAZ and are expressed as fold changes relative to control. Data represent means ± SEM of three independent experiments using triplicate
measurements in each experiment. *P<0.05, **P<0.005 and ***P<0.0005.
851
D.Hirsch etal.
Fig.2. Functional effects of LGR5 silencing on SW480 and HT-29 CRC cells. (A) LGR5 silencing decreases proliferation of SW480 and HT-29 CRC cells
as quantied based on cleavage of WST-1. Columns and error bars represent means ± SEM of three independent experiments using triplicate measurements
in each experiment. (B) Silencing of LGR5 reduces migration in wound healing assays. Columns and error bars represent means ± SEM from one experiment
representative of three independent experiments (n=6 scratches per cell type and time point in each experiment). Scale bars, 100µm. (C) In line with the results
of the wound healing assay, migration of SW480 shLGR5 cells is also decreased in transwell migration assays. Columns and error bars represent means ± SEM
from one experiment representative of three independent experiments using triplicate measurements in each experiment. Scale bars, 200µm. (D) LGR5 silencing
substantially reduces colony formation in vitro. Each 250 shLGR5 and control cells were cultured for 2 weeks. The number of colonies was then assessed using
crystal violet staining. Columns and error bars represent means ± SEM of three independent experiments using triplicate measurements in each experiment.
*P<0.05 and **P<0.005.
852
LGR5 in colorectalcancer
exemplarily validated by immunohistochemistry against two differ-
entially expressed genes, cleaved NOTCH1 and SOX6 (Figure3C).
Lgr5 is overexpressed in AOM/DSS-induced mouse colon tumors
but expression is, like in normal colon epithelium, restricted to a
small percentage ofcells
We next aimed to determine a detailed molecular characterization
of Lgr5 positive and negative cells in primary colon tumors. As the
lack of a reliable LGR5 antibody prevented these analyses in primary
human CRC, we exposed heterozygous Lgr5-EGFP mice, harbor-
ing one EGFP-tagged Lgr5 allele, to a carcinogen-induced inam-
mation-driven colon tumorigenesis model based on AOM and DSS
(Supplementary Figure S2A, available at Carcinogenesis Online)
(6,14). All AOM/DSS-induced tumors were restricted to the colon,
predominantly located in the rectosigmoid colon, sometimes forming
multiple lesions; no lymph node or hematogenous metastases were
detectable (Supplementary Figure S2B, available at Carcinogenesis
Online). Histologically, AOM/DSS-induced colon tumors in Lgr5-
EGFP mice resembled well-differentiated human tubular adenocarci-
nomas, mainly of the intramucosal type (Supplementary Figure S2C,
available at Carcinogenesis Online).
We rst examined expression, distribution and frequency of Lgr5
in AOM/DSS-induced colon tumors. Real-time qRT–PCR analysis
using RNA isolated from histologically dened tumor and non-tumor
regions revealed a signicant overexpression of Lgr5 in AOM/DSS-
induced tumors compared with normal colon mucosa (Figure4A and
Supplementary Figure S3A, available at Carcinogenesis Online).
Immunohistochemistry against GFP showed that, consistent with
previous reports, Lgr5-EGFP expressing cells in normal colons were
located at the crypt bottoms (Supplementary Figure S3B, available
at Carcinogenesis Online) (6). In AOM/DSS-induced tumors, Lgr5-
EGFP expression was found to be restricted to small populations of
scattered cells (Figure4B). Quantication by ow cytometric analysis
revealed that normal colons contained on average 3.8% of Lgr5-EGFP
high cells. This percentage was similar to that of AOM/DSS-induced
tumors, which harbored on average 3.4% of Lgr5-EGFP high cells
(Figure 4C). Supported by the results from our immunohistochem-
istry-based analysis of Lgr5-EGFP expression, this suggests that a
stem cell hierarchy is preserved in AOM/DSS-induced tumors (9,11).
However, there were 3 out of 14 (21.4%) AOM/DSS-induced tumors
without detectable Lgr5-EGFP expressing cells based on GFP expres-
sion, indicating that occasionally also Lgr5-EGFP low cells could
acquire the capacity for tumor initiation and/or maintenance (10).
This is consistent with observations in human CRC samples, in which
LGR5-expressing cells were not detectable in one-third of the ana-
lyzed samples, arguing for a frequent stem cell and a rare non-stem
cell driven carcinogenesis (12). We conrmed by real-time qRT–PCR
that GFP high ow-sorted cells had signicantly higher Lgr5 expres-
sion levels than GFP low ow-sorted cells (Figure4D).
Transcriptome proles of Lgr5 high and low epithelial cells from
AOM/DSS-induced mouse colon tumors are clearly distinct
To examine whether the differential Lgr5 expression levels in nor-
mal mouse colons and AOM/DSS-induced mouse colon tumors
would be reected in specic gene expression proles, we performed
microarray gene expression proling of normal colons and AOM/
DSS-induced tumors ow-sorted into Lgr5 high and low fractions
based on GFP expression. Reassuringly, the previously described
intestinal stem cell-specic genes Lgr5 and Smoc2 were upregulated
in Lgr5 high normal and tumor cells compared with Lgr5 low normal
and tumor cells (12,25). In addition, Lgr5 high tumor cells showed
increased expression of EphB2, which is known to be coexpressed
with Lgr5 (12). Unsupervised hierarchical clustering showed a clear
separation between Lgr5 high and low tumor cells; however, separa-
tion according to Lgr5 expression levels was not as clear in normal
cells (Figure5A). Ingenuity Pathway Analysis revealed that the Wnt
signaling pathway was upregulated in Lgr5 high tumor cells com-
pared with Lgr5 low tumor cells and also with Lgr5 high normal colon
epithelial cells. Gene expression results were exemplarily validated
by immunohistochemistry against Sox6 (Figure5B).
Lgr5 high and low epithelial cells from AOM/DSS-induced mouse
colon tumors are both chromosomallystable
Genomic imbalances inuence gene expression patterns (26). To
exclude that the transcriptional differences between Lgr5 high and
low tumor cells were imposed by distinct patterns of chromosomal
aberrations in the two cell fractions, we additionally performed array
comparative genomic hybridization from the ow-sorted AOM/DSS-
induced mouse colon tumors. All eight tumors were chromosomally
stable, and thus, no difference between Lgr5 high and low cells could
be detected (Supplementary Figure S4, available at Carcinogenesis
Online) (27). This indicates that other mechanisms, for instance
epigenetics, may be the driving force in AOM/DSS-induced mouse
colon tumors (28). In conclusion, these data conclusively indicate
that LGR5 marks stem-like CRC cells and denes a cell compart-
ment in which proliferating, migrating and tumorigenic CRC cells
are enriched. This is consistent with the observed upregulation of
stem cell-related signaling pathways such as NOTCH or Wnt in
LGR5 high CRC cells.
Discussion
Here, we studied the functional and molecular consequences of
LGR5 silencing in CRC cell lines and identied LGR5 as a marker
for stem-like cells in CRC. Based on Lgr5 expression, we dened a
gene expression signature for stem-like cells in CRC using an inam-
mation-driven mouse colon tumorigenesis model based on AOM and
DSS, which mimics sporadic CRC development.
To understand the role of LGR5 in colorectal tumorigenesis, which
is controversial, we silenced its expression in two CRC cell lines
(SW480 and HT-29) (18,29–34). Silencing of LGR5 in these cell
lines resulted in reduced proliferation, migration and colony forma-
tion in vitro as well as reduced tumorigenicity in vivo. This is consist-
ent with previous studies targeting LGR5 in various cancer entities
including basal cell carcinoma, gastric cancer, glioblastoma and CRC
(18,29,31–34). In these cancers, expression of LGR5 was associated
with increased cell proliferation, migration and invasion and decreased
apoptosis. In turn, silencing of LGR5 in these cancers decreased pro-
liferation, colony formation and tumorigenicity and enhanced apop-
tosis. Upregulation of LGR5 in non-tumorigenic NIH3T3 broblasts
Table I. Quantication of tumor initiation in nude mice after subcutaneous injection of CRC cells with differential levels of LGR5 expression
Cell line shLGR5
(no. of tumors/no. of injections)
Control
(no. of tumors/no. of injections)
No. of
injected cells
P value
SW480 4/10 9/10 2000 0.0052
2/5 5/5 20 000
HT-29 8/10 10/10 2000 0.47
Cell line Adherent cells
(no. of tumors/no. of injections)
Spheres
(no. of tumors/no. of injections)
No. of
injected cells
P value
SW480 0/3 2/3 2000 0.40
853
D.Hirsch etal.
Fig.3. Gene expression proling of shLGR5 versus control cells and adherent cells versus spheres of SW480. (A) Venn diagram showing a signicant overlap
of differentially expressed genes when comparing shLGR5 versus control cells and adherent cells versus spheres of SW480 (false discovery rate < 1.0E-103). (B)
Unsupervised hierarchical clustering based on gene expression proles of shLGR5 and control cells as well as adherent cells and spheres from SW480. Samples
cluster by cell type rst. The different LGR5 fractions are clearly separated. (C) In line with our gene expression data, immunohistochemistry against both
cleaved NOTCH1 and SOX6 also shows a lower expression in SW480 shLGR5 than in SW480 control cell derived xenograft tumors. Scale bars, 20µm.
854
LGR5 in colorectalcancer
and in HaCat keratinocytes induced colony formation and promoted
tumorigenicity (18,29).
For reasons that remain to be understood, all those results are in
contrast to the ndings by Walker and colleagues in CRC cell lines
LIM1215 and LIM1899. Upon silencing of LGR5, they reported
increased migration, colony formation and tumorigenicity, and oppo-
site phenotypes when LGR5 was overexpressed (30).
The reduced tumorigenic capacity of SW480 in vivo upon LGR5
silencing, along with the decreased colony formation in vitro, suggests
that LGR5 expression is associated with stem-like properties in this cell
line. The observation that shLGR5 and control HT-29 cells were equally
tumorigenic upon xenotransplantation is somewhat contradictory but
might be either explained by the lower silencing efciency or because
tumorigenicity of HT-29 does not depend on LGR5 expression. Also,
HT-29 has much lower Wnt activity (35). The number of injected cells
might have been too high to reveal differences since HT-29 is a highly
tumorigenic cell line. Alternatively, the reduction of tumor growth rate
but not tumor incidence upon LGR5 silencing in HT-29 might suggest
that LGR5 silencing affects tumor propagation rather than tumor initia-
tion in this cell line. The slower tumor growth rate upon silencing of
LGR5 for both SW480 and HT-29 cells is consistent with previous data
showing that Lgr5 positive cells are actively proliferating (6).
Fig.4. Expression, distribution and frequency of Lgr5 in AOM/DSS-induced mouse colon tumors. (A) Varying degrees of overexpression of Lgr5 in AOM/DSS-
induced colon tumors as determined by real-time qRT–PCR. Lgr5 mRNA levels were normalized to Gapdh and are expressed as fold changes relative to normal
adjacent colon. Horizontal lines represent means. Triplicate measurements were used for each data point. (B) Lgr5-EGFP expression in AOM/DSS-induced
colon tumors is restricted to small populations of scattered cells. Scale bars, 50µm. (C) Quantication of Lgr5-EGFP high cells by ow cytometric analysis
revealed that both normal colons and AOM/DSS-induced tumors contain similar small percentages of Lgr5-EGFP high cells. N, normal colon; T, tumor. (D) Lgr5
expression is enriched in ow-sorted Lgr5-EGFP high normal colon or colon tumor cells compared with respective Lgr5-EGFP low normal colon or colon tumor
cells as determined by real-time qRT–PCR. Lgr5 mRNA levels were normalized to Gapdh. Horizontal lines represent means. Triplicate measurements were used
for each data point. *P<0.05 and **P<0.005.
855
D.Hirsch etal.
Our functional analyses were complemented by studying global
gene expression levels of cell fractions with differential LGR5 expres-
sion. Spheres (in contrast to adherent cells) of SW480 expressed
several stem cell-associated genes including LGR5 and showed an
upregulation of NOTCH signaling. Consistently, spheres appeared to
be more tumorigenic than adherent cells when xenografted in nude
mice. Silencing of LGR5 resulted in the depletion of spheres. In line
with our ndings, LGR5 is upregulated in spheroid cultures of colo-
rectal CSCs and, conversely, becomes downregulated during in vitro
differentiation of these CSCs (32). Taken together, these ndings sug-
gest that LGR5 identies a stem-like cell compartment in CRC, as it
does in normal colorectal epithelium. This has recently been shown
directly by in vivo lineage tracing in mouse intestinal adenomas (11).
NOTCH signaling was downregulated in SW480 CRC cells upon
LGR5 silencing, whereas genes involved in NOTCH signaling were
overexpressed in LGR5 high SW480 spheres. Consistently, Lgr5
deciency in the mouse during intestine development also seems
to have an inhibitory effect on the Notch signaling pathway (36).
In addition, downregulation of NOTCH signaling in CRC cell lines
and primary CRC samples via reduction of NOTCH1 or RBPJk
decreases proliferation, colony formation and tumorigenicity,
whereas upregulation of NOTCH signaling via NOTCH1 results in
opposite changes (3,37,38). This has also been demonstrated for
other tumor entities including pancreatic, lung or breast cancer
(39–41). Consistent with our gene expression analysis, it appears
most likely that the reduced proliferation and migration after LGR5
Fig.5. Gene expression proling of normal mouse colons and AOM/DSS-induced mouse colon tumors ow-sorted for Lgr5-EGFP. (A) Unsupervised
hierarchical clustering based on gene expression proles of Lgr5 high and low fractions from normal colons and AOM/DSS-induced tumors. Samples cluster
by entity (normal colon or tumor) rst. In tumors, the different Lgr5 fractions are clearly separated; however, in normal colons, they are separated by both Lgr5
and individual mice. (B) In line with our gene expression data, Sox6 expression could also be demonstrated by immunohistochemistry (positivity in a small
population of scattered cells in AOM-/DSS-induced tumors). Scale bar, 20µm.
856
LGR5 in colorectalcancer
silencing in CRC cells are a consequence of reduced NOTCH sign-
aling (3).
Hence, our gene expression results substantiate our ndings from
functional assays upon LGR5 silencing in CRC cell lines at a molec-
ular level, suggesting LGR5 as a potential therapeutic target. For
instance, Honokiol inhibits CRC growth by targeting NOTCH signal-
ing in colorectal CSCs (42).
Pursuing stem cells as therapeutic targets might help to overcome
some of the frustrations associated with current cancer treatment regi-
mens. Cataloging gene expression data from these stem cells serves
as a rst step in understanding the molecular features distinguishing
these cell types from the bulk of tumor cells or from normal adult tis-
sue stem cells (43–46).
Despite the advantages of in vitro cultures to analyze features
of stemness potential, such as ease of propagation, there are con-
cerns that molecular changes might be induced when cells are cul-
tured in the absence of their physiological context (13,47). Thus,
we extended our gene expression proling to ex vivo isolated cells.
Normal mouse colons and AOM/DSS-induced mouse colon tumors
from Lgr5-EGFP mice were ow-sorted into Lgr5 high and low epi-
thelial cell fractions. Global gene expression analyses of ow-sorted
fractions revealed an overexpression of the Wnt signaling pathway
in Lgr5 high tumor cells compared with both Lgr5 low tumor cells
and Lgr5 high normal cells. This supports previous ndings showing
that Wnt activity denes colorectal CSCs (48). Although Horst etal.
(49) found that Wnt activity alone might not be sufcient to convey
stem-like potential, our ndings suggest that the combination of both
Wnt signaling and Lgr5 might help to determine cells with stem-like
properties in CRC.
The association of LGR5 and NOTCH signaling seen in cell lines
could not be recapitulated in vivo when ow sorting AOM/DSS-
induced mouse colon tumors for Lgr5. Also, in contrast to ow-
sorted Lgr5 high and low cells from AOM/DSS-induced tumors,
the Wnt signaling pathway was not signicantly altered in our loss-
of-function experiments in cell lines, though tending to be higher
in LGR5 high spheres compared with LGR5 low adherent cells
(P = 0.16). The biological difference of cell lines being cultured
without their physiological context might explain these ndings.
Increasing evidence indicates that not only intrinsic factors but also
extrinsic factors like the microenvironment can inuence the CSC
phenotype. For instance, Vermeulen etal. (48) showed in CRC that
stromal myobroblasts surrounding CSCs not only can maintain
Wnt signaling activity in CSCs but also can activate Wnt signal-
ing in more differentiated tumor cells and thereby induce the CSC
phenotype. Furthermore, there is a methodological difference that
might contribute to the heterogeneity of our ndings in cell lines
and ex vivo isolated tumor cells. In the ex vivo model, ow-sorted
Lgr5 high and low cells were compared, whereas in the cell line
experiment, LGR5 was silenced actively via shRNA and compared
with the original cell population. Overall, the observed heterogene-
ity across the different lines and models reects one of the major
problems in CSC research.
In summary, our comprehensive functional and molecular analy-
sis of LGR5 in CRC cell lines and AOM/DSS-induced mouse colon
tumors conclusively links LGR5 to stem-like cells in CRC. LGR5
did not only serve as a marker for these stem-like CRC cells but was
also of functional relevance for CRC cells, thus representing a poten-
tial therapeutic target, in particular as conditional deletion of Lgr5
in mouse guts does not seem to negatively affect normal intestinal
epithelium (8). To further specify the role of LGR5 in human CRC,
studies of LGR5 in primary human CRC specimens will be needed in
the future and, as a prerequisite, the development of a reliable LGR5
antibody.
Supplementary material
Supplementary Material and methods, Table S1 and Figures S1S4
can be found at http://carcin.oxfordjournals.org/
Funding
Intramural Research Program, National Institutes of Health, National
Cancer Institute; German Academic Exchange Service (D.H.).
Acknowledgements
The authors thank B.Chen for help with gures and IT-related support,
X.Lu for help with cell culture-related experiments, M.E.Jorge (Veterinary
Technician, Animal Facility, National Institutes of Health) for help with nude
mouse tumor measurements, D.Despres and B.Klaunberg (Mouse Imaging
Facility, National Institutes of Health) for advice and assistance with mouse
colonoscopies, B.Taylor (FACS Core Laboratory, Center for Cancer Research,
National Cancer Institute, National Institutes of Health) for expert technical
assistance with ow sorting, K.Wolk (Immunohistochemistry Laboratory,
Institute of Pathology, University Medical Center Mannheim) for help with
immunohistochemical staining and C.A.Klein (Experimental Medicine and
Therapy Research, University of Regensburg) for providing expert advice.
Conict of Interest Statement: None declared.
References
1. Fearon,E.R. etal. (1990) A genetic model for colorectal tumorigenesis.
Cell, 61, 759–767.
2. Ried,T. etal. (1996) Comparative genomic hybridization reveals a specic
pattern of chromosomal gains and losses during the genesis of colorectal
tumors. Genes Chromosomes Cancer, 15, 234–245.
3. Camps,J. et al. (2013) Genetic amplication of the NOTCH modula-
tor LNX2 upregulates the WNT/β-catenin pathway in colorectal cancer.
Cancer Res., 73, 2003–2013.
4. Clarke,M.F. etal. (2006) Cancer stem cells—perspectives on current status
and future directions: AACR Workshop on cancer stem cells. Cancer Res.,
66, 9339–9344.
5. Visvader,J.E. etal. (2008) Cancer stem cells in solid tumours: accumulat-
ing evidence and unresolved questions. Nat. Rev. Cancer, 8, 755–768.
6. Barker,N. et al. (2007) Identication of stem cells in small intestine and
colon by marker gene Lgr5. Nature, 449, 1003–1007.
7. Carmon,K.S. etal. (2011) R-spondins function as ligands of the orphan
receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc.
Natl Acad. Sci. USA, 108, 11452–11457.
8. de Lau,W. etal. (2011) Lgr5 homologues associate with Wnt receptors and
mediate R-spondin signalling. Nature, 476, 293–297.
9. Barker,N. etal. (2009) Crypt stem cells as the cells-of-origin of intestinal
cancer. Nature, 457, 608–611.
10. Visvader,J.E. (2011) Cells of origin in cancer. Nature, 469, 314–322.
11. Schepers,A.G. etal. (2012) Lineage tracing reveals Lgr5+ stem cell activ-
ity in mouse intestinal adenomas. Science, 337, 730–735.
12. Merlos-Suárez,A. etal. (2011) The intestinal stem cell signature identies
colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell, 8,
511–524.
13. Gaiser,T. et al. (2011) Genome and transcriptome proles of CD133-
positive colorectal cancer cells. Am. J.Pathol., 178, 1478–1488.
14. Neufert,C. etal. (2007) An inducible mouse model of colon carcinogenesis
for the analysis of sporadic and inammation-driven tumor progression.
Nat. Protoc., 2, 1998–2004.
15. Bongers,G. et al. (2010) The cytomegalovirus-encoded chemokine recep-
tor US28 promotes intestinal neoplasia in transgenic mice. J. Clin. Invest.,
120, 3969–3978.
16. Camps,J. et al. (2008) Chromosomal breakpoints in primary colon can-
cer cluster at sites of structural variants in the genome. Cancer Res., 68,
1284–1295.
17. Grade,M. etal. (2007) Gene expression proling reveals a massive, ane-
uploidy-dependent transcriptional deregulation and distinct differences
between lymph node-negative and lymph node-positive colon carcinomas.
Cancer Res., 67, 41–56.
18. McClanahan,T. et al. (2006) Identication of overexpression of orphan
G protein-coupled receptor GPR49 in human colon and ovarian primary
tumors. Cancer Biol. Ther., 5, 419–426.
19. Uchida,H. et al. (2010) Overexpression of leucine-rich repeat-contain-
ing G protein-coupled receptor 5 in colorectal cancer. Cancer Sci., 101,
1731–1737.
20. Kai,K. etal. (2009) Maintenance of HCT116 colon cancer cell line con-
forms to a stochastic model but not a cancer stem cell model. Cancer Sci.,
100, 2275–2282.
857
D.Hirsch etal.
21. Yeung,T.M. etal. (2010) Cancer stem cells from colorectal cancer-derived
cell lines. Proc. Natl Acad. Sci. USA, 107, 3722–3727.
22. Kanwar,S.S. etal. (2010) The Wnt/beta-catenin pathway regulates growth
and maintenance of colonospheres. Mol. Cancer, 9, 212.
23. Huang,E.H. etal. (2009) Aldehyde dehydrogenase 1 is a marker for normal
and malignant human colonic stem cells (SC) and tracks SC overpopula-
tion during colon tumorigenesis. Cancer Res., 69, 3382–3389.
24. Fang,X. etal. (2010) ChIP-seq and functional analysis of the SOX2 gene in
colorectal cancers. OMICS, 14, 369–384.
25. Muñoz,J. etal. (2012) The Lgr5 intestinal stem cell signature: robust expres-
sion of proposed quiescent ‘+4’ cell markers. EMBO J., 31, 3079–3091.
26. Ried,T. etal. (2012) The consequences of chromosomal aneuploidy on the
transcriptome of cancer cells. Biochim. Biophys. Acta, 1819, 784–793.
27. Guda,K. etal. (2004) Carcinogen-induced colon tumors in mice are chro-
mosomally stable and are characterized by low-level microsatellite insta-
bility. Oncogene, 23, 3813–3821.
28. Rosenberg,D.W. etal. (2009) Mouse models for the study of colon carcino-
genesis. Carcinogenesis, 30, 183–196.
29. Tanese,K. etal. (2008) G-protein-coupled receptor GPR49 is up-regulated
in basal cell carcinoma and promotes cell proliferation and tumor forma-
tion. Am. J.Pathol., 173, 835–843.
30. Walker,F. et al. (2011) LGR5 is a negative regulator of tumourigenicity,
antagonizes Wnt signalling and regulates cell adhesion in colorectal cancer
cell lines. PLoS One, 6, e22733.
31. Simon,E. (2012) [Abstracts of the 96th Annual Meeting of the German
Society of Pathology. May 31-June 3, 2012. Berlin, Germany]. Pathologe,
33 (suppl. 1), 5–175.
32. Kemper,K. et al. (2012) Monoclonal antibodies against Lgr5 identify
human colorectal cancer stem cells. Stem Cells, 30, 2378–2386.
33. Al-Kharusi,M.R. etal. (2013) LGR5 promotes survival in human colorec-
tal adenoma cells and is upregulated by PGE2: implications for targeting
adenoma stem cells with NSAIDs. Carcinogenesis, 34, 1150–1157.
34. Nakata,S. etal. (2013) LGR5 is a marker of poor prognosis in glioblastoma
and is required for survival of brain cancer stem-like cells. Brain Pathol.,
23, 60–72.
35. Kendziorra,E. etal. (2011) Silencing of the Wnt transcription factor TCF4
sensitizes colorectal cancer cells to (chemo-) radiotherapy. Carcinogenesis,
32, 1824–1831.
36. Garcia,M.I. et al. (2009) LGR5 deciency deregulates Wnt signaling and
leads to precocious Paneth cell differentiation in the fetal intestine. Dev.
Biol., 331, 58–67.
37. Zhang,Y. et al. (2010) Notch1 regulates the growth of human colon can-
cers. Cancer, 116, 5207–5218.
38. Sikandar,S.S. etal. (2010) NOTCH signaling is required for formation and
self-renewal of tumor-initiating cells and for repression of secretory cell
differentiation in colon cancer. Cancer Res., 70, 1469–1478.
39. Bao,B. et al. (2011) Notch-1 induces epithelial-mesenchymal transition
consistent with cancer stem cell phenotype in pancreatic cancer cells.
Cancer Lett., 307, 26–36.
40. Xie,M. et al. (2012) Activation of Notch-1 enhances epithelial-mesen-
chymal transition in getinib-acquired resistant lung cancer cells. J. Cell.
Biochem., 113, 1501–1513.
41. Simmons,M.J. etal. (2012) NOTCH1 inhibition in vivo results in mam-
mary tumor regression and reduced mammary tumorsphere-forming activ-
ity in vitro. Breast Cancer Res., 14, R126.
42. Ponnurangam,S. etal. (2012) Honokiol in combination with radiation tar-
gets notch signaling to inhibit colon cancer stem cells. Mol. Cancer Ther.,
11, 963–972.
43. Reya,T. etal. (2001) Stem cells, cancer, and cancer stem cells. Nature, 414,
105–111.
44. Fabrizi,E. etal. (2010) Therapeutic implications of colon cancer stem cells.
World J.Gastroenterol., 16, 3871–3877.
45. Nguyen,L.V. etal. (2012) Cancer stem cells: an evolving concept. Nat. Rev.
Cancer, 12, 133–143.
46. Marusyk,A. etal. (2012) Intra-tumour heterogeneity: a looking glass for
cancer? Nat. Rev. Cancer, 12, 323–334.
47. Snippert,H.J. et al. (2011) Tracking adult stem cells. EMBO Rep., 12,
113–122.
48. Vermeulen,L. etal. (2010) Wnt activity denes colon cancer stem cells and
is regulated by the microenvironment. Nat. Cell Biol., 12, 468–476.
49. Horst,D. et al. (2012) Differential WNT activity in colorectal cancer con-
fers limited tumorigenic potential and is regulated by MAPK signaling.
Cancer Res., 72, 1547–1556.
Received August, 1, 2013; revised October 29, 2013;
accepted November 6, 2013
858
... While work has focused on defining the roles of TCF7L2, TCF7, and LEF1 in CRC, the role of TCF7L1 remains poorly understood. TCF7L1 has Genes 2023, 14, 481 2 of 16 been shown to play an oncogenic role in CRC by repressing expression of known CRC tumor suppressor ephrin type-B receptor 3 (EPHB3) and the Wnt antagonist Dickkopf4 (DKK4) [9,10]. Our group has also shown that TCF7L1 and TCF7L2/β-catenin complexes dynamically regulate MYC expression through defined WREs during distinct stages of the cell cycle in CRC cells [11]. ...
... Canonical Wnt/β-catenin signaling is fundamental for regulating stem cell selfrenewal and cellular proliferation in intestinal crypts and the Wnt target gene, leucine-richrepeat containing G-protein-coupled receptor 5 (LGR5), has been established as the bona fide cell surface marker of intestinal stem cells [12,13]. Furthermore, LGR5 is recognized as a marker of CRC stem cells and has been implicated in CRC pathogenesis as LGR5 + cells are accepted as the cells-of-origin in CRC [14,15]. Moreover, the intrinsic plasticity of LGR5 − enterocytes to revert to LGR5 + cells and then re-establish the cellular hierarchy upon loss or ablation of LGR5 further implicates LGR5 in both tumor initiation and metastatic outgrowth [16][17][18][19][20]. ...
... Our study further defines a role for TCF7L1 in cancer stem cell biology by demonstrating that TCF7L1 mediates repression of LGR5 expression and that this contributes to reduction of spheroid formation efficiency. LGR5 + stem cells have been implicated in many stages of CRC tumorigenesis, from initiation as the cells-of-origin [14] to outgrowth and maintenance of metastatic lesions [17]. More recently, LGR5 − cancer cells have been distinguished as the majority of migrating and disseminating cells from the primary tumor to the metastatic site, highlighting an important role for LGR5 − cancer cells and their intrinsic capability to re-establish the LGR5 + cancer cell population [20]. ...
Article
Full-text available
Mutations in components of the Wnt/β-catenin signaling pathway drive colorectal cancer (CRC), in part, by deregulating expression of genes controlled by the T-cell factor (TCF) family of transcription factors. TCFs contain a conserved DNA binding domain that mediates association with TCF binding elements (TBEs) within Wnt-responsive DNA elements (WREs). Intestinal stem cell marker, leucine-rich-repeat containing G-protein-coupled receptor 5 (LGR5), is a Wnt target gene that has been implicated in CRC stem cell plasticity. However, the WREs at the LGR5 gene locus and how TCF factors directly regulate LGR5 gene expression in CRC have not been fully defined. Here, we report that TCF family member, TCF7L1, plays a significant role in regulating LGR5 expression in CRC cells. We demonstrate that TCF7L1 binds to a novel promoter-proximal WRE through association with a consensus TBE at the LGR5 locus to repress LGR5 expression. Using CRISPR activation and interference (CRISPRa/i) technologies to direct epigenetic modulation, we demonstrate that this WRE is a critical regulator of LGR5 expression and spheroid formation capacity of CRC cells. Furthermore, we found that restoring LGR5 expression rescues the TCF7L1-mediated reduction in spheroid formation efficiency. These results demonstrate a role for TCF7L1 in repressing LGR5 gene expression to govern the spheroid formation potential of CRC cells.
... An examination of the relevance of the OSAP in independent datasets from Gene Expression Omnibus (GEO), GSE39582 [23], GSE37283 [24], GSE36807 [25], GSE16879 [26], GSE36133 [27], GSE 46200 [28], and GSE37929 [29] was performed using the Array Studio software (Qiagen France SAS, Les Ulis, France). The methodologies employed to process and normalize data and to perform the Hierarchical Clustering Analysis, Principal Component Analysis (PCA), and ssGSEA projections are detailed in the Supplementary Material and Methods section. ...
... Several genes of the OSAP signature are also known to be active in immune cells, unveiling a potential interference of the inflammation index with OSAP scores. To circumvent this problem and assess whether OSAP induction takes place in the initiation of colorectal tumor development, we used the previously reported gene expression profiles of intestinal epithelial stem and precursor cells isolated from either normal colons or AOM/DSSinduced tumors isolated from Lgr5-EGFP mice exposed to azoxymethane/dextran sodium sulfate [28] (AOM/DSS, dataset GSE46200). The transcriptomic analysis was performed from cells which were flow-sorted into Lgr5 high and Lgr5 low fractions based on GFP ex- As we compared homogenous epithelial cell populations, the in vitro-established epithelial OSAP signature was used. ...
... We confirmed that the program was induced in inflamed mucosae of IBD patients and correlated with previously reported stem-cell signatures [29,47,48], suggesting that this cell reprogramming takes place in either intestinal stem or precursor cells. By scoring the OSAP in intestinal epithelial cells that were flow-sorted from chronic-inflammation and sporadic colorectal carcinogenesis mouse models on the basis of lineage-tracing experiments, we confirmed that the score was higher in intestinal progenitor cells isolated from adenomas compared to their normal counterparts in both murine carcinogenesis models [28,29]. Although a signature score depends on the algorithm used and the state of the cells and can only be compared between samples from the same analysis, the positive scores observed in adenomas from AOM/DSS-treated mice supports a preponderant role for inflammation in the activation of OSAP. ...
Article
Full-text available
Simple Summary Chronic inflammation, as observed in Crohn’s disease and ulcerative colitis patients, damages the intestinal mucosa. We reasoned that if a subset of intestinal epithelial cells adapt to this inflammatory stress to survive, this adaptation could contribute to their malignant transformation. We show that human colonic epithelial cells escape chronic inflammation in vitro through a partial genetic reprogramming. By questioning data bases, we confirm that this reprogramming takes place in the inflamed mucosae of patients with Crohn’s disease and ulcerative colitis, and that it is induced in vivo during the early stages of murine intestinal carcinogenesis. These data contribute to understanding the pathology and underline how orchestrators of cellular adaptation might contribute to intestinal homeostasis and, potentially, tumor initiation. Abstract Reactive oxygen species (ROS) are considered to be the main drivers of inflammatory bowel disease. We investigated whether this permanent insult compels intestinal stem cells to develop strategies to dampen the deleterious effects of ROS. As an adverse effect, this adaptation process may increase their tolerance to oncogenic insults and facilitate their neoplastic transformation. We submitted immortalized human colonic epithelial cells to either a mimic of chronic inflammation or to a chemical peroxide, analyzed how they adapted to stress, and addressed the biological relevance of these observations in databases. We demonstrated that cells adapt to chronic-inflammation-associated oxidative stress in vitro through a partial genetic reprogramming. Through a gene set enrichment analysis, we showed that this program is recurrently active in the intestinal mucosae of Crohn’s and ulcerative colitis disease patients and evolves alongside disease progression. Based on a previously reported characterization of intestinal stem and precursor cells using tracing experiments, we lastly confirmed the activation of the program in intestinal precursor cells during murine colorectal cancer development. This adaptive process is thus likely to play a role in the progression of Crohn’s and ulcerative disease, and potentially in the initiation of colorectal cancer.
... These genes locate near spot E and upregulate in LMS5. The GPCR LGR5 as an exception locates in the high expression side of the landscapes in spot C, upregulated in highly cycling CRLM, which agrees with the role of LGR5 as a marker for proliferative stem-like cells in CRC [65,66]. In summary, high-resolution cartography of the CRLM transcriptomes reveals a network of cellular programs governed by genetic lesions (mutations and CNVs) and by epigenetic chromatin remodeling leading to the antagonistic activation of epithelial-like proliferative cancer cells or inflammatory, mesenchyme-like cells. ...
Article
Full-text available
Simple Summary Liver metastasis is a significant factor contributing to mortality associated with colorectal cancer. Establishing the biological mechanisms of metastasis is crucial for refining diagnostics and identifying therapeutic windows for interventions. Currently, little is known of the processes that govern the development of liver metastases, the role of the tumor microenvironment, the role of epigenetics, and potential treatment-induced shaping effects. Machine learning-based bioinformatics has provided an important methodical option to decipher fine-granular details of the transcriptomic landscape of tumor heterogeneity and the underlying molecular mechanisms. Our molecular portrayal method has potential implications for treatment decisions, which may require personalized diagnostics. Abstract The molecular mechanisms of the liver metastasis of colorectal cancer (CRLM) remain poorly understood. Here, we applied machine learning and bioinformatics trajectory inference to analyze a gene expression dataset of CRLM. We studied the co-regulation patterns at the gene level, the potential paths of tumor development, their functional context, and their prognostic relevance. Our analysis confirmed the subtyping of five liver metastasis subtypes (LMS). We provide gene-marker signatures for each LMS, and a comprehensive functional characterization that considers both the hallmarks of cancer and the tumor microenvironment. The ordering of CRLMs along a pseudotime-tree revealed a continuous shift in expression programs, suggesting a developmental relationship between the subtypes. Notably, trajectory inference and personalized analysis discovered a range of epigenetic states that shape and guide metastasis progression. By constructing prognostic maps that divided the expression landscape into regions associated with favorable and unfavorable prognoses, we derived a prognostic expression score. This was associated with critical processes such as epithelial–mesenchymal transition, treatment resistance, and immune evasion. These factors were associated with responses to neoadjuvant treatment and the formation of an immuno-suppressive, mesenchymal state. Our machine learning-based molecular profiling provides an in-depth characterization of CRLM heterogeneity with possible implications for treatment and personalized diagnostics.
... Furthermore, LGR5 is utilized for the prognosis of adversity of cancer, for example, two meta-analyses by Q. and Jiang et al. (2015)were observed with high LGR5 levels associated with shorter overall survival (OS) and diseasefree survival (DFS). Repression of LGR5 via siRNA showed reduced proliferation, migration, and colony formation, and was even observed with apoptosis on LGR5 knockdown (Hirsch et al., 2014). In addition, X. observed enhanced anti-cancer properties in HT29 cells upon LGR5 silencing namely, enhance chemosensitivity, reduced invasive capacity, induction of cell death and reduced stem cell characteristics. ...
... Colorectal cancer stem cells (CSCs), or tumor-initiating cells, mediate drug resistance and relapse through their capacity to either self-renew or differentiate into heterogeneous lineages of tumor cells (1,2). Leucine-rich repeat containing, G protein-coupled Receptor 5 (LGR5), is highly expressed in approximately 60-70% of CRCs (3)(4)(5) and is a validated marker of normal adult intestinal stem cells and functional CSCs (6)(7)(8). Originally identified as a Wnt target gene, LGR5 and related receptors LGR4/6 bind R-spondin (RSPO1-4) growth factors responsible for modulating Wnt/-catenin signaling, an important pathway implicated in CRC and stem cell maintenance (9). ...
Article
LGR5 is highly expressed in colorectal cancer (CRC) and cancer stem cells that play important roles in tumor initiation, progression, and metastasis. Loss of LGR5 has been shown to enhance therapy resistance. However, the molecular mechanisms that mediate this resistance remain elusive. In this study, we demonstrate conversion of LGR5+ CRC cells to an LGR5- state in response to chemotherapy, LGR5-targeted antibody-drug conjugates (ADCs), or LGR5 gene ablation, led to activation of STAT3. Further investigation revealed increased STAT3 activation occurred a result of increased MET activity. LGR5 overexpression decreased MET-STAT3 activity and sensitized CRC cells to therapy. STAT3 inhibition suppressed MET phosphorylation, while constitutively active STAT3 reduced LGR5 levels and increased MET activity, suggesting a potential feedback mechanism. Combination treatment of MET-STAT3 inhibitors with irinotecan or ADCs substantiated synergistic effects in CRC cells and tumor organoids. In CRC xenografts, STAT3 inhibition combined with irinotecan enhanced tumor growth suppression and prolonged survival. These findings suggest a mechanism by which drug-resistant LGR5- CRC cells acquire a survival advantage through activation of MET-STAT3 and provide rationale for new treatment strategies to target CRC.
Preprint
Metastasis is the principal cause of cancer death, yet we lack an understanding of metastatic cell states, their relationship to primary tumor states, and the mechanisms by which they transition. In a cohort of biospecimen trios from same-patient normal colon, primary and metastatic colorectal cancer, we show that while primary tumors largely adopt Lgr5+ intestinal stem-like states, metastases display progressive plasticity. Loss of intestinal cell states is accompanied by reprogramming into a highly conserved fetal progenitor state, followed by non-canonical differentiation into divergent squamous and neuroendocrine-like states, which is exacerbated by chemotherapy and associated with poor patient survival. Using matched patient-derived organoids, we demonstrate that metastatic cancer cells exhibit greater cell-autonomous multilineage differentiation potential in response to microenvironment cues than their intestinal lineage-restricted primary tumor counterparts. We identify PROX1 as a stabilizer of intestinal lineage in the fetal progenitor state, whose downregulation licenses non-canonical reprogramming.
Chapter
Full-text available
Colorectal cancer is the leading cause of mortality. Several studies are focused on understanding the molecular mechanism to develop potential therapeutics to enhance patient health standards. Curcumin is a bioactive component derived from Curcuma Longa implicated with a spectrum of anticancer properties such as antioxidants. It has been proposed to possess promising features in suppressing colon cancer proliferation. However, the pharmacokinetics studies denote that curcumin lacks bioavailability at the target site which leads to systemic metabolism and excretion. So, studies were diverted towards eliciting the importance of structural units and modifying curcumin conformation to improve the anti-cancer effect and bioavailability. Curcumin and its analogues were employed in clinical trials as a cotreatment group to alleviate the side effects of traditional chemotherapy. Here the authors review the recent trends in the usage of curcumin in colorectal cancer and emphasis relevant studies to update the current status of therapeutics.
Article
In recent years, antibody-drug conjugates (ADCs) have emerged as promising anti-cancer therapeutic agents with several having already received market approval for the treatment of solid tumor and hematological malignancies. As ADC technology continues to improve and the range of indications treatable by ADCs increases, the repertoire of target antigens has expanded and will undoubtedly continue to grow. G protein-coupled receptors (GPCRs) are well-characterized therapeutic targets implicated in many human pathologies, including cancer, and represent a promising emerging target of ADCs. In this review, we will discuss the past and present therapeutic targeting of GPCRs and describe ADCs as therapeutic modalities. Moreover, we will summarize the status of existing preclinical and clinical GPCR-targeted ADCs and address the potential of GPCRs as novel targets for future ADC development.
Article
Background & aims: Fibrosis and tissue stiffening are hallmarks of the inflammatory bowel disease (IBD). We have hypothesized that the increased stiffness directly contributes to the dysregulation of the epithelial cell homeostasis in IBD. Here, we aim to determine the impact of tissue stiffening on the fate and function of the intestinal stem cells (ISCs). Methods: We developed a long-term culture system consisting of 2.5-dimensional intestinal organoids grown on a hydrogel matrix with tunable stiffness. Single-cell RNA sequencing provided stiffness-regulated transcriptional signatures of the ISCs and their differentiated progeny. YAP-knockout and YAP-overexpression mice were used to manipulate YAP expression. In addition, we analyzed colon samples from murine colitis models and human IBD samples to assess the impact of stiffness on ISCs in vivo. Results: We demonstrated that increasing the stiffness potently reduced the population of LGR5+ ISCs and KI-67+ proliferating cells. Conversely, cells expressing the stem cell marker, OLFM4, became dominant in the crypt-like compartments and pervaded the villus-like regions. Concomitantly, stiffening prompted the ISCs to preferentially differentiate toward goblet cells. Mechanistically, stiffening increased the expression of cytosolic YAP, driving the extension of OLFM4+ cells into the villus-like regions, while it induced the nuclear translocation of YAP, leading to preferential differentiation of ISCs towards goblet cells. Furthermore, analysis of colon samples from murine colitis models and IBD patients demonstrated cellular and molecular remodeling reminiscent of those observed in vitro. Conclusions: Collectively, our findings highlight that matrix stiffness potently regulates the stemness of ISCs and their differentiation trajectory, supporting the hypothesis that fibrosis-induced gut stiffening plays a direct role in epithelial remodeling in IBD.
Article
Cancer is not a hard-wired phenomenon but an evolutionary disease. From the onset of carcinogenesis, cancer cells continuously adapt and evolve to satiate their ever-growing proliferation demands. This results in the formation of multiple subtypes of cancer cells with different phenotypes, cellular compositions, and consequently displaying varying degrees of tumorigenic identity and function. This phenomenon is referred to as cancer plasticity, during which the cancer cells exist in a plethora of cellular states having distinct phenotypes. With the advent of modern technologies equipped with enhanced resolution and depth, for example, single-cell RNA-sequencing and advanced computational tools, unbiased cancer profiling at a single-cell resolution are leading the way in understanding cancer cell rewiring both spatially and temporally. In this review, the processes and mechanisms that give rise to cancer plasticity include both intrinsic genetic factors such as epigenetic changes, differential expression due to changes in DNA, RNA, or protein content within the cancer cell, as well as extrinsic environmental factors such as tissue perfusion, extracellular milieu are detailed and their influence on key cancer plasticity hallmarks such as epithelial-mesenchymal transition (EMT) and cancer cell stemness (CSCs) are discussed. Due to therapy evasion and drug resistance, tumor heterogeneity caused by cancer plasticity has major therapeutic ramifications. Hence, it is crucial to comprehend all the cellular and molecular mechanisms that control cellular plasticity. How this process evades therapy, and the therapeutic avenue of targeting cancer plasticity must be diligently investigated.
Article
Full-text available
Chromosomal copy number alterations (aneuploidy) define the genomic landscape of most cancer cells, but identification of the oncogenic drivers behind these imbalances remains an unfinished task. In this study, we conducted a systematic analysis of colorectal carcinomas that integrated genomic copy number changes and gene expression profiles. This analysis revealed 44 highly overexpressed genes mapping to localized amplicons on chromosome 13, gains of which occur often in colorectal cancers. RNAi-mediated silencing identified 8 candidates whose loss of function reduced cell viability 20% or more in colorectal cancer cell lines. The functional space of the genes NUPL1, LNX2, POLR1D, POMP, SLC7A1, DIS3, KLF5, and GPR180 was established by global expression profiling after RNAi exposure. One candidate, LNX2, not previously known as an oncogene, was involved in regulating NOTCH signaling. Silencing LNX2 reduced NOTCH levels but also downregulated the transcription factor TCF7L2 and markedly reduced WNT signaling. LNX2 overexpression and chromosome 13 amplification therefore constitutively activates the WNT pathway, offering evidence of an aberrant NOTCH-WNT axis in colorectal cancer.
Article
Full-text available
Stem cell biology has come of age. Unequivocal proof that stem cells exist in the haematopoietic system has given way to the prospective isolation of several tissue-specific stem and progenitor cells, the initial delineation of their properties and expressed genetic programmes, and the beginnings of their utility in regenerative medicine. Perhaps the most important and useful property of stem cells is that of self-renewal. Through this property, striking parallels can be found between stem cells and cancer cells: tumours may often originate from the transformation of normal stem cells, similar signalling pathways may regulate self-renewal in stem cells and cancer cells, and cancer cells may include 'cancer stem cells' — rare cells with indefinite potential for self-renewal that drive tumorigenesis.
Article
Full-text available
Introduction NOTCH activation has been recently implicated in human breast cancers, associated with a poor prognosis, and tumor-initiating cells are hypothesized to mediate resistance to treatment and disease relapse. To address the role of NOTCH1 in mammary gland development, transformation, and mammary tumor-initiating cell activity, we developed a doxycycline-regulated mouse model of NOTCH1-mediated mammary transformation. Methods Mammary gland development was analyzed by using whole-mount analysis and by flow cytometry in nulliparous transgenic mice maintained in the presence/absence of doxycycline (or intracellular NOTCH1). Mammary tumors were examined histologically and immunophenotyped by staining with antibodies followed by flow cytometry. Tumors were transplanted into mammary fat pads under limiting dilution conditions, and tumor-initiating cell frequency was calculated. Mammary tumor cells were also plated in vitro in a tumorsphere assay in the presence/absence of doxycycline. RNA was isolated from mammary tumor cell lines cultured in the presence/absence of doxycycline and used for gene-expression profiling with Affymetrix mouse arrays. NOTCH1-regulated genes were identified and validated by using quantitative real-time polymerase chain reaction (PCR). Mammary tumor-bearing mice were treated with doxycycline to suppress NOTCH1 expression, and disease recurrence was monitored. Results Similar to published studies, we show that constitutive expression of human intracellular NOTCH1 in the developing mouse mammary gland inhibits side branching and promotes luminal cell fate. These mice develop mammary adenocarcinomas that express cytokeratin (CK) 8/18. In vivo limiting-dilution analyses revealed that these mammary tumors exhibit functional heterogeneity and harbor a rare (1/2,978) mammary tumor-initiating cell population. With this dox-regulated NOTCH1 mammary tumor model, we demonstrate that NOTCH1 inhibition results in mammary tumor regression in vivo and prevents disease recurrence in four of six tumors tested. Consistent with the in vivo data, NOTCH1 inhibition reduces mammary tumorsphere activity in vitro. We also identify the embryonic stem cell transcription factor Nanog as a novel NOTCH1-regulated gene in tumorspheres and in mouse and human breast cancer cell lines. Conclusions These data indicate that NOTCH1 inhibition results in mammary tumor regression in vivo and interferes with disease recurrence. We demonstrate that NOTCH1-transformed mouse mammary tumors harbor a rare mammary tumor-initiating population and that NOTCH1 contributes to mammary tumor-initiating activity. This work raises the possibility that NOTCH therapeutics may target mammary tumor-initiating cells in certain human breast cancer subtypes.
Article
Full-text available
The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By “lineage retracing” using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat–containing heterotrimeric guanine nucleotide–binding protein–coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5+ cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5+ cells as well as all other adenoma cell types. The Lgr5+ cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.
Article
Aims: Virtual microscopy is already established in teachings at many universities. Yet, the technique is subject of continuous further development. A variety of features exist, in which the single applications differ. However, which of the multitude of features are considered useful, necessary or redundant is unknown. Therefore, we have investigated the needs of the students with a questionnaire and developed a new application for virtual microscopy out of these data. Methods: In cooperation with the Zentrum für Qualitätssicherung und -entwicklung(-> Englische Bezeichnung erfragen) Mainz a questionnaire with a feature-set was created. 216 students assessed the importance of functions like annotations, additional teaching texts and a broad scope of target devices. Based on this evaluation a new application was constructed. Results: The analysis of the questionnaire revealed that 96,2% of the students consider virtual microscopy as a meaningful learning opportunity and desirable for test preparation. The features annotations (87,3%) and additional teaching texts (52,3%) have also been evaluated positively. Furthermore, many students asked for a quiz mode in the comments section. In contrast, the establishment of a discussion forum was less important to the students (4,2%). These results provided the basis for the development of a novel application in which technologies were used (including HTML1.1, AJAX) to both meet the desire of the students and ensure the expansibility (openlayers) and sustainability of the application. Hence, proprietary technologies like Flash were abandoned. Discussion: The survey revealed the requirements of the users which conformed the basis for the development of a new application. In future innovations the applications should be able to run on all modern browsers without proprietary extension. The course of further developments, e.g. detailed depth portrayal or adventure experience microscopy, should be determined in further questionnaires.
Article
Colorectal carcinomas (CRC) might be organized hierarchically and contain a subpopulation of tumorigenic, putative cancer stem cells that are CD133 positive. We studied the biological and genetic characteristics of such cells in CRC cell lines and primary tumors. Three CRC cell lines were sorted in CD133 positive and negative fractions. The respective genetic aberration profiles were studied using array comparative genomic hybridization (aCGH) and expression profiling. Tumorigenicity for each cellular population was tested by injection into nude mice. Additionally, we compared CD133+ and CD133- cells of 12 primary colorectal tumors using laser capture microdissection and aCGH. Three of five CRC cell lines displayed both CD133+ and CD133- cells, but tumorigenicity of these subfractions did not differ significantly and aCGH revealed essentially identical genomic imbalances. However, 96 genes were differentially expressed between the two populations. Array comparative genomic hybridization analysis after laser capture microdissection of CD133+ and CD133- areas in primary colorectal tumors revealed genetic differences in 7 of 12 cases. The use of cell lines for studying genomic alterations that define cancer stem cell characteristics, therefore, seems questionable. In contrast, CD133+ cells in primary cancer samples showed a unique genomic aberration profile. In conclusion, our data suggest that CD133 positivity defines a genetically distinct cellular compartment in primary CRC, which potentially includes tumor initiating cells.
Article
Cyclooxygenase-2 is overexpressed in the majority of colorectal tumours leading to elevated levels of PGE(2), promoting many hallmarks of cancer. Importantly, PGE(2) is reported to enhance Wnt/β-catenin signalling in colorectal carcinoma cells and in normal haematopoietic stem cells where it promotes stem cell function. Although Wnt signalling plays a crucial role in intestinal stem cells, the relationship between PGE(2) and intestinal stem cells is unclear. Given that the key intestinal cancer stem cell marker LGR5 (Leucine-rich G-protein coupled Receptor 5) is a Wnt target and PGE(2) enhances Wnt signaling, the focus of this study was to investigate whether PGE(2) regulated LGR5 expression in colorectal adenoma cells and whether LGR5 was important for tumour cell survival. PGE(2) upregulated LGR5 protein in adenoma (RG/C2) and carcinoma (DLD1) cell lines. LGR5 knockdown induced cell death in RG/C2 and AA/C1 adenoma cells, suggesting that LGR5 has an important survival-promoting role in adenoma cells. Indeed, we detected LGR5 protein expression in 4/4 human adenoma cell lines. Furthermore, LGR5 siRNA inhibited the survival-promoting effects of PGE(2) in RG/C2, suggesting that PGE(2) promotes adenoma cell survival, at least in part, by increasing LGR5 expression. These studies therefore show the first link between PGE(2) and LGR5 in human colorectal adenoma and carcinoma cells and demonstrate a survival-promoting role of LGR5. As NSAIDs cause adenomas to regress in FAP patients, these studies could have important implications for the mechanism by which NSAIDs are chemopreventive, as lowering PGE(2) levels could reduce LGR5 expression and survival of LGR5+ adenoma stem cells.
Article
The cancer stem cell (CSC) model, in which a small population of cells within a tumor possesses the ability to self-renew and recon-stitute the phenotype of primary tumor, has gained wide accep-tance based on evidence over the past decade. It has also been reported that cancer cell lines contain a CSC subpopulation. How-ever, phenotypic differences between CSCs and non-CSCs in cancer cell lines are not better defined than in primary tumors. Further-more, some cell lines do not have a CSC population, revealed as a side population and expression of CD133. Thus, the identification of CSCs in cancer cell lines remains elusive. Here, we investigated the CSC hierarchy within HCT116 colon cancer cells, which do not have a CD133-positive subpopulation. We examined the expres-sion of alternative CSC markers epithelial specific antigen (ESA) and CD44 in floating-sphere-derived cells, which are known to be the cells of enriching CSCs. Sphere-derived HCT116 cells exhibited heterogeneous expression of ESA and CD44. The two major subpopulations of HCT116 sphere cells (ESA low CD44) ⁄ low and ESA high CD44 high) exhibited a biological ⁄ proliferative hierarchy of sphere-forming and soft agar colony-forming activity. However, there was no difference between the two subpopulations in the incidence of xenograft tumors. When ESA low CD44) ⁄ low cells were allowed to aggregate and re-form floating-spheres, the biologi-cal ⁄ proliferative hierarchy of parental HCT116 spheres was recon-stituted, in terms of ESA and CD44 expression. Thus, HCT116 cells have plasticity when they are set in floating-spheres, suggesting that maintenance of the HCT116 cell line conforms to a stochastic model, not a CSC model.
Article
In colorectal cancer (CRC), a subpopulation of tumor cells, called cancer stem cell (CSC) fraction, is suggested to be responsible for tumor initiation, growth, and metastasis. The search for a reliable marker to identify these CSCs is ongoing as current markers, like CD44 and CD133, are more broadly expressed and therefore are not highly selective and currently also lack function in CSC biology. Here, we analyzed whether the Wnt target Lgr5, which has earlier been identified as a marker for murine intestinal stem cells, could potentially serve as a functional marker for CSCs. Fluorescence-activated cell sorting-based detection of Lgr5, using three newly developed antibodies, on primary colorectal tumor cells revealed a clear subpopulation of Epcam(+) Lgr5(+) cells. Similarly, primary CRC-derived spheroid cultures, known to be enriched for CSCs, contain high levels of Lgr5(+) cells, which decrease upon in vitro differentiation of these CSCs. Selection of the Lgr5(high) CRC cells identified the clonogenic fraction in vitro as well as the tumorigenic population in vivo. Finally, we confirm that Lgr5 expression is dependent on the Wnt pathway and show that Lgr5 overexpression induces clonogenic growth. We thus provide evidence that Lgr5 is, next to a functional intestinal stem cell marker, a selective marker for human colorectal CSCs. STEM CELLS2012;30:2378-2386.