Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Developmental Biology 428 (2017) 273–282

Contents lists available at ScienceDirect

Developmental Biology
journal homepage: www.elsevier.com/locate/developmentalbiology

Review article

Wnt/β-catenin signaling in adult mammalian epithelial stem cells MARK


a,b a,b,c,⁎
Kai Kretzschmar , Hans Clevers
a
Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The
Netherlands
b
Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, The Netherlands
c
Princess Máxima Centre for Pediatric Oncology, 3584 CT Utrecht, The Netherlands

A BS T RAC T

Adult stem cells self-renew and replenish differentiated cells in various organs and tissues throughout a
mammal's life. Over the last 25 years an ever-growing body of knowledge has unraveled the essential regulation
of adult mammalian epithelia by the canonical Wnt signaling with its key intracellular effector β-catenin. In this
review, we discuss the principles of the signaling pathway and its role in adult epithelial stem cells of the
intestine and skin during homeostasis and tumorigenesis. We further highlight the research that led to the
identification of new stem cell markers and methods to study adult stem cells ex vivo.

1. Introduction (Wg), a gene critical in segment polarity during embryonic develop-


ment (Nüsslein-Volhard and Wieschaus, 1980; Rijsewijk et al., 1987).
Stem cells are the cell factories of the mammalian body. From the Further research established that Int-1 belongs to a family of genes
zygotes to the stem cells of the fully-grown animal, all stem cells share highly conserved across the animal kingdom, which is now known as
the capacity to self-renew as well as to produce differentiating daughter the wingless-type mouse mammary tumor virus (MMTV) integration
cells. Yet, as mammalian development progresses, stem cells with site (Wnt) gene family. To date, 19 Wnt genes in the genomes of mouse
decreasing potential to differentiate are produced. In later stages of and human have been described (Clevers and Nusse, 2012). Wnt genes
embryonic development, specialized stem cells are born that reside express signaling molecules that are components of a group of signal
within tissues and organs and fuel their growth and regeneration transduction cascades, known as Wnt signaling pathways. Wnt ligands
throughout adult life. These adult stem cells have the capacity to initiate the pathways through the interaction with cell surface receptors
produce one or more types of differentiated cells. During adult home- of the Frizzled (FZD) family. To activate canonical Wnt signaling with
ostasis, stem cell fate decisions are tightly regulated by extrinsic cues of its key intracellular effector β-catenin, a co-receptor of the low-density
their microenvironment, the so-called stem cell niche, as well as by lipoprotein (LDL) receptor family, LDL-related protein (LRP), is also
intracellular signaling cascades (Watt and Hogan, 2000). In this required (Pinson et al., 2000; Tamai et al., 2000). The other Wnt
review, we discuss one such cascade that is heavily studied as critical signaling pathways that do not involve β-catenin such as the non-
regulator of adult stem cells during homeostasis: the Wnt/β-catenin canonical pathways of Wnt/planar cell polarity (PCP) signaling and
signaling pathway. We further highlight how research on this signaling Wnt/Ca2+ signaling as well as the β-catenin-independent Wnt/LRP6
cascade in mammalian intestine and skin expanded our understanding pathways of Wnt/stabilization of proteins (STOP) signaling and Wnt/
of the underlying mechanisms of epithelial tumor formation and target of rapamycin (TOR) have been reviewed elsewhere (Acebron and
helped to uncover new markers of stem cells and a novel culture Niehrs, 2016; van Amerongen, 2012).
method for adult stem cells. In the absence of Wnt ligands, Frizzled and LRP receptors are
inactive and a destruction complex containing several components
2. The Wnt/β-catenin signaling pathway including adenomatous polyposis coli (APC) and Axin recruit newly
synthesized cytoplasmic β-catenin (Fig. 1). Trapped β-catenin is then
In 1982, Nusse and Varmus described the cloning of a new murine phosphorylated by two kinases of the destruction complex, casein
proto-oncogene that they called integration-1 (Int-1) (Nusse and kinase-1 (CK1) and glycogen synthase kinase-3 (GSK-3). Subsequently,
Varmus, 1982). It later emerged that its fruit fly homolog was wingless phosphorylated β-catenin is recognized by a E3 ligase harboring a β-


Corresponding author at: Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The
Netherlands.
E-mail address: h.clevers@hubrecht.eu (H. Clevers).

http://dx.doi.org/10.1016/j.ydbio.2017.05.015
Received 1 February 2017; Received in revised form 15 May 2017; Accepted 16 May 2017
Available online 17 May 2017
0012-1606/ © 2017 Elsevier Inc. All rights reserved.
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

extracellular Wnt OFF extracellular Wnt ON


Wnt

Frizzled Frizzled

cytoplasmic LRP
LRP P
Dvl P
cytoplasmic Axin
Dvl CK1
Axin GSK-3
-cat APC
CK1 P
GSK-3 P
-cat P
APC
P P -cat
P
-TrCP
-TrCP -cat
Proteasome
-cat -cat
P P U
P U
U

Groucho -cat
TCF TCF
nucleus nucleus

Fig. 1. Schematic of the canonical Wnt signaling, which is inactive in the absence of Wnt ligands (left panel) and active upon binding of Wnt ligands (right panel).

transduction repeat-containing protein (β-TrCP) that ubiquitinates β- Itasaki et al., 2003), block Wnt signaling through interaction with
catenin for proteasomal degradation. membrane-associated LRPs. Both Wnt ligands and LRPs are simulta-
Canonical Wnt signaling remains inactive until Wnt ligands bind neously bound by adenomatosis polyposis down-regulated 1 (APCDD1)
with both Frizzled and LRP receptors causing the phosphorylation of to inhibit Wnt/β-catenin activation (Shimomura et al., 2010). Apart
the latter receptor (Janda et al., 2012) (Fig. 1). Dishevelled (Dvl), as from its role as cytoplasmic effector of canonical Wnt signaling, β-
part of the destruction complex, acts as adaptor for phosphorylated catenin interacts with the cytoplasmic domain of E-cadherin and is also
LRP to recruit the entire intact complex to the plasma membrane (Li an intracellular component of adhesion junctions (Ozawa et al., 1989).
et al., 2012). While the destruction complex is still able to bind β- As such binding of β-catenin by E-cadherin at the plasma membrane
catenin and phosphorylate it, ubiquitination by β-TrCP is inhibited. may therefore reduce the cytoplasmic pool of β-catenin available for
Newly synthesized β-catenin accumulates in the cytoplasm eventually canonical Wnt signaling (Huelsken et al., 1994).
translocating into the nucleus. Nuclear β-catenin then replaces a Wnt agonists, in contrast, potentate signal strength of canonical
repressor of T-cell factor (TCF)/lymphoid enhancer factor (LEF) Wnt signaling. The secreted protein Norrin, for instance, binds
transcription factors named Groucho (Roose et al., 1998; van de Frizzled-4−LRP5 in the absence of Wnt ligands and, subsequently,
Wetering et al., 1991; Verbeek et al., 1995). Interaction of β-catenin activates β-catenin target gene expression (Junge et al., 2009; Xu et al.,
and TCF/LEF recruits transcriptional co-activators and histone modi- 2004). Secreted proteins of the roof plate-specific spondin (R-spondin
fiers such as the ATP-dependent helicase Brahma-related gene 1 or RSPO) family were initially described to be co-expressed with Wnt
(BRG1, also known as SMARCA4), cyclic adenosine monophosphate- ligands and agonists of canonical Wnt signaling (Kazanskaya et al.,
response element (CREB)-binding protein (CBP), p300, B-cell lympho- 2004). Later, research conducted independently by three different
ma 9 (BCL9) and pygopus (Korinek et al., 1997; Molenaar et al., 1996; groups revealed that R-spondins are the long unknown ligands of the
Morin et al., 1997; Schuijers et al., 2014; van de Wetering et al., 1997). leucine rich repeat containing G protein-coupled receptor (LGR) family
This complex is transcriptionally active allowing expression of β- (de Lau et al., 2011; Glinka et al., 2011; Ruffner et al., 2012).
catenin target genes, which mediate an array of developmental and Subsequently, it was discovered that the active LGR−RSPO complex
homeostatic processes, some to be discussed in this review. Wnt/β- binds and inactivates the transmembrane E3 ubiquitin ligases zinc and
catenin signal strength and the resulting transcriptional activity are ring finger 3 (ZNRF3) and ring finger protein 43 (RNF43), which are
highly context dependent. It has been suggested that even low levels of both β-catenin target genes and Wnt antagonists (Hao et al., 2012; Koo
nuclear β-catenin accumulation may be sufficient to induce the et al., 2012). The above described mechanism for the inactivation of
transcriptional program independent of the absolute levels of β-catenin ZNRF3 and RNF43 through LGR−RSPO is prerequisite for the
(Goentoro and Kirschner, 2009). Furthermore, cell type-specific Wnt/ prolonged stabilization of β-catenin, subsequent expression of β-
β-catenin target gene expression may also be determined by splice catenin target gene and stem cell self-renewal (Yan et al., 2017), which
variants of TCF/LEF1 transcription factors present in the transcrip- plays an essential role for adult homeostasis of epithelial tissues. Work
tional complex (Hovanes et al., 2001; Van de Wetering et al., 1996; in the 1990s established that Wnt/β-catenin signaling is not only
Wallmen et al., 2012). required for early embryonic development in vertebrates (Korinek
A variety of extracellular and membrane-associated modulators et al., 1998b), but is also critical for regulating adult homeostasis in
acting either as inhibitors or agonists tightly regulate Wnt/β-catenin mammals. In 1998, a study linked canonical Wnt signaling with the
signaling. Several extracellular secreted molecules are Wnt pathway maintenance of the adult stem cell compartment first using the mouse
inhibitors that directly bind Wnt ligands such as secreted Frizzled- intestine as a model stimulating a new direction of research (Korinek
related proteins (SFRPs) (Leyns et al., 1997; Wang et al., 1997) or Wnt et al., 1998a).
inhibitory factor (WIF) (Hsieh et al., 1999). Other inhibitors, for
instance, members of the Dickkopf family (DKKs) (Glinka et al.,
3. Wnt/β-catenin signaling in intestinal stem cells
1998; Mao et al., 2001), sclerostin (SOST) (Semenov et al., 2005)
and sclerostin domain containing 1 (SOSTDC1) (Ahn et al., 2010;
The intestinal tract can be separated into two major parts, the small

274
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

intestine (subdivided into duodenum, jejunum and ileum) and the


large intestine (consisting of cecum, colon and rectum). To increase the
surface of the absorptive epithelium, the small intestine comprises villi
that harbor differentiated cell types such as enterocytes, enteroendo-
crine cells and goblet cells, while the large intestine is devoid of villi.
Cellular input for the constant loss of differentiated cells is governed by
a compartment of proliferative cells residing in specialized stem cell
niches, called crypts of Lieberkühn, which are embedded in the
mesenchymal tissue of both small and large intestine (Lieberkühn,
1745).
To investigate the role of canonical Wnt signaling for adult home-
ostasis of the gut, Korinek et al. (1998a) inactivated β-catenin target
gene expression through depletion of Tcf7l2, the gene encoding TCF4,
which is specifically expressed in intestinal crypts. Mice lacking TCF4
developed a severe phenotype upon the transition from embryonic
development to postnatal homeostasis of the gut. Tcf7l2-/- mice died
shortly after birth and while the villus compartment of the small
intestine remained unchanged, the entire crypt epithelium with
harboring the adult stem cells was absent (Korinek et al., 1998a).
Overexpression of the Wnt inhibitor DKK1 using transgenic mice or Fig. 3. Crypt bottom of the intestinal epithelium with different cell types described in
adenoviral treatment of wildtype mice completely disrupts proliferation this compartment. *Lrig1 expression specifically in +4 position cells remains contro-
in the adult intestinal crypts (Kuhnert et al., 2004; Pinto et al., 2003) versial.
(Fig. 2A). Later, two different studies inactivated Wnt/β-catenin
signaling postnatally in the intestinal epithelium in vivo (Fevr et al., 1887) and were named crypt base columnar (CBC) cells by Cheng and
2007; van Es et al., 2012a). This resulted in increased intestinal Leblond more than forty years ago (Cheng and Leblond, 1974);
differentiation and complete loss of stem cell proliferation in the crypt however, their existence was neglected until the discovery of Lgr5
demonstrating the critical role of the pathway for adult intestinal brought them back under attention (Fig. 3). To further study these
homeostasis (Fevr et al., 2007; van Es et al., 2012a) (Fig. 2B). To Lgr5+ CBC cells, a genetic mouse models was generated that allowed
further study adult stem cells in the intestine and their specific role of purification of Lgr5+ cells by flow cytometry as well as in vivo lineage-
homeostasis, markers were required. tracing experiments using the inducible Cre/loxP system (Barker et al.,
A screen of previously identified β-catenin target genes provided 2007). When Barker and coworkers injected tamoxifen into double-
several candidate marker genes enriched in putative adult stem cells mutant mice carrying both Lgr5 lineage tracer and LacZ reporter, only
(van de Wetering et al., 2002). The gene Lgr5 was one of them and single Lgr5+ cells in the bottom of the crypts were β-gal+ the following
showed a unique expression pattern marking proliferating cells inter- day. Subsequently, histological analysis performed at later time points,
spersed between Paneth cells in the bottom of intestinal crypts (Barker revealed that the number of β-gal+ intestinal cells dramatically
et al., 2007). These actively cycling cells were described by Josef Paneth increased within five days and labelled clones had expanded out of
140 years ago as “schmale Zellen” (German for slender cells) (Paneth, the crypt bottom reaching high into the villus compartment of the small

Fig. 2. Overexpression of Wnt inhibitor DKK1 (A) or inactivation of the transcription factor TCF4 (B) in the intestinal epithelium causes a loss of the stem cell compartment (positive for
the proliferation marker Ki67 in the left panels and absent in right panels) in the crypt bottom. Images adapted from Pinto et al. (2003) in (A) and from van Es et al. (2012a) in (B).

275
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

intestine (Barker et al., 2007). Further characterization using cell Matrigel (Sato et al., 2009). Providing the embedded single cells with
morphology and co-marker expression confirmed that all differentiated culture medium rich in stem cell niche factors such as epidermal
cell types present in the epithelium of the small intestine including the growth factor (EGF), the bone morphogenetic protein (BMP) inhibitor
absorptive enterocytes as well as the secretory enteroendocrine cells, Noggin and the above discussed Wnt agonist R-spondin, spontaneously
goblet cells and Paneth cells were indeed β-gal+ progeny of Lgr5+ CBC generated three-dimensional self-organizing cell clusters that re-
cells. Long-term lineage tracing showed persistence of the β-gal+ sembled the anatomy of the intestinal epithelium (Sato et al., 2009).
progeny in the intestinal epithelium for the life time of a mouse, These in-vitro grown structures, so-called organoids, not only con-
demonstrating that Lgr5+ cells are bona fide adult stem cells in vivo tained Lgr5+ stem cells, but all differentiated cell types of the intestinal
(Barker et al., 2007). Several follow-up studies utilized multicolor epithelium (Grün et al., 2015; Sato et al., 2009). Using minced pieces of
lineage tracing as well as intravital imaging experiments to define that murine intestinal tissue Kuo and colleagues succeeded in generating
Lgr5+ stem cells divide symmetrically and stochastically compete for organoid cultures, which, however, required mesenchymal cells as
space in the crypt bottom stem cell niche, a process called neutral drift support (Ootani et al., 2009). These organoids contained stem cells
(Lopez-Garcia et al., 2010; Ritsma et al., 2014; Snippert et al., 2010b). expressing Lgr5 as well as Bmi1 that increased in number upon R-
Additional β-catenin target genes were later found to be specific or at spondin treatment. Organoid cultures can be maintained in culture
least enriched in these CBC cells, such as achaete scute-like 2 (Ascl2) long-term and can be used to selectively induce particular types of
(Schuijers et al., 2015; van der Flier et al., 2009), prominin-1/cluster of differentiated intestinal epithelial cells by manipulating intracellular
differentiation 133 (CD133) (Snippert et al., 2009; Zhu et al., 2009) signaling pathways including the Wnt/β-catenin pathway (Basak et al.,
and Musashi-1 (Potten et al., 2003). 2016; Sato et al., 2009; Yin et al., 2014) further highlighting that Wnt-
The discovery of rapidly cycling, but long-lived Lgr5+ stem cells was active intestinal crypt cells such as those expressing Lgr5+ are indeed
in stark contrast to the previous dogma that quiescent, DNA label- stem cells in vivo and ex vivo.
retaining intestinal stem cells reside in the so-called +4 position just Since intestinal stem cell homeostasis is dependent on the activity
above the Paneth cells (Potten et al., 1978) (Fig. 3). However, the of the Wnt/β-catenin signaling cascade, it does not come as a surprise
putative stemness character of the +4 cells had not been assessed that components of the pathway are frequently mutated in colorectal
experimentally before, because of missing specific markers. carcinomas (CRCs). The main molecular mechanism behind tumor
Subsequently, several markers such as B lumphoma Mo-MLV insertion initiation in the intestine are loss-of-function mutations in the APC
region 1 homolog (Bmi1), mouse telomerase reverse transcriptase gene cause cytoplasmic stabilization of β-catenin allowing for its
(mTert), homeodomain-only protein (Hopx) and leucine-rich repeats subsequent nuclear translocation and activation of target gene expres-
and immunoglobulin-like domains protein 1 (Lrig1) were proposed to sion (Korinek et al., 1997; Rubinfeld et al., 1996; Su et al., 1993). In
specifically label quiescent stem cells at the +4 position (Fig. 3). Several rare instances activating mutations in CTNNB1, the gene encoding β-
studies demonstrated that cells expressing these markers are compe- catenin, or inactivating mutations in the AXIN2 gene have been found
tent to regenerate the intestinal epithelium including Lgr5+ stem cells to be tumor-initiating (Lammi et al., 2004; Liu et al., 2000; Morin
during homeostasis and upon injury or are, in contrast to Lgr5+ stem et al., 1997). Crypt stem cells expressing markers such as Lgr5, Bmi1
cells, radiation-resistant (Li et al., 2014; Montgomery et al., 2011; or CD133/Prominin-1 that acquire APC mutations are likely cells-of-
Powell et al., 2012; Sangiorgi and Capecchi, 2008; Takeda et al., 2011; origin of intestinal cancers (Barker et al., 2009; Sangiorgi and
Tian et al., 2011; Yan et al., 2012). However, the interpretation of these Capecchi, 2008; Schepers et al., 2012; Zhu et al., 2009). APC loss-of-
results remains controversial. For instance, the exact expression function in progenitor cells is insufficient to drive intestinal tumor-
pattern of Lrig1 is disputed, as Jensen and colleagues described at igenesis (Barker et al., 2009; Tetteh et al., 2016a). However, when APC
the same time as Powell et al. that Lrig1 is broadly expressed in rapidly is inactivated in the presence of mutations in KRAS, TP53 and SMAD4
cycling cells in the lower intestinal crypt with the highest levels found that are associated with tumor propagation (Drost et al., 2015; Fearon
in Lgr5+ stem cells (Wong et al., 2012). Furthermore, crypt expression and Vogelstein, 1990; Fumagalli et al., 2017), CRCs can also arise from
of the other +4 cell markers Bmi1, mTert and Hopx is broader than differentiated intestinal cells in an experimental setting (Tetteh et al.,
previously anticipated with high levels of these markers indeed being 2016b).
found in Lgr5+ stem cells (Grün et al., 2015; Munoz et al., 2012; Wang
et al., 2013). On the other hand, robust evidence suggests that the 4. Wnt/β-catenin signaling in epidermal stem cells
intestinal epithelium is unexpectedly plastic, which makes drawing
conclusions on a possible stem cell hierarchy difficult (Tetteh et al., Canonical Wnt signaling is also heavily studied in another epithelial
2015). Our group showed that secretory precursor cells expressing the tissue: mammalian epidermis, which comprises a stratified epithelium
Notch ligand Delta-like 1 (Dll1) are competent to replenish the stem called interfollicular epidermis and several specialized appendages,
cell niche upon loss of Lgr5+ stem cells (van Es et al., 2012b). A study hair follicles, sebaceous glands and sweat glands. The stem cell
by Winton and colleagues revealed that a pool of Bmi1+ intestinal compartment of the epidermis is the so-called basal layer that is
label-retaining cells (LRCs) acting as secretory precursors of the embedded in an extracellular matrix (ECM)-protein-rich basement
lineages of Paneth cells and enteroendocrine cells during normal membrane deposited by the dermis, a mesenchymal niche containing
homeostasis, can replenish the entire intestinal epithelium upon different cell types such as fibroblasts. Committed cells leave the basal
damage (Buczacki et al., 2013). Recent work demonstrated that also layer, move into the suprabasal layers and terminally differentiate into
the precursors of the most abundant intestinal epithelial cell type, the specialized epidermal lineages contributing the protective barrier of the
absorptive enterocytes, have the capacity to replace lost Lgr5+ stem skin. For instance, sebocytes produce a moist and oily sebum contain-
cells (Tetteh et al., 2016a). It therefore remains unclear if Lgr5+ CBC ing antimicrobial products, follicular keratinocytes generate hair shafts
cells and +4 cells are distinct stem cell compartments or indeed that collectively built the heat-protective fur coat and interfollicular
interdependent stem cell pools during intestinal homeostasis and keratinocytes express crosslinking structural proteins (keratinization)
regeneration in vivo (Takeda et al., 2011). This unresolved question to eventually building an impenetrable physical barrier of highly
requires further research to uncover the exact nature of the relation- keratinized dead cells, the cornified envelope. While interfollicular
ship of these intestinal stem cell populations. epidermis and sebaceous glands undergo constant cellular turnover,
The remarkable capacity of Lgr5+ CBC cells to renew the intestinal hair follicles self-renew in a cyclic fashion with phases of hair growth
epithelium long-term can not only be observed in vivo. Utilizing the (anagen), regression (catagen) and rest (telogen).
EGFP tag it was possible to purify single Lgr5−EGFP+ stem cells by In 2001, an elegant study provided first evidence for the critical role
flow cytometry and embed them into extracellular matrix (ECM)-rich of Wnt/β-catenin signaling during hair follicle formation and regen-

276
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

eration in murine skin epidermis (Huelsken et al., 2001). In their epidermal lineages (Jaks et al., 2008). While Lgr5 expression in skin
study, Birchmeier and colleagues specifically depleted of β-catenin in development was not detected until late in embryogenesis at embryonic
the epidermal basal layer. This resulted in perturbed hair follicle day (E)18.5, its close relative Lgr6 is expressed as early as E14.5 in hair
morphogenesis during skin development, while maintenance of the placodes forming in the skin (Snippert et al., 2010a). Postnatally, Lgr6
hair follicle stem cell compartment was disrupted postnatally triggering marks actively cycling stem cells in the isthmus, a region of the hair
hair follicle loss in young adult mice (Huelsken et al., 2001). However, follicle just above the bulge, the sebaceous gland and the interfollicular
formation of the stratified epithelium in between hair follicles, called epidermis (Füllgrabe et al., 2015; Kretzschmar et al., 2016; Page et al.,
interfollicular epidermis, was not effected in neonatal mice. Depletion 2013; Snippert et al., 2010a) (Fig. 5). These epidermal compartments
of the genes encoding LEF1, TCF3 and TCF4 resulted in similar are also regenerated long-term by Lgr6+ stem cells, which further
phenotypes as those described for epidermal specific β-catenin deletion contribute to wound repair and new hair follicles arising in adult mouse
(Nguyen et al., 2009; van Genderen et al., 1994). While transgenic mice skin (Füllgrabe et al., 2015; Kretzschmar et al., 2016; Snippert et al.,
overexpressing LEF1, the main transcription factor interacting with β- 2010a). Interestingly, Lgr5 is co-expressed with Lgr4 in the lower hair
catenin to trigger expression of hair differentiation-specific Wnt target follicle, while Lgr6 is not co-expressed with either in the epidermis
genes, under the control of the keratin 14 (K14) promoter (specifically (Snippert et al., 2010a). The reason for this distinct compartmentaliza-
expressed in the epidermal basal layer) showed aberrant formation of tion of the Lgr family members within the epidermis is not known and
hair placodes in epidermal compartments such as the interfollicular requires further investigations.
epidermis (Zhou et al., 1995). These early studies suggested that Wnt/ Since Lgr6 is, in contrast to Lgr5, no direct Wnt/β-catenin target
β-catenin signaling may be essential in epidermal stem cell fate gene, and considering earlier studies showing no obvious effect of
decisions selecting the differentiation towards either the follicular or inactivating β-catenin in the interfollicular epidermis and sebaceous
the epidermal lineage. glands (Huelsken et al., 2001), it remained unclear whether Wnt-
In agreement with this work, several studies by the laboratories of responsive cells existed outside of the lower hair follicle (i.e. hair bulge
Fuchs and Watt demonstrated that Wnt/β-catenin signaling in post- and germ). A study by Nusse and colleagues addressed this issue and
natal epidermis acts as a critical regulator of the lineage selection discovered that stem cells in the basal layer of interfollicular epidermis
program of epidermal stem cells. Different mouse models with genetic expressed the Wnt/β-catenin target gene Axin2 (Lim et al., 2013)
stabilization of β-catenin in the epidermal basal layer showed de novo (Fig. 5). These stem cells not only robustly contributed to epidermal
formation of hair follicles in adult skin (Baker et al., 2010; Gat et al., wound repair, but also required activation of canonical Wnt signaling
1998; Lo Celso et al., 2004; Silva-Vargas et al., 2005). Transient in an autocrine manner to proliferate and maintain adult homeostasis
stabilization of β-catenin in specific regions of the epidermal basal layer of the epidermis. Interestingly, a subsequent study showed that Axin2
of adult mouse skin induced formation of so-called ectopic hair follicles is also expressed by a population of hair follicle stem cells that are
from epidermal compartments such as the interfollicular epidermis maintained by a similar autocrine mechanism, yet, these cells located
and the sebaceous glands, normally avoid of hair follicle differentiation in the outer bulge appear to be quiescent (Lim et al., 2016) (Fig. 5).
(Baker et al., 2010; Silva-Vargas et al., 2005) (Fig. 4). To demonstrate Collectively, these results demonstrate that Wnt/β-catenin signaling
the opposite, the two groups independently generated transgenic mice plays an essential role for the homeostasis of adult skin epidermis.
expressing a N-terminally truncated version of LEF1 (ΔNLEF1) under Some of its components and target genes are further markers of
the control of the K14 promoter that allowed for inhibition of the distinct epidermal stem cell populations that govern cellular input into
transcriptional activity of canonical Wnt signaling, even in the presence the autonomously maintained lineages of epidermal differentiation
of β-catenin in the nucleus (Merrill et al., 2001; Niemann et al., 2002). (Kretzschmar et al., 2016; Page et al., 2013).
Gradually, this resulted in a loss of hair differentiation and a conver- In the epidermis, sustained Wnt/β-catenin signaling is critical for
sion of the hair follicles into cysts, expressing markers of both the cell differentiation into the hair follicle lineage. In line with this
interfollicular epidermis and sebaceous gland (Merrill et al., 2001; observation, activating mutations of pathway components, in particular
Niemann et al., 2002). Critical component of the hair follicle stem cell of β-catenin itself, have been associated with the initiation of hair
niche are mesenchymal cells that aggregate together to form the dermal follicle tumors such as pilomatricomas (Chan et al., 1999; Gat et al.,
papilla (Driskell et al., 2009; Greco et al., 2009; Jahoda et al., 1984; 1998; Lo Celso et al., 2004). The most common skin cancer, the basal
Rompolas et al., 2012). Modulation of Wnt/β-catenin signaling within cell carcinoma (BCC), has been shown to arise from activating
dermal papilla cells has been shown to control hair follicle growth mutations of both Wnt/β-catenin and Hedgehog signaling (Palmer
(Kishimoto et al., 2000; Kitagawa et al., 2009). However, there is et al., 2008; Salto-Tellez et al., 2006; Youssef et al., 2012). The cellular
growing evidence that β-catenin activation within hair follicle stem origin of BCCs remained controversial, since this tumor type resembles
cells is sufficient to drive hair growth (Deschene et al., 2014; undifferentiated follicular and interfollicular basal keratinocytes.
Kretzschmar et al., 2015) (Fig. 4). Collectively, these studies exemplify Several studies by the Blanpain and co-workers suggested that BCCs
the critical role of Wnt/β-catenin signaling for postnatal skin home- are initiated by interfollicular epidermal cells that adopt a hair follicle-
ostasis and epidermal stem cell biology. like phenotype (Youssef et al., 2012; Youssef et al., 2010), while other
Epidermal stem cells were initially believed to be solely localized to reports defined hair follicle stem cells as tumor-initiating cells
a region of the hair follicle enriched for label-retaining cells (LRCs), the (Peterson et al., 2015; Wang et al., 2011). On the other hand,
so-called bulge (Braun et al., 2003; Cotsarelis et al., 1990; Tumbar Grachtchouk and colleagues showed in mice that different BCC
et al., 2004). However, in the past ten years more and more stem cell subtypes arise from different epidermal subcompartments with the
populations have been described in the epidermis (Kretzschmar and interfollicular epidermis being the origin of superficial BCCs and
Watt, 2014). Several components of canonical Wnt pathway have been nodular BCCs developing from the hair follicle (Grachtchouk et al.,
shown to be specifically expressed in epidermal stem cells. A study in 2011). Another type of β-catenin-dependent epidermal tumors are
2008, for instance, found that Lgr5 marks rapidly cycling, Wnt-active trichofolliculomas, which are benign and contain mostly differentiated
stem cells in the hair follicle (Jaks et al., 2008) (Fig. 5). Lgr5 expression cells (Gat et al., 1998), possibly because of activation of vitamin D
did overlap with known bulge stem cell markers such as CD34 and K15, receptor signaling (Palmer et al., 2008). While most β-catenin-depen-
while being absent from LRCs. Yet, Lgr5+ hair follicle stem cells exhibit dent epidermal tumors adopt a HF-like phenotype, inactivating muta-
the highest colony-forming capacity of all tested epidermal stem cell tions in LEF1 trigger formation of sebaceous gland tumors (Niemann
populations tested in vitro. Furthermore, lineage tracing experiments et al., 2002; Niemann et al., 2003; Takeda et al., 2006). Huelsken and
revealed robust contribution of Lgr5+ stem cells to the cellular input in co-workers showed that in squamous cell carcinomas (SCCs) generated
vivo to the hair follicle lineage and in skin reconstitution assays to all in the epidermis of mice using the classical two-step chemical

277
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

Fig. 4. Activation of β-catenin in adult epidermis induces formation of ectopic hair follicle from sebaceous glands and other non-follicular epidermal compartments. Wild-type (A) or
ΔK5ΔNβ-cateninER transgenic mice (B, C) received one to three doses of 1.5 mg 4-hydroxytamoxifen (4-OHT) and tissue was collected one or two weeks later as indicated. Back skin
sections were stained with hematoxylin and eosin (H & E). Lines indicate ectopic HFs and dashed lines demarcate sebaceous glands. Scale bars: 200 µm.
Adapted from Kretzschmar et al. (2016).

carcinogenesis protocol, hair follicle stem cells with high levels of how research into this pathway has uncovered several adult stem cell
nuclear β-catenin acted as tumor-propagating cells (Malanchi et al., markers expressed in these tissues, some of them even functional such
2008). This study suggests that SCCs may also be β-catenin-dependent as Lgr5 (de Lau et al., 2011) and Axin2 (Lim et al., 2013). Based on
epidermal tumors, as epidermal loss of β-catenin caused complete these discoveries, Wnt pathway components and β-catenin target genes
tumor regression. The involvement of Wnt/β-catenin signaling in many were identified as adult stem cell markers in many other epithelial
different types of epidermal tumors demonstrates its critical role for tissues of the mammalian body. Stem cells in the stomach (Barker
neoplasms of the skin. Compartmentalization of epidermal stem cells et al., 2010), kidney (Barker et al., 2012), mammary glands (de Visser
as well as cross-talk with other signaling pathways may underlie et al., 2012; Plaks et al., 2013; Rios et al., 2014; Shackleton et al., 2006;
epidermal tumor heterogeneity arising from altered Wnt/β-catenin Van Keymeulen et al., 2011), cochlea of the ear (Bramhall et al., 2014;
signaling (Kretzschmar et al., 2016; Watt and Collins, 2008). Chai et al., 2012; McLean et al., 2016; Shi et al., 2012), ovary (Flesken-
Nikitin et al., 2013) and taste buds (Ren et al., 2014; Takeda et al.,
5. Concluding remarks 2013; Yee et al., 2013) have been shown to express Lgr5, while Lgr6+
stem cells are described for mammary glands (Blaas et al., 2016), taste
In this review, we have presented an overview over the current buds (Ren et al., 2014), nails (Lehoczky and Tabin, 2015) and cochlea
knowledge about the Wnt/β-catenin signaling cascade and its role for (Zhang et al., 2015). Axin2+ stem and progenitor cells also have been
the epithelia in adult intestine and skin. We have further highlighted identified in mammary gland (van Amerongen et al., 2012), kidney

278
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

Haegebarth, A., Korving, J., Begthel, H., Peters, P.J., Clevers, H., 2007. Identification
of stem cells in small intestine and colon by marker gene Lgr5. Nature 449,
1003–1007.
Basak, O., Beumer, J., Wiebrands, K., Seno, H., van Oudenaarden, A., Clevers, H., 2016.
Induced quiescence of Lgr5+ stem cells in intestinal organoids enables
differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell.
Blaas, L., Pucci, F., Messal, H.A., Andersson, A.B., Josue Ruiz, E., Gerling, M., Douagi, I.,
Spencer-Dene, B., Musch, A., Mitter, R., Bhaw, L., Stone, R., Bornhorst, D., Sesay,
A.K., Jonkers, J., Stamp, G., Malanchi, I., Toftgard, R., Behrens, A., 2016. Lgr6 labels
a rare population of mammary gland progenitor cells that are able to originate
luminal mammary tumours. Nat. Cell Biol. 18, 1346–1356.
Bramhall, N.F., Shi, F., Arnold, K., Hochedlinger, K., Edge, A.S., 2014. Lgr5-positive
supporting cells generate new hair cells in the postnatal cochlea. Stem Cell Rep. 2,
311–322.
Braun, K.M., Niemann, C., Jensen, U.B., Sundberg, J.P., Silva-Vargas, V., Watt, F.M.,
2003. Manipulation of stem cell proliferation and lineage commitment: visualisation
of label-retaining cells in wholemounts of mouse epidermis. Development 130,
5241–5255.
Buczacki, S.J., Zecchini, H.I., Nicholson, A.M., Russell, R., Vermeulen, L., Kemp, R.,
Winton, D.J., 2013. Intestinal label-retaining cells are secretory precursors
expressing Lgr5. Nature 495, 65–69.
Chai, R., Kuo, B., Wang, T., Liaw, E.J., Xia, A., Jan, T.A., Liu, Z., Taketo, M.M., Oghalai,
J.S., Nusse, R., Zuo, J., Cheng, A.G., 2012. Wnt signaling induces proliferation of
sensory precursors in the postnatal mouse cochlea. Proc. Natl. Acad. Sci. USA 109,
8167–8172.
Fig. 5. Epidermal stem cell populations in adult telogen skin (hair follicles are resting) Chan, E.F., Gat, U., McNiff, J.M., Fuchs, E., 1999. A common human skin tumour is
marked by components of the Wnt/β-catenin signaling pathway. caused by activating mutations in beta-catenin. Nat. Genet 21, 410–413.
Cheng, H., Leblond, C.P., 1974. Origin, differentiation and renewal of the four main
epithelial cell types in the mouse small intestine. I. Columnar cell. Am. J. Anat. 141,
(Rinkevich et al., 2014), liver (Wang et al., 2015) and cochlea (Jan 461–479.
et al., 2013). Further, the understanding of the intestinal stem cell Clevers, H., Nusse, R., 2012. Wnt/beta-catenin signaling and disease. Cell 149,
niche requiring Wnt ligands and R-spondins and the ability to purify 1192–1205.
Cotsarelis, G., Sun, T.T., Lavker, R.M., 1990. Label-retaining cells reside in the bulge area
adult intestinal stem cells led to one of the most exiting discovery of the of pilosebaceous unit: implications for follicular stem cells, hair cycle, and skin
last ten year, the generation of organoid cultures (Sato et al., 2009). carcinogenesis. Cell 61, 1329–1337.
Organoid technology has transformed the fields of developmental de Lau, W., Barker, N., Low, T.Y., Koo, B.K., Li, V.S., Teunissen, H., Kujala, P.,
Haegebarth, A., Peters, P.J., van de Wetering, M., Stange, D.E., van Es, J.E.,
biology and stem cell research (Kretzschmar and Clevers, 2016). It is
Guardavaccaro, D., Schasfoort, R.B., Mohri, Y., Nishimori, K., Mohammed, S., Heck,
also a promising application in disease research and clinical therapeu- A.J., Clevers, H., 2011. Lgr5 homologues associate with Wnt receptors and mediate
tics. And yet, it all started from a simple observation in the mouse R-spondin signalling. Nature 476, 293–297.
de Visser, K.E., Ciampricotti, M., Michalak, E.M., Tan, D.W., Speksnijder, E.N., Hau,
intestine linking canonical Wnt signaling with the maintenance of the
C.S., Clevers, H., Barker, N., Jonkers, J., 2012. Developmental stage-specific
adult stem cell compartment (Korinek et al., 1998a). We look forward contribution of LGR5(+) cells to basal and luminal epithelial lineages in the postnatal
to more exiting research shedding light on unknown roles of Wnt/β- mammary gland. J. Pathol. 228, 300–309.
catenin signaling cascade on adult epithelia and their stem cell niches. Deschene, E.R., Myung, P., Rompolas, P., Zito, G., Sun, T.Y., Taketo, M.M., Saotome, I.,
Greco, V., 2014. Beta-Catenin activation regulates tissue growth non-cell
autonomously in the hair stem cell niche. Science 343, 1353–1356.
Acknowledgments Driskell, R.R., Giangreco, A., Jensen, K.B., Mulder, K.W., Watt, F.M., 2009. Sox2-positive
dermal papilla cells specify hair follicle type in mammalian epidermis. Development
136, 2815–2823.
We are grateful to all colleagues in the field who contributed to the Drost, J., van Jaarsveld, R.H., Ponsioen, B., Zimberlin, C., van Boxtel, R., Buijs, A., Sachs,
work described in this review. We apologize to those whose research N., Overmeer, R.M., Offerhaus, G.J., Begthel, H., Korving, J., van de Wetering, M.,
could not be discussed due to space limitations. We are thankful to Kim Schwank, G., Logtenberg, M., Cuppen, E., Snippert, H.J., Medema, J.P., Kops, G.J.,
Clevers, H., 2015. Sequential cancer mutations in cultured human intestinal stem
Boonekamp for critical reading the manuscript. KK is supported by a cells. Nature 521, 43–47.
VENI grant from the Netherlands Organisation for Scientific Fearon, E.R., Vogelstein, B., 1990. A genetic model for colorectal tumorigenesis. Cell 61,
Research (NWO-ZonMW, 016.166.140) and is a long-term fellow of 759–767.
Fevr, T., Robine, S., Louvard, D., Huelsken, J., 2007. Wnt/beta-catenin is essential for
the Human Frontier Science Program Organization (HFSPO, LT771/
intestinal homeostasis and maintenance of intestinal stem cells. Mol. Cell Biol. 27,
2015). HC is named inventor on several patents related to Lgr5 stem- 7551–7559.
cell-based organoid technology. Flesken-Nikitin, A., Hwang, C.I., Cheng, C.Y., Michurina, T.V., Enikolopov, G., Nikitin,
A.Y., 2013. Ovarian surface epithelium at the junction area contains a cancer-prone
stem cell niche. Nature 495, 241–245.
References Füllgrabe, A., Joost, S., Are, A., Jacob, T., Sivan, U., Haegebarth, A., Linnarsson, S.,
Simons, B.D., Clevers, H., Toftgard, R., Kasper, M., 2015. Dynamics of Lgr6(+)
Acebron, S.P., Niehrs, C., 2016. β-catenin-independent roles of Wnt/LRP6 signaling. progenitor cells in the hair follicle, sebaceous gland, and interfollicular epidermis.
Trends Cell Biol. 26, 956–967. Stem Cell Rep. 5, 843–855.
Ahn, Y., Sanderson, B.W., Klein, O.D., Krumlauf, R., 2010. Inhibition of Wnt signaling by Fumagalli, A., Drost, J., Suijkerbuijk, S.J., van Boxtel, R., de Ligt, J., Offerhaus, G.J.,
Wise (Sostdc1) and negative feedback from Shh controls tooth number and Begthel, H., Beerling, E., Tan, E.H., Sansom, O.J., Cuppen, E., Clevers, H., van
patterning. Development 137, 3221–3231. Rheenen, J., 2017. Genetic dissection of colorectal cancer progression by orthotopic
Baker, C.M., Verstuyf, A., Jensen, K.B., Watt, F.M., 2010. Differential sensitivity of transplantation of engineered cancer organoids. Proc. Natl. Acad. Sci. USA 114,
epidermal cell subpopulations to beta-catenin-induced ectopic hair follicle E2357–E2364.
formation. Dev. Biol. 343, 40–50. Gat, U., DasGupta, R., Degenstein, L., Fuchs, E., 1998. De Novo hair follicle
Barker, N., Huch, M., Kujala, P., van de Wetering, M., Snippert, H.J., van Es, J.H., Sato, morphogenesis and hair tumors in mice expressing a truncated beta-catenin in skin.
T., Stange, D.E., Begthel, H., van den Born, M., Danenberg, E., van den Brink, S., Cell 95, 605–614.
Korving, J., Abo, A., Peters, P.J., Wright, N., Poulsom, R., Clevers, H., 2010. Glinka, A., Dolde, C., Kirsch, N., Huang, Y.L., Kazanskaya, O., Ingelfinger, D., Boutros,
Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric M., Cruciat, C.M., Niehrs, C., 2011. LGR4 and LGR5 are R-spondin receptors
units in vitro. Cell Stem Cell 6, 25–36. mediating Wnt/beta-catenin and Wnt/PCP signalling. EMBO Rep. 12, 1055–1061.
Barker, N., Ridgway, R.A., van Es, J.H., van de Wetering, M., Begthel, H., van den Born, Glinka, A., Wu, W., Delius, H., Monaghan, A.P., Blumenstock, C., Niehrs, C., 1998.
M., Danenberg, E., Clarke, A.R., Sansom, O.J., Clevers, H., 2009. Crypt stem cells as Dickkopf-1 is a member of a new family of secreted proteins and functions in head
the cells-of-origin of intestinal cancer. Nature 457, 608–611. induction. Nature 391, 357–362.
Barker, N., Rookmaaker, M.B., Kujala, P., Ng, A., Leushacke, M., Snippert, H., van de Goentoro, L., Kirschner, M.W., 2009. Evidence that fold-change, and not absolute level,
Wetering, M., Tan, S., Van, Es, J.H., Huch, M., Poulsom, R., Verhaar, M.C., Peters, of beta-catenin dictates Wnt signaling. Mol. Cell 36, 872–884.
P.J., Clevers, H., 2012. Lgr5(+ve) stem/progenitor cells contribute to nephron Grachtchouk, M., Pero, J., Yang, S.H., Ermilov, A.N., Michael, L.E., Wang, A., Wilbert,
formation during kidney development. Cell Rep. 2, 540–552. D., Patel, R.M., Ferris, J., Diener, J., Allen, M., Lim, S., Syu, L.J., Verhaegen, M.,
Barker, N., van Es, J.H., Kuipers, J., Kujala, P., van den Born, M., Cozijnsen, M., Dlugosz, A.A., 2011. Basal cell carcinomas in mice arise from hair follicle stem cells

279
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

and multiple epithelial progenitor populations. J. Clin. Invest. 121, 1768–1781. cells establishes intestinal stem cell hierarchy. Stem Cell Rep. 3, 876–891.
Greco, V., Chen, T., Rendl, M., Schober, M., Pasolli, H.A., Stokes, N., Dela Cruz-Racelis, Li, V.S., Ng, S.S., Boersema, P.J., Low, T.Y., Karthaus, W.R., Gerlach, J.P., Mohammed,
J., Fuchs, E., 2009. A two-step mechanism for stem cell activation during hair S., Heck, A.J., Maurice, M.M., Mahmoudi, T., Clevers, H., 2012. Wnt signaling
regeneration. Cell Stem Cell 4, 155–169. through inhibition of beta-catenin degradation in an intact Axin1 complex. Cell 149,
Grün, D., Lyubimova, A., Kester, L., Wiebrands, K., Basak, O., Sasaki, N., Clevers, H., van 1245–1256.
Oudenaarden, A., 2015. Single-cell messenger RNA sequencing reveals rare Lieberkühn, J.N., 1745. Dissertatio de fabrica et actione villorum intestinorum tenuium
intestinal cell types. Nature 525, 251–255. hominis, Leiden.
Hao, H.X., Xie, Y., Zhang, Y., Charlat, O., Oster, E., Avello, M., Lei, H., Mickanin, C., Liu, Lim, X., Tan, S.H., Koh, W.L., Chau, R.M., Yan, K.S., Kuo, C.J., van Amerongen, R.,
D., Ruffner, H., Mao, X., Ma, Q., Zamponi, R., Bouwmeester, T., Finan, P.M., Klein, A.M., Nusse, R., 2013. Interfollicular epidermal stem cells self-renew via
Kirschner, M.W., Porter, J.A., Serluca, F.C., Cong, F., 2012. ZNRF3 promotes Wnt autocrine Wnt signaling. Science 342, 1226–1230.
receptor turnover in an R-spondin-sensitive manner. Nature 485, 195–200. Lim, X., Tan, S.H., Yu, K.L., Lim, S.B., Nusse, R., 2016. Axin2 marks quiescent hair
Hovanes, K., Li, T.W., Munguia, J.E., Truong, T., Milovanovic, T., Lawrence Marsh, J., follicle bulge stem cells that are maintained by autocrine Wnt/beta-catenin signaling.
Holcombe, R.F., Waterman, M.L., 2001. Beta-catenin-sensitive isoforms of lymphoid Proc. Natl. Acad. Sci. USA 113, E1498–E1505.
enhancer factor-1 are selectively expressed in colon cancer. Nat. Genet. 28, 53–57. Liu, W., Dong, X., Mai, M., Seelan, R.S., Taniguchi, K., Krishnadath, K.K., Halling, K.C.,
Hsieh, J.C., Kodjabachian, L., Rebbert, M.L., Rattner, A., Smallwood, P.M., Samos, C.H., Cunningham, J.M., Boardman, L.A., Qian, C., Christensen, E., Schmidt, S.S., Roche,
Nusse, R., Dawid, I.B., Nathans, J., 1999. A new secreted protein that binds to Wnt P.C., Smith, D.I., Thibodeau, S.N., 2000. Mutations in AXIN2 cause colorectal cancer
proteins and inhibits their activities. Nature 398, 431–436. with defective mismatch repair by activating beta-catenin/TCF signalling. Nat.
Huelsken, J., Birchmeier, W., Behrens, J., 1994. E-cadherin and APC compete for the Genet. 26, 146–147.
interaction with beta-catenin and the cytoskeleton. J. Cell Biol. 127, 2061–2069. Lo Celso, C., Prowse, D.M., Watt, F.M., 2004. Transient activation of beta-catenin
Huelsken, J., Vogel, R., Erdmann, B., Cotsarelis, G., Birchmeier, W., 2001. beta-Catenin signalling in adult mouse epidermis is sufficient to induce new hair follicles but
controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell continuous activation is required to maintain hair follicle tumours. Development
105, 533–545. 131, 1787–1799.
Itasaki, N., Jones, C.M., Mercurio, S., Rowe, A., Domingos, P.M., Smith, J.C., Krumlauf, Lopez-Garcia, C., Klein, A.M., Simons, B.D., Winton, D.J, 2010. Intestinal stem cell
R., 2003. Wise, a context-dependent activator and inhibitor of Wnt signalling. replacement follows a pattern of neutral drift. Science 330, 822–825.
Development 130, 4295–4305. Malanchi, I., Peinado, H., Kassen, D., Hussenet, T., Metzger, D., Chambon, P., Huber, M.,
Jahoda, C.A., Horne, K.A., Oliver, R.F., 1984. Induction of hair growth by implantation of Hohl, D., Cano, A., Birchmeier, W., Huelsken, J., 2008. Cutaneous cancer stem cell
cultured dermal papilla cells. Nature 311, 560–562. maintenance is dependent on beta-catenin signalling. Nature 452, 650–653.
Jaks, V., Barker, N., Kasper, M., van Es, J.H., Snippert, H.J., Clevers, H., Toftgard, R., Mao, B., Wu, W., Li, Y., Hoppe, D., Stannek, P., Glinka, A., Niehrs, C., 2001. LDL-
2008. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat. Genet. 40, receptor-related protein 6 is a receptor for Dickkopf proteins. Nature 411, 321–325.
1291–1299. McLean, W.J., McLean, D.T., Eatock, R.A., Edge, A.S., 2016. Distinct capacity for
Jan, T.A., Chai, R., Sayyid, Z.N., van Amerongen, R., Xia, A., Wang, T., Sinkkonen, S.T., differentiation to inner ear cell types by progenitor cells of the cochlea and vestibular
Zeng, Y.A., Levin, J.R., Heller, S., Nusse, R., Cheng, A.G., 2013. Tympanic border organs. Development 143, 4381–4393.
cells are Wnt-responsive and can act as progenitors for postnatal mouse cochlear Merrill, B.J., Gat, U., DasGupta, R., Fuchs, E., 2001. Tcf3 and Lef1 regulate lineage
cells. Development 140, 1196–1206. differentiation of multipotent stem cells in skin. Genes Dev. 15, 1688–1705.
Janda, C.Y., Waghray, D., Levin, A.M., Thomas, C., Garcia, K.C., 2012. Structural basis of Molenaar, M., van de Wetering, M., Oosterwegel, M., Peterson-Maduro, J., Godsave, S.,
Wnt recognition by Frizzled. Science 337, 59–64. Korinek, V., Roose, J., Destree, O., Clevers, H., 1996. XTcf-3 transcription factor
Junge, H.J., Yang, S., Burton, J.B., Paes, K., Shu, X., French, D.M., Costa, M., Rice, D.S., mediates beta-catenin-induced axis formation in Xenopus embryos. Cell 86,
Ye, W., 2009. TSPAN12 regulates retinal vascular development by promoting 391–399.
Norrin- but not Wnt-induced FZD4/beta-catenin signaling. Cell 139, 299–311. Montgomery, R.K., Carlone, D.L., Richmond, C.A., Farilla, L., Kranendonk, M.E.,
Kazanskaya, O., Glinka, A., del Barco Barrantes, I., Stannek, P., Niehrs, C., Wu, W., 2004. Henderson, D.E., Baffour-Awuah, N.Y., Ambruzs, D.M., Fogli, L.K., Algra, S.,
R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Breault, D.T., 2011. Mouse telomerase reverse transcriptase (mTert) expression
Xenopus myogenesis. Dev. Cell 7, 525–534. marks slowly cycling intestinal stem cells. Proc. Natl. Acad. Sci. USA 108, 179–184.
Kishimoto, J., Burgeson, R.E., Morgan, B.A., 2000. Wnt signaling maintains the hair- Morin, P.J., Sparks, A.B., Korinek, V., Barker, N., Clevers, H., Vogelstein, B., Kinzler,
inducing activity of the dermal papilla. Genes Dev. 14, 1181–1185. K.W., 1997. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in
Kitagawa, T., Matsuda, K., Inui, S., Takenaka, H., Katoh, N., Itami, S., Kishimoto, S., beta-catenin or APC. Science 275, 1787–1790.
Kawata, M., 2009. Keratinocyte growth inhibition through the modification of Wnt Munoz, J., Stange, D.E., Schepers, A.G., van de Wetering, M., Koo, B.K., Itzkovitz, S.,
signaling by androgen in balding dermal papilla cells. J. Clin. Endocrinol. Metab. 94, Volckmann, R., Kung, K.S., Koster, J., Radulescu, S., Myant, K., Versteeg, R.,
1288–1294. Sansom, O.J., van Es, J.H., Barker, N., van Oudenaarden, A., Mohammed, S., Heck,
Koo, B.K., Spit, M., Jordens, I., Low, T.Y., Stange, D.E., van de Wetering, M., van Es, A.J., Clevers, H., 2012. The Lgr5 intestinal stem cell signature: robust expression of
J.H., Mohammed, S., Heck, A.J., Maurice, M.M., Clevers, H., 2012. Tumour proposed quiescent '+4' cell markers. EMBO J. 31, 3079–3091.
suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nguyen, H., Merrill, B.J., Polak, L., Nikolova, M., Rendl, M., Shaver, T.M., Pasolli, H.A.,
Nature 488, 665–669. Fuchs, E., 2009. Tcf3 and Tcf4 are essential for long-term homeostasis of skin
Korinek, V., Barker, N., Moerer, P., van Donselaar, E., Huls, G., Peters, P.J., Clevers, H., epithelia. Nat. Genet. 41, 1068–1075.
1998a. Depletion of epithelial stem-cell compartments in the small intestine of mice Niemann, C., Owens, D.M., Hulsken, J., Birchmeier, W., Watt, F.M., 2002. Expression of
lacking Tcf-4. Nat. Genet. 19, 379–383. DeltaNLef1 in mouse epidermis results in differentiation of hair follicles into
Korinek, V., Barker, N., Morin, P.J., van Wichen, D., de Weger, R., Kinzler, K.W., squamous epidermal cysts and formation of skin tumours. Development 129,
Vogelstein, B., Clevers, H., 1997. Constitutive transcriptional activation by a beta- 95–109.
catenin-Tcf complex in APC-/- colon carcinoma. Science 275, 1784–1787. Niemann, C., Unden, A.B., Lyle, S., Zouboulis Ch, C., Toftgard, R., Watt, F.M., 2003.
Korinek, V., Barker, N., Willert, K., Molenaar, M., Roose, J., Wagenaar, G., Markman, M., Indian hedgehog and beta-catenin signaling: role in the sebaceous lineage of normal
Lamers, W., Destree, O., Clevers, H., 1998b. Two members of the Tcf family and neoplastic mammalian epidermis. Proc. Natl. Acad. Sci. USA 100 (Suppl 1),
implicated in Wnt/beta-catenin signaling during embryogenesis in the mouse. Mol. 11873–11880.
Cell Biol. 18, 1248–1256. Nusse, R., Varmus, H.E., 1982. Many tumors induced by the mouse mammary tumor
Kretzschmar, K., Clevers, H., 2016. Organoids: modeling development and the stem cell virus contain a provirus integrated in the same region of the host genome. Cell 31,
niche in a dish. Dev. Cell 38, 590–600. 99–109.
Kretzschmar, K., Cottle, D.L., Schweiger, P.J., Watt, F.M., 2015. The androgen receptor Nüsslein-Volhard, C., Wieschaus, E., 1980. Mutations affecting segment number and
antagonizes Wnt/beta-catenin signaling in epidermal stem cells. J. Invest. Dermatol. polarity in Drosophila. Nature 287, 795–801.
135, 2753–2763. Ootani, A., Li, X., Sangiorgi, E., Ho, Q.T., Ueno, H., Toda, S., Sugihara, H., Fujimoto, K.,
Kretzschmar, K., Watt, F.M., 2014. Markers of epidermal stem cell subpopulations in Weissman, I.L., Capecchi, M.R., Kuo, C.J., 2009. Sustained in vitro intestinal
adult mammalian skin. Cold Spring Harb. Perspect. Med., 4. epithelial culture within a Wnt-dependent stem cell niche. Nat. Med. 15, 701–706.
Kretzschmar, K., Weber, C., Driskell, R.R., Calonje, E., Watt, F.M., 2016. Ozawa, M., Baribault, H., Kemler, R., 1989. The cytoplasmic domain of the cell adhesion
Compartmentalized epidermal activation of beta-Catenin differentially affects lineage molecule uvomorulin associates with three independent proteins structurally related
reprogramming and underlies tumor heterogeneity. Cell Rep. 14, 269–281. in different species. EMBO J. 8, 1711–1717.
Kuhnert, F., Davis, C.R., Wang, H.T., Chu, P., Lee, M., Yuan, J., Nusse, R., Kuo, C.J., Page, M.E., Lombard, P., Ng, F., Gottgens, B., Jensen, K.B., 2013. The epidermis
2004. Essential requirement for Wnt signaling in proliferation of adult small comprises autonomous compartments maintained by distinct stem cell populations.
intestine and colon revealed by adenoviral expression of Dickkopf-1. Proc. Natl. Cell Stem Cell 13, 471–482.
Acad. Sci. USA 101, 266–271. Palmer, H.G., Anjos-Afonso, F., Carmeliet, G., Takeda, H., Watt, F.M., 2008. The vitamin
Lammi, L., Arte, S., Somer, M., Jarvinen, H., Lahermo, P., Thesleff, I., Pirinen, S., D receptor is a Wnt effector that controls hair follicle differentiation and specifies
Nieminen, P., 2004. Mutations in AXIN2 cause familial tooth agenesis and tumor type in adult epidermis. PLoS One 3, e1483.
predispose to colorectal cancer. Am. J. Hum. Genet. 74, 1043–1050. Paneth, J., 1887. Ueber die secernirenden Zellen des Dünndarm-Epithels. Arch. für
Lehoczky, J.A., Tabin, C.J., 2015. Lgr6 marks nail stem cells and is required for digit tip Mikrosk. Anat. 31, 113–192.
regeneration. Proc. Natl. Acad. Sci. USA 112, 13249–13254. Peterson, S.C., Eberl, M., Vagnozzi, A.N., Belkadi, A., Veniaminova, N.A., Verhaegen,
Leyns, L., Bouwmeester, T., Kim, S.H., Piccolo, S., De Robertis, E.M., 1997. Frzb-1 is a M.E., Bichakjian, C.K., Ward, N.L., Dlugosz, A.A., Wong, S.Y., 2015. Basal cell
secreted antagonist of Wnt signaling expressed in the Spemann organizer. Cell 88, carcinoma preferentially arises from stem cells within hair follicle and
747–756. mechanosensory niches. Cell Stem Cell 16, 400–412.
Li, N., Yousefi, M., Nakauka-Ddamba, A., Jain, R., Tobias, J., Epstein, J.A., Jensen, S.T., Pinson, K.I., Brennan, J., Monkley, S., Avery, B.J., Skarnes, W.C., 2000. An LDL-
Lengner, C.J., 2014. Single-cell analysis of proxy reporter allele-marked epithelial receptor-related protein mediates Wnt signalling in mice. Nature 407, 535–538.

280
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

Pinto, D., Gregorieff, A., Begthel, H., Clevers, H., 2003. Canonical Wnt signals are Veenboer, C., Barker, N., Klein, A.M., van Rheenen, J., Simons, B.D., Clevers, H.,
essential for homeostasis of the intestinal epithelium. Genes Dev. 17, 1709–1713. 2010b. Intestinal crypt homeostasis results from neutral competition between
Plaks, V., Brenot, A., Lawson, D.A., Linnemann, J.R., Van Kappel, E.C., Wong, K.C., de symmetrically dividing Lgr5 stem cells. Cell 143, 134–144.
Sauvage, F., Klein, O.D., Werb, Z., 2013. Lgr5-expressing cells are sufficient and Snippert, H.J., van Es, J.H., van den Born, M., Begthel, H., Stange, D.E., Barker, N.,
necessary for postnatal mammary gland organogenesis. Cell Rep. 3, 70–78. Clevers, H., 2009. Prominin-1/CD133 marks stem cells and early progenitors in
Potten, C.S., Booth, C., Tudor, G.L., Booth, D., Brady, G., Hurley, P., Ashton, G., Clarke, mouse small intestine. Gastroenterology 136, 2187–2194, (e2181).
R., Sakakibara, S., Okano, H., 2003. Identification of a putative intestinal stem cell Su, L.K., Vogelstein, B., Kinzler, K.W., 1993. Association of the APC tumor suppressor
and early lineage marker; musashi-1. Differentiation 71, 28–41. protein with catenins. Science 262, 1734–1737.
Potten, C.S., Hume, W.J., Reid, P., Cairns, J., 1978. The segregation of DNA in epithelial Takeda, H., Lyle, S., Lazar, A.J., Zouboulis, C.C., Smyth, I., Watt, F.M., 2006. Human
stem cells. Cell 15, 899–906. sebaceous tumors harbor inactivating mutations in LEF1. Nat. Med. 12, 395–397.
Powell, A.E., Wang, Y., Li, Y., Poulin, E.J., Means, A.L., Washington, M.K., Takeda, N., Jain, R., LeBoeuf, M.R., Wang, Q., Lu, M.M., Epstein, J.A., 2011.
Higginbotham, J.N., Juchheim, A., Prasad, N., Levy, S.E., Guo, Y., Shyr, Y., Aronow, Interconversion between intestinal stem cell populations in distinct niches. Science
B.J., Haigis, K.M., Franklin, J.L., Coffey, R.J., 2012. The pan-ErbB negative regulator 334, 1420–1424.
Lrig1 is an intestinal stem cell marker that functions as a tumor suppressor. Cell 149, Takeda, N., Jain, R., Li, D., Li, L., Lu, M.M., Epstein, J.A., 2013. Lgr5 identifies
146–158. progenitor cells capable of taste bud regeneration after injury. PLoS One 8, e66314.
Ren, W., Lewandowski, B.C., Watson, J., Aihara, E., Iwatsuki, K., Bachmanov, A.A., Tamai, K., Semenov, M., Kato, Y., Spokony, R., Liu, C., Katsuyama, Y., Hess, F., Saint-
Margolskee, R.F., Jiang, P., 2014. Single Lgr5- or Lgr6-expressing taste stem/ Jeannet, J.P., He, X., 2000. LDL-receptor-related proteins in Wnt signal
progenitor cells generate taste bud cells ex vivo. Proc. Natl. Acad. Sci. USA 111, transduction. Nature 407, 530–535.
16401–16406. Tetteh, P.W., Basak, O., Farin, H.F., Wiebrands, K., Kretzschmar, K., Begthel, H., van den
Rijsewijk, F., Schuermann, M., Wagenaar, E., Parren, P., Weigel, D., Nusse, R., 1987. The Born, M., Korving, J., de Sauvage, F., van Es, J.H., van Oudenaarden, A., Clevers, H.,
Drosophila homolog of the mouse mammary oncogene int-1 is identical to the 2016a. Replacement of lost Lgr5-positive stem cells through plasticity of their
segment polarity gene wingless. Cell 50, 649–657. enterocyte-lineage daughters. Cell Stem Cell 18, 203–213.
Rinkevich, Y., Montoro, D.T., Contreras-Trujillo, H., Harari-Steinberg, O., Newman, Tetteh, P.W., Farin, H.F., Clevers, H., 2015. Plasticity within stem cell hierarchies in
A.M., Tsai, J.M., Lim, X., Van-Amerongen, R., Bowman, A., Januszyk, M., mammalian epithelia. Trends Cell Biol. 25, 100–108.
Pleniceanu, O., Nusse, R., Longaker, M.T., Weissman, I.L., Dekel, B., 2014. In vivo Tetteh, P.W., Kretzschmar, K., Begthel, H., van den Born, M., Korving, J., Morsink, F.,
clonal analysis reveals lineage-restricted progenitor characteristics in mammalian Farin, H., van Es, J.H., Offerhaus, G.J., Clevers, H., 2016b. Generation of an
kidney development, maintenance, and regeneration. Cell Rep. 7, 1270–1283. inducible colon-specific Cre enzyme mouse line for colon cancer research. Proc. Natl.
Rios, A.C., Fu, N.Y., Lindeman, G.J., Visvader, J.E., 2014. In situ identification of Acad. Sci. USA 113, 11859–11864.
bipotent stem cells in the mammary gland. Nature 506, 322–327. Tian, H., Biehs, B., Warming, S., Leong, K.G., Rangell, L., Klein, O.D., de Sauvage, F.J.,
Ritsma, L., Ellenbroek, S.I., Zomer, A., Snippert, H.J., de Sauvage, F.J., Simons, B.D., 2011. A reserve stem cell population in small intestine renders Lgr5-positive cells
Clevers, H., van Rheenen, J., 2014. Intestinal crypt homeostasis revealed at single- dispensable. Nature 478, 255–259.
stem-cell level by in vivo live imaging. Nature 507, 362–365. Tumbar, T., Guasch, G., Greco, V., Blanpain, C., Lowry, W.E., Rendl, M., Fuchs, E., 2004.
Rompolas, P., Deschene, E.R., Zito, G., Gonzalez, D.G., Saotome, I., Haberman, A.M., Defining the epithelial stem cell niche in skin. Science 303, 359–363.
Greco, V., 2012. Live imaging of stem cell and progeny behaviour in physiological van Amerongen, R., 2012. Alternative Wnt pathways and receptors. Cold Spring Harb.
hair-follicle regeneration. Nature 487, 496–499. Perspect. Biol., 4.
Roose, J., Molenaar, M., Peterson, J., Hurenkamp, J., Brantjes, H., Moerer, P., van de van Amerongen, R., Bowman, A.N., Nusse, R., 2012. Developmental stage and time
Wetering, M., Destree, O., Clevers, H., 1998. The Xenopus Wnt effector XTcf-3 dictate the fate of Wnt/beta-catenin-responsive stem cells in the mammary gland.
interacts with groucho-related transcriptional repressors. Nature 395, 608–612. Cell Stem Cell 11, 387–400.
Rubinfeld, B., Albert, I., Porfiri, E., Fiol, C., Munemitsu, S., Polakis, P., 1996. Binding of Van de Wetering, M., Castrop, J., Korinek, V., Clevers, H., 1996. Extensive alternative
GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. splicing and dual promoter usage generate Tcf-1 protein isoforms with differential
Science 272, 1023–1026. transcription control properties. Mol. Cell Biol. 16, 745–752.
Ruffner, H., Sprunger, J., Charlat, O., Leighton-Davies, J., Grosshans, B., Salathe, A., van de Wetering, M., Cavallo, R., Dooijes, D., van Beest, M., van Es, J., Loureiro, J.,
Zietzling, S., Beck, V., Therier, M., Isken, A., Xie, Y., Zhang, Y., Hao, H., Shi, X., Liu, Ypma, A., Hursh, D., Jones, T., Bejsovec, A., Peifer, M., Mortin, M., Clevers, H.,
D., Song, Q., Clay, I., Hintzen, G., Tchorz, J., Bouchez, L.C., Michaud, G., Finan, P., 1997. Armadillo coactivates transcription driven by the product of the Drosophila
Myer, V.E., Bouwmeester, T., Porter, J., Hild, M., Bassilana, F., Parker, C.N., Cong, segment polarity gene dTCF. Cell 88, 789–799.
F., 2012. R-Spondin potentiates Wnt/beta-catenin signaling through orphan van de Wetering, M., Oosterwegel, M., Dooijes, D., Clevers, H., 1991. Identification and
receptors LGR4 and LGR5. PLoS One 7, e40976. cloning of TCF-1, a T lymphocyte-specific transcription factor containing a sequence-
Salto-Tellez, M., Peh, B.K., Ito, K., Tan, S.H., Chong, P.Y., Han, H.C., Tada, K., Ong, W.Y., specific HMG box. EMBO J. 10, 123–132.
Soong, R., Voon, D.C., Ito, Y., 2006. RUNX3 protein is overexpressed in human basal van de Wetering, M., Sancho, E., Verweij, C., de Lau, W., Oving, I., Hurlstone, A., van der
cell carcinomas. Oncogene 25, 7646–7649. Horn, K., Batlle, E., Coudreuse, D., Haramis, A.P., Tjon-Pon-Fong, M., Moerer, P.,
Sangiorgi, E., Capecchi, M.R., 2008. Bmi1 is expressed in vivo in intestinal stem cells. van den Born, M., Soete, G., Pals, S., Eilers, M., Medema, R., Clevers, H., 2002. The
Nat. Genet. 40, 915–920. beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal
Sato, T., Vries, R.G., Snippert, H.J., van de Wetering, M., Barker, N., Stange, D.E., van cancer cells. Cell 111, 241–250.
Es, J.H., Abo, A., Kujala, P., Peters, P.J., Clevers, H., 2009. Single Lgr5 stem cells van der Flier, L.G., van Gijn, M.E., Hatzis, P., Kujala, P., Haegebarth, A., Stange, D.E.,
build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, Begthel, H., van den Born, M., Guryev, V., Oving, I., van Es, J.H., Barker, N., Peters,
262–265. P.J., van de Wetering, M., Clevers, H., 2009. Transcription factor achaete scute-like 2
Schepers, A.G., Snippert, H.J., Stange, D.E., van den Born, M., van Es, J.H., van de controls intestinal stem cell fate. Cell 136, 903–912.
Wetering, M., Clevers, H., 2012. Lineage tracing reveals Lgr5+ stem cell activity in van Es, J.H., Haegebarth, A., Kujala, P., Itzkovitz, S., Koo, B.K., Boj, S.F., Korving, J., van
mouse intestinal adenomas. Science 337, 730–735. den Born, M., van Oudenaarden, A., Robine, S., Clevers, H., 2012a. A critical role for
Schuijers, J., Junker, J.P., Mokry, M., Hatzis, P., Koo, B.K., Sasselli, V., van der Flier, the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal. Mol. Cell Biol. 32,
L.G., Cuppen, E., van Oudenaarden, A., Clevers, H., 2015. Ascl2 acts as an R- 1918–1927.
spondin/Wnt-responsive switch to control stemness in intestinal crypts. Cell Stem van Es, J.H., Sato, T., van de Wetering, M., Lyubimova, A., Nee, A.N., Gregorieff, A.,
Cell 16, 158–170. Sasaki, N., Zeinstra, L., van den Born, M., Korving, J., Martens, A.C., Barker, N., van
Schuijers, J., Mokry, M., Hatzis, P., Cuppen, E., Clevers, H., 2014. Wnt-induced Oudenaarden, A., Clevers, H., 2012b. Dll1+ secretory progenitor cells revert to stem
transcriptional activation is exclusively mediated by TCF/LEF. EMBO J. 33, cells upon crypt damage. Nat. Cell Biol. 14, 1099–1104.
146–156. van Genderen, C., Okamura, R.M., Farinas, I., Quo, R.G., Parslow, T.G., Bruhn, L.,
Semenov, M., Tamai, K., He, X., 2005. SOST is a ligand for LRP5/LRP6 and a Wnt Grosschedl, R., 1994. Development of several organs that require inductive
signaling inhibitor. J. Biol. Chem. 280, 26770–26775. epithelial-mesenchymal interactions is impaired in LEF-1-deficient mice. Genes Dev.
Shackleton, M., Vaillant, F., Simpson, K.J., Stingl, J., Smyth, G.K., Asselin-Labat, M.L., 8, 2691–2703.
Wu, L., Lindeman, G.J., Visvader, J.E., 2006. Generation of a functional mammary Van Keymeulen, A., Rocha, A.S., Ousset, M., Beck, B., Bouvencourt, G., Rock, J., Sharma,
gland from a single stem cell. Nature 439, 84–88. N., Dekoninck, S., Blanpain, C., 2011. Distinct stem cells contribute to mammary
Shi, F., Kempfle, J.S., Edge, A.S., 2012. Wnt-responsive Lgr5-expressing stem cells are gland development and maintenance. Nature 479, 189–193.
hair cell progenitors in the cochlea. J. Neurosci. 32, 9639–9648. Verbeek, S., Izon, D., Hofhuis, F., Robanus-Maandag, E., te Riele, H., van de Wetering,
Shimomura, Y., Agalliu, D., Vonica, A., Luria, V., Wajid, M., Baumer, A., Belli, S., M., Oosterwegel, M., Wilson, A., MacDonald, H.R., Clevers, H., 1995. An HMG-box-
Petukhova, L., Schinzel, A., Brivanlou, A.H., Barres, B.A., Christiano, A.M., 2010. containing T-cell factor required for thymocyte differentiation. Nature 374, 70–74.
APCDD1 is a novel Wnt inhibitor mutated in hereditary hypotrichosis simplex. Wallmen, B., Schrempp, M., Hecht, A., 2012. Intrinsic properties of Tcf1 and Tcf4 splice
Nature 464, 1043–1047. variants determine cell-type-specific Wnt/beta-catenin target gene expression.
Silva-Vargas, V., Lo Celso, C., Giangreco, A., Ofstad, T., Prowse, D.M., Braun, K.M., Watt, Nucleic Acids Res. 40, 9455–9469.
F.M., 2005. Beta-catenin and Hedgehog signal strength can specify number and Wang, B., Zhao, L., Fish, M., Logan, C.Y., Nusse, R., 2015. Self-renewing diploid
location of hair follicles in adult epidermis without recruitment of bulge stem cells. Axin2(+) cells fuel homeostatic renewal of the liver. Nature 524, 180–185.
Dev. Cell 9, 121–131. Wang, F., Scoville, D., He, X.C., Mahe, M.M., Box, A., Perry, J.M., Smith, N.R., Lei, N.Y.,
Snippert, H.J., Haegebarth, A., Kasper, M., Jaks, V., van Es, J.H., Barker, N., van de Davies, P.S., Fuller, M.K., Haug, J.S., McClain, M., Gracz, A.D., Ding, S., Stelzner, M.,
Wetering, M., van den Born, M., Begthel, H., Vries, R.G., Stange, D.E., Toftgard, R., Dunn, J.C., Magness, S.T., Wong, M.H., Martin, M.G., Helmrath, M., Li, L., 2013.
Clevers, H., 2010a. Lgr6 marks stem cells in the hair follicle that generate all cell Isolation and characterization of intestinal stem cells based on surface marker
lineages of the skin. Science 327, 1385–1389. combinations and colony-formation assay. Gastroenterology 145 (383–395),
Snippert, H.J., van der Flier, L.G., Sato, T., van Es, J.H., van den Born, M., Kroon- e381–321.

281
K. Kretzschmar, H. Clevers Developmental Biology 428 (2017) 273–282

Wang, G.Y., Wang, J., Mancianti, M.L., Epstein, E.H., Jr., 2011. Basal cell carcinomas spondin ligands during Lgr5+ intestinal stem-cell self-renewal. Nature 545,
arise from hair follicle stem cells in Ptch1(+/-) mice. Cancer Cell 19, 114–124. 238–242.
Wang, S., Krinks, M., Lin, K., Luyten, F.P., Moos, M., Jr., 1997. Frzb, a secreted protein Yee, K.K., Li, Y., Redding, K.M., Iwatsuki, K., Margolskee, R.F., Jiang, P., 2013. Lgr5-
expressed in the Spemann organizer, binds and inhibits Wnt-8. Cell 88, 757–766. EGFP marks taste bud stem/progenitor cells in posterior tongue. Stem Cells 31,
Watt, F.M., Collins, C.A., 2008. Role of beta-catenin in epidermal stem cell expansion, 992–1000.
lineage selection, and cancer. Cold Spring Harb. Symp. Quant. Biol. 73, 503–512. Yin, X., Farin, H.F., van Es, J.H., Clevers, H., Langer, R., Karp, J.M., 2014. Niche-
Watt, F.M., Hogan, B.L., 2000. Out of Eden: stem cells and their niches. Science 287, independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny.
1427–1430. Nat. Methods 11, 106–112.
Wong, V.W., Stange, D.E., Page, M.E., Buczacki, S., Wabik, A., Itami, S., van de Wetering, Youssef, K.K., Lapouge, G., Bouvree, K., Rorive, S., Brohee, S., Appelstein, O., Larsimont,
M., Poulsom, R., Wright, N.A., Trotter, M.W., Watt, F.M., Winton, D.J., Clevers, H., J.C., Sukumaran, V., Van de Sande, B., Pucci, D., Dekoninck, S., Berthe, J.V., Aerts,
Jensen, K.B., 2012. Lrig1 controls intestinal stem-cell homeostasis by negative S., Salmon, I., del Marmol, V., Blanpain, C., 2012. Adult interfollicular tumour-
regulation of ErbB signalling. Nat. Cell Biol. 14, 401–408. initiating cells are reprogrammed into an embryonic hair follicle progenitor-like fate
Xu, Q., Wang, Y., Dabdoub, A., Smallwood, P.M., Williams, J., Woods, C., Kelley, M.W., during basal cell carcinoma initiation. Nat. Cell Biol. 14, 1282–1294.
Jiang, L., Tasman, W., Zhang, K., Nathans, J., 2004. Vascular development in the Youssef, K.K., Van Keymeulen, A., Lapouge, G., Beck, B., Michaux, C., Achouri, Y.,
retina and inner ear: control by Norrin and Frizzled-4, a high-affinity ligand-receptor Sotiropoulou, P.A., Blanpain, C., 2010. Identification of the cell lineage at the origin
pair. Cell 116, 883–895. of basal cell carcinomaNat. Cell Biol. 12, 299–305.
Yan, K.S., Chia, L.A., Li, X., Ootani, A., Su, J., Lee, J.Y., Su, N., Luo, Y., Heilshorn, S.C., Zhang, Y., Chen, Y., Ni, W., Guo, L., Lu, X., Liu, L., Li, W., Sun, S., Wang, L., Li, H., 2015.
Amieva, M.R., Sangiorgi, E., Capecchi, M.R., Kuo, C.J., 2012. The intestinal stem cell Dynamic expression of Lgr6 in the developing and mature mouse cochlea. Front Cell
markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl. Neurosci. 9, 165.
Acad. Sci. USA 109, 466–471. Zhou, P., Byrne, C., Jacobs, J., Fuchs, E., 1995. Lymphoid enhancer factor 1 directs hair
Yan, K.S., Janda, C.Y., Chang, J., Zheng, G.X.Y., Larkin, K.A., Luca, V.C., Chia, L.A., Mah, follicle patterning and epithelial cell fate. Genes Dev. 9, 700–713.
A.T., Han, A., Terry, J.M., Ootani, A., Roelf, K., Lee, M., Yuan, J., Li, X., Bolen, C.R., Zhu, L., Gibson, P., Currle, D.S., Tong, Y., Richardson, R.J., Bayazitov, I.T., Poppleton,
Wilhelmy, J., Davies, P.S., Ueno, H., von Furstenberg, R.J., Belgrader, P., Ziraldo, H., Zakharenko, S., Ellison, D.W., Gilbertson, R.J., 2009. Prominin 1 marks
S.B., Ordonez, H., Henning, S.J., Wong, M.H., Snyder, M.P., Weissman, I.L., Hsueh, intestinal stem cells that are susceptible to neoplastic transformation. Nature 457,
A.J., Mikkelsen, T.S., Garcia, K.C., Kuo, C.J., 2017. Non-equivalence of Wnt and R- 603–607.

282

You might also like