20
INTRODUCTION Tumour is composed of a heterogeneous group of cells with different morphologies and behaviour. Research in cancer biology indicates that several cancers are supported by a small subset of cells with stem cell like properties and are termed as cancer stem cells (CSCs) or tumour initiating cells (TIC). Evidences of CSCs involved in resistance to conventional therapies, leading to metastasis and tumour recurrence is abundant (Beck and Blanpain, 2013; Chandler and Lagasse, 2010; Prince and Ailles, 2008). As early as 1937, Furth and colleagues demonstrated that a single cell was able to produce a haematopoietic malignancy on implantation in mice (Furth et al., 1937). This suggested that certain cells within a tumour may have the ability to give rise to tumour growth (Furth et al., 1937). Later, in 1994, John Dick's group identified human acute myeloid leukaemia-initiating cells using + - CD34 CD38 markers and showed that these cells initiated tumour (Lapidot et al., 1994). In 1997, Bonnet and Dick showed for the first + - time that the CD34 CD38 population of cells had the self-renewal property. The authors performed limiting dilution assay to show that + - low numbers of CD34 CD38 cells were able to form tumours in NOD/SCID mice, identical to donors; whereas considerably higher Key words: Cancer stem cells (CSCs); EMT, Epithelial to mesenchymal transition; Lineage tracing; β-catenin; NICD, Notch intracellular domain. *Corresponding Author: Sanjeev K. Waghmare, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India. Email: [email protected] Developmental Signalling in Maintenance and Regulation of Cancer Stem Cells Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India Sweta Dash, Raghava Reddy Sunkara and Sanjeev K. Waghmare* Tissue stem cells self-renew throughout the life of an organism thereby maintaining tissue homeostasis and prevent cancer. The major signalling pathways such as Wnt, Notch and Sonic hedgehog control the stem cell regulation and their deregulation leads to cancer. Recent evidences showed that there exists a subset of cells within tumour termed as cancer stem cells (CSCs). These CSCs escape the conventional chemo- radiotherapy and further lead to tumour relapse followed by metastasis. This review focuses on the developmental signalling pathways that are involved in the regulation and maintenance of normal stem cells and CSCs. Understanding the molecular mechanism may be useful to specifically target the CSCs while sparing the normal stem cells to reduce tumorigenecity. Biomed Res J 2015;2(1):37-56 Review

Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

INTRODUCTION

Tumour is composed of a heterogeneous group

of cells with different morphologies and

behaviour. Research in cancer biology

indicates that several cancers are supported by

a small subset of cells with stem cell like

properties and are termed as cancer stem cells

(CSCs) or tumour initiating cells (TIC).

Evidences of CSCs involved in resistance to

conventional therapies, leading to metastasis

and tumour recurrence is abundant (Beck and

Blanpain, 2013; Chandler and Lagasse, 2010;

Prince and Ailles, 2008).

As early as 1937, Furth and colleagues

demonstrated that a single cell was able to

produce a haematopoietic malignancy on

implantation in mice (Furth et al., 1937). This

suggested that certain cells within a tumour

may have the ability to give rise to tumour

growth (Furth et al., 1937). Later, in 1994,

John Dick's group identified human acute

myeloid leukaemia-initiating cells using + -CD34 CD38 markers and showed that these

cells initiated tumour (Lapidot et al., 1994). In

1997, Bonnet and Dick showed for the first + -time that the CD34 CD38 population of cells

had the self-renewal property. The authors

performed limiting dilution assay to show that + -low numbers of CD34 CD38 cells were able

to form tumours in NOD/SCID mice, identical

to donors; whereas considerably higher

Key words: Cancer stem cells (CSCs); EMT, Epithelial to mesenchymal transition; Lineage tracing; β-catenin; NICD, Notch intracellular domain.*Corresponding Author: Sanjeev K. Waghmare, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India.Email: [email protected]

Developmental Signalling in Maintenance and Regulation

of Cancer Stem Cells

Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar,

Navi Mumbai, India

Sweta Dash, Raghava Reddy Sunkara and Sanjeev K. Waghmare*

Tissue stem cells self-renew throughout the life of an organism thereby maintaining tissue homeostasis and

prevent cancer. The major signalling pathways such as Wnt, Notch and Sonic hedgehog control the stem

cell regulation and their deregulation leads to cancer. Recent evidences showed that there exists a subset of

cells within tumour termed as cancer stem cells (CSCs). These CSCs escape the conventional chemo-

radiotherapy and further lead to tumour relapse followed by metastasis. This review focuses on the

developmental signalling pathways that are involved in the regulation and maintenance of normal stem cells

and CSCs. Understanding the molecular mechanism may be useful to specifically target the CSCs while

sparing the normal stem cells to reduce tumorigenecity.

Biomed Res J 2015;2(1):37-56

Review

Page 2: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

+ +numbers of non-CSCs (CD34 CD38 ) were

unable to form tumours (Bonnet and Dick,

1997). These cells were coined as cancer stem

cells. In 2003, Michael Clarke's group

reported the first isolation of CSCs from breast

tumour (Al Hajj et al., 2003). Subsequently,

the presence of CSCs in other solid tumours

like melanomas, hepatocellular carcinoma,

glioblastoma, pancreatic cancer, colorectal

cancer and head and neck cancer have been

identified (Keshet et al., 2008; Li et al., 2007;

Ma et al., 2007; Prince et al., 2007; Ricci-

Vitiani et al., 2007; Singh et al., 2004). The

CSC markers from various cancers are listed in

the Table 1. The characterisation of CSCs uses

various assays that include: sphere-forming

assay, serial transplantation assay in NOD/

SCID mice and in vivo lineage tracing. Serial

transplantation assay, is considered as 'gold

standard' assay, and measures self-renewal as

well as the tumorigenic property of CSCs in

vivo (Al Hajj et al., 2003; Beck and Blanpain,

2013; Bonnet, 1997; Prince et al., 2007).

Recently the strongest evidence for existence

of CSCs has come from the lineage tracing

experiments in mice model for various cancers

such as glioblastoma, skin and colon cancers.

The assay showed that the individual

fluorescent tagged cells have the capability to

give rise to a tumour (Chen et al., 2012;

Driessens et al., 2012; Schepers et al., 2012).

Although many different markers for

CSCs have been identified in tumours of

different tissues, cells isolated by using these

markers are not a pure CSC population. Hence,

one of the major challenges is the isolation of a

pure population of CSCs. Recent study on

quantitative proliferation dynamics of hair

follicle stem cells showed the isolation of stem

cells based on their cell division. This suggests

that it may be possible to isolate pure stem cell

population (Waghmare and Tumbar, 2013;

Waghmare et al., 2008). Another challenge is

to understand how these CSC populations are

regulated and maintained. Therefore, it is

important to study the various signalling

pathways that are crucial for survival of CSC

population.

Biomed Res J 2015;2(1):37-56

38 Developmental signalling in cancer cells

Table 1: Cancer stem cell markers in various cancers

Cancer Cancer stem cell markers

Leukaemia CD34+CD38

- (Bonnet, 1997)

Breast Cancer CD44+CD24- (Hajj et al., 2003); ALDH1+ (Ginesteir et al., 2007); CD133+ (Wright et al., 2008)

Head and Neck

Cancer

CD44+ Lin

- (Prince et al., 2007); A1DH1+ (Clay et al., 2010; Krishnamurthy et al., 2010); CD133

+

(Zhang et al., 2010); CD10+ (Fukusumi et al., 2014); CD98+ (Matens de Kemp et al., 2013)

Pancreatic Cancer CD44+CD24-ESA+ (Li et al., 2007); c-Met (Li et al., 2011)

Liver Cancer CD133+ (Ma et al., 2007); CD90+ (Yang et al., 2008); CD13+ (Haraguchi et al., 2010); OV6+ (Yang et al.,

2008)

Glioblastoma CD133+ (Singh et al., 2004); SSEA1+ (Son et al., 2009), MET (De Bacco et al., 2009)

Melanoma ABCB5+ (Keshet et al., 2008)

Colorectal Cancer CD133+ (Ricci-Vitiani et al., 2007); CD166+ (Dalerba et al., 2007; Vermeulen et al., 2008); Lgr5+ (Barker

et al., 2007; Vermeulen et al., 2008), CD44+ (Haraguchi et al., 2008), CD44v6+ (Todaro et al., 2014)

Page 3: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Embryonic developmental process and

cancer stem cells

Development of an organism is regulated at

the molecular level by various signalling

pathways, and deregulation in these molecular

mechanisms leads to cancer formation. Recent

studies have shown various similarities

between cancer and development. During the

normal developmental process, undifferentia-

ted embryonic stem cells further differentiate

and give rise to the differentiated tissues of an

organism. Similarly in cancer, undifferentia-

ted CSCs are involved in tumour progression

that leads to metastasis (Bellacosa, 2013).

The embryonic stem cells have a core

transcriptional network comprising of

transcription factors like Oct4, Sox2 and

Nanog that contribute to self-renewal and

pluripotency (Boyer et al., 2005). Similarly,

lung CSCs showed elevated levels of Oct4 and

Nanog transcription factors (Chiou et al.,

2010). In head and neck cancer, CD44 variant

CD44v3 was shown to interact with Oct4-

Sox2-Nanog leading to CSC like properties

such as self-renewal and cisplatin resistance

(Bourguignon et al., 2012). Recently, it was

shown that the lineage ablation of Sox2-

expressing cells in both benign and malignant

skin squamous cell carcinomas resulted in

tumour regression indicating an important role

of Sox2 in tumour initiation and CSC

functions. Moreover, chromatin immuno-

precipitation analysis identified Sox2 target

genes involved in controlling tumour stemness

(Boumahdi et al., 2014).

Another important phenomena common

to both the CSCs as wells as the embryonic

stem cells is the occurrence of epithelial to

mesenchymal transition (EMT). During EMT,

the cells lose their polarity and acquire

migration capabilities that results in loss of

epithelial marker E-cadherin and simulta-

neous increase in mesenchymal marker N-

cadherin. During embryogenesis, EMT is

associated with gastrulation required for the

formation of the three germ layers. In cancer,

EMT leads to invasion, metastasis and cancer

stem cell-like phenotype (Kalluri, 2009;

Singh, 2010). A recent study showed that

Twist1, an EMT promoter protein, is expressed

during early stages of tumorigenesis and is

required for the initiation of skin tumours

(Beck et al., 2015).

All these indicate that regulation of

embryonic stem cells and CSCs share similar

mechanisms. Therefore, it suggests that

deregulation of various developmental

pathways are involved in cancer formation and

CSC regulation and maintenance. Hence,

studying the developmental signalling

pathways will shed light on the regulation of

CSCs.

Developmental signalling pathways and

CSCs

The various pathways which are deregulated

in cancer include Wnt, Notch, Hedgehog,

EGFR, PI3K, NFκB, etc. Among these, three

Dash et al. 39

Biomed Res J 2015;2(1):37-56

Page 4: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

well-known pathways such as Wnt, Notch and

Hedgehog play an important role in the

development and normal homeostasis.

Conversely, deregulation of these pathways is

shown in CSC regulation and maintenance

(Ailles, 2012; Purow, 2012).

Wnt Pathway

Wnt pathway is evolutionarily conserved and

is involved in various organisms. It was first

discovered in Drosophila, when a mutation in

wingless (wg) gene led to a distinct phenotype

including absence of wings and halters. Later,

Nusse's group showed that the insertion of

Mouse Mammary Tumour Virus (MMTV) in

mice led to mammary tumour by proviral

activation of the int oncogene. The int

oncogene was later demonstrated as the mouse

homologue of the Drosophila wg gene. From

these two studies, a new nomenclature Wnt

(combination of wg and int) was obtained

(Nusse et al., 1984; Rijswijk et al., 1987;

Sharma, 2013).

There are 19 highly conserved Wnt

ligands discovered till date. These ligands are

secreted hydrophobic glycoproteins found to

be associated with cell membranes and extra-

cellular matrix. In Wnt producing cells, the

endoplasmic reticulum produces Wnt ligands,

lipid modified by porcupine (Mikels, 2006;

Willert et al., 2003). Wnt ligands can act

through two general categories of pathways:

canonical and non-canonical. The canonical

pathway is β-catenin dependent, while the 2+non-canonical pathways include Wnt/Ca and

Wnt/JNK pathways. In the canonical pathway

shown in Fig. 1, Wnt ligands bind to the

conserved cysteine rich domain (CRD) of the

frizzled receptors (Fz) which in turn forms co-

receptors complexes with low-density

lipoprotein like receptors (Lrp5/6). Further,

this interaction recruits the Dishevelled (Dsh)

Figure 1: Wnt Pathway. A) In the absence of the Wnt ligand, β-catenin is phosphorylated by destruction complex (APC,

CK1α, GSK3 and Axin) and is subjected to proteasomal degradation resulting in no transcription of the Wnt target

genes.B) In the presence of the Wnt ligand, the destruction complex is disrupted and thereby β-catenin enters the

nucleus and brings about the transcription of Wnt target genes. APC: Adenomatous Polyposis Coli; CK1α: Casein kinase

1α; GSK3: Glycogen synthase kinase 3; TCF: T cell factor; LEF: Lymphoid enhancing factor; Dsh: Dishevelled; LRP:

Low-density lipoprotein like receptors.

40 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 5: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

protein to the cytoplasmic tail of Fz receptor

and brings about inhibition of destruction

complex surrounding β-catenin. The

components of the destruction complex

comprise of scaffold protein Axin, Glycogen

synthase kinase 3β (GSK3β), Casein kinase 1α

(CK1α) and adenomatous polyposis coli

(APC). In the absence of the Wnt ligands, the

destruction complex hyper-phosphorylates β-

catenin and targets it for proteasomal

degradation by ubiquitination. The binding of

Wnt ligand to Lrp5/6 causes phosphorylation

of the cytoplasmic tail of Lrp6, which in turn

recruits Axin to the receptor complex that

disrupts the destruction complex and stabilises

β-catenin. The stable β-catenin translocates to

the nucleus and binds to the lymphoid

enhancing factor/T-cell factor (LEF/TCF)

thereby transcriptionally activating the

different target genes involved in cell fate

determination during embryonic development

and tissue homeostasis (Mikels, 2006; Willert

et al., 2003).

Wnt signalling in normal development and

cancer

Wnt pathway is involved in different biological

processes such as embryonic development,

self-renewal, proliferation, morphogenesis,

etc. Wnt3a and Wnt1 knock out in mice led to

deficiencies in neural crest derivatives and

neural tube formation during the development

(McMahon et al., 1990; Yoshikawa et al.,

1997). Wnt3 knock out in mice led to early

gastrulation defect and perturbations in the

establishment of apical ectodermal ridge

during development (Liu et al., 1999). Further,

absence of Wnt4 ligand led to defects in female

development, while Wnt7a deletion led to

female infertility in mice (Jeays-Ward et al.,

2004; Parr et al., 1998). Axin1 knockout in

mice led to neuro-ectodermal and cardiac

abnormalities (Zeng et al., 1997). Wnt

signalling was shown to be crucial in hair

follicle development as targeted deletion of β-

catenin in the epidermis led to failure in

placode morphogenesis (Huelsken et al.,

2000). Absence of Lef1 led to defects in the

pro-B-cell proliferation and abnormalities in

several organs like teeth, mammary glands,

whiskers and hair (Reya et al., 2000;

VanGenderen et al., 1994); while the knockout

of Tcf1 led to thymocyte proliferation and

differentiation defects (Schilham et al., 1998).

Using the Wnt reporter, Axin2-LacZ, Wnt

responsive cells were localised to the sub

ventricular zone (SVZ) of the developing brain

and basal layer of the mammary ducts, which

are the stem cells niches. Furthermore, these

Wnt responsive cells showed high sphere

forming ability and were able to differentiate.

Hence, the Wnt pathway plays an important

role in normal development and tissue

homeostasis (Logan, 2004; VanAmerongen et

al., 2009).

There are strong evidences showing

involvement of Wnt pathway in regulation of

various cancers. Frequent somatic mutations

Biomed Res J 2015;2(1):37-56

Dash et al. 41

Page 6: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

in β-catenin were observed in both mice and

human hepatocarcinomas (Coste et al., 1998),

prostate cancers and colon cancers (Voeller et

al., 1998). During intestinal adenoma

initiation, the first step was APC inactivation

followed by β-catenin stabilization, while

progression from adenoma to carcinoma

required the synergistic action of k-ras

activation and β-catenin nuclear localization

(Phelps et al., 2009). β-catenin was shown to

be essential for retaining tumorigenecity of

MDA-MB-231 breast cancer cell lines both in

vivo and in vitro. Further, β-catenin

knockdown cells implanted into mice showed

decrease in the tumour size. In addition, an in

vitro study in breast cancer cell lines showed

reduction in aldehyde dehydrogenase 1

(ALDH1) positive cells (Xu et al., 2015).

Wnt3a expression was associated with EMT

and promoted colon cancer progression (Qi et

al., 2014). Moreover, deletion of Axin1 was

reported in sporadic medulloblastomas and

hepatocellular carcinomas (Dahmen et al.,

2001). Increased expression of Dsh protein in

non-small cell lung carcinoma and meso-

thelioma have been reported (Uematsu et al.,

2003).

Wnt signalling in normal and cancer stem

cell regulation and maintenance

Wnt signalling is important in adult stem

regulation and has been shown to be involved

in stem cell proliferation, self-renewal and

maintenance. In hemato-poietic stem cells

(HSC), overexpression of β-catenin increases

the stem cell pool size suggesting that Wnt

pathway is critical to maintain the

hematopoietic stem cell homeostasis (Reya et

al., 2003). In mice hair follicle stem cells, live

cell imaging showed that β-catenin activation

in hair follicle stem cells was involved in hair

follicle tissue growth (Deschene et al., 2014).

Further, Wnt target gene Lgr5, a G-protein

coupled receptor was identified as an intestinal

stem cell marker indicating an important role

of Wnt pathway in the regulation of intestinal

stem cells (Ailles, 2012, Valkenburg, 2011).

The deletion of Tcf4, a Wnt downstream gene

showed loss of stem cell activity and reduced

proliferation of the intestinal epithelium

(Korinek et al., 1998). In addition, Lgr5 was

identified as a marker of hair follicle stem cells

(Jaks et al., 2008) with multipotent properties.

Moreover, Wnt inhibitor SFRP1 was shown to

play an important role in hematopoietic stem

cell maintenance through extrinsic regulation

(Renstrom et al., 2009). Over-expression of

Sfrp1 led to enhanced mesenchymal stem cell

function in angiogenesis (Dufourcq et al.,

2008). Besides, Sfrp1 was over-expressed in

hair follicle stem cells as compared to the non-

stem cells (Tumbar et al., 2004; Zhang et al.,

2009). Recently, it was shown that Sfrp1 gene

is critical for maintaining proper mammary

gland development wherein loss of Sfrp1

promotes mammosphere formation; however

the role in mammary stem cells needs further

investigation (Gauger et al., 2012).

42 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 7: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

In cancer, various reports have shown that

deregulation of Wnt pathway is crucial for the

CSC regulation. Human head and neck CSCs

treated with Wnt antagonist, secreted frizzled-

related protein 4 (sFRP4), the CSCs showed

reduction in the sphere-forming capacity and

decrease in the stemness markers like CD44

and ALDH1 (Warrier et al., 2014). In another

report, β-catenin was shown to be required for

maintenance of cutaneous CSCs since deletion

of β-catenin led to reduction in the CSCs and

tumour regression (Malanchi et al., 2008).

CSCs isolated from mammary tumours of

radiation treated p53-null mice showed altered

DNA repair in response to radiation as well as

β-catenin activation (Zhang et al., 2010). In

prostate cancer, Wnt signalling induced

tumour initiation, EMT and metastasis.

Additionally, in prostate cancer cell lines and

primary cultures, Wnt3a treatment increased

the self-renewal capacity of putative prostate

CSCs, emphasizing that Wnt signalling plays

an important role in prostate cancer (Barker,

2006; Valkenburg, 2011; Verras et al., 2004).

Moreover, the inactivation of APC in Lgr5-

positive stem cells at the intestinal crypts led to

transformation within days; while inactivation

of APC in progenitors or differentiated cells

did not lead to tumour formation even after 30

weeks (Barker et al., 2009). In addition, the

deletion of CD44, a CSC marker and a Wnt

target gene in mice having heterogeneous APC Min/+

mutation (APC ), attenuates intestinal

tumorigenesis (Zeilstra et al., 2008).

Notch Pathway

Notch gene was first discovered in Drosophila

by Morgan and Bridges where they showed

that a mutation led to wings notching and

hence the name “Notch” was coined (Morgan

and Bridges, 1916; Mohr, 1919; Poulson,

1940). There are four Notch genes, three

Delta-like and two Jagged genes in mammals,

that are translated into different Notch ligands,

Delta and Jagged. Recently, it was shown that

for cell fate determination during

development, complex of Notch receptor-

Delta-Jagged acts in concert (Fiuza, 2004;

Boaretoa et al., 2015).

Since the Notch ligands such as Delta and

Jagged proteins, as well as Notch receptors are

transmembrane proteins, cell-cell contact is

important for the signalling cascade. The

Notch receptors contain an extracellular

subunit, having multiple EGF-like repeats,

and a transmembrane subunit (Wharton et al.,

1985). When the Notch ligand binds to its

receptor, the extracellular domain of the Notch

receptor is dissociated from the trans-

membrane domain and the S2 cleavage site is

exposed (Fig. 2). This site is cleaved by

ADAM (a disintegrin and metalloprotease)

generating an intermediate that is further

cleaved by γ-secretase to generate Notch

Intracellular Domain (NICD). NICD then

translocates to the nucleus where it binds to

ubiquitous transcription factor CSL (CBF-1,

Suppressor of Hairless, Lag-1). This complex

displaces a co-repressor complex containing

Biomed Res J 2015;2(1):37-56

Dash et al. 43

Page 8: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

SKIP, SHARP and histone deacetylases.

Further, it recruits a co-activator complex

containing a MAML (Mastermind-like

protein), p300 and other chromatin modifying

enzymes, thereby bringing about transcription

of different Notch target genes (Ailles, 2012;

Andersson, 2011; Fiuza, 2004).

Notch signalling in normal development

and cancer

Notch pathway is also evolutionarily

conserved and is important in cell to cell

communication that regulates cell fate

determination during development, cell

proliferation, differentiation and apoptosis.

The loss of Notch function in vertebrates is

associated with disruption of neurogenesis,

somite formation, angiogenesis, and lymphoid

development. In Drosophila, Notch is shown

to control the fate of various cell types in the

eye. In vertebrates, Notch is involved in the

establishment of the central and peripheral

nervous systems, spermatogenesis, oogenesis,

myogenesis and imaginal disc development

(Artavanis-Tsakonas et al., 1999). In normal

mammary development, Notch pathway

activation is required for regulation of cell

fate, proliferation and stem cell self-renewal.

The Notch pathway is also shown to be

important for tip-cell formation during

mammalian astrocyte differentiation and

angiogenesis. In vertebrates, the Notch

pathway leads to patterning during inner ear

hair cell formation and insulin-secreting

pancreatic β cell production (Ailles, 2012;

Fortini, 2009).

Notch signalling has been shown to be

involved in various cancers. For instance,

Notch1 regulates breast cancer cells by

inducing Slug expression (Shao et al., 2015).

Notch4 promotes growth of gastric cancer

cells through activation of Wnt1, β-catenin

Figure 2: Notch Pathway. A) In the absence of the Notch ligands (Delta and Jagged), the S2 cleavage site remains

hidden and inaccessible to ADAM. Hence, the NICD is not formed, with consequent no transcription of the Notch target

genes. B) In the presence of the Notch ligands, the conformational change in the intracellular subunit of the Notch

receptor takes place exposing the S2 cleavage site, thus leading to the formation of NICD. Further, NICD translocates the

nucleus and brings about transcription of Notch target genes. NICD: Notch intracellular domain; ADAM: A disintegrin and

metalloprotease; MAML: Mastermind-like protein; SKIP: Ski-interacting protein; SHARP: SMRT associated protein.

44 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 9: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

(Quian et al., 2015) and the downstream target

such as c-myc and cyclin-D1. In mice, Notch1

activation combined with p53 loss showed

synergistic effect in the formation of

Osteogenic sarcoma (Tao et al., 2014).

Notch signalling in normal and cancer stem

cell regulation and maintenance

Notch has been shown to be involved in self-

renewal, proliferation and differentiation of

adult stem cells in various tissues. In mice

mammary stem cells, the knockdown of Cbf-1,

a canonical Notch effector, showed increased

stem cell activity in vivo suggesting a role in

controlling mammary stem cells. (Bouras et

al., 2008). Notch directly targets the crypt base

columnar cells that maintain stem cell activity

in mice (VanDussen et al., 2012). The Notch

activation maintains the slow cycling property

of neural stem cells; however, blocking Notch

resulted in increased number of stem cell

divisions followed by depletion of the stem

cell pool (Chapouton et al., 2010).

Furthermore, constitutive activation of Notch

signalling promotes self-renewal in muscle

stem cells through upregulation of Pax7 (Wen

et al., 2012). Mice with satellite cell specific

deletion of RBP-Jj (recombining binding

protein-Jj), a nuclear factor required for Notch

signalling, showed depletion of the stem cell

pool and their muscles lacked ability to

regenerate in response to injury (Bjornson et

al., 2012). Notch 3 has been shown to be

expressed in stem cells and disruption led to

defective stem cells proliferation (Kitamoto

and Hanaoka, 2010).

Notch signalling plays an important role in

a number of hematopoietic and solid tumours,

but the strongest evidences for its role in CSC

regulation has been shown in breast cancer,

embryonal brain tumours and gliomas (Fan et

al., 2006; Pannuti et al., 2010). In various

human breast cancer cell lines and primary

patient tissues, a significant decrease in

mammosphere formation after Notch

inhibition has been demonstrated (Abel et al.,

2014). Further, studies on the human

mammary mammospheres have shown a

feedback loop between Her2/Neu and Notch,

as well as promotion of a hypoxia resistant

phenotype (Pannuti et al., 2010). In brain

tumours, blockade of Notch led to a 5-fold +reduction in the CD133 cell fraction and total

depletion of the side population cells.

Additionally, differentiated cell growth was

observed after Notch inhibition, but lacked

formation of tumour xenografts efficiently,

indicating that the CSCs required for tumour

propagation were absent (Fan et al., 2006). A

recent study on primary human pancreatic

xenografts showed upregulation of the notch

pathway components in pancreatic CSCs.

Additionally, inhibition of notch pathway

reduced CSC percentage and tumour-sphere

formation significantly (Abel et al., 2014).

Biomed Res J 2015;2(1):37-56

Dash et al. 45

Page 10: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Sonic-hedgehog Pathway

Hedgehog pathway is delineated in

Drosophila and determines the anterior-

posterior orientation of developing structures

(Nusslein-Volhard and Wieschaus, 1980).

Similar to Wnt and Notch, the key components

of the Hedgehog pathway are evolutionarily

conserved, although differences are observed

in the mammalian and Drosophila Hedgehog

signalling. While Drosophila has only one

hedgehog gene, three homologues have been

identified in vertebrates namely, Sonic (Shh),

Desert (Dhh), and Indian hedgehog (Ihh). The

Sonic Hedgehog pathway is extensively

investigated in the vertebrate system (Chen et

al., 2005; Varjosalo et al., 2006). The other

components of the Hedgehog pathway include

patched (Ptc) and smoothened (Smo),

constituting a 12-pass transmembrane

glycoprotein and a 7-pass transmembrane

protein, respectively (Varjosalo, 2008;

Wicking et al., 1999).

In the Sonic-hedgehog pathway

elaborated in Fig. 3, absence of the hedgehog

ligand, Smoothened (Smo) is inhibited by

being bound to Patched (Ptc). When the

hedgehog ligand binds to Ptc, inhibition of

Smo is released which acts on protein complex

comprising of fused (Fu), suppressor of fused

(Sufu) and cos-2-costa-2 (Wicking et al.,

1999; Merchant, 2010). These proteins are

generally bound with Gli thereby inhibiting its

action. Once the complex is disrupted, Gli

translocates to the nucleus and brings about

transcription of different downstream targets

(Sasaki et al., 1999; Ruiz, 2007; Stecca, 2010).

Sonic-hedgehog signalling in normal

development and cancer

In vertebrates, the Sonic Hedgehog (Shh), is

expressed widely throughout the developing

central nervous system (CNS), limb, gut, teeth

and hair-follicle. Dhh is involved in

development of the germline, while the Ihh is

Figure 3: Hedgehog Pathway. A) In the absence of Hedgehog ligands (Indian, Sonic and Dessert), the Patched

receptor (ptc) exerts inhibitory action on the Smoothened receptor (smo). The Gli complex (Gli1 and Gli2) remains in the

cytoplasm followed by no transcription of Hedgehog target genes. B) In the presence of Hedgehog ligands, the inhibitory

action of Patched (ptc) on Smoothened (smo) is released, and hence Gli complex translocates to the nucleus and brings

about transcription of Hedgehog target genes.Fu: Fused; SuFu: Suppressor of Fused; Cos 2: cos-2costa-2.

46 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 11: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

involved in development of the skeletal system

(Bitgood et al., 1996; Wicking et al., 1999).

Shh also plays a role in neural stem cells,

determining the neuronal cell fate (Merkle et

al., 2007). It was demonstrated that during

repair of acute airway injury, the Hedgehog

pathway gets activated in the airway

epithelium (Watkins et al., 2003). Hedgehog

signalling components Ptc, Gli1, and Gli2

were over-expressed when mammary

progenitor cells grow as mammospheres (Liu

et al., 2006). These reports indicate that the

Hedgehog pathway plays a role in normal stem

cell regulation (Ailles, 2012).

Hedgehog signalling is involved in

various cancers. For example, Ptc1 mutation

was observed in patients with medullo-

blastoma and rhabdomyosarcoma (Hahn et al.,

1996; Johnson et al., 1996; Pietsch et al.,

1997). Sufu as well as Smo mutations were

observed in medulloblastoma (Xie et al., 1998;

Taylor et al., 2002); Gli1 and Gli3 mutations

were seen in pancreatic adenocarcinoma; and

Gli1 gene amplification was seen in

glioblastoma (Clement et al., 2007; Jones et

al., 2008). Further, Kern et al showed that Gli

and PI3K/AKT/mTOR signalling act

synergistically to initiate and maintain chronic

lymphocytic leukemia (Kern et al., 2015).

Another report showed that Sonic hedgehog

ligand over-expression led to increased

number and size of intestinal adenomas in

APC (HET) mice, while loss of Indian

Hedgehog almost completely blocks intestinal

adenoma development (Buller et al., 2015).

Sonic-hedgehog signalling in normal and

cancer stem cell regulation and

maintenance

Hedgehog signalling is involved in stem cell

regulation of various tissues. Shh regulates

self-renewal of neural stem cells (Palma et al.,

2005). The components of Hedgehog pathway

such as Ptc, Gli1 and Gli2 are expressed in the

mammary stem cells and down regulated

during differentiation (Liu et al., 2006).

Hedgehog is involved in controlling neural

stem cells through the p53-independent

regulation of Nanog (Po et al., 2005).

In colon carcinoma, Hedgehog signalling

is activated in CSCs with higher expression of

Gli1 and Gli2. In non-small cell lung cancer,

the malignant phenotype of the tumours is

maintained by ligand-dependent Hedgehog

pathway activation (Watkins et al., 2003).

Furthermore, Bmi1, which is a downstream

target of the Hedgehog pathway was activated

in breast CSCs and is also shown to regulate

normal and leukemic stem cells (Liu et al.,

2006; Takebe, 2011).

CONCLUSION

Tumour maintenance and progression is

regulated by a subset of cells that are known as

cancer stem cells (CSCs). Recently, due to

increase in evidences on the existence of

Biomed Res J 2015;2(1):37-56

Dash et al. 47

Page 12: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

CSCs, they have gained more attention but

how these CSCs escape the chemo-

radiotherapy is still unknown. Moreover, how

the CSCs are maintained in the tumour micro-

environment remains elusive. Several reports

showed signalling pathways such as Wnt,

Notch and Sonic-hedgehog are deregulated in

cancer, and also involved in the CSC

regulation and maintenance. In addition,

evidence of cross-talk between the signalling

pathways exists. Therefore, understanding

these signalling pathways at the molecular

level will be of utmost importance The study .

will enable counteracting the issue of

signalling cross-talk, and perhaps, multi

targeted drugs approach can be fruitful.

Hence, further detailed research on

deregulation of the developmental pathways

in CSCs needs to be investigated. Eventually,

elucidation of the signalling mechanisms will

enable to specifically target CSCs without

affecting the normal cells.

ACKNOWLEDGEMENTS

The authors acknowledge Mr. Rahul Sarate

and Mr. Gopal Chovatiya for their

suggestions.

CONFLICT OF INTEREST

The authors claim no conflict of interest.

Biomed Res J 2015;2(1):37-56

REFERENCES

Abel E, Kim E, Wu J, Hynes M, Proctor E, Brendar

F, Simeone D. The Notch Pathway Is

Important in Maintaining the Cancer Stem

Cell Population in Pancreatic Cancer. Plos

One 2014;9: e91983.

Ailles L, Karamboulas C. Developmental signaling

pathways in CSCs of solid tumor. Biochim

Biophys Acta 2012; 1830:2481–2495.

Al-Hajj M, Wicha M, Morrison S, Clarke M.

Prospective identification of tumorigenic

breast cancer cells. Proc Natl Acad Sci 2003;

100:3983–3988.

Andersson EM, Sandberg R, Lendahl U. Notch

signaling: simplicity in design, versatility in

function. Development 2011;138:3593–3612.

Artavanis-Tsakonas S, Rand M, Lake R. Notch

Signaling: Cell Fate Control and Signal

Integration in Development. Science

1999;284:770–776.

Barker N, Clevers H. Mining the Wnt pathway for

cancer therapeutics. Nat Rev 2006;5:

997–1015.

Barker N, Ridgway RA, van Es JH, van de

Wetering M, Begthel H, van den Born M, et al.

Crypt stem cells as the cells-of-origin of

intestinal cancer. Nature 2009;457:608–611.

Barker N, van Es JH, Kuipers J, Kujala P, van den

Born M, Cozijnsen M, et al. Identification of

stem cells in small intestine and colon by

marker gene Lgr5. Nature 2007;449:

1003–1007.

Beck B, Blanpain C. Unravelling cancer stem cell

potential. Nat Rev 2013;13:727–738.

Beck B, Lapouge G, Rorive S, Drogat B,

Desaedelaere K, Delafaille S, et al. Different

levels of twist1 regulate skin tumor initiation,

stemness, and progression. Cell Stem Cell

2015;16:67–79.

48 Developmental signalling in cancer cells

Page 13: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Bellacosa A. Developmental disease and cancer:

biological and clinical overlaps. Am J Med

Genet A 2013;11:2788–2796.

Bitgood MJ, Shen L, McMahon AP. Sertoli cell

signaling by Desert hedgehog regulates the

male germline. Curr. Biol 1996;6:298–304.

Bjornson CR, Cheung TH, Liu L, Tripathi PV,

Steeper KM, Rando TA.Notch signaling is

necessary to maintain quiescence in adult

muscle stem cells. Stem Cells 2012;30:

232–242.

Boaretoa M, Jolly MK, Lu M, Onuchica JN,

Clementia C, Ben-Jacob E. Jagged–Delta

asymmetry in Notch signaling can give rise to

a Sender/Receiver hybrid phenotype. Proc

Natl Acad Sci USA 2015,112:E402–409.

Bonnet D , Dick J. Human acute myeloid leukemia

is organized as a hierarchy that originates from

a primitive hematopoietic cell. Nat Med 1997;

3:730–737

Boumahdi S, Driessens G, Lapouge G, Rorive S,

Nassar D, Le Mercier M, et al. SOX2 controls

tumour initiation and cancer stem-cell

functions in squamous-cell carcinoma. Nature

2014;511:246–250.

Bouras T, Pal B, Vaillant F, Harburg G, Asselin-

Labat ML, ASR, et al. Notch signaling

regulates mammary stem cell function and

luminal cell-fate commitment. Cell Stem Cell

2008;3:429–441.

Bourguignon L, Wong G, Earle C, Chen L.

Hyaluronan-CD44v3 interaction with Oct4-

Sox2-Nanog promotes miR-302 expression

leading to self-renewal, clonal formation, and

cisplatin resistance in CSCs from head and

neck squamous cell carcinoma. J Bio Chem

2012;287:32800–32824.

Boyer L, Lee T, Cole M, Johnstone S, Levine S,

Zucker J, et al. Core Transcriptional

Regulatory Circuitry in Human Embryonic

Stem Cells. Cell 2005;6:947–956.

Büller NV, Rosekrans SL, Metcalfe C, Heijmans J,

van Dop WA, Fessler E, et al. Stromal Indian

hedgehog signaling is required for intestinal

adenoma formation in mice. Gastroenterology

2015;148:170–180.

Chandler J, Lagasse E. Cancerous stem cells:

deviant stem cells with cancer-causing

misbehavior. Stem Cell Res Ther 2010;1:

e1–e9.

Chapouton P, Skupien P, Hesl B, Coolen M, Moore

JC, Madelaine R, et al. Notch activity levels

control the balance between quiescence and

recruitment of adult neural stem cells. J

Neurosci 2010;30:7961–7974.

Chen MH, Gao N, Kawakami T, Chuang PT. Mice

deficient in the fused homolog do not exhibit

phenotypes indicative of perturbed hedgehog

signaling during embryonic development. Mol

Cell Biol 2005;25:7042–7053.

Chen J, McKay R, Yanjio L, Burns D, Parada L. A

restricted cell population propagates

glioblastoma growth after chemotherapy.

Nature 2012;488:522–526.

Chiou S, Wang M, Chou Y, Chen C, Hong C, Hsieh

W, et al. Coexpression of Oct4 and Nanog

enhances malignancy in lung adenocarcinoma

by inducing cancer stem cell–like properties

and epithelial–mesenchymal trans-

differentiation. Can Res 2010;70:1931–1940.

Clay M, Tabor M, Owen J, Carey O, Bradford C,

Wolf G, et al. Single-marker identification of

head and neck squamous cell carcinoma CSCs

with aldehyde dehydrogenase. Head Neck

Biomed Res J 2015;2(1):37-56

Dash et al. 49

Page 14: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

2010;32:1195–1201.

Clement V, Sanchez P, de Tribolet N, Radovanovic

I, Altaba A. HEDGEHOG-GLI1 signaling

regulates human glioma growth, cancer stem

cell self-renewal, and tumorigenicity. Curr

Biol 2007;17:165–172.

Coste A, Romagnolo B, Billuart P, Renard C,

Buendia M, Soubrane O, et al. Somatic

mutations of the β-catenin gene are frequent in

mouse and human hepatocellular carcinomas.

Proc Natl Acad Sci USA 1998;95:8847–8851.

Dahmen RP, Koch A, Denkhaus D, Tonn JC,

Sörensen N, Berthold F, et al. Deletions of

AXIN1, a component of the WNT/wingless

pathway, in sporadic medulloblastomas.

Cancer Res 2001;61:7039–7043.

Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho

RW, et al. Phenotypic characterization of

human colorectal CSCs. Proc Natl Acad Sci

USA 2007;104:10158–10163.

De Bacco F, Casanova E, Medico E, Pellegatta S,

Orzan F, Albano R, et al. The MET oncogene is

a functional marker of a glioblastoma stem cell

subtype.Cancer Res 2012;72:4537–4550.

Deschene ER, Myung P, Rompolas P, Zito G, Sun

TY, Taketo MM, et al. β-Catenin activation

regulates tissue growth non-cell

autonomously in the hair stem cell niche.

Science 2014;343:1353–1356.

Driessens G, Beck B, Blanpain C. Defining the

mode of tumor growth by clonal analysis.

Naure 2012;488:527–530.

Dufourcq P, Descamps B, Tojais NF, Leroux L,

Oses P, Daret D, et al. Secreted frizzled-related

protein-1 enhances mesenchymal stem cell

function in angiogenesis and contributes to

neovessel maturation. Stem Cells 2008;26:

2991–3001.

Fan X, Matsui W, Khaki L, Stearns D, Chun J, Li Y,

Eberhart C. Notch Pathway Inhibition

Depletes Stem-like Cells and Blocks

Engraftment in Embryonal Brain Tumors.

Cancer Res 2006;66:7445–7452.

Fiuza U, Arias A. Cell and molecular biology of

Notch. J Endocrinol 2007;194:459–474.

Fortini ME. Notch signaling: the core pathway and

its posttranslational regulation. Dev Cell 2009;

16:633–647.

Fukusumi T, Ishii H. CD10 as a novel marker of

therapeutic resistance and CSCs in head and

neck squamous cell carcinoma. Br J Can 2014;

3:506–514.

Furth J, Kahn MC, Breedis C. The transmission of

leukaemia of mice with a single cell. Am J

Cancer 1937;31:276–282.

Gauger KJ, Shimono A, Crisi GM, Schneider SS.

Loss of sfrp1 promotes ductal branching in the

murine mammary gland. BMC Dev Biol 2012;

12:25.

Ginestier C, Hur M, Charafe-Jauffret E, Monville

F, Dutcher J, Brown M, et al. ALDH1 is a

marker of normal and malignant human

mammary stem cells and a predictor of poor

clinical outcome. Cell Stem Cell 2007;1:

555–567.

Hahn H, Wicking C, Zaphiropoulous PG, Gailani

MR, Shanley S, Chidambaram A, et al.

Mutations of the human homolog of

Drosophila patched in the nevoid basal cell

carcinoma syndrome. Cell 1996;85:841–851.

Haraguchi N, Ishii H, Mimori K, Tanaka F,

Ohkuma M, Kim HM, et al. CD13 is a

therapeutic target in human liver CSCs. J Clin

Invest 2010;120:3326–3339.

50 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 15: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Haraguchi N, Ohkuma M, Sakashita H Matsuzaki

S, Tanaka F, Mimori K, et al. CD133+CD44+

population efficiently enriches colon cancer

initiating cells. Ann Surg Oncol 2008;15:

2927–2933.

Huelsken J, Vogel R, Brinkmann V, Erdmann B,

Birchmeier C, Birchmeier W. Requirement for

beta-catenin in anterior-posterior axis

formation in mice. J Cell Biol 2000;148:

567–578.

Jaks V, Barker N, Kasper M, van Es JH, Snippert

HJ, Clevers H, Toftgård R.Lgr5 marks cycling,

yet long-lived, hair follicle stem cells. Nat

Genet 2008;40:1291–1299.

Jeays-Ward K, Dandonneau M, Swain A.Wnt4 is

required for proper male as well as female

sexual development. Dev Biol 2004;276:

431–440.

Johnson RL, Rothman AL, Xie J, Goodrich LV,

Bare JW, Bonifas JM, et al. Human homolog

of patched, a candidate gene for the basal cell

nevus syndrome. Science 1996;272:

1668–1671.

Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ,

Angenendt P, et al. Core signaling pathways in

human pancreatic cancers revealed by global

genomic analyses. Science 2008;321:

1801–1806.

Kalluri R, Weinberg R. The basics of epithelial-

mesenchymal transition. J Clin Inv 2009;119:

1420–1428.

Kern D, Regl G, Hofbauer SW, Altenhofer P,

Achatz G, Dlugosz A, et al. Hedgehog/GLI

and PI3K signaling in the initiation and

maintenance of chronic lymphocytic

leukemia. Oncogene 2015; doi:

10.1038/onc.2014.450.

Keshet GI, Goldstein I, Itzhaki O, Cesarkas K,

Shenhav L, Yakirevitch A, et al. MDR1

expression identifies human melanoma stem

cells. Biochem Biophys Res Commun 2008;

368:930–936.

Kitamoto T, Hanaoka K. Notch3 null mutation in

mice causes muscle hyperplasia by repetitive

muscle regeneration. Stem Cells 2010;28:

2205–2216.

Krishnamurthy S, Warner K, Dong Z, Imai A, Nor

C, Ward B, Helman J. Endothelial cell-

initiated signaling promotes the survival and

self-renewal of CSCs. Cancer Res 2010;70:

9969–9978.

Korinek V, Barker N, Moerer P, van Donselaar E,

Huls G, Peters PJ, Clevers H. Depletion of

epithelial stem-cell compartments in the small

intestine of mice lacking Tcf-4. Nat Genet

1998;19:379–383.

Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang

T, Caceres Cortes J, et al. A cell initiating

human acute myeloid leukaemia after

transplantation into SCID mice. Nature 1994;

367:645–648.

Li C, Heidt D, Dalerba P, Burant C, Zhang L, Adsay

V, et al. Identification of pancreatic CSCs.

Cancer Res 2007;67:1030–1037.

Li C, Wu JJ, Hynes M, Dosch J, Sarkar B, Welling

TH, et al. c-Met is a marker of pancreatic CSCs

and therapeutic target. Gastroenterology 2011;

141:2218–2227.

Liu P, Wakamiya M, Shea MJ, Albrecht U,

Behringer RR, Bradley A. Requirement for

Wnt3 in vertebrate axis formation. Nat Genet

1999;22:361–365.

Liu S, Dontu G, Mantle ID, Patel S, Ahn NS,

Jackson KW, et al. Hedgehog signaling and

Biomed Res J 2015;2(1):37-56

Dash et al. 51

Page 16: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Bmi-1 regulate self-renewal of normal and

malignant human mammary stem cells.

Cancer Res 2006;66:6063–6071.

Logan C, Nusse R. Wnt signaling pathway in

development and disease. Annu Rev Cell Dev

Biol 2004;20:781–810.

Ma S, Chan KW, Hu L, Lee TK, Wo JY, Ng IO, et al.

Identification and characterization of

tumorigenic liver cancer stem/progenitor

cells. Gastroenterology 2007;132:2542–2556.

Malanchi I, Peinado H, Kassen D, Hussenet T,

Metzger D, Chambon P, et al. Cutaneous

cancer stem cell maintenance is dependent on

beta-catenin signalling. Nature 2008;452:

650–653.

Matens de Kemp S, Brink A. CD98 marks a

subpopulation of head and neck squamous cell

carcinoma cells with stem cell properties. Stem

Cell Res 2013;3:477–488.

McMahon AP, Bradley A. The Wnt-1 (int-1) proto-

oncogene is required for development of a

large region of the mouse brain. Cell

1990;62:1073–1085.

Merchant A, Matsui W (2010). Targeting

Hedgehog, a cancer stem cell pathway. Clin

Cancer Res 2010;16:3130–3140.

Merkle FT, Mirzadeh Z, Alvarez-Buylla A. Mosaic

organization of neural stem cells in the adult

brain. Science 2007;317:381–384.

Mikels A, R Nusse. Wnt as ligands: processing,

secretion and reception. Oncogene 2006;25:

7461–7468.

Mohr OL. Character changes caused by mutation

of an entire region of a chromosome in

Drosophila. Genetics 1919;4:275–282.

Morgan TH, Bridges CB. Sex-linked inheritance in

Drosophila. Publs Carnegie Instn 1916;237:

1–88.

Nusse R, van Ooyen A, Cox D, Fung YK, Varmus

H. Mode of proviral activation of a putative

mammary oncogene (int-1) on mouse

chromosome 15. Nature 1984;307:131–136.

Nusslein-Volhard C, Wieschaus E. Mutations

affecting segment number and polarity in

Drosophila. Nature 1980;287:795–801.

Palma V, Lim DA, Dahmane N, Sánchez P, Brionne

TC, Herzberg CD, et al. Sonic hedgehog

controls stem cell behavior in the postnatal and

adult brain. Development 2005;132:335–344.

Pannuti A, Foreman K, Rizzo P, Osipo C, Golde T,

Osborne B, Miele L. Targeting CSCs through

Notch Signaling. Clin Cancer Res 2010;16:

3141–3152.

Parr BA, McMahon AP. Sexually dimorphic

development of the mammalian reproductive

tract requires Wnt-7a. Nature 1998;395:

707–710.

Phelps RA, Chidester S, Dehghanizadeh S, Phelps

J, Sandoval IT, Rai K, et al. A two-step model

for colon adenoma initiation and progression

caused by APC loss. Cell 2009;137:623–634.

Pietsch T, Waha A, Koch A, Kraus J, Albrecht S,

Tonn J, et al. Medulloblastomas of the

desmoplastic variant carry mutations of the

human homologue of Drosophila patched.

Cancer Res 1997;57:2085–2088.

Prince M, Sivanandan R, Wolf GT, Kaplan MJ,

Kaczorouski A, Dalerba P, et al. Identification

of a subpopulation of cells with cancer stem

cell properties in head and neck squamous cell

carcinoma. Proc Natl Acad Sci USA 2007;104:

973–978.

Prince M, Allies L. CSCs in Head and Neck

Squamous Cell Cancer. J Clin Onc 2008;26:

52 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 17: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

2871–2875.

Po A, Ferretti E, Miele E, De Smaele E, Paganelli

A, Canettieri G, et al. Hedgehog controls

neural stem cells through p53-independent

regulation of Nanog. EMBO 2010;29:

2646–2658.

Poulson DF. The effects of certain X-chromosome

deficiencies on the embryonic development of

Drosophila melanogaster. J Exp Zool 1940;83:

271–325.

Purow B. Notch inhibition as a promising new

approach to cancer therapy. Adv Exp Med Biol

2012;727:305–319.

Qian C, Liu F, Ye B, Zhang X, Liang Y, Yao J.

Notch4 promotes gastric cancer growth

through activation of Wnt1/β-catenin

signaling. Mol Cell Biochem 2015;401:

165–174.

Qi L, Sun B, Liu Z, Cheng R, Li Y, Zhao X. Wnt3a

expression is associated with epithelial-

mesenchymal transition and promotes colon

cancer progression. J Exp Clin Cancer Res

2014;33:107.

Renstrom J, Istvanffy R, Gauthier K, Shimono A,

Mages J, Jardon-Alvarez A, et al. Secreted

Frizzled-Related Protein 1 extrinsically

regulates cycling activity and maintenance of

hematopoietic stem cells. Cell Stem Cell 2009;

5:157–167.

Reya T, O'Riordan M, Okamura R, Devaney E,

Willert K, Nusse R, Grosschedl R. Wnt

signaling regulates B lymphocyte

proliferation through a LEF-1 dependent

mechanism. Immunity 2000;13:15–24.

Reya T, Duncan AW, Ailles L, Domen J, Scherer

DC, Willert K, et al. A role for Wnt signalling

in self-renewal of haematopoietic stem cells.

Nature 2003;423:409–414.

Ricci-Vitiani L, Lombardi DG, Pilozzi E Biffoni

M, Todaro M, Peschle C, De Maria R.

Identification and expansion of human colon-

cancer-initiating cells. Nature 2007;445:

111–115.

Rijswijk F, Schuermann M, Wagenaar E, Parren P,

Weighel D, Nusse R. The Drosophila homolog

of the mouse mammary oncogene int-1 is

identical to the segment polarity gene

wingless. Cell 1987;50:649–657.

Sasaki H, Nishizaki Y, Hui C, Nakafuku M,

Kondoh H. Regulation of Gli2 and Gli3

activities by an amino-terminal repression

domain: implication of Gli2 and Gli3 as

primary mediators of Shh signaling.

Development 1999;126:3915–3924.

Schepers A, Snippert H, Stange D, Born M,

Wetering M, Clever H. Lineage tracing reveals

Lgr5+ stem cell activity in mouse intestinal

adenomas. Science 2012;337:730–735.

Schilham MW, Wilson A, Moerer P, Benaissa-

Trouw BJ, Cumano A, Clevers HC. Critical

involvement of Tcf-1 in expansion of

thymocytes. J Immunol 1998;161:3984–3991.

Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S,

et al. Notch1 signaling regulates the epithelial-

mesenchymal transition and invasion of breast

cancer in a Slug-dependent manner. Mol

Cancer 2015;14:28.

Sharma R. Wingless to Wnt: discovery of

conserved cell signalling gene family in the

animal kingdom. Curr Sci 2013;104:

1140–1141.

Shih I, Wang T. Notch signaling: γ-secretase

inhibitors, and cancer therapy. Cancer Res

2007;67:1879–1882.

Biomed Res J 2015;2(1):37-56

Dash et al. 53

Page 18: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Singh SK, Hawkins C, Clarke ID, Squire JA,

Bayani J, Hide T, et al. Identification of human

brain tumor initiating cells. Nature 2004;432:

396–401.

Singh A, Settleman J. EMT, CSCs and drug

resistance: an emerging axis of evil in the war

on cancer. Oncogene 2010;29:4741–4751.

Son MJ, Woolard K, Nam DH, Lee J, Fine HA.

SSEA-1 is an enrichment marker for tumor-

initiating cells in human glioblastoma. Cell

Stem Cell 2009;4:440–452.

Stecca B, Ruiz I, Altaba A. Context-dependent

regulation of the GLI code in cancer by

HEDGEHOG and Non-HEDGEHOG signals.

J Mol Cell Biol 2010;2:84–95.

Takebe N, Harris P, Warren R, Ivy S. Targeting

CSCs by inhibiting Wnt, Notch and Hedgehog

pathways. Nat Rev 2011;8:97–106.

Tao J, Jiang MM, Jiang L, Salvo JS, Zeng HC,

Dawson B, et al. Notch activation as a driver of

osteogenic sarcoma. Cancer Cell 2014;26:

390–401.

Taylor MD, Liu L, Raffel C, Hui CC, Mainprize

TG, Zhang X, et al. Mutations in SUFU

predispose to medulloblastoma. Nat Genet

2002;31:306–310.

Todaro M, Gaggianesi M, Catalano V, Benfante A,

Iovino F, Biffoni M, et al. CD44v6 is a marker

of constitutive and reprogrammed cancer stem

cells driving colon cancer metastasis. Cell

Stem Cell 2014;14:342–356.

Tumbar T, Guasch G, Greco V, Blanpain C, Lowry

WE, Rendl M, Fuchs E. Defining the epithelial

stem cell niche in skin. Science 2004;303:

359–363.

Uematsu K, He B, You L, Xu Z, McCormick Fa,

Jablons DM. Activation of the Wnt pathway in

non-small cell lung cancer: evidence of

dishevelled overexpression. Oncogene 2003;

22:7218–7221.

Uematsu K, Kanazawa S, You L, He B, Xu Z, Li K,

et al. Wnt pathway activation in

mesothelioma: evidence of Dishevelled

overexpression and transcriptional activity of

beta-catenin. Cancer Res 2003;63:4547–4551.

Valkenburg K, Graveel C, Zylstra-Diegel C, Zhong

Z, Williams B. Wnt/β-catenin signaling in

normal and CSCs. Cancers 2011;3:

2050–2079.

VanAmerongen R, Nusse R. Towards an integrated

view of Wnt signaling in development.

Development 2009;136:3205–3214.

VanDussen KL, Carulli AJ, Keeley TM, Patel SR,

Puthoff BJ, Magness ST, et al. Notch signaling

modulates proliferation and differentiation of

intestinal crypt base columnar stem cells.

Development 2012;139:488–497.

VanGenderen C, Okamura RM, Fariñas I, Quo RG,

Parslow TG, Bruhn L, Grosschedl R.

Development of several organs that require

inductive epithelial-mesenchymal interactions

is impaired in LEF-1-deficient mice. Genes

Dev 1994;8:2691–703.

Varjosalo M, Li SP, Taipale J. Divergence of

hedgehog signal transduction mechanism

between Drosophila and mammals. Dev Cell

2006;10:177–186.

Varjosalo M, Taipale J. Hedgehog: functions and

mechanisms. Gen Dev 2008;22:2454–2472.

Verras M, Brown J, Li X, Nusse R, Sun Z. Wnt3a

growth factor induces androgen receptor

mediated transcription and enhances cell

growth in human prostate cancer cells. Cancer

Res 2004;64:8860–8866.

54 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56

Page 19: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Vermeulen L, Todaro M, de Sousa Mello F, Sprick

MR, Kemper K, Perez Alea M, et al. Single-

cell cloning of colon CSCs reveals a multi-

lineage differentiation capacity. Proc Natl

Acad Sci USA 2008;105:13427–13432.

Voeller J, Truica C, Gelmann E. β-Catenin

Mutations in Human Prostate Cancer. Cancer

Res 1998;58:2520–2523.

Waghmare SK, Bansal R, Lee J, Zhang YV,

McDermitt DJ, Tumbar T.Quantitative

proliferation dynamics and random

chromosome segregation of hair follicle stem

cells. EMBO 2008;27:1309–1320.

Waghmare SK, Tumbar T. Adult hair follicle stem

cells do not retain the older DNA strands in

vivo during normal tissue homeostasis.

Chromosome Res. 2013;21:203–212.

Warrier S, Bhuvanalakshmi G, Arfuso F, Rajan G,

Millward M, Dharmarajan A. Cancer stem-

like cells from head and neck cancers are

chemosensitized by the Wnt antagonist,

sFRP4, by inducing apoptosis, decreasing

stemness, drug resistance and epithelial to

mesenchymal transition. Can Gen Ther 2014;

21:381–388.

Watkins DN, Berman DM, Burkholder SG, Wang

B, Beachy PA, Baylin SB.Hedgehog

signalling within airway epithelial progenitors

and in small-cell lung cancer. Nature 2003;

422:313–317.

Wen Y, Bi P, Liu W, Asakura A, Keller C, Kuang S.

Constitutive Notch activation upregulates

Pax7 and promotes the self-renewal of skeletal

muscle satellite cells. Mol Cell Biol 2012;32:

2300–2311.

Wicking C, Smyth I, Bale A. The hedgehog

signalling pathway in tumorigenesis and

development. Oncogene 1999;18:7844–7851.

Willert K, Brown JD, Danenberg E, Duncan AW,

Weissman IL, Reya T, et al. Wnt proteins are

lipid-modified and can act as stem cell growth

factors. Nature 2003;423:448–452.

Wharton KA, Johansen KM, Xu T, Artavanis-

Tsakonas S. Nucleotide sequence from the

neurogenic locus Notch implies a gene product

that shares homology with proteins containing

EGF-like repeats. Cell 1985;43:567–581.

Wright M, Calcagno AM, Salcido CD, Carlson

MD, Ambudkar SV, Varticovski L. Brca1

breast tumors contain distinct CD44+/CD24-

and CD133+ cells with cancer stem cell

characteristics. Breast Cancer Res 2008;

10:R10.

Xie J, Murone M, Luoh SM, Ryan A, Gu Q, Zhang

C, et al. Activating Smoothened mutations in

sporadic basal-cell carcinoma. Nature 1998;

391:90–92.

Xu J, Prosperi JR, Choudhury N, Olopade OI, Goss

KH. β-Catenin Is Required for the

Tumorigenic Behavior of Triple-Negative

Breast Cancer Cells. PLoS One 2015;10:

e0117097.

Yang ZF, Ho DW, Ng MN, Lau CK, Yu WC, Ngai P,

et al. Significance of CD90+ CSCs in Human

Liver Cancer. Cancer Cell 2008;13:153–166.

Yang W, Yan HX, Chen L, Liu Q, He YQ, Yu LX, et

al. Wnt/β-catenin signaling contributes to

activation of normal and tumorigenic liver

progenitor cells. Cancer Res 2008;68:

4287–4295.

Yoshikawa Y, Fujimori T, McMahon AP, Takada S.

Evidence that absence of Wnt-3a signaling

promotes neuralization instead of paraxial

mesoderm development in the mouse. Dev

Biomed Res J 2015;2(1):37-56

Dash et al. 55

Page 20: Developmental Signalling in Maintenance and Regulation of ...downloads.nmims.edu/science/journal/04-Dash-et-al.pdf · developmental signalling pathways that are involved in the regulation

Biol 1997;183:234–242.

Zhang Q, Shi S, Yen Y, Brown J, Ta JQ, Le AD. A

subpopulation of CD133 (+) cancer stem-like

cells characterized in human oral squamous

cell carcinoma confers resistance to

chemotherapy. Cancer Lett 2010;289:

151–160.

Zhang M, Atkinson RL, Rosen JM. Selective

targeting of radiation-resistant tumor-

initiating cells. Proc Natl Acad Sci USA

2010;107:3522–3527.

Zhang YV, Cheong J, Ciapurin N, McDermitt DJ,

Tumbar T. Distinct self-renewal and

differentiation phases in the niche of

infrequently dividing hair follicle stem cells.

Cell Stem Cell 2009;5:267–278.

Zeng L, Fagotto F, Zhang T, Hsu W, Vasicek TJ,

Perry WL, et al. The mouse Fused locus

encodes Axin, an inhibitor of the Wnt signaling

pathway that regulates embryonic axis

formation. Cell 1997;90:181–192.

Zeilstra J, Joosten SP, Dokter M, Verwiel E,

Spaargaren M, Pals ST. Deletion of the Wnt

target and cancer stem cell marker CD44 in

Apc(Min/+) mice attenuates intestinal

tumorigenesis. Cancer Res 2008;68:

3655–3661.

56 Developmental signalling in cancer cells

Biomed Res J 2015;2(1):37-56