SlideShare a Scribd company logo
1 of 38
Download to read offline
Wnt -4 Stimulation
of Human Bone
Marrow Stromal
Stem Cells and
Mesenchymal
Stem Cells of the
Maxilla

Gayathri Vijayakumar
Poly ID# 0408513
Guided Studies- Fall 2010


Nicola C. Partridge, Ph.D.
Professor, Chair
Basic Science & Craniofacial
Biology
E-mail: ncp234@nyu.edu


Michael D Turner, D.D.S., M.D.
Assistant Professor
Oral and Maxillofacial Surgery
E-mail: mdt4@nyu.edu
2|Page




TABLE OF CONTENTS

INTTRODUCTION ....................................................................................................................... 3

OVERVIEW ................................................................................................................................... 4

   WNT PROTEINS ...................................................................................................................................... 4

   WNT SECRETION & EXTRACELLULAR TRANSPORT ..................................................................... 7

   WNT RECEPTION.................................................................................................................................... 8

   WNT SIGNAL TRANSDUCTION PATHWAYS .................................................................................... 9

   REGULATION OF WNT SIGNALING.................................................................................................. 14

   WNT SIGNALING IN BONE FORMATION ......................................................................................... 15

   WNT-4 ..................................................................................................................................................... 17

   PTH & WNT PROTEINS (WNT-4) ........................................................................................................ 20

   WNT PROTEINS & STEM CELLS................................................................................................................. 24

   ROLE OF WNT SIGNALING IN MESENCHYMAL STEM CELLS                                       ....................................................................... 26


CONCLUSION ............................................................................................................................ 30

REFERENCE ............................................................................................................................... 31
3|Page


   1. INTRODUCTION
Wnt proteins are palmitoylated and glycosylated ligands that play a well established role in

embryonic patterning, cell proliferation and cell determination [1][2], besides playing a

significant role in cell cycle arrest, differentiation, apoptosis and tissue homeostasis. As a result,

aberrations in the Wnt signaling pathway are associated with birth defects as well as a multitude

of diseases, most notably cancer [3]. Wnt proteins can be categorized into two types: the

canonical and non-canonical.


Wnts    have     important           roles    in   regulating    many    aspects      of skeletal      development,

from limb formation        to    chondrogenesis         and osteoblast maturation        [4].    In     addition   to

promoting osteoblast maturation, Wnts may play a role in lineage commitment of mesenchymal

precursor cells by preventing adipogenesis is the hypothesized default pathway for mesenchymal

stem        cells that          do           not     receive        proper         inductive          signals      to

become osteoblasts, chondrocytes, myocytes, or other mesodermal cells [5].


Parathyroid hormone (PTH) regulates calcium homeostasis in addition to stimulating bone

formation      [6].      Human and           experimental       animal   studies      have      shown      that PTH

increases osteoblast activity and number via increased differentiation and survival, increases

bone mass and bone strength, and decreases fracture rate [7]. Due to its anabolic actions, PTH is

currently the only clinically available anabolic agent used to effectively treat osteoporosis. [8][9].

In vivo microarray analysis has shown that PTH regulates molecules of the Wnt signaling

pathway in bone [10]. PTH has been demonstrated to stimulate bone lining osteoblasts to

produce non-canonical Wnt-4 in vivo, and exogenous Wnt-4 has been shown to regulate

osteoblast differentiation through non-canonical Wnt signaling pathway [11].
4|Page


Wnt signaling has been implicated in the control over various types of stem cells and may act as

a niche factor to maintain stem cells in a self-renewing state [12]. Recent studies have shown that

Wnt-3a stimulates proliferation while inhibiting osteogenic differentiation of hMSCs [17], while

non-canonical Wnt-5a enhanced osteogenic differentiation but had no effect on hMSC

proliferation [18]. Most significantly, Wnt-4 has been shown to stimulate osteogenesis in hMSCs

isolated from craniofacial tissue through a novel p38 non-canonical Wnt signaling pathway that

is a known pathway associated with osteogenic differentiation [19][20].


Preliminary findings from Dr. Nicola C. Partridge’s lab at the Department of Basic Science and

Craniofacial Biology at NYU-CD have shown that Wnt-4 is stimulated by PTH in vivo [11], and

that Wnt-4 enhances proliferation and osteogenesis of mouse bone marrow stromal stem cells

(BMSSC) in vitro. From this it is hypothesized that Wnt-4 has an important function in

proliferation and differentiation of BMSSC.


The specific aims of this study are to:


    1) Assess the Wnt-4 effect on proliferation on human bone marrow stromal stem cells

    2) Assess the Wnt-4 effect on differentiation of human bone marrow stromal stem cells

    3) Assess the Wnt-4 effect on the proliferation and differentiation of human dental pulp

        stem cells.

   2. Overview

2.1 Wnt Protiens

Together with the other families of secreted factors such as FGF, TGF-beta, and Hedgehog

proteins,Wnt proteins are implicated in a wide variety of biological processes. The first Wnt gene
5|Page


mouse Wnt-1, was discovered in 1982 as a proto-oncogene activated by integration of mouse

mammary tumor virus in mammary tumors [21]. With the molecular identification of the

Drosophila segment polarity gene wingless (wg) as the orthologue of Wnt-1 [22][23] and the

phenotypic analysis of Wnt-1 mutations in the mouse [24], it became clear that Wnt genes are

important regulators of many developmental decisions [25][26].


Table 1 [21] Phenotypes of Wnt mutations in mouse, Drosophila, and C. elegans

Gene Organism Phenotype              Organism                    Phenotype
Wnt-1                                Mouse                       Loss     of    midbrain    and
                                                                 cerebellum
Wnt-2                                Mouse                       Placental defects
Wnt-3A                               Mouse                       Lack of caudal somites and
                                                                 tailbud
Wnt-4                                Mouse                       Kidney defects
Wnt-7A                               Mouse                       Ventralization of limbs
Wingless                             Drosophila                  Segment       polarity,   limb
                                                                 development, many others
Dwnt-2                               Drosophila                  Muscle       defects,    testis
                                                                 development
lin-44                               C.elegans                   Defects in asymmetric cell
                                                                 divisions
mom-2                                C. elegans                  Defects in endoderm induction
                                                                 and spindle orientation



The Wnts comprise a large family of protein ligands that affect diverse processes such as

embryonic induction, generation of cell polarity and the specification of cell fate. In addition to

influencing developmental processes, recent studies point to a key role for Wnt signaling during

adult homeostasis in the maintenance of stem cell pluripotency [27]. Wnts are defined by amino

acid sequence rather than by functional properties [28-29]. As many as 19 mammalian Wnt

homologues are known and are expressed in temporal spatial patterns. Shared features of all

Wnts include a signal sequence for secretion, several highly charged amino acid residues, and
6|Page


many glycosylation sites. Wnt proteins also display a characteristic distribution of 22 cysteine

residues. overexpression in tissue culture cells, several different N-linked glycosylated

intermediate Wnt protein products are observed in cell lysates [30-32], suggesting that Wnt

protein processing and secretion are highly regulated processes.


Even though the primary amino acid sequence of Wnts suggests that they should be soluble,

secreted Wnt proteins are hydrophobic and are mostly found associated with cell membranes and

extracellular matrix (ECM), the reason being that Wnt proteins are lipid modified by the

attachment of a palmitate on the first conserved cysteine residue within the protein family and on

a serine in the middle of the protein which may be necessary for:


       Wnt signaling as well as secretion

       May be necessary for their glycosylation.

       Might also aid in Wnt transport between cells as glycosylation might increase Wnt

        interactions with heparin sulfate proteoglycans (HSPGs) present on the surface of Wnt

        responding cells.

       Potentially anchoring Wnt proteins into the membrane for sustained signaling. [33-34].


There are a total of 7 related Wnt genes in the Drosophila genome and 19 in the human genome

(some with multiple isoforms) that generally have close orthologues in mice. Orthologous Wnt

gene products (proteins with the same function in different species) are often very highly

conserved. [35] Based on their ability to induce secondary body axis in Xenopus embryos, the

Wnt family of proteins is grouped into two functional classes. The Wnt-1 class (Wnts-1, 2, 3, 3a,

7b, 8, 8b, 10b) is able and Wnt-5a (Wnts-4, 5a, 5b, 6, 7a, 11) is unable to induce a secondary

body axis [36].
7|Page


2.2 Wnt Secretion and Extracellular Transport

Two recent genetic screens have identified the multipass transmembrane protein Wntless

(Wls)/Evenness interrupted (Evi)/Mom-3 as acting in the secretory pathway to promote the

release of Wnts from producing cells [37]. A model of Wnt secretion and realease is shown in

figure 1.Exogenously derived lipoproteins termed ―argosomes‖ are implicated in moving Wnts

and other lipid-modified proteins such as Hedgehogs [38]. A model is proposed wherein

palmitoylated proteins associate with lipoprotein particles on the extracellular face of cells.

Traffic of Wnt proteins from one cell to the next requires this association [38]. In addition,

transcytosis may regulate Wnt movement. It has been proposed that the retromer complex

promotes the association of secreted Wnts with other proteins required for ligand transport, such

as lipoprotein particles. [39]




Figure 1 [40]: Model of Wnt secretion and release. Wnt is lipid modified in the ER and is transported to the Golgi,
where it binds to Wls. Next, the Wnt–Wls complex is transported to the plasma membrane and Wnt is released (1).
8|Page


The hydrophobic Wnt protein either remains associated with the plasma membrane or binds directly to lipoprotein
particles or HSPGs to facilitate spreading and gradient formation (2). After the release of Wnt, Wls is internalized
through AP-2/clathrin-mediated endocytosis (3) and is transported back to the TGN through a retromer-dependent
trafficking step (4). An alternative possibility is that Wnt is not released at the plasma membrane, but is
reinternalized together with Wls (5). Dissociation of Wnt from Wls may take place in endosomes, after which Wnt
is released (possibly in association with lipoprotein particles) through a recycling endosomal pathway (6). Also in
this scenario, Wls is recycled back to the TGN through a retromer-dependent transport step (7). Wnt may also be
reinternalized through a Reggie-1/Flotillin-2 dependent pathway (8), which may lead to the release of a more
mobile, micelle-like form of Wnt or to the association of Wnt with lipoprotein particles (6).


2.3 Wnt Reception

The     seven-pass       transmembrane         protein

Frizzled (Fz) protein was first receptor found

to transduce a Wnt signal [41]. . Fz proteins

contain     a    large     extracellular      domain

containing a conserved motif comprised of

10 cysteine residues called the cysteine-rich

domain (CRD) that has been shown to bind
                                                               Figure 2 [21]: Structure of Frizzleds and FRPs. Frizzled
to multiple Wnts with high affinity. At the                    receptors are characterized by an N-terminal signal peptide,
                                                               a cysteine-rich ligand-binding domain (CRD) followed by a
cytoplasmic side, Fzs may interact directly                    hydrophilic linker,seven transmembrane regions, and a
                                                               cytoplasmic tail. FRPs are secreted proteins with a CRD
                                                               similar to Frizzleds. In addition, they contain a region with
with the Dishevelled protein, a known
                                                               similarity to netrins, secreted proteins involved in axon
                                                               guidance.
mediator of Wnt signaling [42-44].


Following Wnt binding, it is thought that Fzs form a co-receptor complex with single-pass

transmembrane proteins of the low-density lipoprotein (LDL) family called Lrp5 and -6 to

transduce the canonical Wnt signal. Lrp5 and -6 proteins have a relatively small intracellular

domain and a large extracellular domain containing several potential protein interaction

domains [45].
9|Page


There are other proteins with known Wnt-binding

domains that can serve as receptors for Wnt ligands.

The single-pass tyrosine kinase Ror2, although

structurally distinct from Fz receptors, is involved

in other forms of Wnt signaling. Another well-

characterized Wnt-binding domain is the Wnt

inhibitory factor (WIF) module, which is also found

in the cell surface atypical receptor tyrosine kinase       Figure 3: Different receptors that Wnt proteins
                                                            can bind to.
Ryk [46][47].                                               http://www.stanford.edu/group/nusselab/cgi-
                                                            bin/wnt/receptors
2.4 Wnt Signal Transduction Pathways

Wnt proteins signal through the canonical and the non-canonical pathways which are composed

of three independent signal transduction pathways (the Wnt/β-catenin pathway, the

Wnt/Ca+2 pathways, or the Wnt/planar polarity pathway) that are used to regulate the expression

of different genes. The Wnt/β-catenin pathway is commonly referred to as the canonical

pathway. It promotes cell fate determination, proliferation, and survival by increasing β-

catenin levels and altering gene expression through Lef/Tcf transcription factors [48]. The non-

canonical    Wnt/Ca+2 pathway stimulates heterotrimeric G proteins, increases intracellular

calcium levels, decreases cyclic GMP levels, and activates protein kinase C to induce NF-

AT and other transcription factors [49]. The non-canonical Wnt/planar polarity pathway activates

Rho/Rac GTPases and Jun N-terminal kinase to modulate cytoskeletal organization and gene

expression. Distinct Wnt ligands probably act through specific Frizzled (Fzd) receptors to initiate

each [49].
10 | P a g e




                       Figure 4: Wnt Signaling Pathways and their Implications



The canonical Wnt signaling pathway (see Figure 5) is activated when Wnts interact with

Lrp/Fzd receptor complexes as shown in the middle portion of figure 4. Receptor engagement

activates an unknown kinase(s) (K) that phosphorylates the cytoplasmic tail of Lrp5/6. These

phosphorylated residues (P) serve as docking sites for Axin and the APC, Dsh, β-catenin

complex. A GSK3β binding protein (GBP) is also mobilized after receptor ligation and excludes

GSK3β from the proximal receptor complex. β-Catenin thereby escapes phosphorylation events

that normally promote its ubiquitination (U) by E2 ligases and degradation by the proteosome

(right side of this figure). As β-catenin levels rise above those needed to bridge cadherins to the
11 | P a g e


actin cytoskeleton (lower left of this figure), some β-catenin molecules travel to the nucleus

where they interact with Lef1/Tcf transcription factors to either increase or decrease the

expression of specific target genes. β-Catenin displaces nuclear co-repressors (CoR) from

Lef1/Tcf to facilitate the expression of genes involved in cell cycle progression (e.g. cyclin D1)

and survival (e.g. c-myc). β-Catenin and Lef1/Tcf suppress other genes, such as osteocalcin

(OCN) and E-cadherin, through unknown mechanisms, but that may involve interactions with

other transcription factors (TF). Soluble antagonists block the canonical Wnt signaling pathway

and            promote     β-catenin        degradation        via        two       mechanisms.




                         Figure 5 [49]: The Canonical Wnt Signaling Pathway

Soluble frizzled related proteins (Sfrp) bind free Wnt molecules and compete with surface

receptors (top right of figure 5). In contrast, Dkks interact with extracellular domains in Lrp5/6
12 | P a g e


and recruit them to complexes containing Krm (top left of figure 5). This trimolecular complex is

internalized to lysosomes where Lrp5/6 are either degraded or recycled to the surface. Dkk

therefore decreases Lrp5/6 cell surface expression to regulate Wnt signaling [49].




                             Figure 6 [55]: Nuclear activity of ß-catenin



Accumulated ß-catenin then translocates to the nucleus, replaces Groucho from TCF, and

activates target genes. ß-catenin forms a complex with TCF and the transcription factors Brg1

and CBP. Lgs and Pygo also bind to -catenin, possibly driving its nuclear localization in addition

to playing a direct role in transcriptional activation. Negative regulation of signaling is provided

by NLK (Nemo-like kinase) which phosphorylates TCF, and ICAT (inhibitor of catenin) and

Chibby, which are antagonists of ß-catenin. In addition to TCF, two other DNA-binding proteins

have been shown to associate with ß-catenin: Pitx2 and Prop1. In the case of Prop1, ß-catenin

can act as a transcriptional activator or repressor of specific genes, depending on the co-factors
13 | P a g e


present. The participation of any particular ß-catenin complex in transcriptional regulation is

highly cell type-dependent.[55]


The non-canonical pathway can be broadly classified into 2 branches based upon phenotypic

response; the Planar Cell Polarity (PCP) pathway and the Wnt/ Ca2+ pathway [50]. However

some authors have classified the pathways as Wnt/calcium signaling, Wnt/PCP signaling,

Wnt/JNK signaling and Wnt/Rho signaling [56]. The PCP pathway is involved in cellular

asymmetry, and it is this cellular asymmetry that controls the rigid architectural orientation of

epithelial tissues and sensory organs (e.g. inner ear cochlea), as well as the morphology and the

migratory processes of mesodermal cells undergoing gastrulation. Activation of PCP signaling

occurs basically through the binding of Wnts to Frizzled (Fz) receptors alone, without LRP co-

receptor involvement. These signals activate Dishevelled (Dvl), which in turn leads to the

activation of the GTPases Rho and Rac. Activated Rac subsequently stimulates JNK activation

[50].


The second branch of non-canonical signaling – the Wnt/Ca2+ pathway – is characterized by

Wnt-Fzd-induced PLC (phospholipase C) activation and the resultant increase of cytoplasmic

Ca2+ levels. These Ca2+ fluxes activate several Ca2+-responsive proteins, such as PKC (protein

kinase C) and CaMKII(calcium/calmodulin-dependent kinase II). CaMKII has been shown to

activate the transcription factor NFAT, TAK1 (TGF-beta activated kinase), and NLK (Nemo-like

kinase, all of which have the net effect of decreasing intracellular cGMP and consequently,

antagonizing Wnt/beta-catenin/TCF signaling [51-52]. The Wnt/Ca2+ signaling pathway is

involved in regulating cellular adhesion, cytoskeletal rearrangements, and other developmental

processes, such as dorsoventral patterning and tissue separation in embryos [53].
14 | P a g e




                                  Figure 7 [54]: Non- canonical pathways



2.5 Regulation of Wnt Signaling:

Wnt signaling is tightly regulated by members of several families of secreted antagonists


    1. Wnt signaling requires interaction with frizzled receptors (Fz) and the presence of a

        single-pass transmembrane molecule of the Lrp family (Lrp5 or 6) which can be inhibited

        by members of the secreted frizzled-related protein (Sfrp) family and Wnt inhibitory

        factor 1 (WIF-1).
15 | P a g e


    2. Dkk1 binds to Lrp with high affinity and to another class of transmembrane molecules,

        the Kremens. Dkk1 promotes the internalization of Lrp and makes it unavailable for Wnt

        reception by forming a complex with Lrp and Kremen [57].

    3. Lrp coreceptor activity is also inhibited by members of sclerostin (SOST gene product).

    4. Chibby is a nuclear antagonist that binds to the C terminus of β-catenin [58].

    5. Another β-catenin binding protein, ICAT, can block the binding of β-catenin to TCF and

        also   can   lead   to   the   dissociation   of   complexes   between β-catenin,   LEF,

        and CBP/p300 [56].

    6. TCF can be phosphorylated by the mitogen-activated protein (MAP) kinase-related

        protein kinase NLK/Nemo.

    7. The phosphorylation of TCF/LEF by activated Nemo is thought to diminish the DNA-

        binding affinity of the β-catenin/TCF/LEF complex, thereby affecting transcriptional

        regulation of Wnt target genes [56].


2.6 Wnt Signaling in Bone formation:

The Wnt signal transduction pathway has been implicated in bone formation: patients suffering

from osteoporosis–pseudoglioma syndrome have an inactivating mutation in the Wnt co-

receptor LRP5 [13], whereas an activating LRP5 mutation is associated with high bone mass

syndrome [14] [15]. Analysis of LRP5-deficient mice revealed a decreased number

of osteoblasts suggesting that Wnt signaling stimulates bone formation at the level of

osteoprogenitor proliferation [16].
16 | P a g e




Figure 8 [56]: Role of canonical Wnt signaling in skeletal development. Mesenchymal stem cells have the ability
to differentiate into chondrocytes or osteoblasts, depending on the environmental cues. Canonical Wnt signaling
is regulated to control the lineage progression between chondrocyte and osteoblast. If there is inadequate
canonical Wnt signaling, differentiation towards chondrocyte lineage is encouraged. However, the maturation of
chondrocytes requires the presence of canonical Wnt signaling. Canonical Wnt signaling is also required for the
progression of osteoblast progenitor cells toward osterix positive osteoblasts and then osteocalcin-positive
osteoblasts. Canonical Wnt signaling represses osteoclastogenesis by increasing the osteoprotegrin (OPG)
expression, therefore the OPG/RANKL ratio. Green plus signs indicate positive effects of Wnt; red circle minus
signs indicate inhibitory effects of physiological canonical Wnt signaling

The effects of Wnt signaling on chondrogenesis are complex and have been implicated in the

regulation of chondrogenic differentiation and hypertrophy [56]. Chondrocyte-specific

inactivationof β-catenin using Col2a1-Cre transgene leads to decreased chondrocyte proliferation

and delayed hypertrophic chondrocyte differentiation [59].                    In another loss-of-function

model, Dermo 1-Cre transgene was used to delete the β-catenin gene in the mesenchymal

precursors of both chondrocytes and osteoblasts. It was shown that there is a significant delay

in chondrocyte maturation in conditional knockout embryos [60]. At the same time Activation

of β-cateninin limb and head mesenchyme repressed the expression of Sox9, a factor essential

for chondrogenesis, thereby preventing mesenchymal cells from differentiating into skeletal

precursors [59]. In the absence of β-catenin, the expression of early osteoblast markers, such

as collagen I, osterix, and osteocalcin was greatly diminished [60].
17 | P a g e




        Figure 9 [49]: Effects of Wnt signaling on osseous cells. The canonical Wnt signaling pathway
        promotes the proliferation, expansion and survival of pre- and immature osteoblasts. Dkks,
        Sfrps, and Wif-1 antagonize Wnt signaling in osteoblasts to facilitate death of immature cells,
        but they may also downregulate the pathway in mature cells to induce terminal differentiation.

Wnt-4:

Wnt-4 is a signaling factor with multiple roles in organogenesis, a deficiency that leads to

abnormal development of the kidney, pituitary gland, female reproductive system, and mammary

gland. Wnt4 is conserved throughout vertebrates and was originally studied in non-mammalian

vertebrates such as zebrafish, chicken and xenopus [61]. Several studies have tried to gauge the

varied functions of this protein, giving the impression that this is indeed a highly conserved

protein essential for proper organ formation and development. Some of the findings are as

follows:


    1. It      is   essential   for   nephrogenesis.   The   Wnt-4 gene      is   expressed    in   the

        assembling nephrons and the medullary stromal cells during kidney development The
18 | P a g e


        role   of   Wnt-4 signaling     in   controlling mesenchyme to epithelium transformation

        and kidney tubule induction is well established [63].

    2. Wnt-4 signaling plays also a role in the determination of the fate of smooth muscle

        cells in the medullary stroma of the developing kidney. Smooth muscle α-actin (α-SMA)

        is markedly reduced in the absence of its signaling [62].

    3. Wnt-4 plays an important role in female sexual development and ovarian function. Wnt-4

        deficiency leads disturbed development of the internal genitalia in mouse and human, and

        to a dramatic reduction of mouse oocytes.            The expression of        Wnt-4 protein

        in human fetal ovaries was high during mid-pregnancy, when new follicles are also

        rapidly being formed. This implies that Wnt-4 may have a substantial role in regulation

        of the follicle formation in human ovaries [64].

    4. In the reproductive system, Wnt-4 is specifically involved in Müllerian duct formation,

        sex-specific blood     vessel formation, oocyte maintenance        and     repression     of

        steroidogenesis. Partial XY male-to-female sex-reversal is observed in mice lacking Wnt-

        4 as well as in one human patient carrying a Wnt-4 point mutation [61].

    5. During mammalian embryogenesis, Wnt-4 is expressed in the gonads of both sexes

        before sex determination events take place and is subsequently down-regulated in the

        male gonad [61].

    6. Wnt4 regulates mammary gland development in response to hormonal changes that take

        place during pregnancy and may also play a part in mammary gland tumorigenesis.

        Expression of Wnt4 is inversely correlated with cell proliferation in the mouse mammary

        C57 mg cell line, suggesting that Wnt4 participates in restricting cell proliferation. On the

        other hand, the elevated expression of theWNT4 gene in fibroadenomas, and occasionally
19 | P a g e


        also in malignant tumor tissue, implies that changes in WNT4 signaling are associated

        with abnormal cell proliferation in human breast cells [68].

    7. Screening of the multiple adult human tissues           Figure 10 [65]. WNT4 transcripts in human adult
                                                               (A) and embryonic (B) tissues. Three mRNA bands,
        revealed three human WNT4 mRNA bands of 1.5,           1.5, 2.4 and 4.3 kb in size, are seen in the adult
                                                               tissues and one, 2.4 kb, in the embryonic liver and
        2.4, and 4.3 kb in size (Figure 8). The most           kidney tissues. The ages of the embryos from 6 to
                                                               12 weeks are indicated below the sample rows. The
        common 1.5 kb transcript was detected in a             arrows show the positions of ribosomal
                                                               RNA.
        number         of         tissues:       the adrenal

        gland, placenta, liver, mammary

        gland, prostate, spinalcord, stomach, thyroid, trach

        ea, skeletal muscle and small intestine, the signal

        being strongest in the adrenal gland and placental

        samples.

    8. The involvement of Wnt-4 in normal mammary

        gland and ovary development suggests that Wnt-

        4 germline mutations may be associated with

        the human cancer predisposition [65].

    9. Wnt-4 is critical for development of the adrenal gland, the zona glomerulosa of which is

        incomplete at birth in Wnt-4-deficient mice, probably as a result of significantly reduced

        aldosterone production. In addition, Wnt-4 represses the migration of steroidogenic

        adrenal precursors into the gonad [66]. In mouse spleen, Wnt1and Wnt-4 signaling

        regulates differentiation of the thymocytes, the number of which is decreased in

        compound mutants [67].
20 | P a g e




      Figure 11 [65]. Mapping of the human WNT4 locus to chromosome region 1p36.12 by prometaphase FISH. (A)
      Hybridization signals for WNT4 (green signals) and BAC RP11-285H13 (red signals). (B) Chromosomal
      location of BAC RP11-285H13 to the region 1p36.11a-1p36.11b and BAC RP11-145C4 to the region 1p36.13c-
      1p36.13d (C) Hybridization signals for WNT4 (green signals) and BAC RP11-145C4 (red signals).




2.7 PTH and Wnt Proteins (WNT-4)
PTH is a single chain polypeptide with 84 amino acids and is the principle regulator of

calcium homeostasis in vertebrates. However, although continuous infusion of PTH induces

bone loss, intermittent administration of PTH results in bone formation [69]. Binding

of PTH and PTHrP to PTH1R activates two signaling pathways in osteoblasts: the PKA pathway

which is responsible for the majority of the calciotropic and skeletal actions of PTH, the PKC

pathway leading to accumulation of 1, 4, 5-inositol triphosphate and increased intracellular

calcium. This pathway has been found to regulate IGF-binding protein-5 [70].


A number of studies suggest that the PTH and Wnt signaling pathways do indeed overlap.

Following treatment of osteoblastic cells with N-terminal PTH, an increase in β-catenin that was

measured in whole cell lysates by Western blot was seen [71].
21 | P a g e




                                Figure 12: PTH regulation of Wnt-4


Furthermore, PTH increased the level of β-catenin expression in mouse osteoblastic cells

(MC3T3-E1) via both PKA and PKC signaling pathways [74]. More recently, PTH was shown to

activate β-catenin signaling in osteoblasts in vitro and in vivo by direct recruitment of LRP6 to

PTH/PTH1R complex. In vivo studies confirmed that intermittent PTH treatment led to an

increase in amount of β catenin in osteoblasts (immunohistochemical analysis with antibody

to β-catenin) with a concurrent increase in bone formation in rat [75].

Partridge et al. also demonstrated a link between PTH and Wnt4 expression in bone [72]. In

vivo microarray     analysis     of     intermittent    and     continuous PTH 1–34      showed
22 | P a g e


that PTH regulated Wnt4 in bone. PTH was shown to stimulate Wnt4 primarily through

the PKA pathway and Wnt4 treatment in osteoblasts induced early expression of bone marker

genes and stimulated key canonical Wnt pathway genes. This finding suggested cross-talk

between the Wnt signaling cascades [73].

Previous studies at Dr. Partridge’s lab at the Department of Basic Science & Craniofacial

Biology at NYU-CD has showed that Wnt-4 is significantly regulated by PTH in vivo after

anabolic and catabolic protocols in bone lining osteoblasts. As pointed out earlier Partridge et

al. demonstrated that PTH stimulated Wnt-4 expression in cultured osteoblastic cells and that

this stimulation is PKA dependent and is a primary response to PTH. It has also been shown

that exogenous Wnt-4 enhances bone marker gene expression during osteoblast

differentiation by activating the non-canonical Wnt/Ca2+ and Wnt/PCP signaling pathways

but does not significantly stimulate the canonical Wnt/ß-catenin pathway. Most significantly,

Wnt-4 has been shown to stimulate osteogenesis in hMSCs isolated from craniofacial tissue

through a novel p38 non-canonical Wnt signaling pathway that is a known pathway

associated with osteogenic differentiation [19][20]. Therefore it can be hypothesized that non-

canonical Wnt signaling plays a significant role in bone and that Wnt-4 may be an important

molecule in PTH’s anabolic effect.[72]


Real time TR-PCR results have shown that PTH stimulates Wnt-4 mRNA expression in all

phases of osteoblastic differentiation but is greatest in the mineralization phase and maximal

stimulation occurs 8h after PTH treatment. Partridge et al. has demonstrated that non-

canonical Wnt-4 does not increase pre-osteoblastic proliferation or the expression of bone

marker genes at proliferation day 7, but enhances bone marker gene expression (runx2,

osterix, osteocalcin, alkaline phosphatase, MMP-13) significantly in the absence of
23 | P a g e


differentiation-promoting factors such as ascorbate. Continuous treatment of rat primary

osteobalsts with Wnt-4 in an osteogenic environment showed that there was increased

osteocalcin mRNA and alkaline phosphatase expression in late stage differentiation with

increased mineralized nodules. In addition there was increased relative expression of runx2

and osterix mRNA during osteoblast differentiation. It was also found that Wnt-4 acute

treatment in the early stages of primary cell differentiation was much more effective at

stimulating the relative gene expression of runx2, osterix, osteocalcin, alkaline phosphatase,

and collagen-1a mRNA expression. Together these suggest that Wnt-4 may promote the

differentiation of osteoblasts as well as uncommitted cells in the bone environment as part of

PTH’s anabolic effect.[72]




               Figure 13: Effect of WNT-4 on osteoblast proliferation & differentiation
24 | P a g e


 2.8 Wnt Proteins and Stem Cells

 The Wnt Signaling pathway has been found to play a very important role in maintaining the

 potency of stem cells and stem cell fate determination. Stem cells from different locations

 interpret Wnt in different ways which reflects an activation of distinct genetic programs in

 response to the same signal. In addition, the time point during development at which a stem cell

 is challenged by Wnt signals determines whether the cell responds by self-renewal or

 differentiation. Besides the cell-intrinsic cues that influence the biological activity of Wnt in

 distinct stem and progenitor cell types, the same type of stem cell might respond in different

 ways to Wnts, depending on its extracellular microenvironment (illustrated in Figure 14 and 15)

 [88].




Figure 14. [88] Cell-intrinsic differences among stem cells
                                                                      Figure 15. [88] The effect of canonical Wnt on a particular
influence the biological function of Wnts. (a) Different stem cell
                                                                      type     of   stem cell      is   context-dependent. (a) In  a
types differentially respond to canonical Wnt signaling and
                                                                      microenvironment X, Wnt activity is modulated by the
undergo either self-renewal or lineage commitment. The
                                                                      factor(s) X. In this context, Wnt signaling elicits self-
differential responsiveness of stem cells is presumably due to
                                                                      renewal. (b) However, the very same stem cell in
distinct cell-intrinsic determinants (indicated in the figure by
                                                                      microenvironment Y, in which Wnts are modulated by the
differently colored cells). (b) A stem cell type of a given cell
                                                                      factor(s) Y, responds to Wnt signaling by adopting a specific
lineage (indicated in various grades of green) can integrate
                                                                      cell fate rather than by self-renewing. Thus, the biological
canonical Wnt signaling in different ways, depending on its cell-
                                                                      activity of Wnt in a particular microenvironment is influenced
intrinsic properties which change over time. At different stages of
                                                                      by the convergence of Wnt signaling with other signal
development, Wnt promotes stem cell self-renewal or lineage
                                                                      transduction pathways.
commitment, or the cell loses its ability to respond to Wnt.
25 | P a g e


Some of the findings regarding the role of Wnt signaling in stem cells are as follows:


    1. In embryonic stem cells, over expression of Wnt1 or of stabilized β-catenin results in the
        inhibition of neural differentiation [77].

    2. Treatment of haematopoietic stem cells with Wnt proteins and sustained expression of β-

        catenin promotes self-renewal in long-term cultures and increases the reconstitution of

        haematopoietic lineages in vivo, which could have been mediated by Notch1 and

        the transcription    factor      HoxB4.     However,     conditional    ablation    of β-catenin in

        haematopoietic stem cells did not impair haematopoiesis and lymphopoiesis, suggesting

        that β-catenin is not required for self-renewal and development of haematopoietic stem

        cells under physiological conditions [78].

    3. Wnts play major roles during central nervous system (CNS) development. Ablation

        of wnt1 results     in     severe    defects    of     the midbrain,    the cerebellum and        the

        developing spinal        cord,   while    ablation   of wnt3A results    in   a    total   loss   of

        the hippocampus. The defects observed in Wnt mutants are possibly explained by

        perturbed proliferation of stem or progenitor cells in the ventricular zone [78] [79] [80].

    4. Wnt/β-catenin signaling is not only essential for the homeostasis of the intestinal

        epithelium; sustained β-catenin activity has also been implicated in the formation

        of colon carcinoma [78] [81].

    5. In vivo manipulation of genes encoding Wnt signaling components indicates that ß–

        catenin deficient stem cells fail to differentiate into follicular keratinocytes and instead

        adopt an epidermal fate and thus play a very important role in the fate determination

        process of epidermal stem cells [82].
26 | P a g e


    6. Wnt signaling has been implicated in the early stages of neural crest development, such

         as neural crest induction and melanocyte formation. In neural crest stem cells, the genetic

         ablation of β-catenin results in lack of melanocytes and sensory neural cells in dorsal root

         ganglia. In fact, NCSCs lacking β-catenin emigrate and proliferate normally but are

         unable to acquire a sensory neuronal fate. Constitutive expression of β-catenin in neural

         stem/progenitor cells results in the expansion of the entire neural tube, supporting a role

         of β-catenin in progenitor proliferation [83].

    7.   In a recent study from Nusse's laboratory, Axin2 gain-of-function mice were used to

         demonstrate that Wnt3A functions as a rate-limiting, self-renewal factor to clonally

         expand mammary stem cells (MaSC) [84].


2.9 Role of Wnt Signaling in Mesenchymal Stem Cells:
Mesenchymal stem cells (MSCs), otherwise termed as mesenchymal progenitor cells or marrow

stromal cells are adherent, fibroblast-like population with the potential for extensive self-renewal

and multilineage differentiation. Under appropriate culture conditions, MSCs are capable of

giving rise to osteoblasts, adipocytes, chondrocytes and myoblasts. Their multipotency, ease of

isolation and ready availability make MSCs particularly suited for tissue engineering and gene

therapy applications [76].


MSCs express a number of Wnt ligands such as Wnt-2, Wnt-4, Wnt-5a, Wnt-11 and several Wnt

receptors as well as various co-receptors and Wnt inhibitors [85]. Exogenous application of Wnt-

3a to cell cultures expands the multi-potential population of MSCs and this proliferative is

presumably achieved by the up-regulation of cyclin D1 and c-myc both of which drive cell cycle

progression to promote growth [17] [86].
27 | P a g e




               Figure 16: Wnt-4 signaling in the bone as part of PTH’s anabolic effect
28 | P a g e


In previous studies done in Dr. Partridge’s lab by Bergenstock et al., it was found that Wnt-4

increased cell proliferation and CFU-F numbers by inhibiting mouse BMSSC apoptosis and

stimulating their rate of growth. mBMSSCs treated with Wnt-4 in osteogenic and non-

osteogenic cultures showed a significant increase in osteocalcin bone marker gene expression

and a significant reduction of adipocyte marker genes (ap2, pparϒ and C/EBPα). It was found

that Wnt-4 stimulated the Wnt/ß-catenin pathway in proliferating mBMSSCs after Wnt-4

treatment. Wnt-4 did not alter ß-catenin expression but stimulated the phosphorylation of

CamKII (non-canonical Wnt/Ca2+) in differentiating mBMSSCs while inhibiting JNK

phophorylation (non-canonical Wnt/PCP) in proliferating mBMSSCs. This suggests the

divergent effect of Wnt-4 depending on the developmental stage of the cell. Since Wnt-4 is

regulated by PTH in bone, this suggests two functions for Wnt-4 in the stem cell environment

as part of PTH’s anabolic effects. The first being to act on stem cells and still uncommitted

osteoprogenitor cells to expand cell numbers which would eventually be released into

osteogenic differentiation.


The mechanism of signaling switch, that determines whether canonical or non-canonical Wnts

increase stem cell proliferation or stimulate differentiation in certain tissues is not well

understood. Now the question arises how Wnt-4 stimulates the canonical and non-canonical

pathways.      One   paper    suggests    that   the    balance    and    coordination   between

nuclear/transcriptionally active beta-catenin and cytoplasmic/cytoskeletal beta-catenin couples

canonical and non-canonical Wnt signaling. Another suggestion is that the coactivators CBP and

p300, part of the Wnt signaling network of proteins, plays the integrator role [87].
29 | P a g e


CREB binding protein (CBP) and p300 are

key regulators of RNA polymeraseII-mediated

transcription that encode highly related protein

accetlytransferases that bind a variety of

transcriptional regulators and other proteins.

As pointed out earlier, research done in this

lab has pointed out that continuous treatment

of mBMSSCs with Wnt-4 activates Wnt/beta-

catenin pathway in the proliferation stage in a

significant manner and then the Wnt/Ca2+

pathway is activated in differentiation, but
                                                   Figure 17 [87]: A. model of coactivator usage. Antagonizing the
                                                   CBP/beta-catenin interaction leads to the downregulation of genes
beta-catenin continues to be produced albeit       that are critical for stem cell/progenitor cell maintenance and
                                                   proliferation (left arm of pathway). This also pushes the cell to
not in a significant manner as in the              utilize p300 as its coactivator. The switch to p300/beta-catenin-
                                                   mediated transcription is the first critical step to initiate a
proliferation stage. Perhaps the co-activators     differentiative program. B. a subset of the gene expression cassette
                                                   that is regulated by the CBP/beta-catenin arm is critical for the
CBP and p300 along with nuclear beta-catenin       maintenance of potency and proliferation (e.g. Oct4, survivin, etc.).
                                                   Other genes that are similarly regulated by CBP/beta-catenin (e.g.
plays a role in making the switch from             hNkd and axin2) are involved in the negative feedback of this arm of
                                                   the pathway, and initiate differentiation via a switch to the
proliferation to differentiation.                  p300/beta-catenin arm.


A recent paper has described that that CBP/beta-catenin mediated transcription is critical for

stem cell/ progenitor cell maintenance and proliferation, whereas a switch to p300/beta-

catenin mediated transcription is the initial critical step to initiate differentiation and a decrease

in cellular potency. A subset of the gene expression cassette (e.g. Oct4, surviving, etc.) is critical

for the maintenance of potency and proliferation, other genes such as hNkd and axin2 that are

regulated in this manner are negative regulators that stop proliferation, exit cell cycle and initiate
30 | P a g e


the process of differentiation [87]. The shift from the canonical to the non-canonical pathway

relies on the activation of the PKC pathway. PKC phosphorylation of Ser89 of p300 increases

the affinity of p300 for beta-catenin both in vivo an in vitro and thus the switch occurs and the

differentiation pathway is initiated (shown in Figure 17) [87]. If this is true in the case of Wnt-4

is still to be investigated.


    3. Conclusion:

Wnt-4 is significantly regulated by PTH as part of its anabolic effect in the bone environment.

Preliminary studies have revealed that Wnt-4 expression is stimulated in osteoblasts in a PKA

dependent manner and it is a primary response to PTH. It has been found to promote osteoblast

differentiation by enhancing bone marker gene expression by activating the non-canonical

pathways.


In mBMSSCs, Wnt-4 has been found to stimulate proliferation by activating the canonical

pathway and then induce differentiation along the osteoblast lineage by stimulating the non-

canonical pathway.


Next we are going to assess if the Wnt-4 can induce proliferation and differentiation along the

osteoblast lineage in human BMSSCs and human dental pulp stem cells (hDPSCs).
31 | P a g e


    4. REFERENCE:


    1. Roles of Wnt signaling in bone formation and resorption, Yasuhiro Kobayashi, Kazuhiro
        Maeda     and      Naoyuki      Takahashi,    Japanese    Dental    Science      Review
        Volume 44, Issue 1, July 2008, Pages 76-82
    2. Roles of Wnt proteins in neural development and maintenance, Ardem Patapoutian and
        Louis F Reichardt, Current Opinion in Neurobiology Volume 10, Issue 3, 1 June 2000,
        Pages 392-399
    3. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two
        coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews
        Volume 62, Issue 12, 30 September 2010, Pages 1149-1155, Stem Cell Gene
        Manipulation and Delivery as Systemic Therapeutics
    4. Wnt signaling in osteoblasts and bone diseases, Jennifer J. Westendorf, Rachel A. Kahler
        and Tania M. Schroeder, Gene Volume 341, 27 October 2004, Pages 19-39
    5. Inhibition of Adipogenesis by Wnt Signaling, Sarah E. Ross, Nahid Hemati, Kenneth A.
        Longo, Christina N. Bennett, Peter C. Lucas, Robin L. Erickson and Ormond A.
        MacDougald, Science 11 August 2000: Vol. 289 no. 5481 pp. 950-953
    6. Anabolic actions of parathyroid hormone on bone, Dempster DW, Cosman F., Parisien
        M., Shen V., Lindsay r., Endocr Rev 14:690-709 (1993).
    7. In vivo studies of anabolic action of PTH and Wnt signaling pathway in bone, Yanfei L.
        Ma, Nalini H. Kulkarni, Qing Qiang Zeng, Tao Wei, David L. Halladay, Masahiko Sato,
        Henry U. Bryant and Jude E. Onyia, Bone Volume 43, Supplement 1, October 2008,
        Page    S37,    International   Conference   on Osteoporosis and Bone Research    2008,
        International Conference on Osteoporosis and Bone Research 2008
    8. Cosman F (2005a): Anabolic therapy for osteoporosis: parathyroid hormone. Curr
        Osteoporos Rep 3: 143-9
    9. Cosman F (2005b): The prevention and treatment of osteoporosis: a review. MedGenMed
        7:73.
    10. Determination of dual effects of parathyroid hormone on skeletal gene expression in vivo
        y microarray and network analysis, Li X, Liu H, Qin L, Tamasi J, Bergenstock M,
        Shapses S, Feyen JH, Notterman DA, Partridge NC, J Biol Chem, (2007), 282:33086-97.
32 | P a g e


    11. Parathyroid hormone stimulation of noncanonical Wnt signaling in bone, Bergenstock
        MK, Partridge NC, Ann N Y Acad Sci. 2007 Nov;1116:354-9.
    12. Wnt signaling and stem cell control, Roel Nusse, Cell Research (2008) 18:523-527
    13. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development, Y.
        Gong, R.B. Slee, N. Fukai, G. Rawadi, S. Roman-Roman, A.M. Reginato et al.,Cell,
        107 (2001), pp. 513–523.
    14. High bone density due to a mutation in LDL-receptor-related protein 5., L.M. Boyden, J.
        Mao, J. Belsky, L. Mitzner, A. Farhi, M.A. Mitnick et al., N. Engl. J. Med.346 (2002),
        pp. 1513–1521.
    15. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant
        high-bone-mass trait., R.D. Little, J.P. Carulli, R.G. Del Mastro, J. Dupuis, M. Osborne,
        C. Folz et al., Am. J. Hum. Genet. 70 (2002), pp. 11–19.
    16. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent
        embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor., M. Kato,
        M.S. Patel, R. Levasseur, I. Lobov, B.H. Chang, D.A. Glass, II et al, J.
        Cell Biol. 157 (2002), pp. 303–314.
    17. Wnt-3a promotes proliferation and suppresses osteogenic differentiation of adult human
        mesenchymal stem cells, Boland GM, Perkins G, Hall DJ, Tuan RS, (2004), J Cell
        Biochem 93:1210-30.
    18. Canonical and Non –Canonical Wnts differentially affect the development potential of
        primary isolate of human bone marrow mesenchymal stem cells, Baksh D., Tuan RS.,
        (2007), J Cell Physiol 212:817-26.
    19. Noncanonical Wnt-4 signaling enhances bone regeneration of mesenchymal stem cells in
        craniofacial defects through activation of p38 MAPK., Chang J, Sonoyama W, Wang
        Z, Jin Q, Zhang C, Krebsbach PH, Giannobile W, Shi S, Wang CY, J Biol Chem. 2007
        Oct 19;282(42):30938-48. Epub 2007 Aug 24.
    20. Activation of p38 mitogen-activated protein kinase and c-Jun-NH2-terminal kinase by
        BMP-2 and their implication in the stimulation of osteoblastic cell differentiation,
        Guicheux J, Lemonnier J, Ghayor C, Suzuki A, Palmer G, Caverzasio J, J Bone Miner
        Res. 2003 Nov;18(11):2060-8.
33 | P a g e


    21. Mechanisms of Wnt signaling in development, Andreas Wodarz, Roel Nusse, Annu. Rev.
        Cell Dev. Biol. 1998. 14:59-88
    22. Cabrera CV, Alonso MC, Johnston P, Phillips RG, Lawrence PA. 1987. Phenocopies
        induced with antisense RNA identify the wingless gene. Cell 50:659–63
    23. Rijsewijk F, Schuermann M, Wagenaar E, Parren P, Weigel D, Nusse R. 1987. The
        Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment
        polarity gene wingless. Cell 50:649–57.
    24. McMahon AP, Bradley A. 1990. The Wnt-1 (int-1) proto-oncogene is required for
        development of a large region of the mouse brain. Cell 62:1073–85
    25. Nusse R, Varmus HE. 1982. Many tumors induced by the mouse mammary tumor virus
        contain a provirus integrated in the same region of the host genome. Cell 31:99–109
    26. Nusse R, Varmus HE. 1992. Wnt genes. Cell 69:1073–87
    27. Reya T, Clevers H. (2005). Wnt signalling in stem cells and cancer. Nature 434: 843–85
    28. Nusse R, Varmus HE. Wnt genes. Cell 1992; 69:1073-1087.
    29. Miller JR. The Wnts. Genome Biol 2002; 3:REVIEWS3001.
    30. Mason JO, Kitajewski J, Varmus HE. Mutational analysis of mouse Wnt-1 identifies two
        temperature-sensitive alleles and attributes of Wnt-1 protein essential for transformation
        of a mammary cell line. Mol Biol Cell 1992; 3:521-533.
    31. Reichsman F, Smith L, Cumberledge S. Glycosaminoglycans can modulate extracellular
        localization of the wingless protein and promote signal transduction. J Cell Biol 1996;
        135:819-827.
    32. Tanaka K, Kitagawa Y, Kadowaki T. Drosophila segment polarity gene product
        porcupine stimulates the posttranslational N-glycosylation of wingless in the endoplasmic
        reticulum. J Biol Chem 2002; 277:12816-12823.
    33. Willert K, Brown JD, Danenberg E, et al. Wnt proteins are lipidmodified and can act as
        stem cell growth factors. Nature 2003; 423:448-452
    34. Takada R, Satomi Y, Kurata T, et al. Monounsaturated fatty acid modification of Wnt
        protein: its role in Wnt secretion. Dev Cell 2006; 11:791-801
    35. Wnt Protein Family, Benjamin N. R. Cheyette* and Randall T. Moon, Encyclopedia of
        Hormones , Pages 665-674
34 | P a g e


    36. Westendorf JJ, Kahler RA, Schroeder TM (2004): Wnt signaling in osteoblasts and bone
        diseases. Gene 341:19-39.
    37. Ching W, Nusse R. (2006). A dedicated Wnt secretion factor. Cell 125: 432–433.
    38. Panakova D, Sprong H, Marois E, Thiele C, Eaton S. Lipoprotein particles are required
        for Hedgehog and Wingless signalling. Nature 2005; 435:58-65.
    39. Verges M, Luton F, Gruber C, et al. The mammalian retromer regulates transcytosis of
        the polymeric immunoglobulin receptor. Nat Cell Biol 2004; 6:763-769
    40. Sailing with the Wnt: Charting the Wnt processing and secretion route, Magdalena J.
        Lorenowicz and Hendrik C. Korswagen, Experimental Cell Research Volume 315, Issue
        16, 1 October 2009, Pages 2683-2689
    41. Wnts as ligands: processing, secretion and reception, AJ Mikels and R Nusse, Oncogene
        (2006) 25, 7461–746
    42. Hsieh JC, Rattner A, Smallwood PM, Nathans J. Biochemical characterization of Wnt-
        frizzled interactions using a soluble, biologically active vertebrate Wnt protein. Proc Natl
        Acad Sci USA 1999; 96:3546-3551.
    43. Wu CH, Nusse R. Ligand receptor interactions in the Wnt signaling pathway in
        Drosophila. J Biol Chem 2002; 277:41762-41769.
    44. Chen W, ten Berge D, Brown J, et al. Dishevelled 2 recruits betaarrestin 2 to mediate
        Wnt5A-stimulated endocytosis of Frizzled, Science 2003; 301:1391-1394.
    45. He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in
        Wnt/beta-catenin signaling: arrows point the way. Development 2004; 131:1663-1677.
    46. Yoda A, Oishi I, Minami Y. Expression and function of the Rorfamily receptor tyrosine
        kinases during development: lessons from genetic analyses of nematodes, mice, and
        humans. J Recept Signal Transduct Res 2003; 23:1-15.
    47. Kroiher M, Miller MA, Steele RE. Deceiving appearances: signaling by ―dead‖ and
        ―fractured‖ receptor protein-tyrosine kinases. Bioessays 2001; 23:69-76.
    48. Behrens et al., 1996 J. Behrens, J.P. von Kries, M. Kuhl, L. Bruhn, D. Wedlich, R.
        Grosschedl and W. Birchmeier, Functional interaction of beta-catenin with the
        transcription factor LEF-1, Nature 382 (1996), pp. 638–642.
    49. Wnt signaling in osteoblasts and bone diseases, Jennifer J. Westendorf, Rachel A. Kahler
        and Tania M. Schroeder, Gene Volume 341, 27 October 2004, Pages 19-39.
35 | P a g e


    50. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two
        coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews
        Volume 62, Issue 12, 30 September 2010, Pages 1149-1155.
    51. T. Saneyoshi, S. Kume, Y. Amasaki and K. Mikoshiba, The Wnt/calcium pathway
        activates NF-AT and promotes ventral cell fate in Xenopus embryos,Nature 417 (2002),
        pp. 295–299
    52. H.Y. Wang and C.C. Malbon, Wnt signaling, Ca2+, and cyclic GMP: visualizing
        Frizzled functions, Science 300 (2003), pp. 1529–1530.
    53. A.D. Kohn and R.T. Moon, Wnt and calcium signaling: beta-catenin-independent
        pathways, Cell Calcium 38 (2005), pp. 439–446
    54. Wnt signaling pathway and lung disease, Michelle Van Scoyk, Jessica Randall, Amen
        Sergew, Lisa M. Williams, Meredith Tennis and Robert A. Winn, Translational Research
        Volume 151, Issue 4, April 2008, Pages 175-180
    55. Wnt Signaling:Multiple Pathways,Multiple Receptors, andMultiple Transcription
        Factors, Michael D. Gordon and Roel Nusse, THE JOURNAL OF BIOLOGICAL
        CHEMISTRY VOL. 281, NO. 32, pp. 22429–22433, August 11, 2006.
    56. Wnt signaling and skeletal development, Fei Liu, Sean Kohlmeier and Cun-Yu Wang,
        Cellular Signalling Volume 20, Issue 6, June 2008, Pages 999-1009
    57. Kremen proteins are Dickkopf receptors that regulate Wnt/ß-catenin signaling, B. Mao,
        W. Wu, G. Davidson, J. Marhold, M. Li, B.M. Mechler, H. Delius, D. Hoppe, P. Stannek,
        C. Walter, A. Glinka and C. Niehrs, Nature 417 (6889) (2002), p. 664
    58. Chibby, a nuclear β-catenin-associated antagonist of the Wnt/Wingless pathway, K.
        Takemaru, S. Yamaguchi,         Y.S.    Lee, Y.       Zhang, R.W. Carthew and R.T.
        Moon, Nature 422 (6934) (2003), p. 905.
    59. H. Akiyama, J.P. Lyons, Y. Mori-Akiyama, X. Yang, R. Zhang, Z. Zhang, J.M. Deng,
        M.M.    Taketo,   T.   Nakamura,       R.R.   Behringer,     P.D.      McCrea     and   B.   de
        Crombrugghe, Interactions     between     Sox9    and      β-catenin    control    chondrocyte
        differentiation, Genes Dev. 18 (9) (2004), p. 1072.
    60. Sequential roles of Hedgehog and Wnt signaling in osteoblast development, H. Hu, M.J.
        Hilton, X. Tu, K. Yu, D.M. Ornitz and F. Long, Development 132 (1) (2005), p. 49.
36 | P a g e


    61. Wnt4 action in gonadal development and sex determination, Pascal Bernard and Vincent
        R. Harley, The International Journal of Biochemistry & Cell Biology Volume 39, Issue 1,
        2007, Pages 31-43
    62. Wnt-4 signaling is involved in the control of smooth muscle cell fate via Bmp-4 in the
        medullary stroma of the developing kidney, Petri Itäranta, Lijun Chi, Tiina Seppänen,
        Mikael Niku, Juha Tuukkanen, Hellevi Peltoketo and Seppo Vainio, Developmental
        BiologyVolume 293, Issue 2, 15 May 2006, Pages 473-483
    63. Stark et al., 1994 K. Stark, S. Vainio, G. Vassileva and A.P. McMahon, Epithelial
        transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4
        Nature 372 (1994), pp. 679–683.
    64. Wnt4 is expressed in human fetal and adult ovaries and its signaling contributes to
        ovarian cell survival, Minna Jääskeläinen, Renata Prunskaite-Hyyryläinen, Florence
        Naillat, Helka Parviainen, Mikko Anttonen, Markku Heikinheimo, Annikki Liakka,
        Roxana Ola, Seppo Vainio, Tommi E. Vaskivuo and Juha S. Tapanainen, Molecular and
        Cellular Endocrinology Volume 317, Issues 1-2, 12 April 2010, Pages 106-111
    65. Characterization and expression of the human WNT4; lack of associated germline
        mutations in high—to moderate—risk breast and ovarian cancer, Hellevi Peltoketo ,
        Minna Allinen, Jaana Vuosku, Sonja Kujala, Tuija Lundan, Annamari Salminen, Robert
        Winqvist and Seppo Vainio, Cancer Letters Volume 213, Issue 1, 15 September 2004,
        Pages 83-90
    66. M. Heikkilä, H. Peltoketo, J. Leppäluoto, M. Ilves, O. Vuolteenaho and S. Vainio, Wnt-4
        deficiency    alters   mouse    adrenal    cortex    function,   reducing      aldosterone
        production. Endocrinology 143 (2002), pp. 4358–4365.
    67. T. Mulroy, J.A. McMahon, S.J. Burakoff, A.P. McMahon and J. Sen, Wnt-1 and Wnt-4
        regulate thymic cellularity. Eur. J. Immunol. 32 (2002), pp. 967–971.
    68. E.L. Huguet, J.A. McMahon, A.P. McMahon, R. Bicknell and A.L. Harris, Differential
        expression of human Wnt genes 2, 3, 4, and 7B in human breast cell lines and normal and
        disease states of human breast tissue. Cancer Res. 54 (1994), pp. 2615–2621.
    69. Tam et al., 1982 C.S. Tam, J.N. Heersche, T.M. Murray and J.A. Parsons, Parathyroid
        hormone stimulates the bone apposition rate independently of its resorptive action:
37 | P a g e


        differential        effects         of          intermittent        and         continuous
        administration, Endocrinology 110 (1982), pp. 506–512.
    70. Wu et al., 2003 X.B. Wu, Y. Li, A. Schneider, W. Yu, G. Rajendren, J. Iqbal, M.
        Yamamoto, M. Alam, L.J. Brunet, H.C. Blair, M. Zaidi and E. Abe, Impaired osteoblastic
        differentiation,   reduced    bone formation,   and    severe   osteoporosis   in   noggin-
        overexpressing mice, J. Clin. Invest. 112 (2003), pp. 924–934.
    71. Kulkarni et al., 2005 N.H. Kulkarni, D.L. Halladay, R.R. Miles, L.M. Gilbert, C.A.
        Frolik, R.J. Galvin, T.J. Martin, M.T. Gillespie and J.E. Onyia, Effects of parathyroid
        hormone on Wnt signaling pathway in bone, J. Cell. Biochem. 95 (2005), pp. 1178–1190
    72. Bergenstock and Partridge, 2007 M.K. Bergenstock and N.C. Partridge, Parathyroid
        hormone stimulation of noncanonical Wnt signaling in bone, Ann. N. Y. Acad.
        Sci. 1116 (2007), pp. 354–359
    73. Is Wnt signalling the final common pathway leading to bone formation?, Frances
        Milat and Kong Wah Ng, Molecular and Cellular Endocrinology Volume 310, Issues 1-2,
        30 October 2009, Pages 52-62
    74. Tobimatsu et al., 2006 T. Tobimatsu, H. Kaji, H. Sowa, J. Naito, L. Canaff, G.N. Hendy,
        T. Sugimoto and K. Chihara, Parathyroid hormone increases beta-catenin levels through
        Smad3 in mouse osteoblastic cells, Endocrinology 147 (2006), pp. 2583–2590
    75. Wan et al., 2008 M. Wan, C. Yang, J. Li, X. Wu, H. Yuan, H. Ma, X. He, S. Nie, C.
        Chang and X. Cao, Parathyroid hormone signaling through low-density lipoprotein-
        related protein 6, Genes Dev. 22 (2008), pp. 2968–2979
    76. Wnt signaling controls the fate of mesenchymal stem cells, Ling Ling, Victor Nurcombe
        and Simon M. Cool, Gene Volume 433, Issues 1-2, 15 March 2009, Pages 1-7
    77. L. Haegele, B. Ingold, H. Naumann, G. Tabatabai, B. Ledermann and S.
        Brandner, Wnt signalling inhibits neural differentiation of embryonic stem cells by
        controlling bone morphogenetic protein expression, Mol cell Neurosci 24 (2003), pp.
        696–708.
    78. Wnt signaling and the regulation of stem cell function, Maurice Kléber and Lukas
        Sommer, Current Opinion in Cell Biology Volume 16, Issue 6, December 2004, Pages
        681-687.
38 | P a g e


    79. M. Ikeya, S.M. Lee, J.E. Johnson, A.P. McMahon and S. Takada, Wnt signalling required
        for expansion of neural crest and CNS progenitors, Nature389 (1997), pp. 966–970.
    80. S.M. Lee, S. Tole, E. Grove and A.P. McMahon, A local Wnt-3a signal is required for
        development of the mammalian hippocampus, Development 127(2000), pp. 457–467.
    81. M. van de Wetering, E. Sancho, C. Verweij, W. de Lau, I. Oving, A. Hurlstone, K. van
        der Horn, E. Batlle, D. Coudreuse and A.P. Haramis et al., The β-catenin/TCF-4 complex
        imposes a crypt progenitor phenotype on colorectal cancer cells, Cell 111 (2002), pp.
        241–250
    82. J. Huelsken, R. Vogel, B. Erdmann, G. Cotsarelis and W. Birchmeier, β-Catenin controls
        hair follicle morphogenesis and stem cell differentiation in the skin, Cell 105 (2001), pp.
        533–545.
    83. Wnt signaling in neuroprotection and stem cell differentiation, Enrique M. Toledo,
        Marcela    Colombres     and   Nibaldo    C.   Inestrosa,   Progress    in   Neurobiology
        Volume 86, Issue 3, November 2008, Pages 281-296.
    84. Y.A. Zeng and R. Nusse, Wnt proteins are self-renewal factors for mammary stem cells
        and promote their long-term expansion in culture, Cell Stem Cell 6 (2010), pp. 568–577
    85. S.L. Etheridge, G.J. Spencer, D.J. Heath and P.G. Genever, Expression profiling and
        functional analysis of wnt signaling mechanisms in mesenchymal stem cells, Stem Cells.
        22 (2004), pp. 849–860
    86. S.H. Baek et al., Regulated subset of G1 growth-control genes in response to
        derepression by the Wnt pathway, Proc. Natl. Acad. Sci. U. S. A. 100 (2003), pp. 3245–
        3250.
    87. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two
        coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews,
        Volume 62, Issue 12, 30 September 2010, Pages 1149-1155.
    88. Wnt signaling and the regulation of stem cell function, Maurice Kléber and Lukas
        Sommer, Current opinion in Cell Biology, Volume 16, Issue 6, December 2004, Pages
        681-687.

More Related Content

What's hot

Micro RNAs and ageing Dr Felekkis 2014 (1)
Micro RNAs and ageing Dr Felekkis 2014 (1)Micro RNAs and ageing Dr Felekkis 2014 (1)
Micro RNAs and ageing Dr Felekkis 2014 (1)Marios Kyriazis
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicineSpringer
 
Molecular characterization of brain and other structures in vertebrates and i...
Molecular characterization of brain and other structures in vertebrates and i...Molecular characterization of brain and other structures in vertebrates and i...
Molecular characterization of brain and other structures in vertebrates and i...M. Luisetto Pharm.D.Spec. Pharmacology
 
Hinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsHinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsAndrew Hinton
 
Notch signaling pathway
Notch signaling pathwayNotch signaling pathway
Notch signaling pathwayAhmedMajid19
 
刊2013Q405-CYTOO專刊(確定版)
刊2013Q405-CYTOO專刊(確定版)刊2013Q405-CYTOO專刊(確定版)
刊2013Q405-CYTOO專刊(確定版)Shelly Shen
 
Selman A. Wakman award for Carol A. Gross
Selman A. Wakman award for Carol A. GrossSelman A. Wakman award for Carol A. Gross
Selman A. Wakman award for Carol A. GrosskokilaM9
 
Knockout mice
Knockout miceKnockout mice
Knockout miceSam Khoo
 
2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generationNatalia Muñoz Roa
 
Signal Transduction and Second Messengers
Signal Transduction and Second MessengersSignal Transduction and Second Messengers
Signal Transduction and Second MessengersSaranraj P
 
Comparative genomics
Comparative genomicsComparative genomics
Comparative genomicsprateek kumar
 
SINGH K.P. and BRILL J.A. 2014
SINGH K.P. and BRILL J.A. 2014SINGH K.P. and BRILL J.A. 2014
SINGH K.P. and BRILL J.A. 2014Kevin Singh
 
Mitochondrial genetics and management
Mitochondrial genetics and managementMitochondrial genetics and management
Mitochondrial genetics and managementEmmanuel Aguon
 
Seminario sara y Juan D
Seminario sara y Juan DSeminario sara y Juan D
Seminario sara y Juan DSaraGmez58
 
Ernie ppt
Ernie pptErnie ppt
Ernie pptcbsua
 

What's hot (20)

Micro RNAs and ageing Dr Felekkis 2014 (1)
Micro RNAs and ageing Dr Felekkis 2014 (1)Micro RNAs and ageing Dr Felekkis 2014 (1)
Micro RNAs and ageing Dr Felekkis 2014 (1)
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
 
Molecular characterization of brain and other structures in vertebrates and i...
Molecular characterization of brain and other structures in vertebrates and i...Molecular characterization of brain and other structures in vertebrates and i...
Molecular characterization of brain and other structures in vertebrates and i...
 
Hinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsHinton et al 2012 Adv Genetics
Hinton et al 2012 Adv Genetics
 
Apoptosis
ApoptosisApoptosis
Apoptosis
 
Notch signaling pathway
Notch signaling pathwayNotch signaling pathway
Notch signaling pathway
 
Emt and ecm 2013
Emt and ecm 2013Emt and ecm 2013
Emt and ecm 2013
 
Hedgehog pathway
Hedgehog pathwayHedgehog pathway
Hedgehog pathway
 
刊2013Q405-CYTOO專刊(確定版)
刊2013Q405-CYTOO專刊(確定版)刊2013Q405-CYTOO專刊(確定版)
刊2013Q405-CYTOO專刊(確定版)
 
Selman A. Wakman award for Carol A. Gross
Selman A. Wakman award for Carol A. GrossSelman A. Wakman award for Carol A. Gross
Selman A. Wakman award for Carol A. Gross
 
Knockout mice
Knockout miceKnockout mice
Knockout mice
 
2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation
 
Signal Transduction and Second Messengers
Signal Transduction and Second MessengersSignal Transduction and Second Messengers
Signal Transduction and Second Messengers
 
Comparative genomics
Comparative genomicsComparative genomics
Comparative genomics
 
SINGH K.P. and BRILL J.A. 2014
SINGH K.P. and BRILL J.A. 2014SINGH K.P. and BRILL J.A. 2014
SINGH K.P. and BRILL J.A. 2014
 
Mitochondrial genetics and management
Mitochondrial genetics and managementMitochondrial genetics and management
Mitochondrial genetics and management
 
The absence of mitochondrial ferritin in mice reduces male fertility
The absence of mitochondrial ferritin in mice reduces male fertilityThe absence of mitochondrial ferritin in mice reduces male fertility
The absence of mitochondrial ferritin in mice reduces male fertility
 
Nrneph.2014.170
Nrneph.2014.170Nrneph.2014.170
Nrneph.2014.170
 
Seminario sara y Juan D
Seminario sara y Juan DSeminario sara y Juan D
Seminario sara y Juan D
 
Ernie ppt
Ernie pptErnie ppt
Ernie ppt
 

Viewers also liked

Digitalni fotoaparat canon power shot a40
Digitalni fotoaparat canon power shot a40Digitalni fotoaparat canon power shot a40
Digitalni fotoaparat canon power shot a40sopmceri
 
Host modulation therapy: An indispensable part of perioceutics
Host modulation therapy: An indispensable part of perioceuticsHost modulation therapy: An indispensable part of perioceutics
Host modulation therapy: An indispensable part of perioceuticsMinkle Gulati
 
Collaborative Solutions e-Health event - WWWMachealth
Collaborative Solutions e-Health event - WWWMachealthCollaborative Solutions e-Health event - WWWMachealth
Collaborative Solutions e-Health event - WWWMachealthCollaborative Solutions
 
Collaborative Solutions eHealth Event - Specialist Information Services
Collaborative Solutions eHealth Event - Specialist Information ServicesCollaborative Solutions eHealth Event - Specialist Information Services
Collaborative Solutions eHealth Event - Specialist Information ServicesCollaborative Solutions
 
Collaborative Solutions eHealth Event - iOmniscient
Collaborative Solutions eHealth Event - iOmniscientCollaborative Solutions eHealth Event - iOmniscient
Collaborative Solutions eHealth Event - iOmniscientCollaborative Solutions
 
Worksample1 110305233348-phpapp02 [autosaved]
Worksample1 110305233348-phpapp02 [autosaved]Worksample1 110305233348-phpapp02 [autosaved]
Worksample1 110305233348-phpapp02 [autosaved]Shadid Al
 

Viewers also liked (9)

Tecnoconnect finaldraft
Tecnoconnect finaldraftTecnoconnect finaldraft
Tecnoconnect finaldraft
 
Digitalni fotoaparat canon power shot a40
Digitalni fotoaparat canon power shot a40Digitalni fotoaparat canon power shot a40
Digitalni fotoaparat canon power shot a40
 
Host modulation therapy: An indispensable part of perioceutics
Host modulation therapy: An indispensable part of perioceuticsHost modulation therapy: An indispensable part of perioceutics
Host modulation therapy: An indispensable part of perioceutics
 
Collaborative Solutions e-Health event - WWWMachealth
Collaborative Solutions e-Health event - WWWMachealthCollaborative Solutions e-Health event - WWWMachealth
Collaborative Solutions e-Health event - WWWMachealth
 
DIA DE LA MUJER 2014
DIA DE LA MUJER 2014DIA DE LA MUJER 2014
DIA DE LA MUJER 2014
 
Collaborative Solutions eHealth Event - Specialist Information Services
Collaborative Solutions eHealth Event - Specialist Information ServicesCollaborative Solutions eHealth Event - Specialist Information Services
Collaborative Solutions eHealth Event - Specialist Information Services
 
Collaborative Solutions eHealth Event - iOmniscient
Collaborative Solutions eHealth Event - iOmniscientCollaborative Solutions eHealth Event - iOmniscient
Collaborative Solutions eHealth Event - iOmniscient
 
Proposal print
Proposal printProposal print
Proposal print
 
Worksample1 110305233348-phpapp02 [autosaved]
Worksample1 110305233348-phpapp02 [autosaved]Worksample1 110305233348-phpapp02 [autosaved]
Worksample1 110305233348-phpapp02 [autosaved]
 

Similar to A Review on the role of Wnt-4 in Stem Cell Biology

Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biology
Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biologyPresentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biology
Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biologyGayathri Vijayakumar
 
Sequence And Sequence Of Proteins
Sequence And Sequence Of ProteinsSequence And Sequence Of Proteins
Sequence And Sequence Of ProteinsLindsey Rivera
 
1536709-wnt canonical pathway-greta brezgyte
1536709-wnt canonical pathway-greta brezgyte1536709-wnt canonical pathway-greta brezgyte
1536709-wnt canonical pathway-greta brezgyteGreta Brezgyte
 
Early development of invertebrates
Early development of invertebratesEarly development of invertebrates
Early development of invertebratesMerlyn Denesia
 
Korc Poster Final 11 23 10
Korc Poster Final 11 23 10Korc Poster Final 11 23 10
Korc Poster Final 11 23 10Jack Crawford
 
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...Yara Tarabay
 
Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Jacob Wong
 
Cer cor11 franco-cercor-bhr199
Cer cor11 franco-cercor-bhr199Cer cor11 franco-cercor-bhr199
Cer cor11 franco-cercor-bhr199shiraknafo
 
Transgenic pig (1) (1)
Transgenic pig (1) (1)Transgenic pig (1) (1)
Transgenic pig (1) (1)jain_sonia
 
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences Magazine
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences MagazineCell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences Magazine
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences MagazineThe Lifesciences Magazine
 
tissue engineering
tissue engineering tissue engineering
tissue engineering boris saha
 
Daniel Mejía Arrieta Mutation in DNA Replication
Daniel Mejía Arrieta Mutation in DNA ReplicationDaniel Mejía Arrieta Mutation in DNA Replication
Daniel Mejía Arrieta Mutation in DNA ReplicationDaniel Mejia
 
Symptoms And Treatment Of Trypanosoma Brucei
Symptoms And Treatment Of Trypanosoma BruceiSymptoms And Treatment Of Trypanosoma Brucei
Symptoms And Treatment Of Trypanosoma BruceiJennifer Baker
 
Transgenic pig
Transgenic pig Transgenic pig
Transgenic pig jain_sonia
 

Similar to A Review on the role of Wnt-4 in Stem Cell Biology (20)

WNT 4 gene
WNT 4 geneWNT 4 gene
WNT 4 gene
 
Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biology
Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biologyPresentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biology
Presentation On Wnt 4 and rhe role of HDAC4 & SIRT1 in bone biology
 
Glioma Essay
Glioma EssayGlioma Essay
Glioma Essay
 
Sequence And Sequence Of Proteins
Sequence And Sequence Of ProteinsSequence And Sequence Of Proteins
Sequence And Sequence Of Proteins
 
nikolakopoulou et al., 2006b
nikolakopoulou et al., 2006bnikolakopoulou et al., 2006b
nikolakopoulou et al., 2006b
 
1536709-wnt canonical pathway-greta brezgyte
1536709-wnt canonical pathway-greta brezgyte1536709-wnt canonical pathway-greta brezgyte
1536709-wnt canonical pathway-greta brezgyte
 
mmmmm
mmmmmmmmmm
mmmmm
 
Early development of invertebrates
Early development of invertebratesEarly development of invertebrates
Early development of invertebrates
 
Korc Poster Final 11 23 10
Korc Poster Final 11 23 10Korc Poster Final 11 23 10
Korc Poster Final 11 23 10
 
Schnatwinkel-2008
Schnatwinkel-2008Schnatwinkel-2008
Schnatwinkel-2008
 
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
 
Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013
 
Cer cor11 franco-cercor-bhr199
Cer cor11 franco-cercor-bhr199Cer cor11 franco-cercor-bhr199
Cer cor11 franco-cercor-bhr199
 
Transgenic pig (1) (1)
Transgenic pig (1) (1)Transgenic pig (1) (1)
Transgenic pig (1) (1)
 
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences Magazine
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences MagazineCell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences Magazine
Cell Division: Mitosis, Meiosis & Cytokinesis | The Lifesciences Magazine
 
PROTEINS (Molecular Biology)
PROTEINS (Molecular Biology)PROTEINS (Molecular Biology)
PROTEINS (Molecular Biology)
 
tissue engineering
tissue engineering tissue engineering
tissue engineering
 
Daniel Mejía Arrieta Mutation in DNA Replication
Daniel Mejía Arrieta Mutation in DNA ReplicationDaniel Mejía Arrieta Mutation in DNA Replication
Daniel Mejía Arrieta Mutation in DNA Replication
 
Symptoms And Treatment Of Trypanosoma Brucei
Symptoms And Treatment Of Trypanosoma BruceiSymptoms And Treatment Of Trypanosoma Brucei
Symptoms And Treatment Of Trypanosoma Brucei
 
Transgenic pig
Transgenic pig Transgenic pig
Transgenic pig
 

A Review on the role of Wnt-4 in Stem Cell Biology

  • 1. Wnt -4 Stimulation of Human Bone Marrow Stromal Stem Cells and Mesenchymal Stem Cells of the Maxilla Gayathri Vijayakumar Poly ID# 0408513 Guided Studies- Fall 2010 Nicola C. Partridge, Ph.D. Professor, Chair Basic Science & Craniofacial Biology E-mail: ncp234@nyu.edu Michael D Turner, D.D.S., M.D. Assistant Professor Oral and Maxillofacial Surgery E-mail: mdt4@nyu.edu
  • 2. 2|Page TABLE OF CONTENTS INTTRODUCTION ....................................................................................................................... 3 OVERVIEW ................................................................................................................................... 4 WNT PROTEINS ...................................................................................................................................... 4 WNT SECRETION & EXTRACELLULAR TRANSPORT ..................................................................... 7 WNT RECEPTION.................................................................................................................................... 8 WNT SIGNAL TRANSDUCTION PATHWAYS .................................................................................... 9 REGULATION OF WNT SIGNALING.................................................................................................. 14 WNT SIGNALING IN BONE FORMATION ......................................................................................... 15 WNT-4 ..................................................................................................................................................... 17 PTH & WNT PROTEINS (WNT-4) ........................................................................................................ 20 WNT PROTEINS & STEM CELLS................................................................................................................. 24 ROLE OF WNT SIGNALING IN MESENCHYMAL STEM CELLS ....................................................................... 26 CONCLUSION ............................................................................................................................ 30 REFERENCE ............................................................................................................................... 31
  • 3. 3|Page 1. INTRODUCTION Wnt proteins are palmitoylated and glycosylated ligands that play a well established role in embryonic patterning, cell proliferation and cell determination [1][2], besides playing a significant role in cell cycle arrest, differentiation, apoptosis and tissue homeostasis. As a result, aberrations in the Wnt signaling pathway are associated with birth defects as well as a multitude of diseases, most notably cancer [3]. Wnt proteins can be categorized into two types: the canonical and non-canonical. Wnts have important roles in regulating many aspects of skeletal development, from limb formation to chondrogenesis and osteoblast maturation [4]. In addition to promoting osteoblast maturation, Wnts may play a role in lineage commitment of mesenchymal precursor cells by preventing adipogenesis is the hypothesized default pathway for mesenchymal stem cells that do not receive proper inductive signals to become osteoblasts, chondrocytes, myocytes, or other mesodermal cells [5]. Parathyroid hormone (PTH) regulates calcium homeostasis in addition to stimulating bone formation [6]. Human and experimental animal studies have shown that PTH increases osteoblast activity and number via increased differentiation and survival, increases bone mass and bone strength, and decreases fracture rate [7]. Due to its anabolic actions, PTH is currently the only clinically available anabolic agent used to effectively treat osteoporosis. [8][9]. In vivo microarray analysis has shown that PTH regulates molecules of the Wnt signaling pathway in bone [10]. PTH has been demonstrated to stimulate bone lining osteoblasts to produce non-canonical Wnt-4 in vivo, and exogenous Wnt-4 has been shown to regulate osteoblast differentiation through non-canonical Wnt signaling pathway [11].
  • 4. 4|Page Wnt signaling has been implicated in the control over various types of stem cells and may act as a niche factor to maintain stem cells in a self-renewing state [12]. Recent studies have shown that Wnt-3a stimulates proliferation while inhibiting osteogenic differentiation of hMSCs [17], while non-canonical Wnt-5a enhanced osteogenic differentiation but had no effect on hMSC proliferation [18]. Most significantly, Wnt-4 has been shown to stimulate osteogenesis in hMSCs isolated from craniofacial tissue through a novel p38 non-canonical Wnt signaling pathway that is a known pathway associated with osteogenic differentiation [19][20]. Preliminary findings from Dr. Nicola C. Partridge’s lab at the Department of Basic Science and Craniofacial Biology at NYU-CD have shown that Wnt-4 is stimulated by PTH in vivo [11], and that Wnt-4 enhances proliferation and osteogenesis of mouse bone marrow stromal stem cells (BMSSC) in vitro. From this it is hypothesized that Wnt-4 has an important function in proliferation and differentiation of BMSSC. The specific aims of this study are to: 1) Assess the Wnt-4 effect on proliferation on human bone marrow stromal stem cells 2) Assess the Wnt-4 effect on differentiation of human bone marrow stromal stem cells 3) Assess the Wnt-4 effect on the proliferation and differentiation of human dental pulp stem cells. 2. Overview 2.1 Wnt Protiens Together with the other families of secreted factors such as FGF, TGF-beta, and Hedgehog proteins,Wnt proteins are implicated in a wide variety of biological processes. The first Wnt gene
  • 5. 5|Page mouse Wnt-1, was discovered in 1982 as a proto-oncogene activated by integration of mouse mammary tumor virus in mammary tumors [21]. With the molecular identification of the Drosophila segment polarity gene wingless (wg) as the orthologue of Wnt-1 [22][23] and the phenotypic analysis of Wnt-1 mutations in the mouse [24], it became clear that Wnt genes are important regulators of many developmental decisions [25][26]. Table 1 [21] Phenotypes of Wnt mutations in mouse, Drosophila, and C. elegans Gene Organism Phenotype Organism Phenotype Wnt-1 Mouse Loss of midbrain and cerebellum Wnt-2 Mouse Placental defects Wnt-3A Mouse Lack of caudal somites and tailbud Wnt-4 Mouse Kidney defects Wnt-7A Mouse Ventralization of limbs Wingless Drosophila Segment polarity, limb development, many others Dwnt-2 Drosophila Muscle defects, testis development lin-44 C.elegans Defects in asymmetric cell divisions mom-2 C. elegans Defects in endoderm induction and spindle orientation The Wnts comprise a large family of protein ligands that affect diverse processes such as embryonic induction, generation of cell polarity and the specification of cell fate. In addition to influencing developmental processes, recent studies point to a key role for Wnt signaling during adult homeostasis in the maintenance of stem cell pluripotency [27]. Wnts are defined by amino acid sequence rather than by functional properties [28-29]. As many as 19 mammalian Wnt homologues are known and are expressed in temporal spatial patterns. Shared features of all Wnts include a signal sequence for secretion, several highly charged amino acid residues, and
  • 6. 6|Page many glycosylation sites. Wnt proteins also display a characteristic distribution of 22 cysteine residues. overexpression in tissue culture cells, several different N-linked glycosylated intermediate Wnt protein products are observed in cell lysates [30-32], suggesting that Wnt protein processing and secretion are highly regulated processes. Even though the primary amino acid sequence of Wnts suggests that they should be soluble, secreted Wnt proteins are hydrophobic and are mostly found associated with cell membranes and extracellular matrix (ECM), the reason being that Wnt proteins are lipid modified by the attachment of a palmitate on the first conserved cysteine residue within the protein family and on a serine in the middle of the protein which may be necessary for:  Wnt signaling as well as secretion  May be necessary for their glycosylation.  Might also aid in Wnt transport between cells as glycosylation might increase Wnt interactions with heparin sulfate proteoglycans (HSPGs) present on the surface of Wnt responding cells.  Potentially anchoring Wnt proteins into the membrane for sustained signaling. [33-34]. There are a total of 7 related Wnt genes in the Drosophila genome and 19 in the human genome (some with multiple isoforms) that generally have close orthologues in mice. Orthologous Wnt gene products (proteins with the same function in different species) are often very highly conserved. [35] Based on their ability to induce secondary body axis in Xenopus embryos, the Wnt family of proteins is grouped into two functional classes. The Wnt-1 class (Wnts-1, 2, 3, 3a, 7b, 8, 8b, 10b) is able and Wnt-5a (Wnts-4, 5a, 5b, 6, 7a, 11) is unable to induce a secondary body axis [36].
  • 7. 7|Page 2.2 Wnt Secretion and Extracellular Transport Two recent genetic screens have identified the multipass transmembrane protein Wntless (Wls)/Evenness interrupted (Evi)/Mom-3 as acting in the secretory pathway to promote the release of Wnts from producing cells [37]. A model of Wnt secretion and realease is shown in figure 1.Exogenously derived lipoproteins termed ―argosomes‖ are implicated in moving Wnts and other lipid-modified proteins such as Hedgehogs [38]. A model is proposed wherein palmitoylated proteins associate with lipoprotein particles on the extracellular face of cells. Traffic of Wnt proteins from one cell to the next requires this association [38]. In addition, transcytosis may regulate Wnt movement. It has been proposed that the retromer complex promotes the association of secreted Wnts with other proteins required for ligand transport, such as lipoprotein particles. [39] Figure 1 [40]: Model of Wnt secretion and release. Wnt is lipid modified in the ER and is transported to the Golgi, where it binds to Wls. Next, the Wnt–Wls complex is transported to the plasma membrane and Wnt is released (1).
  • 8. 8|Page The hydrophobic Wnt protein either remains associated with the plasma membrane or binds directly to lipoprotein particles or HSPGs to facilitate spreading and gradient formation (2). After the release of Wnt, Wls is internalized through AP-2/clathrin-mediated endocytosis (3) and is transported back to the TGN through a retromer-dependent trafficking step (4). An alternative possibility is that Wnt is not released at the plasma membrane, but is reinternalized together with Wls (5). Dissociation of Wnt from Wls may take place in endosomes, after which Wnt is released (possibly in association with lipoprotein particles) through a recycling endosomal pathway (6). Also in this scenario, Wls is recycled back to the TGN through a retromer-dependent transport step (7). Wnt may also be reinternalized through a Reggie-1/Flotillin-2 dependent pathway (8), which may lead to the release of a more mobile, micelle-like form of Wnt or to the association of Wnt with lipoprotein particles (6). 2.3 Wnt Reception The seven-pass transmembrane protein Frizzled (Fz) protein was first receptor found to transduce a Wnt signal [41]. . Fz proteins contain a large extracellular domain containing a conserved motif comprised of 10 cysteine residues called the cysteine-rich domain (CRD) that has been shown to bind Figure 2 [21]: Structure of Frizzleds and FRPs. Frizzled to multiple Wnts with high affinity. At the receptors are characterized by an N-terminal signal peptide, a cysteine-rich ligand-binding domain (CRD) followed by a cytoplasmic side, Fzs may interact directly hydrophilic linker,seven transmembrane regions, and a cytoplasmic tail. FRPs are secreted proteins with a CRD similar to Frizzleds. In addition, they contain a region with with the Dishevelled protein, a known similarity to netrins, secreted proteins involved in axon guidance. mediator of Wnt signaling [42-44]. Following Wnt binding, it is thought that Fzs form a co-receptor complex with single-pass transmembrane proteins of the low-density lipoprotein (LDL) family called Lrp5 and -6 to transduce the canonical Wnt signal. Lrp5 and -6 proteins have a relatively small intracellular domain and a large extracellular domain containing several potential protein interaction domains [45].
  • 9. 9|Page There are other proteins with known Wnt-binding domains that can serve as receptors for Wnt ligands. The single-pass tyrosine kinase Ror2, although structurally distinct from Fz receptors, is involved in other forms of Wnt signaling. Another well- characterized Wnt-binding domain is the Wnt inhibitory factor (WIF) module, which is also found in the cell surface atypical receptor tyrosine kinase Figure 3: Different receptors that Wnt proteins can bind to. Ryk [46][47]. http://www.stanford.edu/group/nusselab/cgi- bin/wnt/receptors 2.4 Wnt Signal Transduction Pathways Wnt proteins signal through the canonical and the non-canonical pathways which are composed of three independent signal transduction pathways (the Wnt/β-catenin pathway, the Wnt/Ca+2 pathways, or the Wnt/planar polarity pathway) that are used to regulate the expression of different genes. The Wnt/β-catenin pathway is commonly referred to as the canonical pathway. It promotes cell fate determination, proliferation, and survival by increasing β- catenin levels and altering gene expression through Lef/Tcf transcription factors [48]. The non- canonical Wnt/Ca+2 pathway stimulates heterotrimeric G proteins, increases intracellular calcium levels, decreases cyclic GMP levels, and activates protein kinase C to induce NF- AT and other transcription factors [49]. The non-canonical Wnt/planar polarity pathway activates Rho/Rac GTPases and Jun N-terminal kinase to modulate cytoskeletal organization and gene expression. Distinct Wnt ligands probably act through specific Frizzled (Fzd) receptors to initiate each [49].
  • 10. 10 | P a g e Figure 4: Wnt Signaling Pathways and their Implications The canonical Wnt signaling pathway (see Figure 5) is activated when Wnts interact with Lrp/Fzd receptor complexes as shown in the middle portion of figure 4. Receptor engagement activates an unknown kinase(s) (K) that phosphorylates the cytoplasmic tail of Lrp5/6. These phosphorylated residues (P) serve as docking sites for Axin and the APC, Dsh, β-catenin complex. A GSK3β binding protein (GBP) is also mobilized after receptor ligation and excludes GSK3β from the proximal receptor complex. β-Catenin thereby escapes phosphorylation events that normally promote its ubiquitination (U) by E2 ligases and degradation by the proteosome (right side of this figure). As β-catenin levels rise above those needed to bridge cadherins to the
  • 11. 11 | P a g e actin cytoskeleton (lower left of this figure), some β-catenin molecules travel to the nucleus where they interact with Lef1/Tcf transcription factors to either increase or decrease the expression of specific target genes. β-Catenin displaces nuclear co-repressors (CoR) from Lef1/Tcf to facilitate the expression of genes involved in cell cycle progression (e.g. cyclin D1) and survival (e.g. c-myc). β-Catenin and Lef1/Tcf suppress other genes, such as osteocalcin (OCN) and E-cadherin, through unknown mechanisms, but that may involve interactions with other transcription factors (TF). Soluble antagonists block the canonical Wnt signaling pathway and promote β-catenin degradation via two mechanisms. Figure 5 [49]: The Canonical Wnt Signaling Pathway Soluble frizzled related proteins (Sfrp) bind free Wnt molecules and compete with surface receptors (top right of figure 5). In contrast, Dkks interact with extracellular domains in Lrp5/6
  • 12. 12 | P a g e and recruit them to complexes containing Krm (top left of figure 5). This trimolecular complex is internalized to lysosomes where Lrp5/6 are either degraded or recycled to the surface. Dkk therefore decreases Lrp5/6 cell surface expression to regulate Wnt signaling [49]. Figure 6 [55]: Nuclear activity of ß-catenin Accumulated ß-catenin then translocates to the nucleus, replaces Groucho from TCF, and activates target genes. ß-catenin forms a complex with TCF and the transcription factors Brg1 and CBP. Lgs and Pygo also bind to -catenin, possibly driving its nuclear localization in addition to playing a direct role in transcriptional activation. Negative regulation of signaling is provided by NLK (Nemo-like kinase) which phosphorylates TCF, and ICAT (inhibitor of catenin) and Chibby, which are antagonists of ß-catenin. In addition to TCF, two other DNA-binding proteins have been shown to associate with ß-catenin: Pitx2 and Prop1. In the case of Prop1, ß-catenin can act as a transcriptional activator or repressor of specific genes, depending on the co-factors
  • 13. 13 | P a g e present. The participation of any particular ß-catenin complex in transcriptional regulation is highly cell type-dependent.[55] The non-canonical pathway can be broadly classified into 2 branches based upon phenotypic response; the Planar Cell Polarity (PCP) pathway and the Wnt/ Ca2+ pathway [50]. However some authors have classified the pathways as Wnt/calcium signaling, Wnt/PCP signaling, Wnt/JNK signaling and Wnt/Rho signaling [56]. The PCP pathway is involved in cellular asymmetry, and it is this cellular asymmetry that controls the rigid architectural orientation of epithelial tissues and sensory organs (e.g. inner ear cochlea), as well as the morphology and the migratory processes of mesodermal cells undergoing gastrulation. Activation of PCP signaling occurs basically through the binding of Wnts to Frizzled (Fz) receptors alone, without LRP co- receptor involvement. These signals activate Dishevelled (Dvl), which in turn leads to the activation of the GTPases Rho and Rac. Activated Rac subsequently stimulates JNK activation [50]. The second branch of non-canonical signaling – the Wnt/Ca2+ pathway – is characterized by Wnt-Fzd-induced PLC (phospholipase C) activation and the resultant increase of cytoplasmic Ca2+ levels. These Ca2+ fluxes activate several Ca2+-responsive proteins, such as PKC (protein kinase C) and CaMKII(calcium/calmodulin-dependent kinase II). CaMKII has been shown to activate the transcription factor NFAT, TAK1 (TGF-beta activated kinase), and NLK (Nemo-like kinase, all of which have the net effect of decreasing intracellular cGMP and consequently, antagonizing Wnt/beta-catenin/TCF signaling [51-52]. The Wnt/Ca2+ signaling pathway is involved in regulating cellular adhesion, cytoskeletal rearrangements, and other developmental processes, such as dorsoventral patterning and tissue separation in embryos [53].
  • 14. 14 | P a g e Figure 7 [54]: Non- canonical pathways 2.5 Regulation of Wnt Signaling: Wnt signaling is tightly regulated by members of several families of secreted antagonists 1. Wnt signaling requires interaction with frizzled receptors (Fz) and the presence of a single-pass transmembrane molecule of the Lrp family (Lrp5 or 6) which can be inhibited by members of the secreted frizzled-related protein (Sfrp) family and Wnt inhibitory factor 1 (WIF-1).
  • 15. 15 | P a g e 2. Dkk1 binds to Lrp with high affinity and to another class of transmembrane molecules, the Kremens. Dkk1 promotes the internalization of Lrp and makes it unavailable for Wnt reception by forming a complex with Lrp and Kremen [57]. 3. Lrp coreceptor activity is also inhibited by members of sclerostin (SOST gene product). 4. Chibby is a nuclear antagonist that binds to the C terminus of β-catenin [58]. 5. Another β-catenin binding protein, ICAT, can block the binding of β-catenin to TCF and also can lead to the dissociation of complexes between β-catenin, LEF, and CBP/p300 [56]. 6. TCF can be phosphorylated by the mitogen-activated protein (MAP) kinase-related protein kinase NLK/Nemo. 7. The phosphorylation of TCF/LEF by activated Nemo is thought to diminish the DNA- binding affinity of the β-catenin/TCF/LEF complex, thereby affecting transcriptional regulation of Wnt target genes [56]. 2.6 Wnt Signaling in Bone formation: The Wnt signal transduction pathway has been implicated in bone formation: patients suffering from osteoporosis–pseudoglioma syndrome have an inactivating mutation in the Wnt co- receptor LRP5 [13], whereas an activating LRP5 mutation is associated with high bone mass syndrome [14] [15]. Analysis of LRP5-deficient mice revealed a decreased number of osteoblasts suggesting that Wnt signaling stimulates bone formation at the level of osteoprogenitor proliferation [16].
  • 16. 16 | P a g e Figure 8 [56]: Role of canonical Wnt signaling in skeletal development. Mesenchymal stem cells have the ability to differentiate into chondrocytes or osteoblasts, depending on the environmental cues. Canonical Wnt signaling is regulated to control the lineage progression between chondrocyte and osteoblast. If there is inadequate canonical Wnt signaling, differentiation towards chondrocyte lineage is encouraged. However, the maturation of chondrocytes requires the presence of canonical Wnt signaling. Canonical Wnt signaling is also required for the progression of osteoblast progenitor cells toward osterix positive osteoblasts and then osteocalcin-positive osteoblasts. Canonical Wnt signaling represses osteoclastogenesis by increasing the osteoprotegrin (OPG) expression, therefore the OPG/RANKL ratio. Green plus signs indicate positive effects of Wnt; red circle minus signs indicate inhibitory effects of physiological canonical Wnt signaling The effects of Wnt signaling on chondrogenesis are complex and have been implicated in the regulation of chondrogenic differentiation and hypertrophy [56]. Chondrocyte-specific inactivationof β-catenin using Col2a1-Cre transgene leads to decreased chondrocyte proliferation and delayed hypertrophic chondrocyte differentiation [59]. In another loss-of-function model, Dermo 1-Cre transgene was used to delete the β-catenin gene in the mesenchymal precursors of both chondrocytes and osteoblasts. It was shown that there is a significant delay in chondrocyte maturation in conditional knockout embryos [60]. At the same time Activation of β-cateninin limb and head mesenchyme repressed the expression of Sox9, a factor essential for chondrogenesis, thereby preventing mesenchymal cells from differentiating into skeletal precursors [59]. In the absence of β-catenin, the expression of early osteoblast markers, such as collagen I, osterix, and osteocalcin was greatly diminished [60].
  • 17. 17 | P a g e Figure 9 [49]: Effects of Wnt signaling on osseous cells. The canonical Wnt signaling pathway promotes the proliferation, expansion and survival of pre- and immature osteoblasts. Dkks, Sfrps, and Wif-1 antagonize Wnt signaling in osteoblasts to facilitate death of immature cells, but they may also downregulate the pathway in mature cells to induce terminal differentiation. Wnt-4: Wnt-4 is a signaling factor with multiple roles in organogenesis, a deficiency that leads to abnormal development of the kidney, pituitary gland, female reproductive system, and mammary gland. Wnt4 is conserved throughout vertebrates and was originally studied in non-mammalian vertebrates such as zebrafish, chicken and xenopus [61]. Several studies have tried to gauge the varied functions of this protein, giving the impression that this is indeed a highly conserved protein essential for proper organ formation and development. Some of the findings are as follows: 1. It is essential for nephrogenesis. The Wnt-4 gene is expressed in the assembling nephrons and the medullary stromal cells during kidney development The
  • 18. 18 | P a g e role of Wnt-4 signaling in controlling mesenchyme to epithelium transformation and kidney tubule induction is well established [63]. 2. Wnt-4 signaling plays also a role in the determination of the fate of smooth muscle cells in the medullary stroma of the developing kidney. Smooth muscle α-actin (α-SMA) is markedly reduced in the absence of its signaling [62]. 3. Wnt-4 plays an important role in female sexual development and ovarian function. Wnt-4 deficiency leads disturbed development of the internal genitalia in mouse and human, and to a dramatic reduction of mouse oocytes. The expression of Wnt-4 protein in human fetal ovaries was high during mid-pregnancy, when new follicles are also rapidly being formed. This implies that Wnt-4 may have a substantial role in regulation of the follicle formation in human ovaries [64]. 4. In the reproductive system, Wnt-4 is specifically involved in Müllerian duct formation, sex-specific blood vessel formation, oocyte maintenance and repression of steroidogenesis. Partial XY male-to-female sex-reversal is observed in mice lacking Wnt- 4 as well as in one human patient carrying a Wnt-4 point mutation [61]. 5. During mammalian embryogenesis, Wnt-4 is expressed in the gonads of both sexes before sex determination events take place and is subsequently down-regulated in the male gonad [61]. 6. Wnt4 regulates mammary gland development in response to hormonal changes that take place during pregnancy and may also play a part in mammary gland tumorigenesis. Expression of Wnt4 is inversely correlated with cell proliferation in the mouse mammary C57 mg cell line, suggesting that Wnt4 participates in restricting cell proliferation. On the other hand, the elevated expression of theWNT4 gene in fibroadenomas, and occasionally
  • 19. 19 | P a g e also in malignant tumor tissue, implies that changes in WNT4 signaling are associated with abnormal cell proliferation in human breast cells [68]. 7. Screening of the multiple adult human tissues Figure 10 [65]. WNT4 transcripts in human adult (A) and embryonic (B) tissues. Three mRNA bands, revealed three human WNT4 mRNA bands of 1.5, 1.5, 2.4 and 4.3 kb in size, are seen in the adult tissues and one, 2.4 kb, in the embryonic liver and 2.4, and 4.3 kb in size (Figure 8). The most kidney tissues. The ages of the embryos from 6 to 12 weeks are indicated below the sample rows. The common 1.5 kb transcript was detected in a arrows show the positions of ribosomal RNA. number of tissues: the adrenal gland, placenta, liver, mammary gland, prostate, spinalcord, stomach, thyroid, trach ea, skeletal muscle and small intestine, the signal being strongest in the adrenal gland and placental samples. 8. The involvement of Wnt-4 in normal mammary gland and ovary development suggests that Wnt- 4 germline mutations may be associated with the human cancer predisposition [65]. 9. Wnt-4 is critical for development of the adrenal gland, the zona glomerulosa of which is incomplete at birth in Wnt-4-deficient mice, probably as a result of significantly reduced aldosterone production. In addition, Wnt-4 represses the migration of steroidogenic adrenal precursors into the gonad [66]. In mouse spleen, Wnt1and Wnt-4 signaling regulates differentiation of the thymocytes, the number of which is decreased in compound mutants [67].
  • 20. 20 | P a g e Figure 11 [65]. Mapping of the human WNT4 locus to chromosome region 1p36.12 by prometaphase FISH. (A) Hybridization signals for WNT4 (green signals) and BAC RP11-285H13 (red signals). (B) Chromosomal location of BAC RP11-285H13 to the region 1p36.11a-1p36.11b and BAC RP11-145C4 to the region 1p36.13c- 1p36.13d (C) Hybridization signals for WNT4 (green signals) and BAC RP11-145C4 (red signals). 2.7 PTH and Wnt Proteins (WNT-4) PTH is a single chain polypeptide with 84 amino acids and is the principle regulator of calcium homeostasis in vertebrates. However, although continuous infusion of PTH induces bone loss, intermittent administration of PTH results in bone formation [69]. Binding of PTH and PTHrP to PTH1R activates two signaling pathways in osteoblasts: the PKA pathway which is responsible for the majority of the calciotropic and skeletal actions of PTH, the PKC pathway leading to accumulation of 1, 4, 5-inositol triphosphate and increased intracellular calcium. This pathway has been found to regulate IGF-binding protein-5 [70]. A number of studies suggest that the PTH and Wnt signaling pathways do indeed overlap. Following treatment of osteoblastic cells with N-terminal PTH, an increase in β-catenin that was measured in whole cell lysates by Western blot was seen [71].
  • 21. 21 | P a g e Figure 12: PTH regulation of Wnt-4 Furthermore, PTH increased the level of β-catenin expression in mouse osteoblastic cells (MC3T3-E1) via both PKA and PKC signaling pathways [74]. More recently, PTH was shown to activate β-catenin signaling in osteoblasts in vitro and in vivo by direct recruitment of LRP6 to PTH/PTH1R complex. In vivo studies confirmed that intermittent PTH treatment led to an increase in amount of β catenin in osteoblasts (immunohistochemical analysis with antibody to β-catenin) with a concurrent increase in bone formation in rat [75]. Partridge et al. also demonstrated a link between PTH and Wnt4 expression in bone [72]. In vivo microarray analysis of intermittent and continuous PTH 1–34 showed
  • 22. 22 | P a g e that PTH regulated Wnt4 in bone. PTH was shown to stimulate Wnt4 primarily through the PKA pathway and Wnt4 treatment in osteoblasts induced early expression of bone marker genes and stimulated key canonical Wnt pathway genes. This finding suggested cross-talk between the Wnt signaling cascades [73]. Previous studies at Dr. Partridge’s lab at the Department of Basic Science & Craniofacial Biology at NYU-CD has showed that Wnt-4 is significantly regulated by PTH in vivo after anabolic and catabolic protocols in bone lining osteoblasts. As pointed out earlier Partridge et al. demonstrated that PTH stimulated Wnt-4 expression in cultured osteoblastic cells and that this stimulation is PKA dependent and is a primary response to PTH. It has also been shown that exogenous Wnt-4 enhances bone marker gene expression during osteoblast differentiation by activating the non-canonical Wnt/Ca2+ and Wnt/PCP signaling pathways but does not significantly stimulate the canonical Wnt/ß-catenin pathway. Most significantly, Wnt-4 has been shown to stimulate osteogenesis in hMSCs isolated from craniofacial tissue through a novel p38 non-canonical Wnt signaling pathway that is a known pathway associated with osteogenic differentiation [19][20]. Therefore it can be hypothesized that non- canonical Wnt signaling plays a significant role in bone and that Wnt-4 may be an important molecule in PTH’s anabolic effect.[72] Real time TR-PCR results have shown that PTH stimulates Wnt-4 mRNA expression in all phases of osteoblastic differentiation but is greatest in the mineralization phase and maximal stimulation occurs 8h after PTH treatment. Partridge et al. has demonstrated that non- canonical Wnt-4 does not increase pre-osteoblastic proliferation or the expression of bone marker genes at proliferation day 7, but enhances bone marker gene expression (runx2, osterix, osteocalcin, alkaline phosphatase, MMP-13) significantly in the absence of
  • 23. 23 | P a g e differentiation-promoting factors such as ascorbate. Continuous treatment of rat primary osteobalsts with Wnt-4 in an osteogenic environment showed that there was increased osteocalcin mRNA and alkaline phosphatase expression in late stage differentiation with increased mineralized nodules. In addition there was increased relative expression of runx2 and osterix mRNA during osteoblast differentiation. It was also found that Wnt-4 acute treatment in the early stages of primary cell differentiation was much more effective at stimulating the relative gene expression of runx2, osterix, osteocalcin, alkaline phosphatase, and collagen-1a mRNA expression. Together these suggest that Wnt-4 may promote the differentiation of osteoblasts as well as uncommitted cells in the bone environment as part of PTH’s anabolic effect.[72] Figure 13: Effect of WNT-4 on osteoblast proliferation & differentiation
  • 24. 24 | P a g e 2.8 Wnt Proteins and Stem Cells The Wnt Signaling pathway has been found to play a very important role in maintaining the potency of stem cells and stem cell fate determination. Stem cells from different locations interpret Wnt in different ways which reflects an activation of distinct genetic programs in response to the same signal. In addition, the time point during development at which a stem cell is challenged by Wnt signals determines whether the cell responds by self-renewal or differentiation. Besides the cell-intrinsic cues that influence the biological activity of Wnt in distinct stem and progenitor cell types, the same type of stem cell might respond in different ways to Wnts, depending on its extracellular microenvironment (illustrated in Figure 14 and 15) [88]. Figure 14. [88] Cell-intrinsic differences among stem cells Figure 15. [88] The effect of canonical Wnt on a particular influence the biological function of Wnts. (a) Different stem cell type of stem cell is context-dependent. (a) In a types differentially respond to canonical Wnt signaling and microenvironment X, Wnt activity is modulated by the undergo either self-renewal or lineage commitment. The factor(s) X. In this context, Wnt signaling elicits self- differential responsiveness of stem cells is presumably due to renewal. (b) However, the very same stem cell in distinct cell-intrinsic determinants (indicated in the figure by microenvironment Y, in which Wnts are modulated by the differently colored cells). (b) A stem cell type of a given cell factor(s) Y, responds to Wnt signaling by adopting a specific lineage (indicated in various grades of green) can integrate cell fate rather than by self-renewing. Thus, the biological canonical Wnt signaling in different ways, depending on its cell- activity of Wnt in a particular microenvironment is influenced intrinsic properties which change over time. At different stages of by the convergence of Wnt signaling with other signal development, Wnt promotes stem cell self-renewal or lineage transduction pathways. commitment, or the cell loses its ability to respond to Wnt.
  • 25. 25 | P a g e Some of the findings regarding the role of Wnt signaling in stem cells are as follows: 1. In embryonic stem cells, over expression of Wnt1 or of stabilized β-catenin results in the inhibition of neural differentiation [77]. 2. Treatment of haematopoietic stem cells with Wnt proteins and sustained expression of β- catenin promotes self-renewal in long-term cultures and increases the reconstitution of haematopoietic lineages in vivo, which could have been mediated by Notch1 and the transcription factor HoxB4. However, conditional ablation of β-catenin in haematopoietic stem cells did not impair haematopoiesis and lymphopoiesis, suggesting that β-catenin is not required for self-renewal and development of haematopoietic stem cells under physiological conditions [78]. 3. Wnts play major roles during central nervous system (CNS) development. Ablation of wnt1 results in severe defects of the midbrain, the cerebellum and the developing spinal cord, while ablation of wnt3A results in a total loss of the hippocampus. The defects observed in Wnt mutants are possibly explained by perturbed proliferation of stem or progenitor cells in the ventricular zone [78] [79] [80]. 4. Wnt/β-catenin signaling is not only essential for the homeostasis of the intestinal epithelium; sustained β-catenin activity has also been implicated in the formation of colon carcinoma [78] [81]. 5. In vivo manipulation of genes encoding Wnt signaling components indicates that ß– catenin deficient stem cells fail to differentiate into follicular keratinocytes and instead adopt an epidermal fate and thus play a very important role in the fate determination process of epidermal stem cells [82].
  • 26. 26 | P a g e 6. Wnt signaling has been implicated in the early stages of neural crest development, such as neural crest induction and melanocyte formation. In neural crest stem cells, the genetic ablation of β-catenin results in lack of melanocytes and sensory neural cells in dorsal root ganglia. In fact, NCSCs lacking β-catenin emigrate and proliferate normally but are unable to acquire a sensory neuronal fate. Constitutive expression of β-catenin in neural stem/progenitor cells results in the expansion of the entire neural tube, supporting a role of β-catenin in progenitor proliferation [83]. 7. In a recent study from Nusse's laboratory, Axin2 gain-of-function mice were used to demonstrate that Wnt3A functions as a rate-limiting, self-renewal factor to clonally expand mammary stem cells (MaSC) [84]. 2.9 Role of Wnt Signaling in Mesenchymal Stem Cells: Mesenchymal stem cells (MSCs), otherwise termed as mesenchymal progenitor cells or marrow stromal cells are adherent, fibroblast-like population with the potential for extensive self-renewal and multilineage differentiation. Under appropriate culture conditions, MSCs are capable of giving rise to osteoblasts, adipocytes, chondrocytes and myoblasts. Their multipotency, ease of isolation and ready availability make MSCs particularly suited for tissue engineering and gene therapy applications [76]. MSCs express a number of Wnt ligands such as Wnt-2, Wnt-4, Wnt-5a, Wnt-11 and several Wnt receptors as well as various co-receptors and Wnt inhibitors [85]. Exogenous application of Wnt- 3a to cell cultures expands the multi-potential population of MSCs and this proliferative is presumably achieved by the up-regulation of cyclin D1 and c-myc both of which drive cell cycle progression to promote growth [17] [86].
  • 27. 27 | P a g e Figure 16: Wnt-4 signaling in the bone as part of PTH’s anabolic effect
  • 28. 28 | P a g e In previous studies done in Dr. Partridge’s lab by Bergenstock et al., it was found that Wnt-4 increased cell proliferation and CFU-F numbers by inhibiting mouse BMSSC apoptosis and stimulating their rate of growth. mBMSSCs treated with Wnt-4 in osteogenic and non- osteogenic cultures showed a significant increase in osteocalcin bone marker gene expression and a significant reduction of adipocyte marker genes (ap2, pparϒ and C/EBPα). It was found that Wnt-4 stimulated the Wnt/ß-catenin pathway in proliferating mBMSSCs after Wnt-4 treatment. Wnt-4 did not alter ß-catenin expression but stimulated the phosphorylation of CamKII (non-canonical Wnt/Ca2+) in differentiating mBMSSCs while inhibiting JNK phophorylation (non-canonical Wnt/PCP) in proliferating mBMSSCs. This suggests the divergent effect of Wnt-4 depending on the developmental stage of the cell. Since Wnt-4 is regulated by PTH in bone, this suggests two functions for Wnt-4 in the stem cell environment as part of PTH’s anabolic effects. The first being to act on stem cells and still uncommitted osteoprogenitor cells to expand cell numbers which would eventually be released into osteogenic differentiation. The mechanism of signaling switch, that determines whether canonical or non-canonical Wnts increase stem cell proliferation or stimulate differentiation in certain tissues is not well understood. Now the question arises how Wnt-4 stimulates the canonical and non-canonical pathways. One paper suggests that the balance and coordination between nuclear/transcriptionally active beta-catenin and cytoplasmic/cytoskeletal beta-catenin couples canonical and non-canonical Wnt signaling. Another suggestion is that the coactivators CBP and p300, part of the Wnt signaling network of proteins, plays the integrator role [87].
  • 29. 29 | P a g e CREB binding protein (CBP) and p300 are key regulators of RNA polymeraseII-mediated transcription that encode highly related protein accetlytransferases that bind a variety of transcriptional regulators and other proteins. As pointed out earlier, research done in this lab has pointed out that continuous treatment of mBMSSCs with Wnt-4 activates Wnt/beta- catenin pathway in the proliferation stage in a significant manner and then the Wnt/Ca2+ pathway is activated in differentiation, but Figure 17 [87]: A. model of coactivator usage. Antagonizing the CBP/beta-catenin interaction leads to the downregulation of genes beta-catenin continues to be produced albeit that are critical for stem cell/progenitor cell maintenance and proliferation (left arm of pathway). This also pushes the cell to not in a significant manner as in the utilize p300 as its coactivator. The switch to p300/beta-catenin- mediated transcription is the first critical step to initiate a proliferation stage. Perhaps the co-activators differentiative program. B. a subset of the gene expression cassette that is regulated by the CBP/beta-catenin arm is critical for the CBP and p300 along with nuclear beta-catenin maintenance of potency and proliferation (e.g. Oct4, survivin, etc.). Other genes that are similarly regulated by CBP/beta-catenin (e.g. plays a role in making the switch from hNkd and axin2) are involved in the negative feedback of this arm of the pathway, and initiate differentiation via a switch to the proliferation to differentiation. p300/beta-catenin arm. A recent paper has described that that CBP/beta-catenin mediated transcription is critical for stem cell/ progenitor cell maintenance and proliferation, whereas a switch to p300/beta- catenin mediated transcription is the initial critical step to initiate differentiation and a decrease in cellular potency. A subset of the gene expression cassette (e.g. Oct4, surviving, etc.) is critical for the maintenance of potency and proliferation, other genes such as hNkd and axin2 that are regulated in this manner are negative regulators that stop proliferation, exit cell cycle and initiate
  • 30. 30 | P a g e the process of differentiation [87]. The shift from the canonical to the non-canonical pathway relies on the activation of the PKC pathway. PKC phosphorylation of Ser89 of p300 increases the affinity of p300 for beta-catenin both in vivo an in vitro and thus the switch occurs and the differentiation pathway is initiated (shown in Figure 17) [87]. If this is true in the case of Wnt-4 is still to be investigated. 3. Conclusion: Wnt-4 is significantly regulated by PTH as part of its anabolic effect in the bone environment. Preliminary studies have revealed that Wnt-4 expression is stimulated in osteoblasts in a PKA dependent manner and it is a primary response to PTH. It has been found to promote osteoblast differentiation by enhancing bone marker gene expression by activating the non-canonical pathways. In mBMSSCs, Wnt-4 has been found to stimulate proliferation by activating the canonical pathway and then induce differentiation along the osteoblast lineage by stimulating the non- canonical pathway. Next we are going to assess if the Wnt-4 can induce proliferation and differentiation along the osteoblast lineage in human BMSSCs and human dental pulp stem cells (hDPSCs).
  • 31. 31 | P a g e 4. REFERENCE: 1. Roles of Wnt signaling in bone formation and resorption, Yasuhiro Kobayashi, Kazuhiro Maeda and Naoyuki Takahashi, Japanese Dental Science Review Volume 44, Issue 1, July 2008, Pages 76-82 2. Roles of Wnt proteins in neural development and maintenance, Ardem Patapoutian and Louis F Reichardt, Current Opinion in Neurobiology Volume 10, Issue 3, 1 June 2000, Pages 392-399 3. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews Volume 62, Issue 12, 30 September 2010, Pages 1149-1155, Stem Cell Gene Manipulation and Delivery as Systemic Therapeutics 4. Wnt signaling in osteoblasts and bone diseases, Jennifer J. Westendorf, Rachel A. Kahler and Tania M. Schroeder, Gene Volume 341, 27 October 2004, Pages 19-39 5. Inhibition of Adipogenesis by Wnt Signaling, Sarah E. Ross, Nahid Hemati, Kenneth A. Longo, Christina N. Bennett, Peter C. Lucas, Robin L. Erickson and Ormond A. MacDougald, Science 11 August 2000: Vol. 289 no. 5481 pp. 950-953 6. Anabolic actions of parathyroid hormone on bone, Dempster DW, Cosman F., Parisien M., Shen V., Lindsay r., Endocr Rev 14:690-709 (1993). 7. In vivo studies of anabolic action of PTH and Wnt signaling pathway in bone, Yanfei L. Ma, Nalini H. Kulkarni, Qing Qiang Zeng, Tao Wei, David L. Halladay, Masahiko Sato, Henry U. Bryant and Jude E. Onyia, Bone Volume 43, Supplement 1, October 2008, Page S37, International Conference on Osteoporosis and Bone Research 2008, International Conference on Osteoporosis and Bone Research 2008 8. Cosman F (2005a): Anabolic therapy for osteoporosis: parathyroid hormone. Curr Osteoporos Rep 3: 143-9 9. Cosman F (2005b): The prevention and treatment of osteoporosis: a review. MedGenMed 7:73. 10. Determination of dual effects of parathyroid hormone on skeletal gene expression in vivo y microarray and network analysis, Li X, Liu H, Qin L, Tamasi J, Bergenstock M, Shapses S, Feyen JH, Notterman DA, Partridge NC, J Biol Chem, (2007), 282:33086-97.
  • 32. 32 | P a g e 11. Parathyroid hormone stimulation of noncanonical Wnt signaling in bone, Bergenstock MK, Partridge NC, Ann N Y Acad Sci. 2007 Nov;1116:354-9. 12. Wnt signaling and stem cell control, Roel Nusse, Cell Research (2008) 18:523-527 13. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development, Y. Gong, R.B. Slee, N. Fukai, G. Rawadi, S. Roman-Roman, A.M. Reginato et al.,Cell, 107 (2001), pp. 513–523. 14. High bone density due to a mutation in LDL-receptor-related protein 5., L.M. Boyden, J. Mao, J. Belsky, L. Mitzner, A. Farhi, M.A. Mitnick et al., N. Engl. J. Med.346 (2002), pp. 1513–1521. 15. A mutation in the LDL receptor-related protein 5 gene results in the autosomal dominant high-bone-mass trait., R.D. Little, J.P. Carulli, R.G. Del Mastro, J. Dupuis, M. Osborne, C. Folz et al., Am. J. Hum. Genet. 70 (2002), pp. 11–19. 16. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor., M. Kato, M.S. Patel, R. Levasseur, I. Lobov, B.H. Chang, D.A. Glass, II et al, J. Cell Biol. 157 (2002), pp. 303–314. 17. Wnt-3a promotes proliferation and suppresses osteogenic differentiation of adult human mesenchymal stem cells, Boland GM, Perkins G, Hall DJ, Tuan RS, (2004), J Cell Biochem 93:1210-30. 18. Canonical and Non –Canonical Wnts differentially affect the development potential of primary isolate of human bone marrow mesenchymal stem cells, Baksh D., Tuan RS., (2007), J Cell Physiol 212:817-26. 19. Noncanonical Wnt-4 signaling enhances bone regeneration of mesenchymal stem cells in craniofacial defects through activation of p38 MAPK., Chang J, Sonoyama W, Wang Z, Jin Q, Zhang C, Krebsbach PH, Giannobile W, Shi S, Wang CY, J Biol Chem. 2007 Oct 19;282(42):30938-48. Epub 2007 Aug 24. 20. Activation of p38 mitogen-activated protein kinase and c-Jun-NH2-terminal kinase by BMP-2 and their implication in the stimulation of osteoblastic cell differentiation, Guicheux J, Lemonnier J, Ghayor C, Suzuki A, Palmer G, Caverzasio J, J Bone Miner Res. 2003 Nov;18(11):2060-8.
  • 33. 33 | P a g e 21. Mechanisms of Wnt signaling in development, Andreas Wodarz, Roel Nusse, Annu. Rev. Cell Dev. Biol. 1998. 14:59-88 22. Cabrera CV, Alonso MC, Johnston P, Phillips RG, Lawrence PA. 1987. Phenocopies induced with antisense RNA identify the wingless gene. Cell 50:659–63 23. Rijsewijk F, Schuermann M, Wagenaar E, Parren P, Weigel D, Nusse R. 1987. The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell 50:649–57. 24. McMahon AP, Bradley A. 1990. The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain. Cell 62:1073–85 25. Nusse R, Varmus HE. 1982. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 31:99–109 26. Nusse R, Varmus HE. 1992. Wnt genes. Cell 69:1073–87 27. Reya T, Clevers H. (2005). Wnt signalling in stem cells and cancer. Nature 434: 843–85 28. Nusse R, Varmus HE. Wnt genes. Cell 1992; 69:1073-1087. 29. Miller JR. The Wnts. Genome Biol 2002; 3:REVIEWS3001. 30. Mason JO, Kitajewski J, Varmus HE. Mutational analysis of mouse Wnt-1 identifies two temperature-sensitive alleles and attributes of Wnt-1 protein essential for transformation of a mammary cell line. Mol Biol Cell 1992; 3:521-533. 31. Reichsman F, Smith L, Cumberledge S. Glycosaminoglycans can modulate extracellular localization of the wingless protein and promote signal transduction. J Cell Biol 1996; 135:819-827. 32. Tanaka K, Kitagawa Y, Kadowaki T. Drosophila segment polarity gene product porcupine stimulates the posttranslational N-glycosylation of wingless in the endoplasmic reticulum. J Biol Chem 2002; 277:12816-12823. 33. Willert K, Brown JD, Danenberg E, et al. Wnt proteins are lipidmodified and can act as stem cell growth factors. Nature 2003; 423:448-452 34. Takada R, Satomi Y, Kurata T, et al. Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Dev Cell 2006; 11:791-801 35. Wnt Protein Family, Benjamin N. R. Cheyette* and Randall T. Moon, Encyclopedia of Hormones , Pages 665-674
  • 34. 34 | P a g e 36. Westendorf JJ, Kahler RA, Schroeder TM (2004): Wnt signaling in osteoblasts and bone diseases. Gene 341:19-39. 37. Ching W, Nusse R. (2006). A dedicated Wnt secretion factor. Cell 125: 432–433. 38. Panakova D, Sprong H, Marois E, Thiele C, Eaton S. Lipoprotein particles are required for Hedgehog and Wingless signalling. Nature 2005; 435:58-65. 39. Verges M, Luton F, Gruber C, et al. The mammalian retromer regulates transcytosis of the polymeric immunoglobulin receptor. Nat Cell Biol 2004; 6:763-769 40. Sailing with the Wnt: Charting the Wnt processing and secretion route, Magdalena J. Lorenowicz and Hendrik C. Korswagen, Experimental Cell Research Volume 315, Issue 16, 1 October 2009, Pages 2683-2689 41. Wnts as ligands: processing, secretion and reception, AJ Mikels and R Nusse, Oncogene (2006) 25, 7461–746 42. Hsieh JC, Rattner A, Smallwood PM, Nathans J. Biochemical characterization of Wnt- frizzled interactions using a soluble, biologically active vertebrate Wnt protein. Proc Natl Acad Sci USA 1999; 96:3546-3551. 43. Wu CH, Nusse R. Ligand receptor interactions in the Wnt signaling pathway in Drosophila. J Biol Chem 2002; 277:41762-41769. 44. Chen W, ten Berge D, Brown J, et al. Dishevelled 2 recruits betaarrestin 2 to mediate Wnt5A-stimulated endocytosis of Frizzled, Science 2003; 301:1391-1394. 45. He X, Semenov M, Tamai K, Zeng X. LDL receptor-related proteins 5 and 6 in Wnt/beta-catenin signaling: arrows point the way. Development 2004; 131:1663-1677. 46. Yoda A, Oishi I, Minami Y. Expression and function of the Rorfamily receptor tyrosine kinases during development: lessons from genetic analyses of nematodes, mice, and humans. J Recept Signal Transduct Res 2003; 23:1-15. 47. Kroiher M, Miller MA, Steele RE. Deceiving appearances: signaling by ―dead‖ and ―fractured‖ receptor protein-tyrosine kinases. Bioessays 2001; 23:69-76. 48. Behrens et al., 1996 J. Behrens, J.P. von Kries, M. Kuhl, L. Bruhn, D. Wedlich, R. Grosschedl and W. Birchmeier, Functional interaction of beta-catenin with the transcription factor LEF-1, Nature 382 (1996), pp. 638–642. 49. Wnt signaling in osteoblasts and bone diseases, Jennifer J. Westendorf, Rachel A. Kahler and Tania M. Schroeder, Gene Volume 341, 27 October 2004, Pages 19-39.
  • 35. 35 | P a g e 50. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews Volume 62, Issue 12, 30 September 2010, Pages 1149-1155. 51. T. Saneyoshi, S. Kume, Y. Amasaki and K. Mikoshiba, The Wnt/calcium pathway activates NF-AT and promotes ventral cell fate in Xenopus embryos,Nature 417 (2002), pp. 295–299 52. H.Y. Wang and C.C. Malbon, Wnt signaling, Ca2+, and cyclic GMP: visualizing Frizzled functions, Science 300 (2003), pp. 1529–1530. 53. A.D. Kohn and R.T. Moon, Wnt and calcium signaling: beta-catenin-independent pathways, Cell Calcium 38 (2005), pp. 439–446 54. Wnt signaling pathway and lung disease, Michelle Van Scoyk, Jessica Randall, Amen Sergew, Lisa M. Williams, Meredith Tennis and Robert A. Winn, Translational Research Volume 151, Issue 4, April 2008, Pages 175-180 55. Wnt Signaling:Multiple Pathways,Multiple Receptors, andMultiple Transcription Factors, Michael D. Gordon and Roel Nusse, THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 32, pp. 22429–22433, August 11, 2006. 56. Wnt signaling and skeletal development, Fei Liu, Sean Kohlmeier and Cun-Yu Wang, Cellular Signalling Volume 20, Issue 6, June 2008, Pages 999-1009 57. Kremen proteins are Dickkopf receptors that regulate Wnt/ß-catenin signaling, B. Mao, W. Wu, G. Davidson, J. Marhold, M. Li, B.M. Mechler, H. Delius, D. Hoppe, P. Stannek, C. Walter, A. Glinka and C. Niehrs, Nature 417 (6889) (2002), p. 664 58. Chibby, a nuclear β-catenin-associated antagonist of the Wnt/Wingless pathway, K. Takemaru, S. Yamaguchi, Y.S. Lee, Y. Zhang, R.W. Carthew and R.T. Moon, Nature 422 (6934) (2003), p. 905. 59. H. Akiyama, J.P. Lyons, Y. Mori-Akiyama, X. Yang, R. Zhang, Z. Zhang, J.M. Deng, M.M. Taketo, T. Nakamura, R.R. Behringer, P.D. McCrea and B. de Crombrugghe, Interactions between Sox9 and β-catenin control chondrocyte differentiation, Genes Dev. 18 (9) (2004), p. 1072. 60. Sequential roles of Hedgehog and Wnt signaling in osteoblast development, H. Hu, M.J. Hilton, X. Tu, K. Yu, D.M. Ornitz and F. Long, Development 132 (1) (2005), p. 49.
  • 36. 36 | P a g e 61. Wnt4 action in gonadal development and sex determination, Pascal Bernard and Vincent R. Harley, The International Journal of Biochemistry & Cell Biology Volume 39, Issue 1, 2007, Pages 31-43 62. Wnt-4 signaling is involved in the control of smooth muscle cell fate via Bmp-4 in the medullary stroma of the developing kidney, Petri Itäranta, Lijun Chi, Tiina Seppänen, Mikael Niku, Juha Tuukkanen, Hellevi Peltoketo and Seppo Vainio, Developmental BiologyVolume 293, Issue 2, 15 May 2006, Pages 473-483 63. Stark et al., 1994 K. Stark, S. Vainio, G. Vassileva and A.P. McMahon, Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4 Nature 372 (1994), pp. 679–683. 64. Wnt4 is expressed in human fetal and adult ovaries and its signaling contributes to ovarian cell survival, Minna Jääskeläinen, Renata Prunskaite-Hyyryläinen, Florence Naillat, Helka Parviainen, Mikko Anttonen, Markku Heikinheimo, Annikki Liakka, Roxana Ola, Seppo Vainio, Tommi E. Vaskivuo and Juha S. Tapanainen, Molecular and Cellular Endocrinology Volume 317, Issues 1-2, 12 April 2010, Pages 106-111 65. Characterization and expression of the human WNT4; lack of associated germline mutations in high—to moderate—risk breast and ovarian cancer, Hellevi Peltoketo , Minna Allinen, Jaana Vuosku, Sonja Kujala, Tuija Lundan, Annamari Salminen, Robert Winqvist and Seppo Vainio, Cancer Letters Volume 213, Issue 1, 15 September 2004, Pages 83-90 66. M. Heikkilä, H. Peltoketo, J. Leppäluoto, M. Ilves, O. Vuolteenaho and S. Vainio, Wnt-4 deficiency alters mouse adrenal cortex function, reducing aldosterone production. Endocrinology 143 (2002), pp. 4358–4365. 67. T. Mulroy, J.A. McMahon, S.J. Burakoff, A.P. McMahon and J. Sen, Wnt-1 and Wnt-4 regulate thymic cellularity. Eur. J. Immunol. 32 (2002), pp. 967–971. 68. E.L. Huguet, J.A. McMahon, A.P. McMahon, R. Bicknell and A.L. Harris, Differential expression of human Wnt genes 2, 3, 4, and 7B in human breast cell lines and normal and disease states of human breast tissue. Cancer Res. 54 (1994), pp. 2615–2621. 69. Tam et al., 1982 C.S. Tam, J.N. Heersche, T.M. Murray and J.A. Parsons, Parathyroid hormone stimulates the bone apposition rate independently of its resorptive action:
  • 37. 37 | P a g e differential effects of intermittent and continuous administration, Endocrinology 110 (1982), pp. 506–512. 70. Wu et al., 2003 X.B. Wu, Y. Li, A. Schneider, W. Yu, G. Rajendren, J. Iqbal, M. Yamamoto, M. Alam, L.J. Brunet, H.C. Blair, M. Zaidi and E. Abe, Impaired osteoblastic differentiation, reduced bone formation, and severe osteoporosis in noggin- overexpressing mice, J. Clin. Invest. 112 (2003), pp. 924–934. 71. Kulkarni et al., 2005 N.H. Kulkarni, D.L. Halladay, R.R. Miles, L.M. Gilbert, C.A. Frolik, R.J. Galvin, T.J. Martin, M.T. Gillespie and J.E. Onyia, Effects of parathyroid hormone on Wnt signaling pathway in bone, J. Cell. Biochem. 95 (2005), pp. 1178–1190 72. Bergenstock and Partridge, 2007 M.K. Bergenstock and N.C. Partridge, Parathyroid hormone stimulation of noncanonical Wnt signaling in bone, Ann. N. Y. Acad. Sci. 1116 (2007), pp. 354–359 73. Is Wnt signalling the final common pathway leading to bone formation?, Frances Milat and Kong Wah Ng, Molecular and Cellular Endocrinology Volume 310, Issues 1-2, 30 October 2009, Pages 52-62 74. Tobimatsu et al., 2006 T. Tobimatsu, H. Kaji, H. Sowa, J. Naito, L. Canaff, G.N. Hendy, T. Sugimoto and K. Chihara, Parathyroid hormone increases beta-catenin levels through Smad3 in mouse osteoblastic cells, Endocrinology 147 (2006), pp. 2583–2590 75. Wan et al., 2008 M. Wan, C. Yang, J. Li, X. Wu, H. Yuan, H. Ma, X. He, S. Nie, C. Chang and X. Cao, Parathyroid hormone signaling through low-density lipoprotein- related protein 6, Genes Dev. 22 (2008), pp. 2968–2979 76. Wnt signaling controls the fate of mesenchymal stem cells, Ling Ling, Victor Nurcombe and Simon M. Cool, Gene Volume 433, Issues 1-2, 15 March 2009, Pages 1-7 77. L. Haegele, B. Ingold, H. Naumann, G. Tabatabai, B. Ledermann and S. Brandner, Wnt signalling inhibits neural differentiation of embryonic stem cells by controlling bone morphogenetic protein expression, Mol cell Neurosci 24 (2003), pp. 696–708. 78. Wnt signaling and the regulation of stem cell function, Maurice Kléber and Lukas Sommer, Current Opinion in Cell Biology Volume 16, Issue 6, December 2004, Pages 681-687.
  • 38. 38 | P a g e 79. M. Ikeya, S.M. Lee, J.E. Johnson, A.P. McMahon and S. Takada, Wnt signalling required for expansion of neural crest and CNS progenitors, Nature389 (1997), pp. 966–970. 80. S.M. Lee, S. Tole, E. Grove and A.P. McMahon, A local Wnt-3a signal is required for development of the mammalian hippocampus, Development 127(2000), pp. 457–467. 81. M. van de Wetering, E. Sancho, C. Verweij, W. de Lau, I. Oving, A. Hurlstone, K. van der Horn, E. Batlle, D. Coudreuse and A.P. Haramis et al., The β-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells, Cell 111 (2002), pp. 241–250 82. J. Huelsken, R. Vogel, B. Erdmann, G. Cotsarelis and W. Birchmeier, β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin, Cell 105 (2001), pp. 533–545. 83. Wnt signaling in neuroprotection and stem cell differentiation, Enrique M. Toledo, Marcela Colombres and Nibaldo C. Inestrosa, Progress in Neurobiology Volume 86, Issue 3, November 2008, Pages 281-296. 84. Y.A. Zeng and R. Nusse, Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture, Cell Stem Cell 6 (2010), pp. 568–577 85. S.L. Etheridge, G.J. Spencer, D.J. Heath and P.G. Genever, Expression profiling and functional analysis of wnt signaling mechanisms in mesenchymal stem cells, Stem Cells. 22 (2004), pp. 849–860 86. S.H. Baek et al., Regulated subset of G1 growth-control genes in response to derepression by the Wnt pathway, Proc. Natl. Acad. Sci. U. S. A. 100 (2003), pp. 3245– 3250. 87. The Wnt signaling pathway in cellular proliferation and differentiation: A tale of two coactivators, Jia-Ling Teo and Michael Kahn, Advanced Drug Delivery Reviews, Volume 62, Issue 12, 30 September 2010, Pages 1149-1155. 88. Wnt signaling and the regulation of stem cell function, Maurice Kléber and Lukas Sommer, Current opinion in Cell Biology, Volume 16, Issue 6, December 2004, Pages 681-687.