Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Received: 22 October 2021 | Revised: 10 February 2022 | Accepted: 7 March 2022

DOI: 10.1002/cbin.11797

Cell Biology
REVIEW International

Wnt signaling pathway: A comprehensive review

Rabia Hayat1 | Maleeha Manzoor2 | Ali Hussain3

1
Institute of Evolution and Marine
Biodiversity, Ocean University of China, Abstract
Qingdao, China
Wnt signaling is an evolutionary cell‐to‐cell coordination mechanism and it is highly
2
Department of Zoology, Government College
University, Faisalabad, Pakistan
critical for a variety of physiological processes of an organism's body, including stem
3
Department of Wildlife and Ecology, cell regeneration, proliferation, division, migration, polarity of a cell, determining fate
University of Veterinary and Animal Sciences, of the cell and specification of neural crest, neural symmetry and morphogenesis.
Lahore, Pakistan
Wnts are extracellular secreted glycol proteins, consisted of a family of 19 human
Correspondence proteins that represent the complex nature of the regulatory structure and
Maleeha Manzoor, Department of Zoology,
physiological efficiency of signaling. Moreover, a Wnt/β‐catenin‐dependent path-
Government College University, Faisalabad,
Pakistan. way and the β‐catenin‐independent pathway that is further classified into the Planar
Email: maleehamanzoor@gcuf.edu.pk
Cell Polarity and Wnt/Ca2+ pathways have been established as key signaling nodes
downstream of the frizzled (Fz/Fzd) receptor, and these nodes are extensively
analyzed at biochemical and molecular levels. Genetic and epigenetic activities that
ultimately characterize the pathway and its subsequent responses contribute to
Wnt‐β‐catenin signaling pathway hypo or hyper‐activation and is associated with
the variety of human disorders progression most significantly cancers. Recognizing
how this mechanism operates is crucial to the advancement of cancer prevention
therapies or regenerative medicine methods.

KEYWORDS
cancer, Wnt proteins, Wnt signaling, Wnt/Ca2+, Wnt/β‐catenin‐dependent

1 | INTRODUCTION multiple fields, that is, evolutionary biologists to cellular biologists as


well as from embryologists to challenging biochemists, have
Extensive research work has been dedicated to analyze the signaling contributed important knicks and knacks to crack the biological
pathways and molecular processes that regulate an organism's puzzles of Wnt signaling. And if we developed deeper insights about
growth in the new era of molecular medicine. In the current research, Wnt proteins biological functions, we can more effectively identify
this focus is profoundly intertwined and leads to the idea that and understand the abnormal signaling which ultimately help us to
identifying the processes that govern natural development will raise recognize its role in the etiology of several diseases like structural and
our expectations enormously to deter and manage the pleiotropic metabolic disturbances to various types of tumors (Nusse &
disorders that appear when such pathways go faulty. The Wnt Clevers, 2017, Wiese et al., 2018). The title Wnt has been derived
signaling is among the key pathways about which much work has by combining designations of wingless (Wg) polarity gene of
been put into delineating (Zhan et al., 2017). An image of a signaling Drosophila segment and homolog of vertebrate, integrated or int‐1
cascade that is entirely crucial for the developmental process of all (Holstein, 2012; Logan & Nusse, 2004; Wodarz & Nusse, 1998). From
complex organisms along with the development and repair of an evolutionary point of view, Wnt signaling is highly conserved and
different tissue types has been drawn over 40 years of scientific contributes significantly to embryogenesis, homeostasis and regen-
investigations in various model organisms. While our knowledge of eration of adult tissues (Nusse & Clevers, 2017). It is also crucial for
this complex pathway is indeed very limited, without such initial maintaining genetic stability as well as play major role in determining
efforts, we might not be here as we are currently. Researchers from fate of the cell, differentiation, apoptosis and motility of cells along

Cell Biol Int. 2022;1–15. wileyonlinelibrary.com/journal/cbin © 2022 International Federation for Cell Biology | 1
2 | Cell Biology HAYAT ET AL.

International
with stem cell maintenance (Clevers, 2006). A variety of disorders, Various inhibitors, such as the secreted Fz associated proteins (SFRP)
namely embryonic deformities, degenerative diseases, diabetes, 1‐4 family and Wnt inhibitor factor (WIF) that serve like dissolvable
cancers, and autoimmune diseases are linked with abnormal regula- scavanger of Wnt ligands, also strictly control the Wnt signaling
tion of Wnt signaling (Clevers & Nusse, 2012; Kahn, 2014; Kumar pathways (Fan et al., 2017). Other molecules adhere to LRP 5/6,
et al., 2016; Zhan et al., 2017). including the Dickkopf (DKK) group constituents and sclerostin, and
Wnt signaling is categorized into two pathways. One is the intervene with the downstream activation passage of Wnt. This
β‐catenin dependent pathway that is also regarded as the canonical results in blocking their potential to communicate with Wnt‐Fz
or Wnt/β‐catenin dependent pathway, whereas the second pathway (Nusse & Varmus, 2012, Voronkov & Krauss, 2013). It is believed that
is the β‐catenin‐independent signaling that is also called as non‐ dysregulation of Wnt canonical pathway leads to tumor progression,
canonical pathway. This non‐canonical pathway has two classes chronic inflammation and neurodegenerative disorders, thus natural
+2
including Wnt/planar cell polarity (PCP) and Wnt/calcium (Wnt/Ca ) antagonists are now being studied for health applications (Cui
pathway, which can antagonize Wnt/β‐catenin dependent signaling et al., 2017).
in certain cases (Grumolato et al., 2010). The Wnt/β‐catenin
dependent signaling primarily involves in regulation of cell prolifera-
tion, while polarity and mobility of the cell are regulated by 2 | C O M PO NE N TS O F W NT S I GN A L I N G
Wnt/β‐catenin independent pathway. (Amin & Vincan, 2012;
Gajos‐Michniewicz & Czyz, 2020; Komiya & Habas, 2008). The Wnt signaling type components are diverse and comprise a
Wnts are secretory proteins which are integral for the cell‐to‐cell broad group of secreted proteins for activation of signaling
communication. Many intracellular signal transduction cascades are cascades (evolutionary conserved). 19 Wnt proteins are docu-
activated by the extracellular Wnt signal, the Wnt/β‐catenin‐ mented so far in vertebrates as ligands (Langton et al., 2016).
signaling and β‐catenin‐independent signaling (Baarsma & These proteins interact with more than 15 types of cell membrane
Konigshoff, 2017; Habas & Dawid, 2005). Wnt genes expressed receptors and co‐receptors, namely Fz, LRP5/6 coreceptor (Janda
and translated into Wnts. Wnt proteins are cysteine‐rich glycopro- et al., 2017; Teratani et al., 2018) and RYK receptor tyrosine kinase
teins. Secretion of these proteins by cells into the extracellular matrix and Ror1 and Ror2 (Receptor tyrosine kinase‐like orphan receptors
of near surroundings cause receptor‐mediated cell signaling (Niehrs & 1 and 2) (Foulquier et al., 2018). Among them it took more than a
Acebron, 2012). This protein family consists of approximately 19 decade to describe Fz as Wnt protein expression receptors.
secreted cysteine‐rich glycoproteins. These Wnts are of 350–400 Additionally, numerous downstream signaling mechanisms are
amino acids in length. These Wnt proteins are conserved in different triggered by other potential receptors and their co‐receptors such
species ranging across invertebrates to mammals (Cadigan & as single transmembrane receptor tyrosine kinase, PTK7 (protein
Nusse, 1997; Pai et al., 2017). Binding of Wnt and Fz family receptor tyrosine kinase 7), and GPCRs (G‐protein coupled receptor), and so
involves the cysteine‐rich extracellular N‐terminal domain of Fz. This forth (Farin et al., 2016). Furthermore, interrelated intracellular
domain is a member of such receptor family which is coupled with intermediaries and many types of endogenous blockers including
G‐protein (Pai et al., 2017). Additionally, co‐receptors for modulating DKK and sFRP are also present. Different components of Wnt
Wnt signaling are also necessary for the interface of Wnt and Fz. For signaling are Wnt ligands, Alternative ligands, Extracellular
example, the low‐density lipoprotein‐related protein5/6 (LRP5/6) is modulators, Fzd receptors, LRP receptors, Alternative receptors,
needed for the regulation of the canonical Wnt signaling (Catalano Signaling intermediates, β‐catenin destruction complex, Cellular
et al., 2020; Komiya & Habas, 2008). trafficking and distribution, Effector, Transcription factors and
With the involvement of a broad variety of secretory Wnt Intracellular modulators.
antagonists, one primary layer of Wnt signaling regulation exists in
the extracellular environment. The signal is directed toward
phosphoprotein Disheveled (Dsh/Dvl) in the cytoplasmic matrix after 3 | MECHANISM
coupling of Wnt and receptor complex as documented previously
that Dsh could communicate in a direct way with Fz (Dejana, 2010; 3.1 | Transcription and translocation
Wallingford & Habas, 2005). The Wnt signal splits into at least three
distinct steps at the point of Dsh, including canonical, PCP, and Wnt/ Wnt signaling starts by coupling Wnt protein to Fz family receptor's
Ca2+ cascades. Dsh is considered a significant downstream part of N‐terminal domain. Wnt proteins are preliminary activation sources
this process of transduction and is the 1st cytoplasmic protein to be of the signaling pathway. Such receptors subsequently spread across
pivotally active in all three main Wnt signaling cascades. It is still the plasma membrane seven times and form a separate family of
uncertain, although, how the regulation and channeling of the signal GPCRs (Bryja et al., 2007). The contact of Wnt protein with the Fz
into these pathways are controlled by Dsh protein (Catalano receptor, co‐receptors are crucial for Wnt signaling activation. The
et al., 2020; Komiya & Habas, 2008). lipoprotein (LRP)‐5/6. RTK and ROR2 are some examples of such
In rats, humans, zebrafish, Xenopus laevis and D. melanogaster receptors. A signal is sent toward the phosphoprotein Dsh/Dvl in the
Wnt proteins are found to be conserved across species (Nusse, 2005). cytoplasm when the receptor is activated.
HAYAT ET AL. Cell Biology | 3
International
The direct connection of Fz and Dsh helps to carry this signal. In invertebrates as LRP5 and ‐6 and in Drosophila as Arrow (Wehrli
almost all species, Dsh proteins have been identified and all have the et al., 2000). In a study, two monoclonal antibodies that can work in
highly conserved protein domains including a DIX amino‐terminal opposition to LRP6 along surprising potential by disrupting signaling
domain, a core PDZ region and a DEP carboxy‐terminal region. Such via certain Wnt proteins and increase signaling through others were
distinct realms are crucial as this Wnt signal is capable of going out identified. Such results indicate that Lrp6 have distinct binding
into several paths after Dsh, and each route interfaces with a distinct positions for various types of Wnt proteins, since these antibodies
variety of the three domains (Habas & Dawid, 2005). couple with nonoverlapping domains of LRP6 proteins (He
et al., 2004).
Signaling via dimeric Wnt receptors, much like most signal
3.2 | Extracellular regulators transduction mechanisms, entails a ligand‐persuaded structural
alteration in receptors accompanied by phosphorylation of main
The 19 human Wnt proteins are very promiscuous and each is selected proteins. The bonding of Axin to the LRP6 cytoplasmic tail is
capable of binding with more than one of the ten members of the Fz a necessary stage in signaling. Axin‐LRP6 coupling is controlled by at
family, compatible with comparatively high sustainability values of least two distinct kinases including GSK3 and CK1γ, via LPR6 tail
the binding interfaces. Norrin ligand can also have function for phosphorylation. In a variety of Wnt signaling elements, namely
activating the Wnt/β‐catenin path via coupling with the Wnt β‐catenin, Axin, APC, and possibly LRPs, serine is phosphorylated by
receptors, however, Norrin‐Fz is specialized to make signaling GSK3 in the PPPSP motif of these Wnt components. In membrane,
complex (Komiya & Habas, 2008). Norrin protein in sequence CK1γ is stabilized via its palmitoylated C terminal and play its role in
analysis reveals no associations with the Wnts. For overexpression phosphorylation of these proteins on sites next to the motif of PPPSP
and purification, Norrin also faces major challenges as Wnt. (Davidson et al., 2005). Whether or how Wnt stimulates these
Expression analysis in Escherichia coli and identification of the crystal protein kinases is not evident, while introducing Wnt protein to cells
structure of Norrin showed fusion with the maltose‐binding protein causes phosphorylation due to CK1γ in mins, indicating an immediate
C‐terminus have reportedly been uncovered and studied. This signal reaction.
complex shows a dimer made of two interconnected monomers
(Chang et al., 2015; Clevers, 2004). This dimmer locked together
through three intermolecular disulfide bridges. The structural 4 | EXTRACELLULAR D ISTRIBUTIO N AND
similarity of Norrin was predicted to match with the cysteine‐knot MOVEM ENT
cytokines, as well as the crystal structure indicated that it comprises
this basic configuration. Nonetheless, it is totally surprising and As best shown by Wg, Wnt proteins can act as morphogens intended to
unusual such arrangement of these three intermolecular disulfide interact both with short and long signaling. The concern of its
bonds. In Wnts proteins' C‐terminal domain, the detection of the distribution and dissemination across the soluble extracellular environ-
cytokine‐type fold shows a previously unknown structural resem- ment is raised due to Wg lipidation. Even so, purified Wnt3a through
blance of Wnt and Norrin. The additional structural similarities artificial liposomal packaging shows enhanced activity (Morrell
resulting from future investigations of the alignment of the signaling et al., 2008). Two separate Wg secretory routes have been hypothe-
complexes Fz and LRP5/6 including these two distinctive ligands are sized, though not entirely corroborated, for short and long‐distance
deemed very fascinating (Shen et al., 2015). signaling. To conceal lipid alterations within (Katanaev et al., 2008), or
attach to lipoprotein molecules, Wg can form multimers that might be
implicated in long‐distance Wg signaling (Panakova et al., 2005). Wg
3.3 | Heterodimeric structure of Wnt receptors secretion for long‐distance is directly promoted by the reggie‐1/flotillin‐
2 (membrane microdomain protein) (Katanaev et al., 2008). In the
Wnt proteins adhere to the complex of the heterodimeric receptor, concentration of Wg morphogen, Wg receptors and heparan sulfate
containing an Fz as well as an LRP5/6 receptor protein when proteoglycans including Dally and Dally‐like proteins show significant
interfering with target cells. Seven‐transmembrane receptors are the functions via the modulation of destruction, diffusion, endo/trans‐
10 Fz proteins of mammals having broad extracellular N‐terminal cytosis of Wg, and can act as important co‐receptors of low affinity in
cysteine‐rich regions (Bhanot et al., 1996), which offer a principal site Wg signaling (Glinka et al., 2011).
for Wnt adhesion (Dann et al., 2001). Multiple binding interfaces are
seen in the configuration of the CRD as it adheres to the Wnt, one of
which includes a hydrophobic furrow that attach to the Wnt molecule 5 | WNT: A DEVELOPMENTAL SIGNALING
of lipid (Niehrs & Acebron, 2012). The relationship between Wnt‐Fz PAT HWA Y
is chaste as a solitary Wnt can attach to several Fz proteins (Bhanot
et al., 1996) and vise‐versa, that too confirmed through Wnt‐CRD The Wnt signaling is specifically categorized into two groups based
complex form (Niehrs & Acebron, 2012). Fzs communicate with an on the participation of the β‐catenin a key intracellular molecule. The
LRP5 family's single‐pass transmembrane molecule recognized in canonical Wnt pathway and the Noncanonical Wnt pathway are
4 | Cell Biology HAYAT ET AL.

International
additionally classified as the PCP and Wnt/Ca2+ pathways. The surface receptors occurred, CK1 and GSK‐3 progressively phospho-
canonical Wnt signaling is thoroughly studied and described the rylate β‐catenin at serine/threonine residues positioned at the
signaling cascade, which depends on β‐catenin stabilization leading to N‐terminal protein region. Then, sequentially this phosphorylated
transcriptional activation (Rao & Kuhl, 2010). In specific, most β‐catenin undergoes ubiquitination for its degradation via E3
research is focusing on the pathway for Wnt/β‐catenin, the well‐ ubiquitin ligase β‐TrCP (Azzolin et al., 2014; Stamos & Weis, 2013).
known and defined arbitrator in the regulation of proliferation and As transcriptional repressors that suppress transcription of the gene,
migration of cell (Wray & Hartmann, 2012). transcription factor T‐cell factor (TCF) and Groucho generate a
The amounts of β‐catenin molecules are lower in a normal constitutive repressor complex when β‐catenin do not degrade.
condition lacking Wnt ligand (Figure 1a), but cytoplasmic catenin Decreased levels of nuclear β‐catenin could trigger and control
interacts with the nominal “destruction complex.” Structural protein, transcription of the target gene (Nusse & Clevers, 2017).
Axin, adenomatous polyposis coli (APC) of tumor suppressor, STK The cytoplasmic component of FZD could attach to DVL in the
(serine/threonine kinases), GSK‐3, CK1 (casein kinase 1, protein existence of Wnt ligands (Figure 1b), which will then offer a site for
phosphatase 2A, and β‐TrCP (β‐transducing repeat‐containing E3 the destruction of β‐catenin (Gammons & Bienz, 2018). The
ubiquitin‐protein ligase) are constituted of this destruction complex suppression of β‐catenin breakdown results once the complex is
(Stamos & Weis, 2013). When ligation of Wnt ligands with the cell employed, where phosphorylation of LRP5/6 via CK‐1 alpha occurs

F I G U R E 1 Canonical pathway signaling Wnt. (a) Signaling via complex of the Fz receptor and LRP5/6 coreceptor in the presence of Wnt
stimulation causes concurrent CK1 and GSK3‐β mediated LRP6 phosphorylation, allowing the diffusion of an Axin‐containing protein complex
toward the plasma membrane from the cytosol. Dvl is attracted to the membrane as well and couples to Fz while with phosphorylated LRP5/6
Axin binds. This complex established at Fz/LRP5/6 of the membrane stimulates β‐cat stabilization by either sequestration and/or Axin
deprivation. In terms of regulating the transcriptional activation of target genes, β‐catenin diffuses into the nucleus where it combines with Lef/
Tcf family subunits to form a composite. (b) The activity of the destruction complex consisting of CKI alpha, GSK3β, APC, Axin in the lack of Wnt
ligand produces a hyperphosphorylated β‐catenin, which is a subject for ubiquitination and proteasome degradation. β‐catenin is destroyed
following phosphorylation and ubiquitination via proteosome. Dvl offers a medium to degrade β‐catenin for selection. The TCF/Groucho
constitutive repressor complex represses the Wnt target genes
HAYAT ET AL. Cell Biology | 5
International
and allowing LRP5/6 to interact with the destruction complex (Niehrs also regulates the transcription of activated T‐cell dependent nuclear
& Shen, 2010). Accumulation of β‐catenin in cytoplasm matrix makes factor. Increasing evidence is pointing to the signaling pathway of
its diffusion to the nucleus. Within the nucleus, it triggers cellular Wnt/PCP as a crucial facilitator in the regulation of kidney and renal
responses via TCF and/or lymphoid enhancer factor through disease development (Matsuyama et al., 2014). This pathway in
controlling its target gene expression. These factors are the activators nephrogenesis is implicated in EMT (Burn et al., 2011). Moreover, a
of the Wnt pathways (Lee et al., 2014). variety of non‐canonical Wnt ligands that have been conventionally
Other signaling channels are activated by non‐canonical Wnt described can adversely impact the signaling pathway of canonical
proteins (e.g., Wnt4, Wnt5a, and Wnt11), which can also control Wnt/β‐catenin.
either transcriptional or non‐transcriptional actions in cells. The non‐
canonical route involves the PCP, also recognized as the signaling
pathway for Wnt/c‐Jun N‐terminal kinase (JNK) and the signaling 6 | OTHER WNT SI GNALI NG P ATHWAYS
pathway for Wnt/Ca2+, receptor‐like tyrosine kinase. Wnt/PCP
(shown in Figure 2) ligands namely Wnt5a, Wnt7, and Wnt11 attach It has been shown that the microtubule cytoskeleton, during neuronal
to the FZD receptor. Activation of small GTPases (Rho and Rac migration and synapse formation, is regulated by Wnt signaling and
family), and JNK mediated by DVL exert the downstream effectors this process relies on GSK3 and Dsh action (Salinas, 2007). One more
that guide cytoskeletal structure and direct cell polarization within Wnt channel by utilizing the Ryk receptor and proto‐oncoprotein Src
the epithelial sheet plane via regulation of the target gene governs the neurons repulsion during axonal coordination (Surmann‐
transcription. The small GTPase Rac and Rho collectively activate Schmitt et al., 2012). The Rap1‐GTPase that leads to cytoskeletal
kinases like Rho and JNK that contribute to downstream (Yang & regulation is inhibited by Wnt signaling through casein kinase during
Mlodzik, 2015) c‐Jun N‐terminal kinase expression and transcription gastrulation (Semenov et al., 2007). Wnt signaling through Dsh and
factor 2 activation (Sokol, 2015). GSK3 negatively controls the TSC2 tumor suppressor for cell growth
The Wnt/Ca2+ pathway (Figure 3), which produces Ca2+ through to control mechanistic target of rapamycin (Inoki, 2006). For
G‐proteins, is just another non‐canonical signaling pathway (Nusse & epithelial polarization and cell mobility, cell polarity and the
Clevers, 2017). The Wnt/Ca2+ signaling pathway regulates phospho- microtubule cytoskeleton are modulated by Wnt signaling through
lipase C (PLC) activation, calcium responsive calmodulin kinase II Dsh, aPKC, Par3, Par6, and LGl (Dollar et al., 2005). Wnt signaling
(CAMK II) and protein kinase C (PKC) enzymes activation, as well as it by PKA as well as the transcription factor CREB activates

F I G U R E 2 Wnt is translocated by Fz
independent of LRP5/6 in the non‐canonical
Wnt/PCP signaling pathway, leads to Dsh
activation. Dsh modulates the Rho activation
via Daam1, which then causes Rho kinase
(ROCK) activation. Via the actin‐binding
protein Profilin, Daam1 also regulates the
polymerization of actin. Rac activation is also
mediated by Dsh, which triggers JNK in
response. For cytoskeletal alterations during
gastrulation, Rock, JNK, and Profilin activation
are combined for Polarization and
translocation of the cell
6 | Cell Biology HAYAT ET AL.

International
F I G U R E E 3 Non‐canonical Wnt/Ca2+
signaling pathway. Wnt signaling via Fz
facilitates Dsh activation via G‐protein
activation. Phosphodiesterase PDE suppresses
the PKG and in consequence suppress the
emancipate of Ca2+, is triggered by
Disheveled. By PLC Dsh activates IP3, leading
to the emancipation of intracellular Ca2+,
which regulates CamK11 and calcineurin in
response. To control the fates of a ventral cell,
NF‐AT is stimulated by calcineurin. TAK and
NLK activation occur due to CamK11, which
impedes the activity of β‐catenin/TCF in
controlling the formation of the dorsal axis
negatively. During gastrulation activation of
CDC42 by PKC and DAG occur to initiate
tissue differentiation and cell movements

muscle‐specific gene expression of MyoD and Myf5 during myogen- to affect adjacent Wnt responsive cells across different sites. In
esis (Chen et al., 2005). Wnt signaling through the use of the ROR2 several species, Wnt/β‐catenin signaling is useful in identifying
receptor ultimately activates proto‐cadherin PAPC expression the dorso‐ventral and anteroposterior body axes (Genikhovich &
through the CDC42 and JNK channels in gastrulation (Schambony Technau, 2017; Niehrs, 2010). Wnt3a and Wnt4 have been reported
& Wedlich, 2007). These new pathways lead to complexity of Wnt to expressed in mice at the stage of 8‐cells (Lloyd et al., 2003).
signaling which may affect a variety of cellular processes, and Expression of Fz5 has been observed in the distal visceral endoderm
research to explain how these pathways are interconnected remains (dVE) and later in the anterior visceral endoderm (aVE) at the stage of
crucial. peri‐gastrula. While Fz10 was observed to be expressed in the
delamination of the primitive streak of the epiblast. Additionally, it
has been indicated that expression of Fz7 was observed in the
7 | I N D U C E D CE L L RE S P O N S E S extraembryonic area of pre‐gastrula and the epiblast of 6–7 days
post‐coitum (dpc) embryos, being more anteriorly bound at 7.5 dpc
7.1 | Role of Wnt in embryonic development (Kemp et al., 2007). The signaling by Wnt had also been associated
with parameters controlling the differentiation of the germ layer in
Wnt pathways are crucial for the embryonic development and stem the growing embryo. This was shown by the signaling pathway of
cell proliferation (Kerekes et al., 2021). Multiple signaling pathways Wnt/β‐catenin that mediated the expression of SRY HMG‐box 17
regulate the processes of Neural Crust (NC) induction and specifica- transcription factor (Sox17) for visceral endoderm patterning as well
tion, while Wnt/β‐catenin signaling has a continual role in activating as definite endoderm development. Wnt ligand‐dependent β‐catenin
and maintaining the gene regulatory network during NC induction pathway then stimulates a relatively distinctive transcriptional
and pigment cell specification (Sutton et al., 2021). Components of cascade at the initiation of gastrulation that regulates differentiation
Wnt family exhibit expression at the initial stage of embryonic of the anteroposterior axis. In this case, the pathway of
growth. One of several conserved communication networks that Wnt/β‐catenin is blocked anteriorly, resulting in the head structure
dictate embryo development is the Wnt/β‐catenin signaling formation, and subsequently activated posteriorly to form tail
(Sanz‐Ezquerro et al., 2017; Tepekoy et al., 2015). It contains Wnt structure (Green et al., 2015). The spatial and temporal
ligands that are produced by cells generating Wnt and that function Wnt/β‐catenin transcriptional activities regulation is central to
HAYAT ET AL. Cell Biology | 7
International
such patterning cases by the intervention of secretory antagonists, The Wnt/β‐catenin dependent pathway is implicated in the
epigenetic regulation, activities of various Tcf/Lef factors and the regulation of both normal and cancer cell proliferation (Holland
fusion of signaling with other pathways of development process. et al., 2013; Teo & Kahn, 2010; Valkenburg et al., 2011). Cancer stem
Wnt/β‐catenin signaling also have major role in the morphogenesis cell biochemistry is distinct from that of most tumor cells. To facilitate
of several tissues originating from the three germ layers, apart from their unregulated expansion, longevity, and proliferation, these so‐called
its early functions in forming the body axes of embryo (Steinhart & Wnt‐addicted cells derail and rely on consistent Wnt pathway
Angers, 2018). regulation. In cancer, via mutations in the sequence of downstream
oncogenes and tumor suppressor genes, Wnt signaling could work
independently without the involvement of typical triggers, it becomes
7.2 | Role of Wnt in cell fate specification chronically activated even if a signal is not received by the regular
receptor. In addition, the binding of the β‐catenin to transcription
Wnt signaling regulates cell fate decisions in diverse contexts during factors like the TCF4 enzyme causes the required gene activation by the
development, and loss of Wnt signaling in the cranial mesenchyme molecules. This binding is highly inhibited by LF3 in in vitro experiments,
results in a robust and binary cell fate switch from cranial bone to in cell lines, and decreased tumor growth in mice models. It stopped the
ectopic cartilage. Genetic interaction between the Wnt and ERK replication process and also decrease their ability to spread, all without
signaling pathways to repress Sox9 and ERK signaling is activated disturbing normal cells (Willert & Jones, 2006).
downstream of Wnt signaling during calvarial osteoblast fate
decisions in vivo (Ibarra et al., 2021)
In a study, 57 genes in 4 functionally linked and enriched classes 7.4 | Role of Wnt in cell migration
were identified in 239 Wnt regulated pathways (Huang et al., 2009).
Groups included are cuticle structural components, transcriptional During the development process of an embryo, cell migration
factors, signal proteins, and protein kinases. In the lateral hypodermal facilitates the body axis establishment, the development of tissues,
cells and the development, 13 out of 57 genes (about 24%) are the initiation of limbs, and many other processes. Wnt signaling,
expressed (Bruders et al., 2013). The major group of enriched genes, the especially at the stage of convergence and extension, supports to
cuticle, contains such encoding parts of the outer surface of the worm, instigate these processes. During gastrulation, for appropriate
which is synthesized by hypodermal cells. A study identified a group of convergence and extension, mediation from PCP along with the
cuticular collagen genes whose expression is usually observed in the canonical Wnt pathway is necessary. The Wnt/Ca2+ signaling, which
mid‐L4 stage in the larva. But in the case of ectopic Wnt signaling these inhibits convergence and extension when triggered, additionally
genes showed expression earlier. Reduced activity for these genes controls convergence and extension. In subsequent developmental
induced a deficiency in the structure of the adult cuticle. Finding of this stages, Wnt signaling also promotes cell migration by the regulation
study is in accordance with previous study and supports the theory of of the migration activity of neuroblasts, neural crest cells, tracheal
unrecognized function of Wnt signaling in adult cuticle synthesis cells, and myocytes (Schambony & Wedlich, 2013). For example,
(Jackson et al., 2014; Van Camp et al., 2014). myogenesis was found to be caused by Wnt‐1 and Wnt‐3 in chicken
and by Wg in Drosophila melanogaster. Wnts perform an additional
function in regulating the motility of myocytes, in contrast to
7.3 | Role of Wnt in cell proliferation determining the action. In the rat, Wnt‐5A and Wnt‐3A triggered in
cardiac myocytes the N‐cadherin expression and fibroblasts expres-
To control the proliferation and differentiation of cells, Wnt/β‐ sion of E‐cadherin and M‐cadherin. In the presence of fibroblasts,
catenin signaling pathway act as a central dogma (Kahn, 2014). This increased cell adhesion mediated by cadherin appears to be a
mechanism is regulated by canonical signaling of Wnt, which tends to requirement for the myocyte aggregates formation. Wnt‐5A/Wnt‐3A
increase β‐catenin in the nucleus and cytoplasm. Transcriptional triggers then reduce the mobility of rat's cardiac myocytes and
regulation of a variety of proteins including cyclin D1 and c‐myc, facilitate their accumulation. The regulation of myocyte migration by
which regulate the transition from G1 to S phase of the cell cycle, can DWnt‐2 in the male reproductive tract was seen in Drosophila
indeed be triggered by elevated β‐catenin. DNA replication and melanogaster (Von Maltzahn et al., 2012). Non‐canonical Wnt
mitosis triggered by S‐phase initiation. These events are essentially signaling playing role in regeneration of the intestinal epithelium,
necessary for the proliferation of the cells (Kaldis & Pagano, 2009). and identify enteroendocrine cell‐released ligands as critical coordi-
This controlled spike in proliferation is closely associated with cell nators of intestinal stem cell migration (D. J. K. Hu, Yun, et al., 2021).
differentiation, so they also divide as the stem cells proliferate.
Throughout embryonic development, it promotes the overall growth
and development of complex tissue structures. This is evident in 7.5 | Role of Wnt in bone formation
mechanisms including the circulatory system where Wnt3a con-
tributes to the hematopoietic stem cells proliferation and propagation Mutations which cause the loss in function in LRP5, a coreceptor of
needed for the production of red blood cells (Rasmussen et al., 2018). Wnt, contributed to decreased bone mass in humans, indicated the
8 | Cell Biology HAYAT ET AL.

International
potential contribution of Wnt signaling in regulating bone formation. signaling, two Wnt5a analog small peptides, Foxy‐5, and Box‐5,
Several results obtained from comprehensive studies on mouse and were already produced, thus mitigating tumor growth in selective
animal models with Wnt signaling agent mutations have established tumors. The first findings of the Foxy‐5 phase I clinical study show
that Wnt signaling has possible effects in both normal and strong therapeutic efficacy (Andersson et al., 2015).
pathophysiological cases of bone formation (Gong et al., 2001; Kato The advent of both Wnt5a agonists and antagonists, the
et al., 2002; Mani et al., 2007). The Wnts serve for cells of osteoblast characterization of responsive tumor subgroups for anti‐secretion
precursor and foster their division via the β‐canonical pathway into treatment, additionally direct medical trials in the context of tailored
mature osteoblasts. In contrast, by monitoring the ratio of the approaches. Though, as evidence of selective medical studies shows,
receptor activator of NF‐kB ligand (RANKL)/osteoprotegerin (OPG) the risk of significant site‐specific adverse impacts around stem cell
by a similar mechanism in mature osteoblasts, Wnts inhibit bone loss. niche areas in the body tends to be addressed in tailor studies and
In comparison, scientific investigations have revealed that the non‐ research on drug safety assessments (Zhan et al., 2017). As Wnt
canonical β‐catenin‐independent signaling pathway activation im- signaling has crucial roles in carcinogenesis, metastasis, cancer
proves the development of osteoclasts in mice macrophage cultures recurrence, and chemotherapy resistance. Based on these facts,
activated by RANKL (Glass et al., 2005; Maeda, 2007; Spencer Wnt represents an appealing therapeutic target for cancer treatment
et al., 2006). These findings show that in many facets of bone (Fatima et al., 2021). Any change in Wnt signaling pathway can cause
development and bone resorption, Wnt‐mediated signaling is following types of cancer:
activated.

8.1.1 | Pancreatic cancer


8 | CLINICAL IMPLICATIONS
Pancreatic ductal adenocarcinoma (PDAC) is currently the third
Dysregulation of Wnt signal transduction can contribute to the leading cause of cancer mortality in the United States (Siegel
progression of human diseases, despite the valuable and varying et al., 2021). WNT ligand signaling plays key roles in PDAC initiation,
physiological processes of Wnt signaling. Therefore, these kinds of progression, dissemination, and therapeutic resistance. Therapeutic
diseases, Wnt signaling pathway could also serve as an effective interest is a genetically defined subset of PDAC known to have
biomarker, and targeting this pathway is a beneficial option for increased WNT/β‐catenin transcriptional activity, growth depen-
therapeutic purposes. Present methods of targeting the Wnt signaling dency on WNT ligand signaling, and response to pharmacologic
therefore, mainly concentrated on interventions against cancer. inhibitors of the WNT pathway (Aguilera & Dawson, 2021).
Conversely, it is possible that abnormal Wnt signaling can be
explored as a therapeutic target for other disorders, like neuro-
degenerative disorders, chronic degenerative bone diseases, and 8.1.2 | Lung cancer (LC)
cardiovascular problems, with the advancement of our understanding
of such pathways. LC occupies the first place in global cancer incidence and death rate.
Development of lungs cancer is closely related to the Wnt/β‐catenin
signaling pathway, and its key molecules, such as Wnt1, β‐catenin,
8.1 | Role of Wnt in cancer cyclin D1, and SOX‐2, are expressed to varying degrees in the lung
cancer tissue. Development of novel inhibitors that selectively and
Wnt signaling is among the main development and halting cascades precisely target the Wnt/β‐catenin signaling pathway is still a
and is closely allied with cancer. Its role in oncogenesis has been promising strategy to generate unique LC treatment options that
more notably characterized for colorectal cancer, although in many will benefit LC patients (Zhu et al., 2021).
other cancer domains, abnormal Wnt signaling is also greatly involved
(Zhan et al., 2017). Many morphological and epigenetic experiments
have attempted to classify new elements of the Wnt pathway and 8.1.3 | Breast cancer
their roles in recent years. It became clear with the advancement of
sequencing technologies and the detailed structural analysis of Wnt/β‐catenin signaling pathway along with other components such
cancer genomes that Wnt pathway mutations have shown frequent as Axin2, APC, casein kinase Iα, GSK‐3β, protein phosphatase 2A,
association with cancer progression in humans (Dahmen et al., 2001). FZD7, LRP5, and LRP6 involved in breast cancer metastasis and
The Wnt signaling action in the field of cancer biology is strikingly progression because of its transcriptional control on epithelial to
nuanced and continues just slightly grasped because key constituents mesenchymal transition. Newer approaches using natural, microbial
of the pathway are already known (Segditsas & Tomlinson, 2006). In and chemical agents targeting the Wnt pathway show promising
cancer progression and development, a profound information of non‐ results to treat breast cancer. Targeted therapy involving drug
canonical Wnt signaling has contributed to revolutionary treatment combinations is studied in the preclinical and clinical‐stage to
interventions. To either trigger or suppress Wnt5a dependent increase effectiveness against breast cancer (Raut et al., 2022)
HAYAT ET AL. Cell Biology | 9
International
8.1.4 | Ovarian cancer required for the survival of neurons and neurogenesis but also plays a
vital role in controlling synaptic plasticity and the stability and
Ovarian cancer is one of the most common gynecologic malignancies functionality of the blood‐brain barrier (Nusse & Clevers, 2017).
in women and has a poor prognosis (Shu et al., 2022). Understanding Besides, Wnt/β‐catenin signaling activity reduces the generation of
the mechanisms of ovarian cancer progression and metastasis is amyloid‐β and hyperphosphorylation of tau protein in the brain.
critically important for developing novel therapies (Tian et al., 2022). Wnt/β‐catenin signaling is substantially inhibited by many pathogenic
HOXB8 promotes cell proliferation, migration and invasion through pathways in the AD brain. Therefore, a remarkable potential for
modulating Wnt/β‐catenin and STAT3 signaling pathways in ovarian the rational design of innovative AD strategies is to restore
cancer, suggesting that HOXB8 may provide a promising target for Wnt/β‐catenin signaling (Jia et al., 2019). Parkinson's disease (PD)
the therapy of ovarian cancer (Liu et al., 2022). is the most common disorder of movement (Giovannini et al., 2021).
The level of Wnt/β‐catenin signaling activation could explain the high
phenotypical heterogeneity of autism spectrum disorders and be
8.2 | Role of Wnt in type 2 diabetes instrumental in the development of new diagnostics tools and
therapies (Caracci et al., 2021).
Type 2 diabetes mellitus is among the utmost predominant ailments
that induce decreased insulin release and enhanced peripheral insulin
resistance, associated with increased glucose levels or hyperglycemia, 8.4 | Role of Wnt in rheumatoid arthritis (RA)
and maybe lethal if left unchecked. As Wnt signaling contributes to
insulin levels, it is believed that the defective pathway is responsible RA is the deadliest autoimmune disease and is controlled by many
for diabetes progression. For example, Wnt5b overexpression can factors like genetic makeup, environmental constituents, and
enhance the susceptibility because of its function in adipocyte dysfunctional signaling pathways. RA usually results in inflammation
differentiation, since obesity and type 2 diabetes are co‐morbid and severe deformities in joints of the various parts of the body (Salt
(Welters et al., 2008). Wnt signaling is a powerful mitochondrial & Crofford, 2012). One of the signaling pathway that is playing a vital
biogenesis modulator that inevitably causes an increase in the role in controlling RA is the WNT signaling path. But there are not
development of DNA and cellular damage found to increase reactive enough clear experimental proofs that will explain the complete
oxygen species (ROS). This disruption caused by ROS is important involvement of this pathway in RA (Miao et al., 2013, 2015).
since it is known to induce immediate resistance to hepatic insulin, or Many clinical studies have been held on a various number of RA
insulin resistance caused by damage. Genetic changes in transcription patients and all these studies enlighten the involvement of one of
factors linked with Wnt signaling like TCF7L2 are associated with genes of Wnt gene family which is Wnt7b. The expression of these
enhanced sensitivity (Schinner, 2009). A Chinese herbal medicine genes is upregulated in such patients. These studies also show the
(TSF) helps in the treatment of diabetic kidney disease. This may elevated level of production of some inflammatory proteins and
alleviate myocardial fibrosis in KKAy mouse models by inhibiting agents like TNF‐α, IL‐1β, and IL6 in RA synovium. An interesting point
TGF‐β/Smad and Wnt/β‐catenin signaling pathways. These findings rise after these studies is that these inflammatory agent's TNF‐α,
may increase our knowledge regarding effect of TSF in diabetes IL‐1β, and IL6 are produced in a massive amount in normal synovial
(L. Hu, Wang, et al., 2021). cells which were transfected by the Wnt7b genes. This shows a
linkage between the inflammatory mediators and Wnt genes. These
elevated levels of inflammation proteins also scale up the expression
8.3 | Role of Wnt in neuronal disorder level of Wnt/Fz.
A comparative study conducted between RA patient and other
Enhanced Wnt1 expression can contribute to synaptic reorganization patients who do not have much difference in the number of various
and plasticity in schizophrenia patients' brains. The propensity to proteins like the Wnt inducible signaling pathway proteins (WISP),
schizophrenia is linked with many SNPs in Fz3, and GSK3 behavior in ligands, and Fz receptors were observed between them. Unlike the
schizophrenia appears to be changed reportedly (Katsu et al., 2003). patients who don't have RA, the RA patients have higher level of
Mutated β‐catenin signaling can be implied in the progression of the these proteins in their body (Cheon et al., 2004; Nakamura
most prevalent neurological disorder, Alzheimer's disease (AD), et al., 2005).
identified by extracellular deposits of amyloid β‐peptide, intracellular Upregulation and downregulation of different pathways and
hyperphosphorylated tau accumulation, and specific cholinergic factors collectively result in RA and osteoarthritis like the degradation
neuronal loss (Luo et al., 2007). The constituents of presenilin of the matrix that is found in cartilaginous joints due to the
complexes are β‐Catenin and GSK3β, and the lack of Wnt/β‐catenin switching on of Wnt/β‐catenin signaling in chondrocytes, whereas
activity could be underlying the unset and AD progression (Luo the erosion of bones and their remodeling into a completely different
et al., 2007; Mudher & Lovestone, 2002,). AD is a brain‐related morphology occur due to switching off Wnt signals. The correct
disorder linked with aging, with synaptic loss and dementia (Selkoe & expression of different proteins like fibronectin and metalloprotei-
Hardy, 2016). Wnt/β‐catenin signaling in the brain is not only nase which are involved in bone formation is controlled by the Wnt
10 | Cell Biology HAYAT ET AL.

International
signaling pathway (Schett et al., 2008; Shi et al., 2016). Functionally, WNT/β‐catenin signaling. Results indicated the potential role of IT in
a slight divergence in the expression of Wnt signals results in the the treatment of scleroderma and provide novel insight for the
advancement of cell proliferation and differentiation (Cheon et al., 2004). selection of drug therapy for scleroderma (Li et al., 2021). All these
Different in vitro studies have been carried out on the fibroblast‐like studies indicate that these Wnt pathways are playing a lead role in
synoviocytes (FLS) of RS patients. These FLS show a gradual and tissue fibrosis. Keeping in view, researchers put forward many
consistent higher expression level of Wnt5a and Fz5. These studies different approaches that target these pathways and help in the
show a direct relation between FLS, Wnt5a and Fz5. The activation of treatment of SSc. These approaches including tankyrase inhibitors,
these pathways also activates the FLS in RA patients. These activation porcupine inhibitors, and antagonists of cofactor recruitment to
events are not influenced by the inflammatory environment around β‐catenin have prominent effects in animal treatment models
them (Cheon et al., 2004, Shi et al., 2016). Thus for the treatment of (Distler et al., 2013; Konigshoff et al., 2009).
refractory synovitis, all the receptors that are antagonistic to Fz5 can be
considered effective (Sen et al., 2001). Td‐FLS, fd‐FLS, and fibrocytes
from patients with RA expressed similar levels of Wnt5a and a set of 8.6 | Wnt signaling in inflammatory bowel
Wnt5a receptors/coreceptors. Wnt5a stimulated the expression of the disease (IBD)
pro‐inflammatory targets, and in conjunction with SFRP5 inhibited the
gene expression of TCF4 and the protein levels of the canonical IBD is a disorder in which the body's immune response acts against
coreceptor LRP5 (Mahmoud et al., 2021). Knockdown of phospholipase the natural microflora of the intestine. These microflorae have
D1 (PLD1) could reduce RA‐FLSs metastasis as well as inflammatory positive impacts on digestion. This results in the inflamed colon and
response by modulating the activity of NF‐κB and Wnt/β‐catenin small intestine. To date, an involvement of Wnt signaling in IBD
pathways (Zhang et al., 2021). pathogenesis has been broadly recognized (Shi et al., 2016). Some
previous studies indicate the hyperactive response of the Wnt
signaling pathway along with a cascade of other interlinked signaling
8.5 | Wnt signaling in systemic sclerosis (SSc) factors Wnt2, Wnt5a, Wnt5b, Fzd2, Fzd4, Fzd6, LRP6, Dvl, DKK1,
and SFRP1 in both ulcerative colitis (UC) and Crohn disease (CD)
SSc is a lethal autoimmune disease with highest mortality rate. The tissues (Hughes et al., 2011; Tuller et al., 2013). In human UC
main cause of this deadly disease is the hyperactivation of the different microarray analysis of various Wnt pathway genes have
immune system along with the severe damage to the vascular been carried out which indicates the upregulated expression of
muscles and the agglomeration of various excretory proteins in skin Wnt2b, Wnt3a, Wnt5b, Wnt6, Wnt7, Wnt9, Wnt11, Fzd3, Fzd4,
and organs (Antic et al., 2013). The Wnt pathway is clearly elevated in DKK4, and Dvl2 and downregulated mechanics of Fz1 and Fz5 in
SSc and is pro‐fibrotic via activation of canonical Wnt signaling comparison to non‐IBD (You et al., 2008). Another analytical
(Henderson et al., 2021). The regulatory mechanics of various technique known as IHC staining is also performed for the
signaling machinery play a part in the activation of this disease. confirmation which also enlightens that in human UC tissues the
Many events like the upregulated expression of Wnt signaling expression of β‐catenin and Wnt associated cancer genes E‐cadherin,
components Wnt1, Wnt10b, Fzd2, nuclear β‐catenin, and LEF‐1 cyclin D1, and c‐myc expressions was unregulated (Van Dekken
notices in the patients of this disease, these relations directly pointed et al., 2007). Recently a new genome profiling study reported that in
toward skin fibrosis (Leah, 2013; Svegliati et al., 2014). Many in vivo IBD patients there are visible signs of activation of Wnt signals that
studies in mice show two major events one is the activation of are part of the epithelial lining of the intestine and a visible
fibroblast‐specific dominant active β‐catenin and the other is the de‐ suppression in the non‐canonical Wnt signaling pathways. Along
active form of fibroblast‐specific dominant de‐active β‐catenin which with this another important point also reveals that the process of
are involved in the fibrosis. Mice with fibroblast‐specific dominant mesenchymal cells has an antagonistic effect as it induces the non‐
active β‐catenin show abrupt thickening and collagen accumulation in canonical signals and cause the suppression of canonical Wnt
the skin and within two weeks they develop fibrosis and result in the signaling cascade. This results in lowering and decreased inflamma-
differentiation of fibroblasts into myofibroblasts. In contrast, the tion levels in and IBD positive rats (Xing et al., 2015).
deactivation of fibroblast‐specific β‐catenin reduced the bleomycin‐
induced fibrosis (Beyer et al., 2012). In addition, another in vitro study
indicates the involvement of the Wnt3a in the induction of β‐catenin 8.7 | Role of Wnt in cardiovascular diseases
for the stimulation of fibroblast proliferation, contraction of collagen
gel, and differentiation of myofibroblast, and increased expression of Cardiogenesis and heart diseases both are regulated and controlled
profibrotic genes. In contrast, Wnt3a has a repressing power over by canonical and non‐canonical Wnt signaling pathways (Olson &
adipogenesis but has accelerated effects on the myofibroblast Schneider, 2003; Van Gijn et al., 2002). For instance, an in vivo
differentiation in subcutaneous preadipocytes (Antic et al., 2013). disease carried out on mice as a model organism indicates that if the
Another study determined the effect of icaritin has an anti‐skin mice lack Dvl1 are transfected by myocardial infarction have more
fibrotic effect through activation of AMPK signaling and inhibition of chances of infract rupture. Neovascularization in the infarct area is
HAYAT ET AL. Cell Biology | 11
International
also being affected by the Wnt signaling. Proper cardiac function, inhibition of destructive complex function. Moreover, several new
accuracy and maintenance of optimal infarct size are also achieved by cancer medications are nonspecifically engineered to suppress Wnt
the overexpression of sFRP1 (Barandon et al., 2003). Rats used as a signaling, indicating they affect both normal and cancer cells, which
study model for cardiac hypertrophy shows upregulation of Fz2 (Van may lead to severe negative impacts. A major difficulty lies in
Gijn et al., 2002). If the Wnt pathway got disrupted or dysfunctional designing a strategy that is sufficiently successful to target the
due to any reason, it results in cardiovascular inflammation, dysregulated Wnt pathway while restricting danger to the normal
decreasing plasticity of cells, abnormal levels or accumulation of functioning of the Wnt pathway, guaranteeing that its crucial
cholesterol, and abnormal metabolic response to injuries and trauma. position remains unchanged in various essential biological pro-
Another important cofactor of the Wnt signaling pathway known as cesses. Help to effectively understand the potential pathogenic
LRP6 has a major role in the cardio metabolic‐related diseases functions in human disorders with abnormal Wnt signaling, but also
confirmed by human and mice genetical analysis. Different biological to present possibilities for appropriate drug development to target
weapons can be used as a therapeutic agents for the targeting of Wnt the pathways.
signals and controlling all the factors interlinked with this pathway
and result in many deadly and lethal diseases (Gay & Towler, 2017; A UT H O R C O N T R I B U TI O NS
Krishna et al., 2017). Contributed to manuscript writing: Rabia Hayat. Did critical discus-
sions: Maleeha Manzoor. Helped in revision of the manuscript: Ali
Hussain.
9 | C ONC LUS I ON
DATA AVAILABILITY STATEMENT
Wnt signaling is an important pathway crucial for the appropriate Data sharing not applicable—no new data generated.
development of cells at the embryonic and mature stage by
maintaining cell to cell communication. Wnt signaling further ORC I D
subdivided into three pathways each having its usefulness. Several Maleeha Manzoor http://orcid.org/0000-0002-0393-6766
key components of this dynamic system and their potential role
have also characterized. This review provided a comprehensive
RE F ER EN CES
overview of Wnt signaling, its components, mechanisms, its role in
Aguilera, K. Y., & Dawson, D. W. (2021). WNT ligand dependencies in
developmental stages, and its importance for clinical implications. pancreatic cancer. Frontiers in Cell and Developmental Biology, 9,
Wnt/β‐catenin dependent signaling pathway is the most studied 1027.
pathway. Several studies have described the role of different Wnt Amin, N., & Vincan, E. (2012). The Wnt signaling pathways and cell
adhesion. Frontier in Bioscinece, 17, 784–804.
proteins in the progression of several types of cancer, diabetes,
Andersson, T., Axelsson, L., Mohapatra, P., Prasad, C., Soerensen, P. G.,
neuronal disorder, IBS, RA, SSc, and cardiovascular diseases. The Mau‐Soerensen, M., Lassen, U., Molvadgaard, T., Buhl, U., &
study of potential role in these diseases makes the Wnt pathway an Brunner, N. (2015). Targeting the Wnt‐5a signaling pathway as a
important biomarker for establishing novel therapeutic strategies to novel anti‐metastatic therapy. Molecular Cancer Therapeutics, 14,
A116.
treat and prevent such life threatening diseases.
Antic, M., Distler, J. H., & Distler, O. (2013). Treating skin and lung fibrosis
in systemic sclerosis: A future filled with promise? Current Opinion in
Pharmacology, 13, 455–462.
10 | PER SP ECT IV ES Azzolin, L., Panciera, T., Soligo, S., Enzo, E., Bicciato, S., Dupont, S.,
Bresolin, S., Frasson, C., Basso, G., Guzzardo, V., & Fassina, A. (2014).
yap/taz incorporation in the β‐catenin destruction complex
It has been established that for the proper development and orchestrates the Wnt response. Cell, 158, 157–170.
homeostasis of nearly all tissues, Wnt ligands are important. Baarsma, H., & Konigshoff, M. (2017). ‘Wnt‐er is coming’: Wnt signaling in
Genome, biochemical and functional research have highlighted chronic lung diseases. Thorax, 72, 746–759.
Barandon, L., Couffinhal, T., Ezan, J., Dufourcq, P., Costet, P., Alzieu, P.,
several components of Wnt signaling in recent decades and have
Leroux, L., Moreau, C., Dare, D., & Duplàa, C. (2003). Reduction of
uncovered their cellular processes. Major gaps exist, though, infarct size and prevention of cardiac rupture in transgenic mice
especially concerning the signal transduction pathways mediated overexpressing frza. Circulation, 108, 2282–2289.
by Wnt proteins and β‐catenin. For instance, while Wnt's Beyer, C., Schramm, A., Akhmetshina, A., Dees, C., Kireva, T., Gelse, K.,
Sonnylal, S., De Crombrugghe, B., Taketo, M. M., Distler, O., &
configurations in complex with the Fz CRD offered a description
Schett, G. (2012). β‐catenin is a central mediator of pro‐fibrotic Wnt
of the ligand‐receptor binding, it remains an open question that how
signaling in systemic sclerosis. Annals of Rheumatic Diseases, 71,
this is related to possible allosteric alterations in Fz's heptahelical 761–767.
clusters to transmit signals to the intracellular matrix components. It Bhanot, P., Brink, M., Samos, C. H., Hsieh, J. C., Wang, Y., Macke, J. P.,
remains poorly described that how Wnts co‐work with Fz and co‐ Andrew, D., Nathans, J., & Nusse, R. (1996). A new member of the
frizzled family from drosophila functions as a wingless receptor.
receptors LRP5/6, and what is the exact structure of the complexes
Nature, 382, 225–230.
formed. Another poorly known aspect is the recruitment by active Bruders, R., Swalla, B. J., Moroz, L., & Kohn, A. (2013). Gone with the
Fz receptors of Dsh/Dvl proteins, and how this contributes to the Wnts: Genomic insights into Wnt signaling in the ctenophore,
12 | Cell Biology HAYAT ET AL.

International
Pleurobrachia bachei. Integrative and Comparative Biology, 53, Farin, H. F., Jordens, I., Mosa, M. H., Basak, O., Korving, J., Tauriello, D. V.,
e24–14. De Punder, K., Angers, S., Peters, P. J., & Maurice, M. M. (2016).
Bryja, V., Schulte, G., & Arenas, E. (2007). Wnt‐3a utilizes a novel low dose Visualization of a short‐range Wnt gradient in the intestinal stem‐
and rapid pathway that does not require casein kinase 1‐mediated cell niche. Nature, 530, 340–343.
phosphorylation of Dvl to activate β‐catenin. Cellular Signalling, 19, Fatima, I., Barman, S., Rai, R., Thiel, K. W., & Chandra, V. (2021). Targeting
610–616. Wnt signaling in endometrial cancer. Cancers, 13, 2351.
Burn, S., Webb, A., Berry, R., Davies, J., Ferrer‐Vaquer, A., Foulquier, S., Daskalopoulos, E. P., Lluri, G., Hermans, K. C., Deb, A., &
Hadjantonakis, A., Hastie, N. D., & Hohenstein, P. (2011). Calcium/ Blankesteijn, W. M. (2018). Wnt signaling in cardiac and vascular
NFAT signaling promotes early nephrogenesis. Developmental disease. Pharmacology Reviews, 70, 68–141.
Biology, 352, 288–298. Gajos‐Michniewicz, A., & Czyz, M. (2020). Wnt signaling in melanoma.
Cadigan, K. M., & Nusse, R. (1997). Wnt signaling: A common theme in International Journal of Molecular Sciences, 21, 4852.
animal development. Genes and Development, 11, 3286–3305. Gammons, M., & Bienz, M. (2018). Multiprotein complexes governing Wnt
Caracci, M. O., Avila, M. E., Espinoza‐Cavieres, F. A., López, H. R., signal transduction. Current Opinion in Cell Biology, 51, 42–49.
Ugarte, G. D., & De Ferrari, G. V. (2021). Wnt/β‐catenin‐dependent Gay, A., & Towler, D. A. (2017). Wnt signaling in cardiovascular disease:
transcription in autism spectrum disorders. Frontiers in Molecular Opportunities and challenges. Current Opinion in Lipidology, 28,
Neuroscience, 14, 14. 387–396.
Catalano, A., Bellone, F., Morabito, N., & Corica, F. (2020). Sclerostin and Genikhovich, G., & Technau, U. (2017). On the evolution of bilaterality.
vascular pathophysiology. International Journal of Molecular Science, Development, 144, 3392–3404.
21, 4779. Giovannini, D., Andreola, F., Spitalieri, P., Krasnowska, E. K.,
Chang, T. H., Hsieh, F. L., Zebisch, M., Harlos, K., Elegheert, J., & Colini Baldeschi, A., Rossi, S., Sangiuolo, F., Cozzolino, M., &
Jones, E. Y. (2015). Structure and functional properties of Norrin Serafino, A. (2021). Natriuretic peptides are neuroprotective on in
mimic Wnt for signaling with Frizzled4, Lrp5/6, and proteoglycan. vitro models of PD and promote dopaminergic differentiation of
eLife, 4, e06554. hiPSCs‐derived neurons via the Wnt/β‐catenin signaling. Cell Death
Chen, A. E., Ginty, D. D., & Fan, C. M. (2005). Protein kinase a signaling via Discovery, 7(1), 1–14.
creb controls myogenesis induced by Wnt proteins. Nature, 433, Glass, D. A., II, Bialek, P., Ahn, J. D., Starbuck, M., Patel, M. S., Clevers, H.,
317–322. Taketo, M. M., Long, F., McMahon, A. P., Lang, R. A., & Karsenty, G.
Cheon, H., Boyle, D. L., & Firestein, G. S. (2004). Wnt1 inducible signaling (2005). Canonical Wnt signaling in differentiated osteoblasts
pathway protein‐3 regulation and microsatellite structure in arthritis. controls osteoclast differentiation. Developmental Cell, 8, 751–764.
Journal of Rheumatology, 31, 2106–2114. Glinka, A., Dolde, C., Kirsch, N., Huang, Y. L., Kazanskaya, O.,
Clevers, H. (2004). Wnt signaling: Ig‐norrin the dogma. Current Biology, 14, Ingelfinger, D., Boutros, M., Cruciat, C. M., & Niehrs, C. (2011).
436–437. lgr4 and lgr5 are r‐spondin receptors mediating Wnt/β‐catenin and
Clevers, H. (2006). Wnt/β‐catenin signaling in development and disease. Wnt/PCP signaling. EMBO Reports, 12, 1055–1061.
Cell, 127, 469–480. Gong, Y., Slee, R. B., Fukai, N., Rawadi, G., Roman‐Roman, S.,
Clevers, H., & Nusse, R. (2012). Wnt/β‐catenin signaling and disease. Cell, Reginato, A. A., Wang, H., Cundy, T., Glorieux, F. H., Lev, D., &
149, 1192–1205. Zacharin, M. (2001). LDL receptor‐related protein 5 (LRP5) affects
Cui, Y., Zhang, F., Zhu, C., Geng, L., Tian, T., & Liu, H. (2017). upregulated bone accrual and eye development. Cell, 107, 513–523.
lncRNA SNHG1 contributes to progression of non‐small cell Green, D., Whitener, A. E., Mohanty, S., & Lekven, A. C. (2015). Vertebrate
lung cancer through inhibition of miR‐101‐3p and activation of nervous system posteriorization: grading the function of Wnt
Wnt/β‐catenin signaling pathway. Oncotarget, 8, 17785–17794. signaling. Developmental Dynamics, 244, 507–512.
Dahmen, R., Koch, A., Denkhaus, D., Tonn, J., Sorensen, N., Berthold, F., Grumolato, L., Liu, G., Mong, P., Mudbhary, R., Biswas, R., Arroyave, R.,
Behrens, J., Birchmeier, W., Wiestler, O. D., & Pietsch, T. (2001). Vijayakumar, S., Economides, A. N., & Aaronson, S. A. (2010).
Deletions of AXIN1, a component of the Wnt/wingless pathway, in Canonical and noncanonical Wnts use a common mechanism to
sporadic medulloblastomas. Cancer Research, 61, 7039–7043. activate completely unrelated coreceptors. Genes and Development,
Dann, C. E., Hsieh, J. C., Rattner, A., Sharma, D., Nathans, J., & Leahy, D. J. 24, 2517–2530.
(2001). Insights into Wnt binding and signaling from the structures Habas, R., & Dawid, L. B. (2005). Dishevelled and Wnt signaling: Is the
of two frizzled cysteine‐rich domains. Nature, 412, 86–90. nucleus the final frontier? Journal of Biology, 4, 1–4.
Davidson, G., Wu, W., Shen, J., Bilic, J., Fenger, U., Stannek, P., Glinka, A., He, X., Semenov, M., Tamai, K., & Zeng, X. (2004). LDL receptor‐related
& Niehrs, C. (2005). Casein kinase 1 γ couples Wnt receptor proteins 5 and 6 in Wnt/β‐catenin signaling: Arrows point the way.
activation to cytoplasmic signal transduction. Nature, 438, 867–872. Development, 131, 1663–1677.
Dejana, E. (2010). The role of Wnt signaling in physiological and Henderson, J., Wilkinson, S., Przyborski, S., Stratton, R., & O'Reilly, S.
pathological angiogenesis. Circulation Research, 107, 943–952. (2021). microRNA27a‐3p mediates reduction of the Wnt antagonist
Distler, A., Deloch, L., Huang, J., Dees, C., Lin, N. Y., Palumbo‐Zerr, K., sFRP‐1 in systemic sclerosis. Epigenetics, 16(7), 808–817.
Beyer, C., Weidemann, A., Distler, O., Schett, G., & Distler, J. H. Holland, J. D., Klaus, A., Garratt, A. N., & Birchmeier, W. (2013). Wnt
(2013). Inactivation of tankyrases reduces experimental fibrosis by signaling in stem and cancer stem cells. Current Opinion in Cell
inhibiting canonical Wnt signaling. Annals of Rheumatic Diseases, 72, Biology, 25, 254–264.
1575–1580. Holstein, T. W. (2012). The evolution of the Wnt pathway. Cold Spring
Dollar, G. L., Weber, U., Mlodzik, M., & Sokol, S. Y. (2005). Regulation of Harbor Perspectives in Biology, 4, a007922.
lethal giant larvae by dishevelled. Nature, 437, 1376–1380. Hu, D. J. K., Yun, J., Elstrott, J., & Jasper, H. (2021). Non‐canonical Wnt
Fan, J., Wei, Q., Liao, J., Zou, Y., Song, D., Xiong, D., Ma, C., Hu, X., Qu, X., signaling promotes directed migration of intestinal stem cells to sites
Chen, L., Li, L., Yu, Y., Yu, X., Zhang, Z., Zhao, C., Zeng, Z., Zhang, R., of injury. Nature Communications, 12(1), 1–15.
Yan, S., Wu, T., … Tang, H. (2017). Noncanonical Wnt signaling plays Hu, L., Wang, Y., Wan, Y., Ma, L., Zhao, T., & Li, P. (2021). Tangshen
an important role in modulating canonical Wnt‐regulated stemness, formula improves diabetes‐associated myocardial fibrosis by
proliferation and terminal differentiation of hepatic progenitors. regulating TGF‐β/Smads and Wnt/β‐catenin pathways. Frontiers in
Oncotarget, 8, 27105–27119. Medicine, 8, 2022.
HAYAT ET AL. Cell Biology | 13
International
Huang, D. W., Sherman, B. T., Zheng, X., Yang, J., Imamichi, T., feedback suppressor targeting the β‐catenin/Tcf1 complex.
Stephens, R., & Lempicki, R. A. (2009). Extracting biological Molecular and Cellular Biology, 36, 1793–1802.
meaning from large gene lists with david. curr protoc. Current Langton, P. F., Kakugawa, S., & Vincent, J.‐P. (2016). Making, exporting,
Protocols in Bioinformatics, 2713.11. 1‐13.11. 13. and modulating Wnts. Trends in Cell Biology, 26, 756–765.
Hughes, K., Sablitzky, F., & Mahida, Y. (2011). Expression profiling of Wnt Leah, E. (2013). Epigenetic silencing of Wnt inhibition in systemic
family of genes in normal and inflammatory bowel disease primary sclerosis. Nature Reviews Rheumatology, 9, 384–384.
human intestinal myofibroblasts and normal human colonic crypt Lee, Y. S., Cho, Y. S., Lee, G. K., Lee, S., Kim, Y. W., Jho, S., Kim, H. M.,
epithelial cells. Inflammatory Bowel Diseases, 17, 213–220. Hong, S. H., Hwang, J. A., Kim, S. Y., & Hong, D. (2014). Genomic
Ibarra, B., Machen, C., & Atit, R. (2021). Wnt‐dependent activation of ERK profile analysis of diffuse‐type gastric cancers. Genome Biology, 108,
mediates repression of chondrocyte fate during calvarial 1–15.
development. bioRxiv, 9. Li, M., Liu, Q., He, S., Kong, X., Lin, J., Huang, Y., Wu, W., & Wu, J. (2021).
Inoki, K. O.‐L. (2006). TSC2 integrates Wnt and energy signals via a Icaritin inhibits skin fibrosis through regulating AMPK and Wnt/β‐
coordinated phosphorylation by AMPK and GSK3 to regulate. Cell catenin signaling. Cell Biochemistry and Biophysics, 79(2), 231–238.
Growth. Cell, 126, 955–968. Liu, L., Wang, L., & Li, X. (2022). The roles of HOXB8 through activating
Jackson, B. M., Abete‐Luzi, P., Krause, M. W., & Eisenmann, D. M. (2014). Wnt/β‐catenin and STAT3 signaling pathways in the growth,
Use of an activated beta‐catenin to identify Wnt pathway target migration and invasion of ovarian cancer cells. Cytotechnology, 74,
genes in Caenorhabditis elegans, including a subset of collagen genes 1–11.
expressed in late larval development. G3, 4, 733–747. Lloyd, S., Fleming, T. P., & Collins, J. E. (2003). Expression of Wnt genes
Janda, C. Y., Dang, L. T., You, C., Chang, J., de Lau, W., Zhong, Z. A., during mouse preimplantation development. Gene Expression
Yan, K. S., Marecic, O., Siepe, D., Li, X., Moody, J. D., Williams, B. O., Patterns, 3, 309–312.
Clevers, H., Piehler, J., Baker, D., Kuo, C. J., & Garcia, K. C. (2017). Logan, C. Y., & Nusse, R. (2004). The Wnt signaling pathway in
Surrogate Wnt agonists that phenocopy canonical Wnt and β‐ development and disease. Annual Review of Cell and Developmental
catenin signalling. Nature, 545, 234–237. Biology, 20, 781–810.
Jia, L., Pina‐Crespo, J., & Li, Y. (2019). Restoring Wnt/β‐catenin signaling is Luo, J., Chen, J., Deng, Z.‐L., Luo, X., Song, W.‐X., Sharff, K. A., Tang, N.,
a promising therapeutic strategy for Alzheimer's disease. Molecular Haydon, R. C., Luu, H. H., & He, T.‐C. (2007). Wnt signaling and
Brain Research, 12, 1–11. human diseases: What are the therapeutic implications? Laboratory
Kahn, M. (2014). Can we safely target the Wnt pathway? Nature Reviews Investigation, 87, 97–103.
Drug Discovery, 13, 513–532. Maeda, K. (2007). Wnt5a enhances RANKL‐induced osteoclastogenesis.
Kaldis, P., & Pagano, M. (2009). Wnt signaling in mitosis. Developmental Journal of Bone and Mineral Research, 22, s43.
Cell, 17, 749–750. Mahmoud, D. E., Kaabachi, W., Sassi, N., Mokhtar, A., Moalla, M.,
Katanaev, V. L., Solis, G. P., Hausmann, G., Buestorf, S., Katanayeva, N., Ammar, L. B., Jemmali, S., Rekik, S., Tarhouni, L., Kallel‐Sellami, M.,
Schrock, Y., Stuermer, C. A., & Basler, K. (2008). Reggie‐1/flotillin‐2 Cheour, E., & Laadhar, L. (2021). SFRP5 enhances Wnt5a induced‐
promotes secretion of the long‐range signaling forms of Wingless inflammation in rheumatoid arthritis fibroblast‐like synoviocytes.
and Hedgehog in Drosophila. EMBO Journal, 27, 509–521. Frontiers in Immunology, 12, 2356.
Kato, M., Patel, M. S., Levasseur, R., Lobov, L., Chang, B. H. J., Glass, D. A., Mani, A., Radhakrishnan, J., Wang, H., Mani, A., Mani, M.‐A., Nelson‐
Hartmann, C., Li, L., Hwang, T. H., Brayton, C. F., & Lang, R. A. (2002). Williams, C., Carew, K. S., Mane, S., Najmabadi, H., & Wu, D. (2007).
Cbfa1‐independent decrease in osteoblast proliferation, osteopenia, Lrp6 mutation in a family with early coronary disease and metabolic
and persistent embryonic eye vascularization in mice deficient in risk factors. Science, 315, 1278–1282.
Lrp5, a Wnt coreceptor. Journal of Cell Biology, 157, 303–314. Matsuyama, M., Nomori, A., Nakakuni, K., Shimono, A., & Fukushima, M.
Katsu, T., Ujike, H., Nakano, T., Tanaka, Y., Nomura, A., Nakata, K., (2014). Secreted frizzled‐related protein 1 (Sfrp1) regulates the
Takaki, M., Sakai, A., Uchida, N., Imamura, T., & Kuroda, S. (2003). progression of renal fibrosis in a mouse model of obstructive
The human frizzled‐3 (FZD3) gene on chromosome 8p21, a receptor nephropathy. Journal of Biological Chemistry, 289, 31526–31533.
gene for Wnt ligands, is associated with the susceptibility to Miao, C.‐G., Huang, C., Huang, Y., Yang, Y.‐Y., He, X., Zhang, L., Lv, X.‐W.,
schizophrenia. Neuroscience Letters, 353, 53–56. Jin, Y., & Li, J. (2013). Mecp2 modulates the canonical Wnt pathway
Kemp, C. R., Willems, E., Wawrzak, D., Hendrickx, M., Agbor Agbor, T., & activation by targeting SFRP4 in rheumatoid arthritis fibroblast‐like
Leyns, L. (2007). Expression of Frizzled5, Frizzled7, and Frizzled10 synoviocytes in rats. Cellular Signalling, 25, 598–608.
during early mouse development and interactions with canonical Miao, C.‐G., Qin, D., Du, C.‐L., Ye, H., Shi, W.‐J., Xiong, Y.‐Y., Zhang, X.‐L.,
Wnt signaling. Developmental Dynamics, 236, 2011–2019. Yu, H., Dou, J.‐F., & Ma, S.‐T. (2015). Dnmt1 activates the canonical
Kerekes, K., Trexler, M., Bányai, L., & Patthy, L. (2021). Wnt inhibitory Wnt signaling in rheumatoid arthritis model rats via a crucial
factor 1 binds to and inhibits the activity of sonic hedgehog. Cells, functional crosstalk between miR‐152 and the DNMT1, MeCP2.
10(12), 3496. International Immunopharmacology, 28, 344–353.
Komiya, Y., & Habas, R. (2008). Wnt signal transduction pathways. Morrell, N. T., Leucht, P., Zhao, L., Kim, J.‐B., Ten Berge, D.,
Organogenesis, 4, 68–75. Ponnusamy, K., Carre, A. L., Dudek, H., Zachlederova, M., &
Konigshoff, M., Kramer, M., Balsara, N., Wilhelm, J., Amarie, O. V., Mcelhaney, M. (2008). Liposomal packaging generates Wnt protein
Jahn, A., Rose, F., Fink, L., Seeger, W., & Schaefer, L. (2009). Wnt1‐ with in vivo biological activity. PLoS One, 3, e2930.
inducible signaling protein–1 mediates pulmonary fibrosis in mice Mudher, A., & Lovestone, S. (2002). Alzheimer's disease–Do tauists and
and is upregulated in humans with idiopathic pulmonary fibrosis. baptists finally shake hands? Trends in Neurosciences, 25, 22–26.
Journal of Clinical Investigation, 119, 772–787. Nakamura, Y., Nawata, M., & Wakitani, S. (2005). Expression profiles and
Krishna, S. M., Seto, S.‐W., Jose, R. J., Li, J., Morton, S. K., Biros, E., functional analyses of Wnt‐related genes in human joint disorders.
Wang, Y., Nsengiyumva, V., Lindeman, J. H., & Loots, G. G. (2017). American Journal of Pathology, 167, 97–105.
Wnt signaling pathway inhibitor sclerostin inhibits angiotensin Niehrs, C. (2010). On growth and form: A cartesian coordinate system of
ii–induced aortic aneurysm and atherosclerosis. Arteriosclerosis, Wnt and BMP signaling specifies bilaterian body axes. Development,
Thrombosis, and Vascular Biology, 37, 553–566. 137, 845–857.
Kumar, A., Chalamalasetty, R. B., Kennedy, M. W., Thomas, S., Inala, S. N., Niehrs, C., & Acebron, S. P. (2012). Mitotic and mitogenic Wnt signaling.
Garriock, R. J., & Yamaguchi, T. P. (2016). Zfp703 is a Wnt/β‐catenin EMBO Journal, 31, 2705–2713.
14 | Cell Biology HAYAT ET AL.

International
Niehrs, C., & Shen, J. (2010). Regulation of Lrp6 phosphorylation. Cellular Sokol, S. Y. (2015). Spatial and temporal aspects of Wnt signaling and
and Molecular Life Sciences, 67(15), 2551‐2562. planar cell polarity during vertebrate embryonic development.
Nusse, R. (2005). Wnt signaling in disease and in development. Cell Seminars in Cell and Developmental Biology, 42, 78–85.
Research, 15, 28–32. Spencer, G. J., Utting, J. C., Etheridge, S. L., Arnett, T. R., & Genever, P. G.
Nusse, R., & Clevers, H. (2017). Wnt/β‐catenin signaling, disease, and (2006). Wnt signaling in osteoblasts regulates expression of the
emerging therapeutic modalities. Cell, 169, 985–999. receptor activator of NFκB ligand and inhibits osteoclastogenesis in
Nusse, R., & Varmus, H. (2012). Three decades of Wnts: A personal vitro. Journal of Cell Science, 119, 1283–1296.
perspective on how a scientific field developed. EMBO Journal, 31, Stamos, J. L., & Weis, W. I. (2013). The β‐catenin destruction complex.
2670–2684. Cold Spring Harbor Perspectives in Biology, 5, a007898.
Olson, E. N., & Schneider, M. D. (2003). Sizing up the heart: Development Steinhart, Z., & Angers, S. (2018). Wnt signaling in development and tissue
redux in disease. Genes & Development, 17, 1937–1956. homeostasis. Development, 145, 145.
Pai, S. G., Carneiro, B. A., Mota, J. M., Costa, R., Leite, C. A., Barroso‐ Surmann‐Schmitt, C., Sasaki, T., Hattori, T., Eitzinger, N., Schett, G.,
Sousa, R., Kaplan, J. B., Chae, Y. K., & Giles, F. J. (2017). Wnt/beta‐ Von Der Mark, K., & Stock, M. (2012). The Wnt antagonist Wif‐1
catenin pathway: Modulating anticancer immune response. Journal interacts with CTGF and inhibits CTGF activity. Journal of Cellular
of Hematology & Oncology, 10, 1–12. Physiology, 227, 2207–2216.
Panakova, D., Sprong, H., Marois, E., Thiele, C., & Eaton, S. (2005). Sutton, G., Kelsh, R. N., & Scholpp, S. (2021). The role of Wnt/β‐Catenin
Lipoprotein particles are required for Hedgehog and Wingless signalling in neural crest development in zebrafish. Frontiers in Cell
signaling. Nature, 435, 58–65. and Developmental Biology, 9, 3450.
Rao, T. P., & Kuhl, M. (2010). An updated overview on Wnt signaling Svegliati, S., Marrone, G., Pezone, A., Spadoni, T., Grieco, A., Moroncini, G.,
pathways: A prelude for more. Circulation Research, 106, Grieco, D., Vinciguerra, M., Agnese, S., & Jungel, A. (2014). Oxidative
1798–1806. DNA damage induces the ATM‐mediated transcriptional
Rasmussen, M. L., Ortolano, N. A., Romero‐Morales, A. I., & Gama, V. suppression of the Wnt inhibitor WIF‐1 in systemic sclerosis and
(2018). Wnt signaling and its impact on mitochondrial and cell cycle fibrosis. Science Signalling, 7, ra84‐ra84.
dynamics in pluripotent stem cells. Genes, 9, 109. Teo, J.‐L., & Kahn, M. (2010). The Wnt signaling pathway in cellular
Raut, D., Vora, A., & Bhatt, L. K. (2022). The Wnt/β‐catenin pathway in proliferation and differentiation: A tale of two coactivators.
breast cancer therapy: A pre‐clinical perspective of its targeting for Advanced Drug Delivery Reviews, 62, 1149–1155.
clinical translation. Expert Review of Anticancer Therapy, 22, 97–114. Tepekoy, F., Akkoyunlu, G., & Demir, R. (2015). The role of Wnt signaling
Salinas, P. C. (2007). Modulation of the microtubule cytoskeleton: A role members in the uterus and embryo during pre‐implantation and
for a divergent canonical Wnt pathway. Trends in Cell Biology, 17, implantation. Journal of Assisted Reproduction And Genetics, 32,
333–342. 337–346.
Salt, E., & Crofford, L. (2012). Rheumatoid arthritis: New treatments, Teratani, T., Tomita, K., Suzuki, T., Furuhashi, H., Irie, R., Nishikawa, M.,
better outcomes. Nurse Practitioner, 37, 16–22. Yamamoto, J., Hibi, T., Miura, S., Minamino, T., Oike, Y., Hokari, R., &
Sanz‐Ezquerro, J. J., Munsterberg, A. E., & Stricker, S. (2017). Signaling Kanai, T. (2018). Aortic carboxypeptidase‐like protein, a WNT ligand,
pathways in embryonic development. Frontiers in Cell and exacerbates nonalcoholic steatohepatitis. The Journal of Clinical
Developmental Biology, 5, 76. Investigation, 128(4), 1581–1596.
Schambony, A., & Wedlich, D. (2007). Wnt‐5a/Ror2 regulate expression Tian, W., Lei, N., Zhou, J., Chen, M., Guo, R., Qin, B., Li, Y., & Chang, L.
of XPAPC through an alternative noncanonical signaling pathway. (2022). Extracellular vesicles in ovarian cancer chemoresistance,
Developmental Cell, 12, 779–792. metastasis, and immune evasion. Cell Death & Disease, 13(1), 1–12.
Schambony, A., & Wedlich, D. (2013). Wnt signaling and cell migration. In Tuller, T., Atar, S., Ruppin, E., Gurevich, M., & Achiron, A. (2013). Common
M. Kuhl (Ed.), Madame Curie bioscience database. Landes Bioscience. and specific signatures of gene expression and protein–protein
Schett, G., Zwerina, J., & David, J.‐P. (2008). The role of Wnt proteins in interactions in autoimmune diseases. Genes Immunity, 14, 67–82.
arthritis. Nature Clinical Practice Rheumatology, 4, 473–480. Valkenburg, K. C., Graveel, C. R., Zylstra‐Diegel, C. R., Zhong, Z., &
Schinner, S. (2009). Wnt‐signaling and the metabolic syndrome. Hormone Williams, B. O. (2011). Wnt/β‐catenin signaling in normal and cancer
and Metabolic Research, 41, 159–163. stem cells. Cancers, 3, 2050–2079.
Segditsas, S., & Tomlinson, I. (2006). Colorectal cancer and genetic Van Camp, J., Beckers, S., Zegers, D., & Van Hul, W. (2014). Wnt signaling
alterations in the Wnt pathway. Oncogene, 25, 7531–7537. and the control of human stem cell fate. Stem Cell Reviews and
Selkoe, D. J., & Hardy, J. (2016). The amyloid hypothesis of Alzheimer's Reports, 10, 207–229.
disease at 25 years. EMBO Molecular Medicine, 8, 595–608. Van Dekken, H., Wink, J. C., Vissers, K. J., Franken, P. F., Schouten, W. R.,
Semenov, M. V., Habas, R., Macdonald, B. T., & He, X. (2007). Snapshot: Hop, W. C., Kuipers, E. J., Fodde, R., & Van Der Woude, cj (2007).
Noncanonical Wnt signaling pathways. Cell, 131, 1378.e1–1378.e2. Wnt pathway‐related gene expression during malignant progression
Sen, M., Chamorro, M., Reifert, J., Corr, M., & Carson, D. A. (2001). in ulcerative colitis. Acta Histochemica, 109, 266–272.
Blockade of Wnt‐5A/frizzled 5 signaling inhibits rheumatoid Van Gijn, M. E., Daemen, M. J., Smits, J. F., & Blankesteijn, W. M. (2002).
synoviocyte activation. Arthritis & Rheumatology, 44, 772–781. The Wnt‐frizzled cascade in cardiovascular disease. Cardiovascular
Shen, G., Ke, J., Wang, Z., Cheng, Z., Gu, X., Wei, Y., Melcher, K., Xu, H. E., Research, 55, 16–24.
& Xu, W. (2015). Structural basis of the norrin‐frizzled 4 interaction. Von Maltzahn, J., Chang, N. C., Bentzinger, C. F., & Rudnicki, M. A. (2012).
Cell Research, 25, 1078–1081. Wnt signaling in myogenesis. Trends in Cell Biology, 22, 602–609.
Shi, J., Chi, S., Xue, J., Yang, J., Li, F., & Liu, X. (2016). Emerging role and Voronkov, A., & Krauss, S. (2013). Wnt/beta‐catenin signaling and small
therapeutic implication of Wnt signaling pathways in autoimmune molecule inhibitors. Current Pharmaceutical Design, 19, 634–664.
diseases. Journal of Immunology Research, 21, 2016–2535. Wallingford, J. B., & Habas, R. (2005). The developmental biology of
Shu, C., Zheng, X., Wuhafu, A., Cicka, D., Doyle, S., Niu, Q., Fan, D., dishevelled: An enigmatic protein governing cell fate and cell
Qian, K., Ivanov, A. A., Du, Y., Mo, X., & Fu, H. (2022). Acquisition of polarity. Development, 132, 4421–4436.
taxane resistance by p53 inactivation in ovarian cancer cells. Acta Wehrli, M., Dougan, S. T., Caldwell, K., O'Keefe, L., Schwartz, S., Vaizel‐
Pharmacologica Sinica, 1–10. Ohayon, D., Schejter, E., Tomlinson, A., & Dinardo, S. (2000). Arrow
Siegel, R. L., Miller, K. D., Fuchs, H. E., & Jemal, A. (2021). Cancer encodes an LDL‐receptor‐related protein essential for wingless
Statistics, 2021. CA: A Cancer Journal for Clinicians, 71, 7–33. signaling. Nature, 407, 527–530.
HAYAT ET AL. Cell Biology | 15
International
Welters, H. J., Oknianska, A., Erdmann, K. S., Ryffel, G. U., & Morgan, N. G. You, J., Nguyen, A. V., Albers, C. G., Lin, F., & Holcombe, R. F. (2008). Wnt
(2008). The protein tyrosine phosphatase‐BL, modulates pancreatic pathway‐related gene expression in inflammatory bowel disease.
b‐cell proliferation by interaction with the Wnt signaling pathway. Digestive Diseases and Sciences, 53, 1013–1019.
Journal of Endocrinology, 197, 543–552. Zhan, T., Rindtorff, N., & Boutros, M. (2017). Wnt signaling in cancer.
Wiese, K. E., Nusse, R., & Van Amerongen, R. (2018). Wnt signaling: Oncogene, 36, 1461–1473.
Conquering complexity. Development, 145, 145. Zhang, Z., Chen, X., Gao, B., Sun, G., Wang, Y., Wang, J., Zhang, T.,
Willert, K., & Jones, K. A. (2006). Wnt signaling: Is the party in the Qian, H., Zhang, Y., Huang, J., Sun, R., & Zhou, L. (2021). PLD1
nucleus? Genes Development, 20, 1394–1404. knockdown reduces metastasis and inflammation of fibroblast‐like
Wodarz, A., & Nusse, R. (1998). Mechanisms of Wnt signaling in synoviocytes in rheumatoid arthritis by modulating NF‐κB and Wnt/
development. Annual Review of Cell and Developmental Biology, 14, β‐catenin pathways. Autoimmunity, 54(7), 398–405.
Zhu, W., Wang, H., & Zhu, D. (2021). Wnt/β‐catenin signaling pathway in
59–88.
lung cancer. Medicine in Drug Discovery, 100113.
Wray, J., & Hartmann, C. (2012). WNTing embryonic stem cells. Trends in
Cell Biology, 22, 159–168.
Xing, Y., Chen, X., Cao, Y., Huang, J., Xie, X., & Wei, Y. (2015). Expression
of Wnt and Notch signaling pathways in inflammatory bowel disease
How to cite this article: Hayat, R., Manzoor, M., & Hussain, A.
treated with mesenchymal stem cell transplantation: Evaluation in a
rat model. Stem Cell Research & Therapy, 6, 1–10. (2022). Wnt signaling pathway: A comprehensive review. Cell
Yang, Y., & Mlodzik, M. (2015). Wnt‐frizzled/planar cell polarity signaling: Biology International, 1–15.
Cellular orientation by facing the wind (Wnt). Annual Review of Cell https://doi.org/10.1002/cbin.11797
and Developmental Biology, 31, 623–646.

You might also like