ArticlePDF AvailableLiterature Review

Canonical Wnt Signaling in the Pathology of Iron Overload-Induced Oxidative Stress and Age-Related Diseases

Wiley
Oxidative Medicine and Cellular Longevity
Authors:

Abstract and Figures

Iron accumulates in the vital organs with aging. This is associated with oxidative stress, inflammation, and mitochondrial dysfunction leading to age-related disorders. Abnormal iron levels are linked to neurodegenerative diseases, liver injury, cancer, and ocular diseases. Canonical Wnt signaling is an evolutionarily conserved signaling pathway that regulates many cellular functions including cell proliferation, apoptosis, cell migration, and stem cell renewal. Recent evidences indicate that iron regulates Wnt signaling, and iron chelators like deferoxamine and deferasirox can inhibit Wnt signaling and cell growth. Canonical Wnt signaling is implicated in the pathogenesis of many diseases, and there are significant efforts ongoing to develop innovative therapies targeting the aberrant Wnt signaling. This review examines how intracellular iron accumulation regulates Wnt signaling in various tissues and their potential contribution in the progression of age-related diseases.
This content is subject to copyright. Terms and conditions apply.
Review Article
Canonical Wnt Signaling in the Pathology of Iron Overload-
Induced Oxidative Stress and Age-Related Diseases
Austin Armstrong ,
1,2
Ashok Mandala,
1,2
Milan Malhotra,
1,2
and Jaya P. Gnana-Prakasam
1,2
1
Department of Ophthalmology, Saint Louis University, St. Louis, USA
2
Department of Biochemistry & Molecular Biology, Saint Louis University, St. Louis, USA
Correspondence should be addressed to Jaya P. Gnana-Prakasam; jaya.gnanaprakasam@health.slu.edu
Received 15 December 2021; Accepted 4 January 2022; Published 25 January 2022
Academic Editor: Alin Ciobica
Copyright © 2022 Austin Armstrong et al. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
Iron accumulates in the vital organs with aging. This is associated with oxidative stress, inammation, and mitochondrial
dysfunction leading to age-related disorders. Abnormal iron levels are linked to neurodegenerative diseases, liver injury, cancer,
and ocular diseases. Canonical Wnt signaling is an evolutionarily conserved signaling pathway that regulates many cellular
functions including cell proliferation, apoptosis, cell migration, and stem cell renewal. Recent evidences indicate that iron
regulates Wnt signaling, and iron chelators like deferoxamine and deferasirox can inhibit Wnt signaling and cell growth.
Canonical Wnt signaling is implicated in the pathogenesis of many diseases, and there are signicant eorts ongoing to
develop innovative therapies targeting the aberrant Wnt signaling. This review examines how intracellular iron accumulation
regulates Wnt signaling in various tissues and their potential contribution in the progression of age-related diseases.
1. Introduction
A multitude of age-related diseases are associated with dis-
rupted cellular iron homeostasis. Wnt/β-catenin signaling,
also known as canonical Wnt signaling, is a crucial pathway
that mediates cell development and proliferation. Hence,
abnormal Wnt signaling can be detrimental and is implicated
in diseases across dierent tissue types. Interestingly, an
overlap between dysregulated iron homeostasis and aberrant
Wnt signaling exists in pathologies including colorectal
carcinoma (CRC) [13], diabetic retinopathy (DR) [4, 5],
and age-related macular degeneration (AMD) [68]. For this
reason, there has been a recent interest in studying the inter-
play between iron homeostasis and canonical Wnt signaling.
Iron is a necessary dietary component, as it is crucial to
processes ranging from oxygen transport by red blood cells
[9] to fetal neurodevelopment [10]. Cellular iron homeosta-
sis and mitochondrial iron homeostasis are interdependent
as mitochondria must import iron to form ironsulfur clus-
ters and heme, which are critical for vital cellular functions.
Imbalances in iron homeostasis leading to its accumulation
can be particularly dangerous, such as in hereditary hemo-
chromatosis [11]. The toxicity due to iron overload is largely
related to the potential of iron to induce oxidative stress,
mitochondrial dysfunction, and inammation. Iron-
mediated oxidative stress arises because excess iron, a proox-
idant, generates reactive oxygen species (ROS) via the
Fenton reaction [12]. ROS can then aect the integrity of
DNA [13], as well as other important proteins and lipids
involved in cellular function [14]. Iron-mediated mitochon-
drial dysfunction is a result of iron-induced mitochondrial
DNA damage, which correlates with defects in ironsulfur
cluster biogenesis, electron transport chain, and heme
synthesis [15]. Iron-mediated inammation occurs due to
the proinammatory nature of both excess labile iron [16]
and iron bound to ferritin [17, 18], an iron storage protein.
Cellular labile iron promotes inammation via interleukin-
1β(IL-1β) secretion and NLRP3 inammasome stimulation
in human monocytes through the NF-κB pathway [16]. Sim-
ilarly, ferritin has been shown to function as a local cytokine
Hindawi
Oxidative Medicine and Cellular Longevity
Volume 2022, Article ID 7163326, 13 pages
https://doi.org/10.1155/2022/7163326
through NF-κB pathway activation in activated rat stellate
cells, leading to signicant increases in inammatory mole-
cules such as IL-1β[17]. Hyperferritinemia, a condition
characterized by excess ferritin levels, is constitutively linked
to disorders like familial hemophagocytic lymphohistiocyto-
sis [17] and macrophage activation syndrome [18] that are
characterized by overactive inammatory responses. In
addition, research has demonstrated that inammation itself
can lead to iron accumulation in tissues such as the retina
[19] and the liver [20] by upregulating hepcidin and thereby
decreasing ferroportin expression. These ndings indicate a
positive feedback loop, where iron-mediated inammation
leads to further iron accumulation and subsequently
increased iron-mediated inammation, oxidative stress,
and mitochondrial dysfunction.
The rationale for much of the research discussed in the
following sections is that oxidative stress [2123], inamma-
tion [24, 25], and mitochondrial dysfunction [26] have also
been indicated as mediators of abnormal canonical Wnt
signaling. This overlap raises the fundamental question of
our review: what role does iron overload have in regulating
Wnt signaling? While the relationship between iron, Wnt
signaling, and cancer has become increasingly well-
established [2729], this same relationship is less docu-
mented in pathologies outside of cancer. The present review
is aimed at providing an update on the canonical Wnt path-
way with respect to iron-mediated aberrant Wnt/β-catenin
signaling in several tissue-specic contexts.
2. The Canonical Wnt Pathway
2.1. Contextualizing Wnt. The rst Wnt gene was discovered
in 1982 as an oncogene activated in mouse models of virally
induced mammary carcinoma and was named Int-1 [30].
Several years later, an important gene in Drosophila larval
development known as the wingless (Wg) gene was found
to be a homolog of the murine Int-1 [31]. The discovery of
the Wg homolog to mouse Int-1 and subsequent research
[32, 33] has demonstrated that the canonical Wnt pathway
is highly conserved amongst a variety of species. Even com-
plexity within the Wnt gene family seems to be conserved, as
the sea anemone Nematostella vectensis shares 11 of the 12
known Wnt gene subfamilies with humans [32]. Because of
the conserved nature of the Wnt genes and the implication
of Wnt regulation in a variety of physiological events,
Wnt/β-catenin research has led to a plethora of discoveries
over the last four decades.
Today, we know of 19 Wnt genes in the mammalian
genome that encode 12 subfamilies of Wnt proteins (Wnts)
[34]. These Wnts are involved in a range of cell events but
centrally act as growth factors and cause cell proliferation
in a variety of cell types during both embryogenesis and
adult tissue homeostasis [3537]. In addition, Wnts contrib-
ute to the directional organization of proliferating cells by
altering gene expression and aecting cytoskeletal and
mitotic architecture [3843]. All Wnt genes encode glyco-
proteins that are about 40 kDA in size and that contain
many conserved cysteine residues [44]. After translation,
important palmitoylation and glycosylation events occur
on Wnt proteins in the endoplasmic reticulum (ER). Palmi-
toylation involves the attachment of a palmitoleic acid to a
conserved serine residue on Wnt proteins [45, 46] by an
O-acyltransferase known as porcupine [47, 48]. This lipid
modication is necessary for Wnt signaling, as it is used
downstream as a binding site for the Wnt receptor, Frizzled
[49], as well as the Wntless protein that aids in Wnt secre-
tion [50, 51]. Of the two major types of posttranslational
modications in the ER, glycosylation is the one less under-
stood. Wnts vary in the number of glycosylated sites present,
and the presence of these glycans seems to have varying
importance. Site-directed mutagenesis of specic glycosyl-
ated regions impairs secretion in certain Wnts [52], while
in other Wnt isoforms, it does not seem to have any aect
at all [53]. Though further studies are required to determine
the exact function of posttranslational Wnt glycosylation,
glycosylation currently has signicance as a signal for the
secretion of few specic Wnt glycoproteins.
2.2. Wnt Tracking and Secretion. The tracking and secre-
tory pathways are two examples of conserved characteristics
between the human Wnt proteins [54, 55]. As aforemen-
tioned, Wnt tracking and secretion are dependent upon
proper lipid modication by porcupine in the ER [47, 48].
This is because Wntless (Wls), a transmembrane protein,
binds the palmitoleic acid in the ER for proper Wnt track-
ing and subsequent Wnt release [50, 51, 56]. Indeed, knock-
outs of Wls or inhibitors of porcupine result in the
accumulation of Wnt proteins in ER [57]. Once bound to
Wnt, Wls will travel with Wnt through the Golgi and ulti-
mately to the plasma membrane [50, 51, 56]. Here, Wls
may act in one of the two ways. One possibility is that
Wls can leave its respective Wnt glycoprotein and return
to the trans-Golgi network [5860]. This occurs via retro-
grade movement involving clathrin-mediated endocytosis
and retromer retrieval mediated by a specialized ER-
retrieval motif [50, 57, 61, 62]. Alternatively, Wls may be
incorporated into the membrane of an exosome along with
Wnt for secretion [63, 64].
The distance of Wnt secretion and subsequent signaling
is a heavily debated topic, as there is evidence for Wnt target-
ing both local [34, 65] and distant [66, 67] cells. At the Wnt
secreting membrane, varying events can occur depending
on the desired distance of secretion. Wnts can be tethered
for contact-dependent interactions with nearby cells [68]
or used for longer range signaling either through expres-
sion on an exosome with Wntless requiring R-SNARE
Ykt6 activity [69] or by binding to solubilizing molecules,
such as Swim [66]. After Wnt tracking and secretion,
signaling occurs. The known Wnt pathways can be orga-
nized into single canonical Wnt pathway and two
noncanonical Wnt pathways [37]. This review will focus
on the canonical Wnt pathway, which involves the nuclear
localization of active β-catenin, as this is the best studied
pathway in relation to iron homeostasis.
2.3. Wnt/β-Catenin Signaling Cascade. In canonical Wnt
signaling, there is a complex system of transducing factors
that cooperate to instill cell responsiveness to the Wnt
2 Oxidative Medicine and Cellular Longevity
ligands. A central player in this cascade is β-catenin, as it is
the primary regulator in Wnt target gene expression. With
cytosolic accumulation of β-catenin, subsequent nuclear
translocation occurs allowing the protein to bind to the T-
Cell Factor/Lymphoid Enhancer Factor (TCF/LEF) tran-
scription factor family [70, 71]. Wnt target cells exhibit the
surface receptors Frizzled (FZD) and low-density
lipoprotein-related receptor protein 5 or 6 (LRP5/6) to initi-
ate the signal cascade [34]. FZD is a family of cell surface
proteins with seven transmembrane domains that contain
both an extracellular N-terminal cysteine-rich domain that
interacts with Wnt [49, 72, 73], as well as a hydrophobic
groove that binds the lipid modications on Wnt [49].
LRP5 and LRP6 serve as coreceptors necessary for Wnt
transduction that form heterodimers with FZD in the pres-
ence of Wnt [74, 75]. It has been posited that LRPs contain
binding regions for Wnt, as described by several anti-LRP
monoclonal antibody studies [76]. While there are a variety
of other Wnt receptors that are important for noncanonical
signaling [77], the FZD and LRP-mediated pathway will
remain the focus of this review as it is the best understood
with relation to iron.
In the absence of Wnt, cytoplasmic β-catenin is bound
by an intact destruction complex and targeted for
ubiquitin-mediated degradation, preventing β-catenin accu-
mulation and nuclear translocation. This ultimately results
in Wnt target gene repression. The destruction complex
consists of proteins Axin, Adenomatous polyposis coli
(APC), and Wilms tumor gene on the X chromosome
(WTX), casein kinase 1-α(CK1α), glycogen synthase kinase
3-β(GSK-3β), and other factors [34]. Axin is a scaolding
protein that interacts with β-catenin and other members of
the destruction complex [34]. APC and WTX, two tumor
suppressor proteins, are also essential for an eective
destruction complex [34]. CK1αand GSK-3βare serine-
threonine kinases that perform multiple functions related
to Wnt signaling. In terms of the destruction complex, they
phosphorylate Axin-bound β-catenin to target it for ubiqui-
tination [34, 78]. β-Transducin repeat-containing protein
(β-TrCP), a component of the E3 ubiquitin ligase complex,
recognizes phosphorylated β-catenin and catalyzes the addi-
tion of ubiquitin polymers to direct β-catenin for degrada-
tion by proteasomes preventing their nuclear translocation
[78]. The absence of β-catenin in the nucleus leaves the
repressive TCF/LEF complex active, which then permits
Groucho/transducin-like enhancer (Gro/TLE) family
proteins to recruit histone deacetylases that inhibit Wnt
target gene expression [79].
In the presence of Wnt, canonical Wnt stimulation
begins with Wnt ligand facilitating the heterodimerization
of FZD and its coreceptor, such as LRP5 or LRP6. Ligation
induces a conformational change in the receptors and by a
currently unclear mechanism, recruits, and activates GSK-
3βand CK1γ[8082]. At the membrane, GSK-3βand
CK1γphosphorylate the LRP tail at the PPPSP motif [83]
and in regions adjacent to the PPPSP motifs [84], respec-
tively. This sequential phosphorylation of the LRP tail
results in Axin relocation to the plasma membrane and its
physical removal from the cytosol, inhibiting the formation
of the β-catenin destruction complex [34]. β-Catenin then
accumulates in the cytosol and subsequently translocates to
the nucleus. Nuclear β-catenin interacts with the TCF/LEF
interface discussed earlier to activate Wnt target gene
expression [70, 71, 85, 86] as shown in Figure 1.
2.4. Wnt/β-Catenin Modulation. As previously discussed,
Wnt/β-catenin signaling plays a multitude of roles in a host
of tissues throughout life. Wnts generally function as growth
factors to cause proliferation and have more than 100 down-
stream target genes [3537, 87]. Many of these target genes
are important cell cycle regulators including C-Myc [88]
and Cyclin D [89]. Others include those related to angiogen-
esis like VEGF [90], as well as nearly every component of the
Renin-Angiotensin-System (RAS) [91]. Wnt signaling acti-
vates mitochondrial biogenesis, in turn producing elevated
levels of ROS and oxidative damage [26], which is consid-
ered to be the cause for certain pathological consequences.
Given the wide-ranging functions of canonical Wnt signal-
ing, it is logical that dysfunctional Wnt/β-catenin signaling
would have a variety of negative implications. The nature
of this aberrance, however, is not as intuitive. In some
diseases, such as in cases of CRC [92] or AMD [7, 8], Wnt
signaling is pathologically upregulated, while in others, such
as in Norrie disease [93] or osteopenia [94], Wnt signaling is
pathologically downregulated. For this reason, a variety of
positive and negative modulators exist and are important
for the understanding of Wnt signaling.
2.4.1. Positive Modulation. Positive modulators of Wnt/β-
catenin signaling are those that lead to an increase in active
β-catenin translocating to the nucleus and thus increasing
the expression of Wnt target genes. In cases of chronic
wounds [95], vitiligo [96, 97], or other diseases character-
ized by Wnt/β-catenin inactivation, positive Wnt modula-
tors can be protective. However, in the context of diseases
characterized by Wnt/β-catenin activation, positive Wnt
modulators may be harmful. For example, patients being
treated with lithium, a canonical Wnt activator, were
shown to have a signicantly increased chance of develop-
ing renal carcinogenesis [98].
One important positive regulation pathway of vertebrate
Wnt signaling involves the four R-spondin (RSPO) proteins,
which are characterized by two furin domains and a throm-
bospondin domain [99]. In the absence of RSPO ligands, the
disheveled protein promotes the destruction of FZD on the
membrane by the E3 ligases [100]. However, in the presence
of RPSO proteins, the RSPO ligands will bind to leucine-rich
repeat-containing G protein-coupled 5 (Lgr5) family recep-
tors [101103] resulting in the membrane clearance of E3
ligases RNF43 and ZNRF3 [104, 105]. In eect, this pro-
motes FZD accumulation on the membrane and increases
the sensitivity of a cell to Wnt ligands [104, 105]. Norrin is
a second example of a secreted protein that positively
modulates Wnt signaling [106]. It is characterized by its
cysteine-knot motif [107] and, with the help of its coreceptor
tetraspanin 12 [108, 109], acts by binding directly to the
FZD4/LRP5 complex to activate the canonical Wnt pathway
[93, 110, 111].
3Oxidative Medicine and Cellular Longevity
In addition to the well-studied RSPO and Norrin posi-
tive modulators, there are several others that have been dis-
covered over the years. Protein phosphatase-2A (PP2A) is a
serine-threonine phosphatase composed of three subunits
that can positively regulate the canonical Wnt pathway by
dephosphorylating a variety of proteins including β-catenin
[112]. Other examples include microRNAs miR-135a and
miR-135b that directly repress APC expression in colorectal
cancer cells, thereby destabilizing the β-catenin destruction
complex and allowing Wnt target gene expression [113].
Still, others like heparin sulfate proteoglycans serve as cofac-
tors to promote Wnt signaling in the control of C. elegans
mitotic spindle orientation [114], distal-tip cell migration
[115], and neuronal positioning [116].
2.4.2. Negative Modulation. Negative modulators of Wnt/β-
catenin signaling are those that lead to the destruction of β-
catenin and a decrease in the expression of Wnt target genes.
This may be protective in diseases characterized by Wnt/β-
catenin activation, but deleterious during diseases character-
ized by Wnt/β-catenin inactivation [117]. The potentially
deleterious eect of negative modulators can be seen in the
progression of Alzheimers disease, a disease characterized
by Wnt inactivation [118]. Specically, the worsening of
amyloid beta plaque accumulation and subsequent synaptic
loss was observed with the presence of the negative regulator
Dickkopf-1 [119, 120].
A prototypical downregulator of Wnt signaling is
Notum, an extracellular enzyme that inhibits Wnt proteins
by removing their vital palmitoylate residues [121, 122].
Notum also partakes in a negative feedback loop, as the
TCF/LEF family transcription factors have binding sites in
the Notum promoter region to increase its expression [121,
123]. In addition to Notum, extracellular Wnt inhibitors
include Wnt inhibitory factor (WIF) proteins [124, 125]
and secreted FZD-related proteins (SFRPs) [126128] that
directly bind Wnt proteins to inhibit signaling. There is also
another important class of negative Wnt modulators that act
as transmembrane antagonists by binding and subsequently
blocking Wnt receptors from interacting with Wnt. These
include Shisa [129], Wnt-activated inhibitory factor [130],
Adenomatosis polyposis coli downregulated 1 [131], and
Tiki1 [132]. Lastly, miRNAs have been a recent area of
research that have been implicated for negatively modulat-
ing Wnt signaling. Some examples include miR-200a, which
decreases Wnt target gene transcription [133], miR-21,
which directly represses Wnt1 protein production [134],
and miR-184, which targets Wnt coreceptor FZD-7 to pre-
vent Wnt signaling in retinal neovascularization [135].
In addition to the Notum feedback loop, there are several
other negative feedback loops that regulate canonical Wnt
signaling. An important one of these was mentioned in our
discussion of positive Wnt regulation, with regard to the
E3 ligases RNF43 and ZNRF3. These two ligases are known
to control FZD membrane expression by mediating the
ubiquitination of the FZD cytoplasmic loops, which leads
to FZD lysosomal degradation [136, 137]. A third canonical
Wnt signaling negative feedback loop is the conductin/
Axin2 loop. Axin2 is a Wnt target gene, and its expression
mimics Axin in the destruction complex to increase β-
catenin degradation [138, 139], which subsequently downre-
gulates Wnt target gene expression.
2.5. Iron-Mediated Wnt/β-Catenin Signaling during
Pathological Conditions. In the remainder of this review,
we will discuss the recent ndings on how iron modulates
APC Axin
CK1αGSK3β
L
R
P
6
Frizzled
Active destruction complex
APC
Axin GSK3β
CK1α
β-catenin
Frizzled
Wnt
ligands
Fe
Fe
Fe
Fe
Fe
??
p
Inactive destruction complex
β-catenin
pp
Degraded β-catenin
oxidative
stress
β-catenin
TCF/LEF
Axi n2
cMYC
CCND
Renin angiotensin system
OFF STATE ON STATE
Liver Retina
L
R
P
6
Figure 1: A schematic overview of tissue-dependent Wnt signaling during conditions of iron overload.
4 Oxidative Medicine and Cellular Longevity
Wnt/β-catenin signaling leading to multiple disorders as
outlined in Figure 2.
2.5.1. Cancer. The role of Wnt signaling in cancerous pathol-
ogies is historically validated, with the discovery of Wnt
signaling based on experimentation involving mouse models
of breast cancer and cancer-causing retrovirus mouse mam-
mary tumor Virus (MMTV) [140]. In experiments that
lasted until the 1990s, MMTV was found to insert proviral
DNA into specic regions of the mouse genome, inducing
oncogene formation and mammary hyperplasia, and these
genes were all later connected to Wnt signaling [141143].
In addition, the growing clinical signicance of Wnt signal-
ing is also closely related to cancer, as the most well-known
pathology that involves Wnt is familial adenomatous poly-
posis (FAP) [144]. In FAP, mutation in APC and stabiliza-
tion of β-catenin results in increased colonic cell
proliferation, yielding a presentation of colonic polyps asso-
ciated with increased colorectal cancer risk [145147]. In
addition to MMTV studies and FAP being related to cancer,
recent research has demonstrated that Wnt contributes to
gastrointestinal, hematopoietic, breast, skin, brain, and
colonic cancers [144].
With researchers investigating more on the connection
between cancer and Wnt, iron has emerged as a prominent
Wnt regulator in the context of proliferative pathologies.
In studies using APC knockout cell lines Caco-2 and
SW480, it was highlighted that growth on FeSO4-loaded
media and hemin-loaded media, both rich in iron, increased
Wnt signaling [27]. Thus, iron-mediated Wnt signaling
upregulation was demonstrated in cell lines that resembled
FAP-associated cancer cells with the presence of APC
knockout [27]. Another recent study that investigated Nrf2
mutations and their association with hepatocellular carci-
noma reported that alterations in iron homeostasis and
subsequent Wnt signaling activation play a role in the occur-
rence and proliferation of hepatocellular carcinoma [148].
These studies show a potential contribution of iron in Wnt
upregulation during cancerous pathologies.
Two recent reports revealed that iron chelators can
reverse the Wnt activation during cancer. In the rst study,
a specic iron chelator HQBA inhibited Wnt signaling in a
variety of cancer cell lines and inhibited growth of
mammary tumors in MMTV-Wnt1 mouse models of Wnt-
dependent breast cancer and in MMTV-PyMT mouse
models of Wnt-independent breast cancer [29] indicating
the plausible proliferative eects of iron both dependent
and independent of Wnt signaling. The signicance of this
study with relation to iron is garnered from the fact that
HQBA premixed with iron prior to cancer cell line exposure
prevented its ability to reverse Wnt/β-catenin activation in
several tissue-specic cancer cell lines conrming that the
antitumor eect of HQBA is through iron chelation [29].
A second study identied acyl hydrazones as inhibitors
of Wnt/β-catenin signaling by chelating iron. Upon treat-
ment with acyl hydrazones, intracellular iron was chelated,
Wnt/β-catenin activation was reversed, and cell prolifera-
tion signicantly decreased in human CRC cell lines
SW480 and DID-1 [149]. Taken together, these studies
strongly suggest that excess iron upregulates Wnt/β-
catenin signaling in certain cancerous pathologies, and
that iron chelation may be a potential therapeutic strategy
to prevent cancer progression.
2.5.2. Neurodegenerative Disorders. The relationship
between iron overload and aberrant Wnt signaling has been
described in two studies that suggest excess iron in neuronal
cells pathologically increases Wnt/β-catenin signaling. The
rst study discusses posthemorrhagic chronic hydrocephalus
(PHCH), a potentially fatal medical condition often arising
after an intraventricular hemorrhage (IVH) [150]. PHCH
is known to cause an increase in both cerebral spinal uid
(CSF) iron concentration [151] and ferritin content within
the brain [152]. Additionally, PHCH is characterized by
brotic changes, which, in other tissues, are known to be
linked to dysregulated Wnt/β-catenin signaling [153, 154].
As iron accumulation and brotic changes are both major
players in the progression of PHCH, authors investigated
the therapeutic ability of the iron chelator deferoxamine
(DFX) in the treatment of abnormal Wnt signaling in a
PHCH model. The study revealed that by chelating excess
iron from CSF and brain ferritin, DFX normalized the
upregulated Wnt/β-catenin signaling seen in PHCH after
an IVH. This served to broadly improve PHCH occurrence
and severity [155]. Similarly, another recent study reported
that upregulation of Wnt/β-catenin signaling in neural pro-
genitor cells (NPCs) could be normalized by DFX, leading to
an increase in NPC dierentiation and outgrowth [156].
These studies indicate that iron overload is detrimental to
the cells in neural tissues due to Wnt/β-catenin activation,
which can be therapeutically resolved with DFX treatment.
2.5.3. Bone Remodeling. Iron [157, 158] and canonical Wnt
signaling [159] are both important players in the mainte-
nance of bone. While dietary iron is critical for healthy bone
density in populations such as postmenopausal women
[157], iron overload inducing diseases like β-thalassemia
and hemochromatosis are often associated with decreased
bone density and integrity [160]. Indeed, chelation of iron
with deferasirox has been shown to improve bone density
in patients with β-thalassemia [161], suggesting that excess
iron is detrimental to bone health. On the other hand,
normal Wnt signaling is crucial for appropriate bone remod-
eling because Wnt signaling is involved in both osteoblast
dierentiation and osteoclastogenesis [159, 162, 163]. In
addition, osteoporosis, a disease characterized by reduced
bone density due to ineective bone remodeling [164], is
associated with both decreased Wnt signaling and oxidative
stress [165]. For these reasons, a recent article investigated
iron-dependent Wnt signaling in bone marrow stromal cells
dierentiated towards osteoblasts [166]. The study found
that excess iron is detrimental to osteoblast dierentiation,
and that iron chelation using DFX can reverse the negative
eects of iron overload in the same cells. Moreover, the
results conclude that induction of Wnt5a expression by
DFX is the mediator of this recovery, which occurs through
the PI3K and NFAT pathways [166]. These results indirectly
indicate that excess iron reduces Wnt expression in bone;
5Oxidative Medicine and Cellular Longevity
iron chelation by DFX treatment induces Wnt5a expression
to recover the Wnt signaling. However, the study neither
demonstrated directly that iron overload decreases Wnt sig-
naling, nor showed that chelation of iron brings back Wnt
signaling in the osteoblasts. A nal consideration is that
Wnt5a operates through a noncanonical Wnt pathway
[167]. Despite these limitations, we can still conclude that
the induction of Wnt through DFX treatment is therapeutic
for iron-overloaded osteoblasts. Moreover, recent research
on an in vivo iron-induced osteoporotic rat model demon-
strated similar reduction of Wnt signaling that was recov-
ered with DFX in bone tissues [168]. Another in vivo
mouse study suggests that hepcidin-induced osteoporosis,
mediated through iron overload, may also be targeted via
inhibition of Forkhead box O3a to recover canonical Wnt
signaling [169].
Interestingly, while iron overload leads to decreased
activity of the Wnt pathway in osteoblasts, another study
reported that iron overload induces ROS-mediated apopto-
sis and upregulation of the Wnt pathway in bone marrow
mesenchymal cells of patients with myelodysplastic syn-
dromes [170]. This highlights the variation in Wnt signaling
in response to iron overload between dierent cell types even
within the context of a single tissue like bone.
2.5.4. Liver Injury. Chronic iron overload in hepatocytes,
such as in cases of hemochromatosis, is associated with
severe hepatic injury and cancer largely because of iron-
induced steatosis, brosis, inammation, and oxidative
stress [171, 172]. Interestingly, liver-specicβ-catenin
knockout (KO) mice are known to have similar hepatic
pathology resulting from factors such as brosis and oxida-
tive stress [173, 174]. For these reasons, a recent study inves-
tigated the role of iron overload in hepatic pathology of
liver-specicβ-catenin KO mice [175]. This study con-
cluded that following iron overload, liver-specicβ-catenin
KO mice have increased steatohepatitis and brosis that is
preceded by both inammation and oxidative stress. More-
over, they demonstrate that treatment with an antioxidant
can help prevent this disease progression. In eect, the
absence of β-catenin exacerbates the detrimental eects of
hepatic iron overload. The study also showed that iron over-
load led to a decrease in β-catenin in mice where it was not
knocked out. This is postulated to be an initial protective
mechanism, because these mice had less expression of
Cype2e1, a β-catenin target associated with oxidative stress
[175]. However, the delicate nature of Wnt/β-catenin
expression was proven to be detrimental in cases of β-
catenin KO which increased inammation and eventually
lead to Cype2e1 reappearance, and, thus, further decrease
in β-catenin due to prolonged iron overload could prove to
be similarly detrimental in liver tissue. We recently showed
that iron overload in normal mouse liver decreases Wnt
pathway by suppressing Sirtuin 3 signaling [176]. Thus,
studies so far indicate that iron overload downregulates
Wnt signaling in liver contributing towards the progression
of liver brosis.
2.5.5. Ocular Diseases. Visual impairment is a national and a
global health concern that impairs physical and mental
health in aected individuals. The retina is one of the highest
energy consuming organs in the body. Impaired metabolic
changes in eye diseases often drive neuronal and vascular
pathologies [177, 178]. The most prevalent retinal degenera-
tive diseases are age-related macular degeneration and
diabetic retinopathy. Identication of risk factors and the
molecular mechanisms that govern neuronal cell loss and
vascular changes is of interest for translational researchers
and clinicians to discover preventive and interventional
therapeutics for retinal disorders of the eye.
Retina expresses many iron containing proteins like
RPE65, an isomerohydrolase that converts all-trans-retinyl
ester to 11-cis-retinol in the visual cycle [179], fatty acid
desaturase, an enzyme involved in synthesis of membrane
lipids 9 [180], and guanylate cyclase, involved in the synthe-
sis of cGMP, a second messenger in the phototransduction
pathway. So iron is critical for the retinal health. However,
excess iron due to its prooxidant property induces oxidative
stress [181] resulting in impaired retinal function. We have
previously shown that excess iron alters retinal barrier integ-
rity and accelerates the retinal cell loss by augmenting oxida-
tive stress and inammasome activation [4]. Retinal iron
accumulation has been reported in the human patients and
mouse models of AMD and DR [4, 6]. Similarly, multiple
Neurodegenerative
diseases
Tumor cell
proliferation
AGING
Bone remodeling
Liver injury
Ocular disorders
IRON OVERLOAD
Fe
Fe
Fe
FeFe
Fe
Fe
Fe
Fe
Fe
Fe
Fe
Fe
Aberrant Wnt/β-catenin signaling
Fe
Fe
Fe
Fe
Fe
Fe
Fe
Figure 2: Iron accumulation associated with aging modulates canonical Wnt/β-catenin signaling leading to the progression of liver injury,
neurodegenerative diseases, bone remodeling, cancer, and ocular disorders.
6 Oxidative Medicine and Cellular Longevity
studies have conrmed the pathogenic role of canonical Wnt
signaling in the etiology of AMD [7, 8, 182] and DR [5, 183].
Recently, we reported a critical role for iron-induced oxida-
tive stress in the activation of canonical Wnt pathway medi-
ated by peroxisome proliferator-activated receptor- (PPAR-)
alpha signaling in retina [184]. A comprehensive under-
standing of what drives iron overload and the downstream
Wnt signaling in an increasingly wide range of diseases
would help in preventing the progression of diseases at an
earlier stage in the future.
3. Conclusion
A multivariate genomic scan has revealed high levels of iron
in the blood to be intimately associated with reduced health-
span [185]. Also, intake of iron-rich diet and excess iron
supplements are implicated in many age-related disorders.
Iron-mediated oxidative stress induces inammation and
mitochondrial dysfunction thereby playing a critical role in
the progression of cancer, neurodegenerative disorders, oste-
oporosis, liver brosis and steatosis, and ocular diseases.
Similarly, aberrant changes in the canonical Wnt/β-catenin
signaling pathway are a hallmark of cancer, diabetes melli-
tus, and other degenerative disorders. Elucidating the com-
plex interplay between iron and Wnt pathway could lead
to new insights into the mechanisms of disease progression
and enrich our understanding of the aging biology. Control
of body iron stores and Wnt inhibitors can thus serve as
promising clinical targets to overcome the myriad ramica-
tions of aging.
Data Availability
Data supporting this Review are from previously reported
studies, which have been cited.
Conflicts of Interest
The authors declare that they have no competing interests.
AuthorsContributions
A.A., A.M., M.M., and J.P.G wrote, edited, and nalized the
manuscript. Austin Armstrong and Ashok Mandala contrib-
uted equally to this work.
Acknowledgments
The nancial support from the National Eye Institute (R01-
EY031008) and the American Heart Association
(14SDG20510062) is kindly acknowledged.
References
[1] D. N. Seril, J. Liao, K. L. K. Ho, A. Warsi, C. S. Yang, and G. Y.
Yang, Dietary iron supplementation enhances DSS-induced
colitis and associated colorectal carcinoma development in
mice,Digestive Diseases and Sciences, vol. 47, no. 6,
pp. 12661278, 2002.
[2] J. N. Ilsley, G. S. Belinsky, K. Guda et al., Dietary iron pro-
motes azoxymethane-induced colon tumors in mice,Nutri-
tion and Cancer, vol. 49, no. 2, pp. 162169, 2004.
[3] M. J. Brookes, S. Hughes, F. E. Turner et al., Modulation of
iron transport proteins in human colorectal carcinogenesis,
Gut, vol. 55, no. 10, pp. 14491460, 2006.
[4] K. Chaudhary, W. Promsote, S. Ananth et al., Iron overload
accelerates the progression of diabetic retinopathy in associa-
tion with increased retinal renin expression,Scientic
Reports, vol. 8, no. 1, p. 3025, 2018.
[5] Q. Chen and J. X. Ma, Canonical Wnt signaling in diabetic
retinopathy,Vision Research, vol. 139, pp. 4758, 2017.
[6] P. Hahn, A. H. Milam, and J. L. Dunaief, Maculas
aected by age-related macular degeneration contain
increased chelatable iron in the retinal pigment epithelium
and Bruch's membrane,Archives of Ophthalmology,
vol. 121, no. 8, pp. 10991105, 2003.
[7] T. Zhou, Y. Hu, Y. Chen et al., The pathogenic role of the
canonical Wnt pathway in age-related macular degenera-
tion,Investigative Ophthalmology & Visual Science, vol. 51,
no. 9, pp. 43714379, 2010.
[8] J. Tuo, Y. Wang, R. Cheng et al., Wnt signaling in age-
related macular degeneration: human macular tissue and
mouse model,Journal of Translational Medicine, vol. 13,
p. 330, 2015.
[9] N. C. Andrews, Disorders of iron metabolism,The New
England Journal of Medicine, vol. 341, no. 26, pp. 1986
1995, 1999.
[10] M. K. Georgie,The role of iron in neurodevelopment: fetal
iron deciency and the developing hippocampus,Biochemi-
cal Society Transactions, vol. 36, no. 6, pp. 12671271, 2008.
[11] B. K. Crownover and C. J. Covey, Hereditary hemochroma-
tosis,American Family Physician, vol. 87, no. 3, pp. 183190,
2013.
[12] B. Halliwell and J. M. Gutteridge, Oxygen toxicity, oxygen
radicals, transition metals and disease,The Biochemical
Journal, vol. 219, no. 1, pp. 114, 1984.
[13] S. Toyokuni, Iron-induced carcinogenesis: the role of redox
regulation,Free Radical Biology & Medicine, vol. 20, no. 4,
pp. 553566, 1996.
[14] T. S. Koskenkorva-Frank, G. Weiss, W. H. Koppenol, and
S. Burckhardt, The complex interplay of iron metabolism,
reactive oxygen species, and reactive nitrogen species:
insights into the potential of various iron therapies to induce
oxidative and nitrosative stress,Free Radical Biology & Med-
icine, vol. 65, pp. 11741194, 2013.
[15] P. J. Urrutia, N. P. Mena, and M. T. Nunez, The interplay
between iron accumulation, mitochondrial dysfunction, and
inammation during the execution step of neurodegenerative
disorders,Frontiers in Pharmacology, vol. 5, p. 38, 2014.
[16] K. Nakamura, T. Kawakami, N. Yamamoto et al., Activation
of the NLRP3 inammasome by cellular labile iron,Experi-
mental Hematology, vol. 44, no. 2, pp. 116124, 2016.
[17] R. G. Ruddell, D. Hoang-le, J. M. Barwood et al., Ferritin
functions as a proinammatory cytokine via iron-
independent protein kinase C zeta/nuclear factor kappaB-
regulated signaling in rat hepatic stellate cells,Hepatology,
vol. 49, no. 3, pp. 887900, 2009.
[18] C. Rosário, G. Zandman-Goddard, E. G. Meyron-Holtz, D. P.
DCruz, and Y. Shoenfeld, The hyperferritinemic syndrome:
macrophage activation syndrome, Still's disease, septic shock
7Oxidative Medicine and Cellular Longevity
and catastrophic antiphospholipid syndrome,BMC Medi-
cine, vol. 11, no. 1, 2013.
[19] J. P. Gnana-Prakasam, P. M. Martin, B. A. Mysona, P. Roon,
S. B. Smith, and V. Ganapathy, Hepcidin expression in
mouse retina and its regulation via lipopolysaccharide/Toll-
like receptor-4 pathway independent of Hfe,The Biochemi-
cal Journal, vol. 411, no. 1, pp. 7988, 2008.
[20] K. Y. Yeh, M. Yeh, and J. Glass, Hepcidin regulation of fer-
roportin 1 expression in the liver and intestine of the rat,
American Journal of Physiology. Gastrointestinal and Liver
Physiology, vol. 286, no. 3, pp. G385G394, 2004.
[21] H. C. Korswagen, Regulation of the Wnt/beta-catenin path-
way by redox signaling,Developmental Cell, vol. 10, no. 6,
pp. 687-688, 2006.
[22] Y. Funato, T. Michiue, M. Asashima, and H. Miki, The
thioredoxin-related redox-regulating protein nucleoredoxin
inhibits Wnt-beta-catenin signalling through dishevelled,
Nature Cell Biology, vol. 8, no. 5, pp. 501508, 2006.
[23] A. Vikram, Y. R. Kim, S. Kumar et al., Canonical Wnt
signaling induces vascular endothelial dysfunction via
p66Shc-regulated reactive oxygen species,Arteriosclerosis,
Thrombosis, and Vascular Biology, vol. 34, no. 10,
pp. 23012309, 2014.
[24] K. Oguma, H. Oshima, M. Aoki et al., Activated macro-
phages promote Wnt signalling through tumour necrosis
factor-alpha in gastric tumour cells,The EMBO Journal,
vol. 27, no. 12, pp. 16711681, 2008.
[25] B. Ma and M. O. Hottiger, Crosstalk between Wnt/β-
Catenin and NF-κB signaling pathway during inammation,
Frontiers in Immunology, vol. 7, p. 378, 2016.
[26] J. C. Yoon, A. Ng, B. H. Kim, A. Bianco, R. J. Xavier, and S. J.
Elledge, Wnt signaling regulates mitochondrial physiology
and insulin sensitivity,Genes & Development, vol. 24,
no. 14, pp. 15071518, 2010.
[27] M. J. Brookes, J. Boult, K. Roberts et al., A role for iron in
Wnt signalling,Oncogene, vol. 27, no. 7, pp. 966975, 2008.
[28] Z. Chen, J. Sun, T. Li et al., Iron chelator-induced up-
regulation of Ndrg1 inhibits proliferation and EMT process
by targeting Wnt/β-catenin pathway in colon cancer cells,
Biochemical and Biophysical Research Communications,
vol. 506, no. 1, pp. 114121, 2018.
[29] G. S. Coombs, A. A. Schmitt, C. A. Canning et al., Modula-
tion of Wnt/β-catenin signaling and proliferation by a fer-
rous iron chelator with therapeutic ecacy in genetically
engineered mouse models of cancer,Oncogene, vol. 31,
no. 2, pp. 213225, 2012.
[30] R. Nusse and H. E. Varmus, Many tumors induced by the
mouse mammary tumor virus contain a provirus integrated
in the same region of the host genome,Cell, vol. 31, no. 1,
pp. 99109, 1982.
[31] F. Rijsewijk, M. Schuermann, E. Wagenaar, P. Parren,
D. Weigel, and R. Nusse, The Drosophila homolog of the
mouse mammary oncogene int-1 is identical to the segment
polarity gene wingless,Cell, vol. 50, no. 4, pp. 649657, 1987.
[32] A. Kusserow, K. Pang, C. Sturm et al., Unexpected complex-
ity of the Wnt gene family in a sea anemone,Nature,
vol. 433, no. 7022, pp. 156160, 2005.
[33] A. P. McMahon and R. T. Moon, Ectopic expression of the
proto-oncogene int-1 in Xenopus embryos leads to duplica-
tion of the embryonic axis,Cell, vol. 58, no. 6, pp. 1075
1084, 1989.
[34] H. Clevers and R. Nusse, Wnt/β-Catenin Signaling and Dis-
ease,Cell, vol. 149, no. 6, pp. 11921205, 2012.
[35] C. Y. Logan and R. Nusse, The Wnt signaling pathway in
development and disease,Annual Review of Cell and Devel-
opmental Biology, vol. 20, pp. 781810, 2004.
[36] F. J. Staal, F. Weerkamp, M. R. M. Baert et al., Wnt target
genes identied by DNA microarrays in immature CD34+
thymocytes regulate proliferation and cell adhesion,Journal
of Immunology, vol. 172, no. 2, pp. 10991108, 2004.
[37] H. Clevers, Wnt/β-Catenin Signaling in Development and
Disease,Cell, vol. 127, no. 3, pp. 469480, 2006.
[38] C. Niehrs and S. P. Acebron, Mitotic and mitogenic Wnt sig-
nalling,The EMBO Journal, vol. 31, no. 12, pp. 27052713,
2012.
[39] B. Goldstein, H. Takeshita, K. Mizumoto, and H. Sawa, Wnt
signals can function as positional cues in establishing cell
polarity,Developmental Cell, vol. 10, no. 3, pp. 391396,
2006.
[40] Y. L. Huang and C. Niehrs, Polarized Wnt signaling regu-
lates ectodermal cell fate in Xenopus,Developmental Cell,
vol. 29, no. 2, pp. 250257, 2014.
[41] J. Schneider, A. A. Arraf, M. Grinstein, R. Yelin, and T. M.
Schultheiss, Wnt signaling orients the proximal-distal axis
of chick kidney nephrons,Development, vol. 142, no. 15,
pp. 26862695, 2015.
[42] K. M. Loh, R. van Amerongen, and R. Nusse, Generating cel-
lular diversity and spatial form: Wnt signaling and the evolu-
tion of multicellular animals,Developmental Cell, vol. 38,
no. 6, pp. 643655, 2016.
[43] H. Sawa, Control of cell polarity and asymmetric division in
C. elegans,Current Topics in Developmental Biology,
vol. 101, pp. 5576, 2012.
[44] K. Tanaka, Y. Kitagawa, and T. Kadowaki, Drosophila seg-
ment polarity gene product porcupine stimulates the post-
translational N-glycosylation of wingless in the endoplasmic
reticulum,The Journal of Biological Chemistry, vol. 277,
no. 15, pp. 1281612823, 2002.
[45] K. Willert, J. D. Brown, E. Danenberg et al., Wnt proteins are
lipid-modied and can act as stem cell growth factors,
Nature, vol. 423, no. 6938, pp. 448452, 2003.
[46] R. Takada, Y. Satomi, T. Kurata et al., Monounsaturated
fatty acid modication of Wnt protein: its role in Wnt secre-
tion,Developmental Cell,vol. 11, no. 6, pp. 791801, 2006.
[47] K. Hofmann, A superfamily of membrane-bound O-
acyltransferases with implications for wnt signaling,Trends
in Biochemical Sciences, vol. 25, no. 3, pp. 111-112, 2000.
[48] T. Kadowaki, E. Wilder, J. Klingensmith, K. Zachary, and
N. Perrimon, The segment polarity gene porcupine encodes
a putative multitransmembrane protein involved in Wingless
processing,Genes & Development, vol. 10, no. 24, pp. 3116
3128, 1996.
[49] C. Y. Janda, D. Waghray, A. M. Levin, C. Thomas, and K. C.
Garcia, Structural basis of Wnt recognition by Frizzled,Sci-
ence, vol. 337, no. 6090, pp. 5964, 2012.
[50] J. Yu, J. Chia, C. A. Canning, C. M. Jones, F. A. Bard, and
D. M. Virshup, WLS retrograde transport to the endoplas-
mic reticulum during Wnt secretion,Developmental Cell,
vol. 29, no. 3, pp. 277291, 2014.
[51] P. Herr and K. Basler, Porcupine-mediated lipidation is
required for Wnt recognition by Wls,Developmental Biol-
ogy, vol. 361, no. 2, pp. 392402, 2012.
8 Oxidative Medicine and Cellular Longevity
[52] H. Komekado, H. Yamamoto, T. Chiba, and A. Kikuchi,
Glycosylation and palmitoylation of Wnt-3a are coupled to
produce an active form of Wnt-3a,Genes to Cells, vol. 12,
no. 4, pp. 521534, 2007.
[53] X. Tang, Y. Wu, T. Y. Belenkaya et al., Roles of N-
glycosylation and lipidation in Wg secretion and signaling,
Developmental Biology, vol. 364, no. 1, pp. 3241, 2012.
[54] R. Najdi, K. Prott, S. Sprowl et al., A uniform human Wnt
expression library reveals a shared secretory pathway and
unique signaling activities,Dierentiation, vol. 84, no. 2,
pp. 203213, 2012.
[55] K. D. Prott and D. M. Virshup, Precise regulation of por-
cupine activity is required for physiological Wnt signaling,
The Journal of Biological Chemistry, vol. 287, no. 41,
pp. 3416734178, 2012.
[56] G. S. Coombs, J. Yu, C. A. Canning et al., WLS-dependent
secretion of WNT3A requires Ser 209 acylation and vacuolar
acidication,Journal of Cell Science, vol. 123, no. 19,
pp. 33573367, 2010.
[57] N. Moti, J. Yu, G. Boncompain, F. Perez, and D. M. Virshup,
Wnt trac from endoplasmic reticulum to lopodia,PLoS
One, vol. 14, no. 2, article e0212711, 2019.
[58] F. Port, M. Kuster, P. Herr et al., Wingless secretion pro-
motes and requires retromer-dependent cycling of Wntless,
Nature Cell Biology, vol. 10, no. 2, pp. 178185, 2008.
[59] T. Y. Belenkaya, Y. Wu, X. Tang et al., The retromer com-
plex inuences Wnt secretion by recycling wntless from
endosomes to the trans-Golgi network,Developmental Cell,
vol. 14, no. 1, pp. 120131, 2008.
[60] X. Franch-Marro, F. Wendler, S. Guidato et al., Wingless
secretion requires endosome-to-Golgi retrieval of Wntless/
Evi/Sprinter by the retromer complex,Nature Cell Biology,
vol. 10, no. 2, pp. 170177, 2008.
[61] M. Harterink, F. Port, M. J. Lorenowicz et al., ASNX3-
dependent retromer pathway mediates retrograde transport
of the Wnt sorting receptor Wntless and is required for
Wnt secretion,Nature Cell Biology, vol. 13, no. 8, pp. 914
923, 2011.
[62] I. Gasnereau, P. Herr, P. Z. Chia, K. Basler, and P. A. Gleeson,
Identication of an endocytosis motif in an intracellular
loop of Wntless protein, essential for its recycling and the
control of Wnt protein signaling,The Journal of Biological
Chemistry, vol. 286, no. 50, pp. 4332443333, 2011.
[63] C. Korkut, B. Ataman, P. Ramachandran et al., Trans-synap-
tic transmission of vesicular Wnt signals through Evi/
Wntless,Cell, vol. 139, no. 2, pp. 393404, 2009.
[64] R. Nusse and H. Clevers, Wnt/β-Catenin Signaling, Disease,
and Emerging Therapeutic Modalities,Cell, vol. 169, no. 6,
pp. 985999, 2017.
[65] H. F. Farin, I. Jordens, M. H. Mosa et al., Visualization of a
short-range Wnt gradient in the intestinal stem-cell niche,
Nature, vol. 530, no. 7590, pp. 340343, 2016.
[66] K. A. Mulligan, C. Fuerer, W. Ching, M. Fish, K. Willert, and
R. Nusse, Secreted Wingless-interacting molecule (Swim)
promotes long-range signaling by maintaining Wingless sol-
ubility,Proceedings of the National Academy of Sciences of
the United States of America, vol. 109, no. 2, pp. 370377,
2012.
[67] M. Zecca, K. Basler, and G. Struhl, Direct and long-range
action of a wingless morphogen gradient,Cell, vol. 87,
no. 5, pp. 833844, 1996.
[68] C. Alexandre, A. Baena-Lopez, and J. P. Vincent, Patterning
and growth control by membrane-tethered Wingless,
Nature, vol. 505, no. 7482, pp. 180185, 2014.
[69] J. C. Gross, V. Chaudhary, K. Bartscherer, and M. Boutros,
Active Wnt proteins are secreted on exosomes,Nature Cell
Biology, vol. 14, no. 10, pp. 10361045, 2012.
[70] J. Behrens, J. P. von Kries, M. Kühl et al., Functional interac-
tion of beta-catenin with the transcription factor LEF-1,
Nature, vol. 382, no. 6592, pp. 638642, 1996.
[71] M. Molenaar, M. Van De Wetering, M. Oosterwegel et al.,
XTcf-3 transcription factor mediates beta-catenin-induced
axis formation in Xenopus embryos,Cell, vol. 86, no. 3,
pp. 391399, 1996.
[72] P. Bhanot, M. Brink, C. H. Samos et al., A new member of
the frizzled family from Drosophila functions as a Wingless
receptor,Nature, vol. 382, no. 6588, pp. 225230, 1996.
[73] C. E. Dann, J. C. Hsieh, A. Rattner, D. Sharma, J. Nathans,
and D. J. Leahy, Insights into Wnt binding and signalling
from the structures of two Frizzled cysteine-rich domains,
Nature, vol. 412, no. 6842, pp. 8690, 2001.
[74] K. I. Pinson, J. Brennan, S. Monkley, B. J. Avery, and W. C.
Skarnes, An LDL-receptor-related protein mediates Wnt
signalling in mice,Nature, vol. 407, no. 6803, pp. 535538,
2000.
[75] K. Tamai, M. Semenov, Y. Kato et al., LDL-receptor-related
proteins in Wnt signal transduction,Nature, vol. 407,
no. 6803, pp. 530535, 2000.
[76] Y. Gong, E. Bourhis, C. Chiu et al., Wnt isoform-specic
interactions with coreceptor specify inhibition or potentia-
tion of signaling by LRP6 antibodies,PLoS One, vol. 5,
no. 9, article e12682, 2010.
[77] C. Niehrs, The complex world of WNT receptor signalling,
Nature Reviews. Molecular Cell Biology, vol. 13, no. 12,
pp. 767779, 2012.
[78] H. Aberle, A. Bauer, J. Stappert, A. Kispert, and R. Kemler,
Beta-catenin is a target for the ubiquitin-proteasome path-
way,The EMBO Journal, vol. 16, no. 13, pp. 37973804,
1997.
[79] G. Chen and A. J. Courey, Groucho/TLE family proteins and
transcriptional repression,Gene, vol. 249, no. 1-2, pp. 116,
2000.
[80] X. He, M. Semenov, K. Tamai, and X. Zeng, LDL receptor-
related proteins 5 and 6 in Wnt/beta-catenin signaling:
arrows point the way,Development, vol. 131, no. 8,
pp. 16631677, 2004.
[81] K. Tamai, X. Zeng, C. Liu et al., Amechanism for Wnt cor-
eceptor activation,Molecular Cell, vol. 13, no. 1, pp. 149
156, 2004.
[82] J. Mao, J. Wang, B. Liu et al., Low-density lipoprotein
receptor-related protein-5 binds to Axin and regulates the
canonical Wnt signaling pathway,Molecular Cell, vol. 7,
no. 4, pp. 801809, 2001.
[83] X. Zeng, K. Tamai, B. Doble et al., A dual-kinase mechanism
for Wnt co-receptor phosphorylation and activation,
Nature, vol. 438, no. 7069, pp. 873877, 2005.
[84] G. Davidson, W. Wu, J. Shen et al., Casein kinase 1 gamma
couples Wnt receptor activation to cytoplasmic signal trans-
duction,Nature, vol. 438, no. 7069, pp. 867872, 2005.
[85] H. Hikasa, J. Ezan, K. Itoh, X. Li, M. W. Klymkowsky, and
S. Y. Sokol, Regulation of TCF3 by Wnt-dependent
9Oxidative Medicine and Cellular Longevity
phosphorylation during vertebrate axis specication,Devel-
opmental Cell, vol. 19, no. 4, pp. 521532, 2010.
[86] W. Lee, S. Swarup, J. Chen, T. Ishitani, and E. M. Verheyen,
Homeodomain-interacting protein kinases (Hipks) promote
Wnt/Wg signaling through stabilization of beta-catenin/Arm
and stimulation of target gene expression,Development,
vol. 136, no. 2, pp. 241251, 2009.
[87] J. Willert, M. Epping, J. R. Pollack, P. O. Brown, and R. Nusse,
A transcriptional response to Wnt protein in human embry-
onic carcinoma cells,BMC Developmental Biology, vol. 2,
no. 1, p. 8, 2002.
[88] T. C. He, A. B. Sparks, C. Rago et al., Identication of c-
MYC as a target of the APC pathway,Science, vol. 281,
no. 5382, pp. 15091512, 1998.
[89] O. Tetsu and F. McCormick, Beta-catenin regulates expres-
sion of cyclin D1 in colon carcinoma cells,Nature,
vol. 398, no. 6726, pp. 422426, 1999.
[90] X. Zhang, J. P. Gaspard, and D. C. Chung, Regulation of vas-
cular endothelial growth factor by the Wnt and K-ras path-
ways in colonic neoplasia,Cancer Research, vol. 61, no. 16,
pp. 60506054, 2001.
[91] L. Zhou, Y. Li, S. Hao et al., Multiple genes of the renin-
angiotensin system are novel targets of Wnt/β-Catenin sig-
naling,Journal of the American Society of Nephrology,
vol. 26, no. 1, pp. 107120, 2015.
[92] Y. Mori, H. Nagse, H. Ando et al., Somatic mutations of the
APC gene in colorectal tumors: mutation cluster region in the
APC gene,Human Molecular Genetics, vol. 1, no. 4, pp. 229
233, 1992.
[93] Q. Xu, Y. Wang, A. Dabdoub et al., Vascular development in
the retina and inner ear: control by Norrin and Frizzled-4, a
high-anity ligand-receptor pair,Cell, vol. 116, no. 6,
pp. 883895, 2004.
[94] Ö. Uluçkan, M. Jimenez, S. Karbach et al., Chronic skin
inammation leads to bone loss by IL-17-mediated inhibition
of Wnt signaling in osteoblasts,Science Translational Medi-
cine,vol. 8, no. 330, 2016.
[95] Y. Shi, B. Shu, R. Yang et al., Wnt and Notch signaling path-
way involved in wound healing by targeting c-Myc and Hes 1
separately,Stem Cell Research & Therapy, vol. 6, p. 120,
2015.
[96] J. E. Harris, Melanocyte regeneration in vitiligo requires
WNT beneath their Wings,The Journal of Investigative Der-
matology, vol. 135, no. 12, pp. 29212923, 2015.
[97] C. Regazzetti, F. Joly, C. Marty et al., Transcriptional analy-
sis of vitiligo skin reveals the alteration of WNT pathway: a
promising target for repigmenting vitiligo patients,The
Journal of Investigative Dermatology, vol. 135, no. 12,
pp. 31053114, 2015.
[98] M. Zaidan, F. Stucker, B. Stengel et al., Increased risk of solid
renal tumors in lithium-treated patients,Kidney Interna-
tional, vol. 86, no. 1, pp. 184190, 2014.
[99] O. Kazanskaya, A. Glinka, I. del Barco Barrantes, P. Stannek,
C. Niehrs, and W. Wu, R-Spondin 2 is a secreted activator of
Wnt/beta-catenin signaling and is required for Xenopus
myogenesis,Developmental Cell, vol. 7, no. 4, pp. 525534,
2004.
[100] X. Jiang, O. Charlat, R. Zamponi, Y. Yang, and F. Cong,
Dishevelled promotes Wnt receptor degradation through
recruitment of ZNRF3/RNF43 E3 ubiquitin ligases,Molecu-
lar Cell, vol. 58, no. 3, pp. 522533, 2015.
[101] K. S. Carmon, X. Gong, Q. Lin, A. Thomas, and Q. Liu, R-
spondins function as ligands of the orphan receptors LGR4
and LGR5 to regulate Wnt/beta-catenin signaling,Proceed-
ings of the National Academy of Sciences of the United States
of America, vol. 108, no. 28, pp. 1145211457, 2011.
[102] W. De Lau, N. Barker, T. Y. Low et al., Lgr 5 homologues
associate with Wnt receptors and mediate R-spondin signal-
ling,Nature, vol. 476, no. 7360, pp. 293297, 2011.
[103] A. Glinka, C. Dolde, N. Kirsch et al., LGR4 and LGR5 are R-
spondin receptors mediating Wnt/βcatenin and Wnt/PCP
signalling,EMBO Reports, vol. 12, no. 10, pp. 10551061,
2011.
[104] Y. Xie, R. Zamponi, O. Charlat et al., Interaction with both
ZNRF3 and LGR4 is required for the signalling activity of
R-spondin,EMBO Reports, vol. 14, no. 12, pp. 11201126,
2013.
[105] W. de Lau, W. C. Peng, P. Gros, and H. Clevers, The R-spon-
din/Lgr 5/Rnf43 module: regulator of Wnt signal strength,
Genes & Development, vol. 28, no. 4, pp. 305316, 2014.
[106] A. Ohlmann and E. R. Tamm, Norrin: molecular and func-
tional properties of an angiogenic and neuroprotective
growth factor,Progress in Retinal and Eye Research,
vol. 31, no. 3, pp. 243257, 2012.
[107] T. Meitinger, A. Meindl, P. Bork et al., Molecular modelling
of the Norrie disease protein predicts a cystine knot growth
factor tertiary structure,Nature Genetics, vol. 5, no. 4,
pp. 376380, 1993.
[108] H. J. Junge, S. Yang, J. B. Burton et al., TSPAN12 regulates
retinal vascular development by promoting Norrin- but not
Wnt-induced FZD4/beta-catenin signaling,Cell, vol. 139,
no. 2, pp. 299311, 2009.
[109] M. B. Lai, C. Zhang, J. Shi et al., TSPAN12 is a Norrin
co-receptor that amplies Frizzled 4 ligand selectivity and
signaling,Cell Reports, vol. 19, no. 13, pp. 28092822,
2017.
[110] X. Ye, Y. Wang, H. Cahill et al., Norrin, frizzled-4, and Lrp 5
signaling in endothelial cells controls a genetic program for
retinal vascularization,Cell, vol. 139, no. 2, pp. 285298,
2009.
[111] T. H. Chang, F. L. Hsieh, M. Zebisch, K. Harlos, J. Elegheert,
and E. Y. Jones, Structure and functional properties of Nor-
rin mimic Wnt for signalling with Frizzled 4, Lrp 5/6, and
proteoglycan,eLife, vol. 4, 2015.
[112] W. Zhang, J. Yang, Y. Liu et al., PR55 alpha, a regulatory
subunit of PP2A, specically regulates PP2A-mediated beta-
catenin dephosphorylation,The Journal of Biological Chem-
istry, vol. 284, no. 34, pp. 2264922656, 2009.
[113] R. Nagel, C. le Sage, B. Diosdado et al., Regulation of the
adenomatous polyposis coli gene by the miR-135 family in
colorectal cancer,Cancer Research,vol. 68, no. 14,
pp. 57955802, 2008.
[114] M. Schwabiuk, L. Coudiere, and D. C. Merz, SDN-1/synde-
can regulates growth factor signaling in distal tip cell migra-
tions in C. elegans,Developmental Biology, vol. 334, no. 1,
pp. 235242, 2009.
[115] K. Dejima, S. Kang, S. Mitani, P. C. Cosman, and
A. Chisholm, Syndecan denes precise spindle orientation
by modulating Wnt signaling in C. elegans,Development,
vol. 141, no. 22, pp. 43544365, 2014.
[116] K. Saied-Santiago, R. A. Townley, J. D. Attonito et al., Coor-
dination of heparan sulfate proteoglycans with Wnt signaling
10 Oxidative Medicine and Cellular Longevity
to control cellular migrations and positioning inCaenorhab-
ditis elegans,Genetics, vol. 206, no. 4, pp. 19511967, 2017.
[117] M. Kahn, Can we safely target the WNT pathway?,Nature
Reviews. Drug Discovery, vol. 13, no. 7, pp. 513532, 2014.
[118] C. Tapia-Rojas and N. C. Inestrosa, Loss of canonical Wnt
signaling is involved in the pathogenesis of Alzheimer's dis-
ease,Neural Regeneration Research, vol. 13, no. 10,
pp. 17051710, 2018.
[119] A. Caricasole, A. Copani, F. Caraci et al., Induction of Dick-
kopf-1, a negative modulator of the Wnt pathway, is associ-
ated with neuronal degeneration in Alzheimer's brain,The
Journal of Neuroscience, vol. 24, no. 26, pp. 60216027, 2004.
[120] S. A. Purro, E. M. Dickins, and P. C. Salinas, The secreted
Wnt antagonist Dickkopf-1 is required for Amyloid -Medi-
ated Synaptic Loss,The Journal of Neuroscience, vol. 32,
no. 10, pp. 34923498, 2012.
[121] S. Kakugawa, P. F. Langton, M. Zebisch et al., Notum deacy-
lates Wnt proteins to suppress signalling activity,Nature,
vol. 519, no. 7542, pp. 187192, 2015.
[122] A. J. Giraldez, R. R. Copley, and S. M. Cohen, HSPG modi-
cation by the secreted enzyme Notum shapes the Wingless
morphogen gradient,Developmental Cell, vol. 2, no. 5,
pp. 667676, 2002.
[123] X. Zhang, S. M. Cheong, N. G. Amado et al., Notum is
required for neural and head induction via Wnt deacylation,
oxidation, and inactivation,Developmental Cell, vol. 32,
no. 6, pp. 719730, 2015.
[124] J. C. Hsieh, L. Kodjabachian, M. L. Rebbert et al., A new
secreted protein that binds to Wnt proteins and inhibits
their activities,Nature, vol. 398, no. 6726, pp. 431436,
1999.
[125] C. Surmann-Schmitt, N. Widmann, U. Dietz et al., Wif-1 is
expressed at cartilage-mesenchyme interfaces and impedes
Wnt3a-mediated inhibition of chondrogenesis,Journal of
Cell Science, vol. 122, no. 20, pp. 36273637, 2009.
[126] L. Leyns, T. Bouwmeester, S. H. Kim, S. Piccolo, and E. M. De
Robertis, Frzb-1 is a secreted antagonist of Wnt signaling
expressed in the Spemann organizer,Cell, vol. 88, no. 6,
pp. 747756, 1997.
[127] S. Wang, M. Krinks, K. Lin, F. P. Luyten, and M. Moos Jr.,
Frzb, a secreted protein expressed in the Spemann organizer,
binds and inhibits Wnt-8,Cell, vol. 88, no. 6, pp. 757766,
1997.
[128] P. W. Finch, X. He, M. J. Kelley et al., Purication and
molecular cloning of a secreted, Frizzled-related antagonist
of Wnt action,Proceedings of the National Academy of Sci-
ences of the United States of America, vol. 94, no. 13,
pp. 67706775, 1997.
[129] A. Yamamoto, T. Nagano, S. Takehara, M. Hibi, and
S. Aizawa, Shisa Promotes Head Formation through the
Inhibition of Receptor Protein Maturation for the Caudaliz-
ing Factors, Wnt and FGF,Cell, vol. 120, no. 2, pp. 223
235, 2005.
[130] B. Kagermeier-Schenk, D. Wehner, G. Özhan-Kizil et al.,
Waif1/5T4 Inhibits Wnt/β-Catenin Signaling and Activates
Noncanonical Wnt Pathways by Modifying LRP6 Subcellular
Localization,Developmental Cell, vol. 21, no. 6, pp. 1129
1143, 2011.
[131] Y. Shimomura, D. Agalliu, A. Vonica et al., APCDD1 is a
novel Wnt inhibitor mutated in hereditary hypotrichosis
simplex,Nature, vol. 464, no. 7291, pp. 10431047, 2010.
[132] X. Zhang, J. G. Abreu, C. Yokota et al., Tiki 1 is required for
head formation via Wnt cleavage-oxidation and inactiva-
tion,Cell, vol. 149, no. 7, pp. 15651577, 2012.
[133] M. Korpal, E. S. Lee, G. Hu, and Y. Kang, The miR-200 fam-
ily inhibits epithelial-mesenchymal transition and cancer cell
migration by direct targeting of E-cadherin transcriptional
repressors ZEB1 and ZEB2,The Journal of Biological Chem-
istry, vol. 283, no. 22, pp. 1491014914, 2008.
[134] S. T. Hashimi, J. A. Fulcher, M. H. Chang, L. Gov, S. Wang,
and B. Lee, MicroRNA proling identies miR-34a and
miR-21 and their target genes JAG1 and WNT1 in the coor-
dinate regulation of dendritic cell dierentiation,Blood,
vol. 114, no. 2, pp. 404414, 2009.
[135] Y. Takahashi, Q. Chen, R. V. S. Rajala, and J. X. Ma, Micro-
RNA-184 modulates canonical Wnt signaling through the
regulation of frizzled-7 expression in the retina with
ischemia-induced neovascularization,FEBS Letters,
vol. 589, no. 10, pp. 11431149, 2015.
[136] H. X. Hao, Y. Xie, Y. Zhang et al., ZNRF3 promotes Wnt
receptor turnover in an R-spondin-sensitive manner,
Nature, vol. 485, no. 7397, pp. 195200, 2012.
[137] B. K. Koo, M. Spit, I. Jordens et al., Tumour suppressor
RNF43 is a stem-cell E3 ligase that induces endocytosis of
Wnt receptors,Nature, vol. 488, no. 7413, pp. 665669, 2012.
[138] J. Behrens, B. A. Jerchow, M. Würtele et al., Functional inter-
action of an Axin homolog, conductin, with beta-catenin,
APC, and GSK3beta,Science, vol. 280, no. 5363, pp. 596
599, 1998.
[139] E. H. Jho, T. Zhang, C. Domon, C. K. Joo, J. N. Freund, and
F. Costantini, Wnt/beta-catenin/Tcf signaling induces the
transcription of Axin 2, a negative regulator of the signaling
pathway,Molecular and Cellular Biology, vol. 22, no. 4,
pp. 11721183, 2002.
[140] R. Nusse and H. Varmus, Three decades of Wnts: a personal
perspective on how a scienticeld developed,The EMBO
Journal, vol. 31, no. 12, pp. 26702684, 2012.
[141] C. Dickson, R. Smith, S. Brookes, and G. Peters, Tumorigen-
esis by mouse mammary tumor virus: proviral activation of a
cellular gene in the common integration region int-2,Cell,
vol. 37, no. 2, pp. 529536, 1984.
[142] H. Roelink, E. Wagenaar, S. L. Da Silva, and R. Nusse, Wnt-
3, a gene activated by proviral insertion in mouse mammary
tumors, is homologous to int-1/Wnt-1 and is normally
expressed in mouse embryos and adult brain,Proceedings
of the National Academy of Sciences of the United States of
America, vol. 87, no. 12, pp. 45194523, 1990.
[143] D. Gallahan and R. Callahan, Mammary tumorigenesis in
feral mice: identication of a new int locus in mouse mam-
mary tumor virus (Czech II)-induced mammary tumors,
Journal of Virology, vol. 61, no. 1, pp. 6674, 1987.
[144] N. Ghosh, U. Hossain, A. Mandal, and P. C. Sil, The Wnt
signaling pathway: a potential therapeutic target against can-
cer,Annals of the New York Academy of Sciences, vol. 1443,
no. 1, pp. 5474, 2019.
[145] N. S. Fearnhead, M. P. Britton, and W. F. Bodmer, The ABC
of APC,Human Molecular Genetics, vol. 10, no. 7, pp. 721
733, 2001.
[146] P. J. Morin, A. B. Sparks, V. Korinek et al., Activation of
β-Catenin-Tcf signaling in colon cancer by mutations in
β-Catenin or APC,Science, vol. 275, no. 5307,
pp. 17871790, 1997.
11Oxidative Medicine and Cellular Longevity
[147] G. M. Caldwell, C. E. Jones, A. M. Ashley et al., Wnt signal-
ling in adenomas of familial adenomatous polyposis
patients,British Journal of Cancer, vol. 103, no. 6, pp. 910
917, 2010.
[148] J. Tao, Y. Krutsenko, A. Moghe et al., Nuclear factor ery-
throid 2related factor 2 and βCatenin coactivation in hepa-
tocellular cancer: biological and therapeutic implications,
Hepatology, vol. 74, no. 2, pp. 741759, 2021.
[149] S. Song, T. Christova, S. Perusini et al., Wnt inhibitor screen
reveals iron dependence of β-Catenin signaling in cancers,
Cancer Research, vol. 71, no. 24, pp. 76287639, 2011.
[150] A. Whitelaw, Intraventricular haemorrhage and posthae-
morrhagic hydrocephalus: pathogenesis, prevention and
future interventions,Seminars in Neonatology, vol. 6, no. 2,
pp. 135146, 2001.
[151] J. Wu, Y. Hua, R. F. Keep, T. Nakamura, J. T. Ho, and G. Xi,
Iron and iron-handling proteins in the brain after intracere-
bral hemorrhage,Stroke, vol. 34, no. 12, pp. 29642969,
2003.
[152] Z. Chen, C. Gao, Y. Hua, R. F. Keep, K. Muraszko, and G. Xi,
Role of iron in brain injury after intraventricular hemor-
rhage,Stroke, vol. 42, no. 2, pp. 465470, 2011.
[153] P. Cisternas, C. P. Vio, and N. C. Inestrosa, Role of Wnt sig-
naling in tissue brosis, lessons from skeletal muscle and kid-
ney,Current Molecular Medicine, vol. 14, no. 4, pp. 510522,
2014.
[154] C. G. Miao, Y. Y. Yang, X. He et al., Wnt signaling in liver
brosis: progress, challenges and potential directions,Bio-
chimie, vol. 95, no. 12, pp. 23262335, 2013.
[155] H. Meng, F. Li, R. Hu et al., Deferoxamine alleviates chronic
hydrocephalus after intraventricular hemorrhage through
iron chelation and Wnt1/Wnt3a inhibition,Brain Research,
vol. 1602, pp. 4452, 2015.
[156] A. Ziaei, M. R. P. Ardakani, M. S. Hashemi et al., Acute
course of deferoxamine promoted neuronal dierentiation
of neural progenitor cells through suppression of Wnt/β-
catenin pathway: A novel ecient protocol for neuronal dif-
ferentiation,Neuroscience Letters, vol. 590, pp. 138144,
2015.
[157] M. M. Harris, L. B. Houtkooper, V. A. Stanford et al., Dietary
iron is associated with bone mineral density in healthy post-
menopausal women,The Journal of Nutrition, vol. 133,
no. 11, pp. 35983602, 2003.
[158] S. I. Katsumata, R. Katsumata-Tsuboi, M. Uehara, and
K. Suzuki, Severe iron deciency decreases both bone forma-
tion and bone resorption in rats,The Journal of Nutrition,
vol. 139, no. 2, pp. 238243, 2009.
[159] R. Baron and M. Kneissel, WNT signaling in bone homeo-
stasis and disease: from human mutations to treatments,
Nature Medicine, vol. 19, no. 2, pp. 179192, 2013.
[160] V. Jeney, Clinical impact and cellular mechanisms of iron
overload-associated bone loss,Frontiers in Pharmacology,
vol. 8, p. 77, 2017.
[161] M. Casale, S. Citarella, A. Filosa et al., Endocrine function
and bone disease during long-term chelation therapy with
deferasirox in patients with β-thalassemia major,Ameri-
can Journal of Hematology, vol. 89, no. 12, pp. 1102
1106, 2014.
[162] Y. Gong, R. B. Slee, N. Fukai et al., LDL receptor-related pro-
tein 5 (LRP5) aects bone accrual and eye development,Cell,
vol. 107, no. 4, pp. 513523, 2001.
[163] K. Maeda, Y. Kobayashi, N. Udagawa et al., Wnt5a-Ror2 sig-
naling between osteoblast-lineage cells and osteoclast precur-
sors enhances osteoclastogenesis,Nature Medicine, vol. 18,
no. 3, pp. 405412, 2012.
[164] NIH Consensus Development Panel on Osteoporosis Preven-
tion, Diagnosis, and Therapy, March 7-29, 2000: highlights
of the conference,Southern Medical Journal, vol. 94, no. 6,
pp. 569573, 2001.
[165] S. C. Manolagas, From estrogen-centric to aging and oxida-
tive stress: a revised perspective of the pathogenesis of osteo-
porosis,Endocrine Reviews, vol. 31, no. 3, pp. 266300, 2010.
[166] U. Baschant, M. Rauner, E. Balaian et al., Wnt5a is a key tar-
get for the pro-osteogenic eects of iron chelation on osteo-
blast progenitors,Haematologica, vol. 101, no. 12,
pp. 14991507, 2016.
[167] A. J. Mikels and R. Nusse, Puried Wnt5a protein activates
or inhibits β-CateninTCF signaling depending on receptor
context,PLoS Biology, vol. 4, no. 4, article e115, 2006.
[168] W. Xu, R. Yu, X. Zhu et al., Iron-chelating agent can main-
tain bone homeostasis disrupted by iron overload by upregu-
lating Wnt/beta-catenin signaling,BioMed Research
International, vol. 2020, Article ID 8256261, 2020.
[169] G. Li, H. Zhang, J. Wu et al., Hepcidin deciency causes
bone loss through interfering with the canonical Wnt/β-
catenin pathway via Forkhead box O3a,Journal of Ortho-
paedic Translation, vol. 23, pp. 6776, 2020.
[170] L. Huang, Z. Liu, H. Liu et al., Iron overload impairs bone
marrow mesenchymal stromal cells from higher-risk MDS
patients by regulating the ROS-related Wnt/beta-catenin
pathway,Stem Cells International, vol. 2020, Article ID
8855038, 2020.
[171] X. Huang, Iron overload and its association with cancer risk
in humans: evidence for iron as a carcinogenic metal,Muta-
tion Research, vol. 533, no. 1-2, pp. 153171, 2003.
[172] L. Valenti, A. L. Fracanzani, P. Dongiovanni et al., Iron
depletion by phlebotomy improves insulin resistance in
patients with nonalcoholic fatty liver disease and hyperferri-
tinemia: evidence from a case-control study,The American
Journal of Gastroenterology, vol. 102, no. 6, pp. 12511258,
2007.
[173] P. K. Awuah, B. H. Rhieu, S. Singh, A. Misse, and S. P. S.
Monga, β-Catenin loss in hepatocytes promotes hepatocel-
lular cancer after diethylnitrosamine and phenobarbital
administration to mice,PLoS One,vol. 7, no. 6, article
e39771, 2012.
[174] X. F. Zhang, X. Tan, G. Zeng et al., Conditional beta-catenin
loss in mice promotes chemical hepatocarcinogenesis: role of
oxidative stress and platelet-derived growth factor receptor
alpha/phosphoinositide 3-kinase signaling,Hepatology,
vol. 52, no. 3, pp. 954965, 2010.
[175] M. E. Preziosi, S. Singh, E. V. Valore et al., Mice lacking
liver-specicβ-catenin develop steatohepatitis and brosis
after iron overload,Journal of Hepatology, vol. 67, no. 2,
pp. 360369, 2017.
[176] A. Mandala, W. J. Chen, A. Armstrong et al., PPARαagonist
fenobrate attenuates iron-induced liver injury in mice by
modulating the Sirt 3 and β-catenin signaling,American
Journal of Physiology - Gastrointestinal and Liver Physiology,
vol. 321, no. 3, pp. G262G269, 2021.
[177] J. S. Joyal, M. L. Gantner, and L. E. H. Smith, Retinal energy
demands control vascular supply of the retina in
12 Oxidative Medicine and Cellular Longevity
development and disease: the role of neuronal lipid and glu-
cose metabolism,Progress in Retinal and Eye Research,
vol. 64, pp. 131156, 2018.
[178] S. Selvam, T. Kumar, and M. Fruttiger, Retinal vasculature
development in health and disease,Progress in Retinal and
Eye Research, vol. 63, pp. 119, 2018.
[179] G. Moiseyev, Y. Chen, Y. Takahashi, B. X. Wu, and J. X. Ma,
RPE65 is the isomerohydrolase in the retinoid visual cycle,
Proceedings of the National Academy of Sciences of the United
States of America, vol. 102, no. 35, pp. 1241312418, 2005.
[180] H. Chen, T. J. Lukas, N. Du, G. Suyeoka, and A. H. Neufeld,
Dysfunction of the retinal pigment epithelium with age:
increased iron decreases phagocytosis and lysosomal activ-
ity,Investigative Ophthalmology & Visual Science, vol. 50,
no. 4, pp. 18951902, 2009.
[181] B. S. Rogers, R. C. Symons, K. Komeima et al., Dierential
sensitivity of cones to iron-mediated oxidative damage,
Investigative Ophthalmology & Visual Science, vol. 48, no. 1,
pp. 438445, 2007.
[182] A. Vallee, Y. LeCarpentier, R. Vallee, R. Guillevin, and J. N.
Vallée, Circadian Rhythms in Exudative Age-Related Mac-
ular Degeneration: The Key Role of the Canonical WNT/
β-Catenin Pathway,International journal of molecular sci-
ences, vol. 21, no. 3, p. 820, 2020.
[183] Y. Chen, Y. Hu, T. Zhou et al., Activation of the Wnt path-
way plays a pathogenic role in diabetic retinopathy in
humans and animal models,The American Journal of
Pathology, vol. 175, no. 6, pp. 26762685, 2009.
[184] A. Mandala, A. Armstrong, B. Girresch et al., Fenobrate
prevents iron induced activation of canonical Wnt/β-catenin
and oxidative stress signaling in the retina,NPJ Aging And
Mechanisms Of Disease, vol. 6, no. 1, p. 12, 2020.
[185] P. R. Timmers, J. F. Wilson, P. K. Joshi, and J. Deelen,
Multivariate genomic scan implicates novel loci and haem
metabolism in human ageing,Nature Communications,
vol. 11, p. 3570, 2020.
13Oxidative Medicine and Cellular Longevity
... The accumulation of iron due to its impaired homeostasis can be particularly dangerous. The iron toxicity is caused by its potential to induce oxidative stress thought ROS production and consequently inflammation [49]. ROS can affect DNA, protein and lipid integrity damaging cellular functionality [50]. ...
... Equally, ferritin acts as a local cytokine inducing an increase of pro-inflammatory cytokines such as IL-1β [52]. Moreover, inflammation itself induces iron accumulatio in tissues such as liver upregulating hepcidin [53] and thus reducing FPN1 causing the establishment of a vicious circle, where iron-mediated inflammation generates in turn an increase of intracellular iron levels which consequently induces oxidative stress and iron-mediated inflammation [49]. ...
... Recent studies indicate that iron regulates Wnt signaling, and that the iron chelator DFX can inhibit Wnt signaling [91]. Wnt activation is known to enhance inflammatory processes [92], therefore it is implicated in the pathogenesis of several diseases [49]. The relation between iron excess and the alteration of Wnt signaling has been investigated in neurodegenerative disorders. ...
Article
Full-text available
Iron is a crucial element for mammalian cells, considering its intervention in several physiologic processes. Its homeostasis is finely regulated, and its alteration could be responsible for the onset of several disorders. Iron is closely related to inflammation; indeed, during inflammation high levels of interleukin-6 cause an increased production of hepcidin which induces a degradation of ferroportin. Ferroportin degradation leads to decreased iron efflux that culminates in elevated intracellular iron concentration and consequently iron toxicity in cells and tissues. Therefore, iron chelation could be considered a novel and useful therapeutic strategy in order to counteract the inflammation in several autoimmune and inflammatory diseases. Several iron chelators are already known to have anti-inflammatory effects, among them deferiprone, deferoxamine, deferasirox, and Dp44mT are noteworthy. Recently, eltrombopag has been reported to have an important role in reducing inflammation, acting both directly by chelating iron, and indirectly by modulating iron efflux. This review offers an overview of the possible novel biological effects of the iron chelators in inflammation, suggesting them as novel anti-inflammatory molecules.
... In addition, using the cell line LS174T, wild-type APC but mutant beta-catenin also proved responsive, suggesting that the role of Fe is to regulate beta-catenin. The authors therefore speculated that excess Fe could exacerbate tumorigenesis against the background of APC loss, a situation commonly observed in tumors [139][140][141]. ...
Article
Full-text available
Essential trace elements play an important role in human physiology and are associated with various functions regulating cellular metabolism. Non-essential trace elements, on the other hand, often have well-documented toxicities that are dangerous for the initiation and development of diseases due to their widespread occurrence in the environment and their accumulation in living organisms. Non-essential trace elements are therefore regarded as serious environmental hazards that are harmful to health even in low concentrations. Many representatives of these elements are present as pollutants in our environment, and many people may be exposed to significant amounts of these substances over the course of their lives. Among the most common non-essential trace elements are heavy metals, which are also associated with acute poisoning in humans. When these elements accumulate in the body over years of chronic exposure, they often cause severe health damage in a variety of tissues and organs. In this review article, the role of selected essential and non-essential trace elements and their role in the development of exemplary pathophysiological processes in the cardiovascular system will be examined in more detail.
... Desferrioxamine mesylate (DFO) is a kind of chelator that is clinically used to treat iron poisoning and iron overload [4]. Chelates consist of DFO and ferric ion, which could be excreted completely through urine and feces [5]. Therefore, iron deposited in organs could be reduced. ...
Article
Full-text available
Background Previous meta-analysis had concluded that desferrioxamine mesylate (DFO) could effectively treat intracerebral hematoma (ICH) in animal models. We hope to confirm that DFO could treat ICH patients effectively through the systemic review and meta-analysis of clinical researches. Method Data extraction included hematoma volume (HV), reduction of National Institute of Health Stroke Scale (NIHSS) scores, and relative perihematomal edema (RPHE). The standard mean difference (SMD) and 95% confidence interval (95% CI ) were calculated by fixed effects model. I-square ( I ² ) statistic was used to test the heterogeneity. All p values were two-side with a significant level at 0.05. Results Five randomized controlled trials were included in the meta-analysis, which included 239 patients. At 7 days after onset, there was significant difference of RPHE development (− 1.87 (− 2.22, − 1.51) ( I ² = 0, p = 0.639)) and significant difference of HV absorption (− 0.71 (− 1.06, 0.36) ( I ² = 17.5%, p = 0.271)) between DFO and control groups. There was significant difference of reduction of NHISS scores (0.25 (0.05, 0.46) ( I ² = 0, p = 0.992)) between DFO and control groups at 30 days after onset. Conclusion DFO reduced HV and perihematomal edema in ICH patients at 7 days after onset and improve neurological function at 30 days after onset efficiently and safely. DFO might be a new route of improving treatment of ICH.
... The retina is a part of the central nervous system; it contains complex neural circuitry and transduces the converted electrical potentials to the brain [10]. The neuroprotective roles of rapamycin may be a novel therapeutic pathway in ocular neurodegenerative diseases, such as diabetic retinopathy (DR), age-related macular degeneration (AMD), and glaucoma, which share common pathophysiological mechanisms, especially increased and prolonged oxidative stress, which would ultimately result in retinal neuronal death [11][12][13][14]. Recently, a large number of studies have been conducted to elucidate the neuroprotective role of rapamycin and its underlying mechanism(s) in the treatment of ocular degenerative diseases [15][16][17][18][19][20][21][22][23][24][25][26][27]. ...
Article
Full-text available
Recent advances in the research of the mammalian target of the rapamycin (mTOR) signalling pathway demonstrated that mTOR is a robust therapeutic target for ocular degenerative diseases, including age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma. Although the exact mechanisms of individual ocular degenerative diseases are unclear, they share several common pathological processes, increased and prolonged oxidative stress in particular, which leads to functional and morphological impairment in photoreceptors, retinal ganglion cells (RGCs), or retinal pigment epithelium (RPE). mTOR not only modulates oxidative stress but is also affected by reactive oxygen species (ROS) activation. It is essential to understand the complicated relationship between the mTOR pathway and oxidative stress before its application in the treatment of retinal degeneration. Indeed, the substantial role of mTOR-mediated autophagy in the pathogenies of ocular degenerative diseases should be noted. In reviewing the latest studies, this article summarised the application of rapamycin, an mTOR signalling pathway inhibitor, in different retinal disease models, providing insight into the mechanism of rapamycin in the treatment of retinal neurodegeneration under oxidative stress. Besides basic research, this review also summarised and updated the results of the latest clinical trials of rapamycin in ocular neurodegenerative diseases. In combining the current basic and clinical research results, we provided a more complete picture of mTOR as a potential therapeutic target for ocular neurodegenerative diseases.
Article
This study aimed to investigate the mechanism of iron on intestinal epithelium development of suckling piglets. Compared with newborn piglets, 7-day-old and 21-day-old piglets showed changes in the morphology of the jejunum, increased proliferation, differentiated epithelial cells, and expanded enteroids. Intestinal epithelium maturation markers and iron metabolism genes were significantly changed. These results suggest that lactation is a critical stage in intestinal epithelial development, accompanied by changes in iron metabolism. In addition, deferoxamine (DFO) treatment inhibited the activity of intestinal organoids at passage 4 (P4) of 0-day-old piglets, but no significant difference was observed in epithelial maturation markers at passage 1 (P1) and P4, and only argininosuccinate synthetase 1 (Ass1) and β-galactosidase (Gleb) were up-regulated at passage 7 (P7). These results in vitro show that iron deficiency may not directly affect intestinal epithelium development through intestinal stem cells (ISCs). The iron supplementation significantly down-regulated the mRNA expression of interleukin-22 receptor subunit alpha-2 (IL-22RA2) in the jejunum of piglets. Furthermore, the mRNA expression of IL-22 in 7-day-old piglets was significantly higher than that in 0-day-old piglets. Adult epithelial markers were significantly up-regulated in organoids treated with recombinant murine cytokine IL-22. Thus, IL-22 may play a key role in iron-affecting intestinal epithelium development.
Article
It is a great challenge to develop a safe and effective treatment strategy for age-related osteoporosis and fracture healing. As one of the four FOXO transcription factors, FOXO1 is essential for cell proliferation, survival, senescence, energy metabolism, and oxidative stress in various cells. Our previous study demonstrated that specific Foxo1 gene deletion in osteoblasts in young mice results in bone loss while that in aged mice shows the opposite effect. However, the mechanism underlying the differential regulation of bone metabolism by FOXO1 remains to be elucidated. In this study, we generated osteoblast-specific Foxo1 knockout mice by using Foxo1fl/fl and Bglap-Cre mice. In young mice, Foxo1 gene deletion inhibits osteoblast differentiation, leading to a decreased osteoblast number and decreased bone formation rate because of the weakened ability to resist oxidative stress, eventually resulting in bone loss and delayed healing of bone defects. In aged mice, high levels of reactive oxygen species (ROS) promote the diversion of CTNNB1 (β-catenin) from T cell factor 4 (TCF4)- to FOXO1-mediated transcription, thereby inhibiting Wnt/β-catenin signaling and leading to decreased osteogenic activity. Conversely, FOXO1 deficiency indirectly promotes the binding of β-catenin and TCF4 and activates Wnt/β-catenin signaling, thereby alleviating age-related bone loss and improving bone defect healing. Our study proves that FOXO1 has differential effects on bone metabolism in young and aged mice and elucidates its underlying mechanism. Further, this study provides a new perspective on the treatment of age-related osteoporosis.
Article
Full-text available
Iron accumulation is frequently associated with chronic liver diseases. However, our knowledge on how iron contributes to the liver injury is limited. Aberrant Wnt/β-catenin signaling is a hallmark of several hepatic pathologies. We recently reported that peroxisome proliferator activated receptor alpha (PPARα) agonist, fenofibrate prevents iron induced oxidative stress and β-catenin signaling by chelating the iron. Sirtuin3 (Sirt3), a type of NAD+-dependent deacetylase that plays a critical role in metabolic regulation was found to prevent ischemia reperfusion injury by normalizing the Wnt/β-catenin pathway. In the present study, we explored if fenofibrate prevents iron induced liver injury by regulating the Sirt3 and β-catenin signaling. In-vitro and in-vivo iron treatment resulted in the downregulation of PPARα, Sirt3, active β-catenin and its downstream target gene c-Myc in the mouse liver. Pharmacological activation of Sirt3, both invitro and in vivo, by Honokiol (HK), a known activator of Sirt3, abrogated the inhibitory effect of iron overload on active β-catenin expression and prevented the iron induced upregulation of αSMA and TGFβ expression. Intrinsically, PPARα KO mice showed significant downregulation of hepatic Sirt3 levels. In addition, treatment of iron overload mice with PPARα agonist fenofibrate reduced hepatic iron accumulation and prevented iron induced downregulation of liver Sirt3 and active β-catenin, mitigating the progression of fibrosis. Thus, our results establish a novel link between hepatic iron and PPARα, Sirt3 and β-catenin signaling. Further exploration on the mechanisms by which fenofibrate ameliorates iron induced liver injury likely has significant therapeutic impact on iron associated chronic liver diseases.
Article
Full-text available
Accumulating evidence strongly implicates iron in the pathogenesis of aging and disease. Iron levels have been found to increase with age in both the human and mouse retinas. We and others have shown that retinal diseases such as age-related macular degeneration and diabetic retinopathy are associated with disrupted iron homeostasis, resulting in retinal iron accumulation. In addition, hereditary disorders due to mutation in one of the iron regulatory genes lead to age dependent retinal iron overload and degeneration. However, our knowledge on whether iron toxicity contributes to the retinopathy is limited. Recently, we reported that iron accumulation is associated with the upregulation of retinal and renal renin–angiotensin system (RAS). Evidences indicate that multiple genes/components of the RAS are targets of Wnt/β-catenin signaling. Interestingly, aberrant activation of Wnt/β-catenin signaling is observed in several degenerative diseases. In the present study, we explored whether iron accumulation regulates canonical Wnt signaling in the retina. We found that in vitro and in vivo iron treatment resulted in the upregulation of Wnt/β-catenin signaling and its downstream target genes including renin–angiotensin system in the retina. We confirmed further that iron activates canonical Wnt signaling in the retina using TOPFlash T-cell factor/lymphoid enhancer factor promoter assay and Axin2-LacZ reporter mouse. The presence of an iron chelator or an antioxidant reversed the iron-mediated upregulation of Wnt/β-catenin signaling in retinal pigment epithelial (RPE) cells. In addition, treatment of RPE cells with peroxisome proliferator-activated receptor (PPAR) α-agonist fenofibrate prevented iron-induced activation of oxidative stress and Wnt/β-catenin signaling by chelating the iron. The role of fenofibrate, an FDA-approved drug for hyperlipidemia, as an iron chelator has potentially significant therapeutic impact on iron associated degenerative diseases.
Article
Full-text available
The bone marrow microenvironment plays important roles in the progression of the myelodysplastic syndrome (MDS). The higher incidence of ASXL1 and TET2 gene mutations in our iron overload (IO) MDS patients suggests that IO may be involved in the pathogenesis of MDS. The effects of IO damaging bone marrow mesenchymal stromal cells (MSCs) from higher-risk MDS patients were investigated. In our study, IO decreased the quantity and weakened the abilities of proliferation and differentiation of MSCs, and it inhibited the gene expressions of VEGFA, CXCL12, and TGF-β1 in MSCs regulating hematopoiesis. The increased level of reactive oxygen species (ROS) in MSCs caused by IO might be inducing apoptosis by activating caspase3 signals and involving in MDS progression by activating β-catenin signals. The damages of MSCs caused by IO could be partially reversed by an antioxidant or an iron chelator. Furthermore, the MSCs in IO MDS/AML patients had increased levels of ROS and apoptosis, and the expressions of caspase3 and β-catenin were increased even further. In conclusion, IO affects gene stability in higher-risk MDS patients and impairs MSCs by inducing ROS-related apoptosis and activating the Wnt/β-catenin signaling pathway, which could be partially reversed by an antioxidant or an iron chelator.
Article
Full-text available
Ageing phenotypes, such as years lived in good health (healthspan), total years lived (lifespan), and survival until an exceptional old age (longevity), are of interest to us all but require exceptionally large sample sizes to study genetically. Here we combine existing genome-wide association summary statistics for healthspan, parental lifespan, and longevity in a multivariate framework, increasing statistical power, and identify 10 genomic loci which influence all three phenotypes, of which five (near FOXO3, SLC4A7, LINC02513, ZW10, and FGD6) have not been reported previously at genome-wide significance. The majority of these 10 loci are associated with cardiovascular disease and some affect the expression of genes known to change their activity with age. In total, we implicate 78 genes, and find these to be enriched for ageing pathways previously highlighted in model organisms, such as the response to DNA damage, apoptosis, and homeostasis. Finally, we identify a pathway worthy of further study: haem metabolism. Ageing phenotypes are of great interest but are difficult to study genetically, partly due to the sample sizes required. Here, the authors present a multivariate framework to combine GWAS summary statistics and increase statistical power, identifying additional loci enriched for aging.
Article
Full-text available
Background: The incidence of osteoporotic fractures is increasing. In this study, we explored the activities of Wnt/β-catenin signaling in bone tissues with iron accumulation. Methods: We established rat bipedal walking models (RBWM), and a portion of our RBWM rats were intraperitoneally injected with ferric ammonium citrate, normal saline, and deferoxamine. Bone mineral density was measured with a small animal in vivo imaging system. The protein levels of ferritin, TRAP-5B, RANKL, and OPG in serum were measured by the enzyme-linked immunosorbent assay (ELISA). Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to quantify the RNA and protein expression levels of certain regulators involved in Wnt/β-catenin signaling in bone tissues. Results: In the present study, we established a rat bipedal walking model containing 32 bipedal rats, which were randomly classified into four groups, termed as NS, FAC, FAC+NS, and FAC+DFO. Those three experimental groups with FAC injection had significantly lower bone mineral density (BMD) than the control group NS (P < 0.05). The disruption of bone homeostasis and downregulation of Wnt/β-catenin signaling were also observed in the three groups with FAC injection. Moreover, after the injection of deferoxamine, those aberrations in samples with FAC injection seemed repaired as test results returning or getting close to normal ranges. Conclusion: The osteoporosis could be caused by iron overload, which reduced the bone mineral density by disrupting the homeostasis of bone formation and absorption and attenuating the Wnt/β-catenin signaling in bone tissues. The deferoxamine had the potential to improve the bone health by reducing the accumulation of iron and increasing the bone mass, which might be a promising therapeutic solution for osteoporosis.
Article
Full-text available
Objective Hepcidin deficiency is known to cause body iron accumulation and bone microarchitecture defects, but the exact underlying mechanisms of hepcidin deficiency-induced bone loss remain unclear. Our objective was to understand the molecular mechanism of hepcidin deficiency-induced bone loss. Methods The bone phenotypes of wild type (WT) and hepcidin knockout (Hepcidin-KO) mice were measured by microcomputed tomography. The osteoclastic marker of the bone was measured by tartrate-resistant acid phosphatase staining. The osteoblastic marker of the bone was measured by immunostaining of osteocalcin. Primary osteoblastic and osteoclastic differentiation was performed using bone marrow cells. The mature osteoclast was determined by tartrate-resistant acid phosphatase staining, pit formation assay and relative gene expression. The mature osteoblast was determined by alkaline phosphatase activity, alkaline phosphatase staining, Alizarin Red staining and relative gene expression. The protein expression of β-catenin, TCF4/TCF7L2 and Forkhead box O3a (FOXO3a) was measured by Western blot and their combination by co-immunoprecipitation. In vivo study was performed by tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice. Results We found that Hepcidin-KO mice exhibited iron accumulation and bone loss compared with WT mice. The osteoclastic differentiation of bone marrow-derived macrophages from Hepcidin-KO mice was not significantly different from that of bone marrow–derived macrophages from WT mice. However, the osteoblastic differentiation of bone marrow–derived mesenchymal stem cells from Hepcidin-KO mice was obviously decreased compared with that of bone marrow–derived mesenchymal stem cells from WT mice. Furthermore, it was confirmed in this study that upon hepcidin deficiency, β-catenin, TCF4/TCF7L2 and FOXO3a expression in bone tissues was not altered, but β-catenin combination with TCF4/TCF7L2 was strongly inhibited by β-catenin combination with FOXO3a, indicating that the canonical Wnt/β-catenin pathway was affected. Tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice resulted in bone mass recovery. Conclusion These findings suggested that hepcidin deficiency might cause bone loss by interfering with the canonical Wnt/β-catenin pathway via FOXO3a, and FOXO3a inhibition would be a possible approach to treat hepcidin deficiency-induced bone loss.
Article
Full-text available
: Age-related macular degeneration (AMD) is considered as the main worldwide cause of blindness in elderly adults. Exudative AMD type represents 10 to 15% of macular degeneration cases, but is the main cause of vision loss and blindness. Circadian rhythm changes are associated with aging and could further accelerate it. However, the link between circadian rhythms and exudative AMD is not fully understood. Some evidence suggests that dysregulation of circadian functions could be manifestations of diseases or could be risk factors for the development of disease in elderly adults. Biological rhythms are complex systems interacting with the environment and control several physiological pathways. Recent findings have shown that the dysregulation of circadian rhythms is correlated with exudative AMD. One of the main pathways involved in exudative AMD is the canonical WNT/β-catenin pathway. Circadian clocks have a main role in some tissues by driving the circadian expression of genes involved in physiological and metabolic functions. In exudative AMD, the increase of the canonical WNT/β-catenin pathway is enhanced by the dysregulation of circadian rhythms. Exudative AMD progression is associated with major metabolic reprogramming, initiated by aberrant WNT/β-catenin pathway, of aerobic glycolysis. This review focuses on the interest of circadian rhythm dysregulation in exudative AMD through the aberrant upregulation of the canonical WNT-catenin pathway.
Article
Full-text available
The role of the evolutionarily conserved Wnt signaling pathway is well documented in several cellular processes, such as cell proliferation, differentiation, cell motility, and maintenance of the stem cell niche. The very first indication that aberrant Wnt signaling can cause carcinogenesis came from a finding that the mutation of the adenomatous polyposis coli gene (APC) predisposes a person to colorectal carcinoma. Later, with progressing research it became clear that abnormal activation or mutation of the genes related to this pathway could drive tumorigenesis. Here, we review recent advances in research regarding Wnt signaling regulation and its role in several cancer subtypes. Additionally, the utility of Wnt pathway–targeted cancer therapy intervention is also highlighted, with an overview of current approaches to target the Wnt pathway in oncogenesis and the future scopes and challenges associated with them.
Article
Background: Hepatocellular cancer (HCC) remains a major unmet clinical need. Although activating CTNNB1 mutations are seen in prominent subsets of HCC cases, these by themselves are insufficient for hepatocarcinogenesis. Co-expression of mutant CTNNB1 with clinically relevant co-occurrence has yielded HCCs. Here, we identify cooperation between β-catenin and Nrf2 signaling in HCC. Methods: Public HCC datasets were assessed for concomitant presence of CTNNB1 mutations and either mutations in NFE2L2 or KEAP1, or Nrf2 activation by gene signature. HCC development in mice and similarity to human HCC subsets was assessed following co-expression of T41A-CTNNB1 with either WT-, G31A- or T80K-NFE2L2. Based on mTORC1 activation in CTNNB1-mutated HCCs, response of preclinical HCC to mTOR inhibitor was investigated. Results: Overall, 9% of HCC cases showed concomitant CTNNB1 mutations and Nrf2 activation, subsets of which were due to mutations in NFE2L2/KEAP1. Co-expression of mutated-CTNNB1 with mutant-NFE2L2 but not WT-NFE2L2 led to HCC development and mortality by 12-14 weeks. These HCCs were positive for β-catenin targets like Glutamine synthetase and Cyclin-D1, and Nrf2 targets like NAD(P)H Quinone Dehydrogenase 1 and peroxiredoxin 1. RNA-seq and pathway analysis showed high concordance of preclinical HCC to human HCC subset showing activation of unique (Iron Homeostasis and Glioblastoma Multiforme signaling) and expected (Glutamine Metabolism) pathways. NFE2L2-CTNNB1 HCC mice were treated with mTOR inhibitor everolimus (5mg/kg diet ad libitum), which led to >50% decrease in tumor burden. Conclusion Co-activation of β-catenin and Nrf2 is evident in 9% of all human HCCs. Co-expression of mutant-NFE2L2 and mutant-CTNNB1 led to clinically relevant HCC development in mice, which responded to mTOR inhibitors. Thus, this model has both biological and therapeutic implications.
Article
Disorders of vascular structure and function play a central role in a wide variety of CNS diseases. Mutations in the Frizzled-4 (Fz4) receptor, Lrp5 coreceptor, or Norrin ligand cause retinal hypovascularization, but the mechanisms by which Norrin/Fz4/Lrp signaling controls vascular development have not been defined. Using mouse genetic and cell culture models, we show that loss of Fz4 signaling in endothelial cells causes defective vascular growth, which leads to chronic but reversible silencing of retinal neurons. Loss of Fz4 in all endothelial cells disrupts the blood brain barrier in the cerebellum, whereas excessive Fz4 signaling disrupts embryonic angiogenesis. Sox17, a transcription factor that is upregulated by Norrin/Fz4/Lrp signaling, plays a central role in inducing the angiogenic program controlled by Norrin/Fz4/Lrp. These experiments establish a cellular basis for retinal hypovascularization diseases due to insufficient Frizzled signaling, and they suggest a broader role for Frizzled signaling in vascular growth, remodeling, maintenance, and disease