ArticlePDF AvailableLiterature Review

DYRK1A: the double-edged kinase as a protagonist in cell growth and tumorigenesis

Authors:

Abstract and Figures

DYRK1A (dual-specificity tyrosine-regulated kinase 1A) is a kinase with multiple implications for embryonic development, especially in the nervous system where it regulates the balance between proliferation and differentiation of neural progenitors. The DYRK1A gene is located in the Down syndrome critical region and may play a significant role in the developmental brain defects, early neurodegeneration, and cancer susceptibility of individuals with this syndrome. DYRK1A is also expressed in adults, where it might participate in the regulation of cell cycle, survival, and tumorigenesis, thus representing a potential therapeutic target for certain types of cancer. However, the final readout of DYRK1A overexpression or inhibition depends strongly on the cellular context, as it has both tumor suppressor and oncogenic activities. Here, we will discuss the functions and substrates of DYRK1A associated with the control of cell growth and tumorigenesis with a focus on the potential use of DYRK1A inhibitors in cancer therapy. http://www.tandfonline.com/eprint/T5DXnvz6IfzbNm2CtwUF/full
Content may be subject to copyright.
This article was downloaded by: [78.25.98.238]
On: 20 August 2015, At: 13:47
Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place,
London, SW1P 1WG
Click for updates
Molecular & Cellular Oncology
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/kmco20
DYRK1A: the double-edged kinase as a protagonist in
cell growth and tumorigenesis
P Fernández-Martíneza, C Zahonerob & P Sánchez-Gómezb
a Instituto de Medicina Molecular Aplicada; Universidad CEU-San Pablo; Madrid, Spain
b Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC; Madrid, Spain
Published online: 30 Jan 2015.
To cite this article: P Fernández-Martínez, C Zahonero & P Sánchez-Gómez (2015) DYRK1A: the double-edged
kinase as a protagonist in cell growth and tumorigenesis, Molecular & Cellular Oncology, 2:1, e970048, DOI:
10.4161/23723548.2014.970048
To link to this article: http://dx.doi.org/10.4161/23723548.2014.970048
PLEASE SCROLL DOWN FOR ARTICLE
Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in
the publications on our platform. Taylor & Francis, our agents, and our licensors make no representations or
warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Versions
of published Taylor & Francis and Routledge Open articles and Taylor & Francis and Routledge Open Select
articles posted to institutional or subject repositories or any other third-party website are without warranty
from Taylor & Francis of any kind, either expressed or implied, including, but not limited to, warranties of
merchantability, fitness for a particular purpose, or non-infringement. Any opinions and views expressed in this
article are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The
accuracy of the Content should not be relied upon and should be independently verified with primary sources
of information. Taylor & Francis shall not be liable for any losses, actions, claims, proceedings, demands,
costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in
connection with, in relation to or arising out of the use of the Content.
This article may be used for research, teaching, and private study purposes. Terms & Conditions of access and
use can be found at http://www.tandfonline.com/page/terms-and-conditions
It is essential that you check the license status of any given Open and Open Select article to confirm
conditions of access and use.
DYRK1A: the double-edged kinase as a
protagonist in cell growth and tumorigenesis
P Fern
andez-Martínez
1
, C Zahonero
2
, and P S
anchez-G
omez
2,
*
1
Instituto de Medicina Molecular Aplicada; Universidad CEU-San Pablo; Madrid, Spain;
2
Neuro-oncology Unit; Instituto de Salud Carlos III-UFIEC; Madrid, Spain
Keywords: cancer, cell proliferation, cell differentiation, DYRK1A, neural progenitors
DYRK1A (dual-specicity tyrosine-regulated kinase 1A) is a
kinase with multiple implications for embryonic
development, especially in the nervous system where it
regulates the balance between proliferation and
differentiation of neural progenitors. The DYRK1A gene is
located in the Down syndrome critical region and may play a
signicant role in the developmental brain defects, early
neurodegeneration, and cancer susceptibility of individuals
with this syndrome. DYRK1A is also expressed in adults, where
it might participate in the regulation of cell cycle, survival,
and tumorigenesis, thus representing a potential therapeutic
target for certain types of cancer. However, the nal readout
of DYRK1A overexpression or inhibition depends strongly on
the cellular context, as it has both tumor suppressor and
oncogenic activities. Here, we will discuss the functions and
substrates of DYRK1A associated with the control of cell
growth and tumorigenesis with a focus on the potential use
of DYRK1A inhibitors in cancer therapy.
Introduction
DYRK (dual-specificity tyrosine-regulated kinase) family
members represent a subfamily of protein kinases that have been
identified in distantly related organisms such as yeast, Drosophila,
and human. Seven mammalian Dyrk-related kinases have been
identified: DYRK1A, DYRK1B, DYRK1C, DYRK2, DYRK3,
DYRK4A, and DYRK4B.
1,2
The DYRK proteins are dual-speci-
ficity protein kinases that autophosphorylate a conserved tyrosine
(Y) residue in their own activation loop but phosphorylate their
substrates at serine (S) or threonine (T) residues.
3,4
The Y auto-
phosphorylation occurs during translation and induces kinase
activation; however, once the protein is fully translated, kinase
activity becomes restricted to S and T residues and no longer
depends on Y phosphorylation.
5,6
DYRK1A is the most extensively studied among this family of
kinases because its gene maps to human chromosome 21 within
the Down syndrome critical region (DSCR).
7-9
Moreover, this
kinase is overexpressed in the brain of patients with Down syn-
drome (DS) and many of its known substrates have been linked
to neuropathologic traits of this syndrome.
10,11
In fact, accumu-
lating evidence in experimental models suggests that DYRK1A
inhibitors can reverse some of the neurologic alterations associ-
ated with its overexpression and therefore could be of use in indi-
viduals with DS.
12
These aspects have been extensively reviewed
elsewhere. However, there are other less well-known facets of
DYRK1A, such as its participation in cancer. Many of its down-
stream targets are associated with the control of cell growth and
survival, especially in the nervous system, where it has been
mostly studied, but also in other tissues. Interestingly, in the can-
cer context, DYRK1A activity might be linked to both oncogene-
sis and tumor suppression. Here, we will try to summarize all
known implications of DYRK1A function in cell growth and
cancer with the aim of understanding this dichotomy, which
could have clinically relevant implications for the development of
therapeutic DYRK1A inhibitors.
Role of DYRK1A in Embryonic and Adult
Neurogenesis
Vertebrate Dyrk1A is expressed ubiquitously in a broad spec-
trum of embryonic tissues at different stages of development but
also in some adult tissues, most prevalently in heart, lung, brain,
and skeletal muscle.
7-9,13
In mice, the absence of Dyrk1A is lethal
at the embryonic stage. Heterozygous animals are viable but have
a reduced size at birth that is maintained through adulthood. This
reduction is more noticeable in organs such as the brain and liver.
Heterozygous mice show decreased neonatal viability, a reduced
number of neurons in brain areas, alterations in motor and devel-
opment, dopaminergic deficiency, and impairment in spatial learn-
ing development.
14,15
Conversely, transgenic mice overexpressing
Dyrk1A also present a neurodevelopmental delay and motor and
cognitive deficits.
16-18
These data reflect the extreme gene dosage
sensitivity of this protein and its relevance during neural system
(NS) development, where it controls proliferation, neurogenesis,
neural differentiation, cell death, and synaptic plasticity.
19,20
Drosophila Dyrk1A mutants (minibrain, mnb
-/-
flies) develop a
smaller adult brain (especially the optic lobes and the central brain
hemispheres), which appears to be caused by altered neuroblast
© P Fern
andez-Martínez, C Zahonero, and P S
anchez-G
omez
*Correspondence to: P S
anchez-G
omez; Email: psanchezg@isciii.es
Submitted: 06/13/2014; Revised: 09/03/2014; Accepted: 09/03/2014
http://dx.doi.org/10.4161/23723548.2014.970048
This is an Open Access article distributed under the terms of the Creative
Commons Attribution-Non-Commercial License (http://creativecommons.
org/licenses/by-nc/3.0/), which permits unrestricted non-commercial use,
distribution, and reproduction in any medium, provided the original work is
properly cited. The moral rights of the named author(s) have been asserted.
www.tandfonline.com e970048-1Molecular & Cellular Oncology
Molecular & Cellular Oncology 2:1, e970048; January/February/March 2015; Published with license by Taylor & Francis Group, LLC
REVIEW
Downloaded by [78.25.98.238] at 13:47 20 August 2015
proliferation during postembryonic neurogenesis.
21
Interestingly,
recent evidence suggests that this function is not restricted to the
NS as Mnb is also required for normal leg and wing growth con-
trol.
22
Moreover, truncation of the human DYRK1A gene causes
microcephaly,
23
further supporting an evolutionary conserved
function of this kinase during brain development. There appears to
be a dynamic spatiotemporal expression pattern of Dyrk1A that is
tightly controlled during vertebrate NS development. There is a
transient peak of expression of mouse Dyrk1A immediately before
the transition from proliferating to neurogenic divisions.
24
After
that, its expression is maintained in neural progenitors (NPs),
although at a lower level. Later on, Dyrk1A is upregulated in new-
born postmitotic neurons and downregulated as the neuron begins
to migrate away from the ventricular zone. Once the migrating neu-
ron reaches its target position, Dyrk1A is again expressed before the
final differentiation and dendrite formation occurs.
25,26
These
changes in Dyrk1A expression reinforce the notion that it works as
an inhibitor of cell cycle progression. In fact, in utero electropora-
tion of Dyrk1A in the embryonic mouse neocortex inhibits cell pro-
liferation by inducing the nuclear export and degradation of cyclin
D1.
27
A more recent study indicates that DyrkA kinase activity is
responsible for the stabilization of cellular cyclin D1 and the degra-
dation of p27 (a cyclin-dependent kinase [CDK] inhibitor) in
mouse and human cells.
28
Other authors have shown that upregula-
tion of Dyrk1A induces proliferation arrest of embryonic NPs. Con-
versely, its loss of function causes overproliferation and cell death in
the embryonic chick spinal cord and mouse telencephalon.
29
These
authors suggest that Dyrk1A is both necessary and sufficient for
transcriptional upregulation of the expression of p27. Furthermore,
Dyrk1A phosphorylates p53 in rat embryonic hippocampal
progenitors H19–7 cells, which leads to a
robust induction of p21.
30
These results
support a model in which Dyrk1A impairs
cell cycle progression of embryonic progen-
itors, especially at the transition from G1/
G0 to S phase (summarized in Fig. 1). In
contrast with these data, mnb overexpres-
sion promotes organ growth through inhi-
bition of the Salvador-Warts-Hippo
(SWH) pathway, also called the Hippo
pathway,
22
a known inhibitor of prolifera-
tion and inducer of apoptosis in flies and
mammals.
31-34
Whether these differences
are species- or tissue-specific is not known,
but throughout the text we will see more
examples of different, and even opposite,
readouts of Dyrk1A functions in different
contexts. Although there is no clear expla-
nation for this behavior this gene is
extremely dosage dependent so one could
hypothesize that changes in the level or the
duration of Dyrk1A expression could have
different consequences. There are even
cases in which both downregulation and
overexpression of DYRK1A have the same
readout.
35
Moreover, a recent study using
single-cell image analysis has shown that Dyrk1A mediates a dose-
dependent increase in the duration of the G1 phase via direct phos-
phorylation and subsequent degradation of cyclinD1, directing
PC12 cells into a reversible arrested state. In contrast, knockdown
or kinase inhibition of Dyrk1A greatly increased cyclinD1 protein
levels and split cells into 2 fates, with one subpopulation (with low
p21 expression) shortening the G1 phase and the other (with high
p21 expression) entering a persistent arrested state that differs from
the normal quiescence state in which expression of both proteins is
low.
36
Thus, both upregulation and downregulation of Dyrk1A
levels could lead to cell cycle exit (transient or irreversible, respec-
tively) and have similar consequences on tissue growth.
In addition to directly controlling the cell cycle machinery, it
has been suggested that overexpression of Dyrk1A is necessary to
induce neural differentiation, although it is not clear whether it is
sufficient for this final outcome.
27,29
Regarding the mechanism,
Yang and coworkers have shown that Dyrk1A activity is induced
during in vitro differentiation of hippocampal progenitor cells,
leading to the stimulation of cAMP responsive element binding
protein (CREB) transcriptional activity.
37
Another group has
suggested that Dyrk1A overexpression potentiates nerve growth
factor (NGF)-mediated PC12 neuronal differentiation by upre-
gulating the Ras/MAP kinase signaling pathway.
38
More
recently, it has been proposed that Dyrk1A phosphorylates
Notch in the nuclear compartment and is co-expressed with the
Notch ligand Delta1 in single NPs. Furthermore, Dyrk1A sup-
presses Notch signaling and reduces its capacity to sustain tran-
scription in neural cells, counteracting its antidifferentiative
actions.
29,39
Another interesting possibility would be that
Dyrk1A influences neuronal differentiation through the
Figure 1. DYRK1A activity blocks cell cycle progression. DYRK1A is considered primarily an inhibitor
of proliferation due to its capacity to either block (red lines) cell cycle promoters (green boxes), or
activate (green arrows) cell cycle inhibitors (red boxes).
e970048-2 Volume 2 Issue 1Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
modulation of RE1-silencing transcription factor (REST, also
known as neuron-restrictive silencer factor or NRSF). REST is a
zinc finger transcription factor that silences a range of neuronal
genes in differentiated non-neuronal tissues and NPs.
40
Further-
more, the dissociation of REST and its co-repressors from the
RE1 sites is both necessary and sufficient to trigger the transition
from pluripotent embryonic stem cells to NPs, and from these to
mature neurons.
41
Moreover, REST is degraded in the G2 phase
and this is necessary for the derepression of Mad2, an essential
component of the spindle assembly checkpoint.
42
Reduced
expression of REST has been observed in cultured fetal DS brain
cell-derived neurospheres
43
as well as in the brains of DS mouse
models.
44,45
By inhibiting REST expression, DYRK1A might
inhibit the G2-M checkpoint (Fig. 1) as well as promote some of
the neural differentiation defects observed in DS.
In contrast to the antiproliferative and prodifferentiative capac-
ity of Dyrk1A during CNS development, we recently suggested
that this kinase sustains adult NP self-renewal.
46
In our experience,
Dyrk1A protein is actively distributed during adult NP cell divi-
sion and the inherited kinase acts as an inhibitor of epidermal
growth factor receptor (EGFR) degradation by phosphorylating
Sprouty2 (Spry2), modulator of receptor tyrosine kinases (RTK)
turnover. Interestingly DYRK1A phosphorylates T75 on Spry2
and impairs its inhibitory activity on extracellular signal-regulated
kinase (ERK) signaling downstream of fibroblast growth factor
(FGF).
47
However, Spry2 also phosphorylates and sequesters Cbl,
a major effector of EGFR degradation.
48
The data on adult pro-
genitors suggest that Dyrk1A phosphorylation could be beneficial
for the positive function of Spry2 downstream of EGFR but coun-
teractive for the inhibitory function of this protein on FGF signal-
ing (Fig. 3). Therefore, high expression of Dyrk1A should
correlate with general activation of RTK function. It would be
interesting to test whether phosphorylation by Dyrk1A modulates
the binding of Spry2 to regulatory proteins, as it has been
described for phosphorylation of other Spry2 residues.
49
In fact,
Dyrk1A overexpression decreases the interaction of Spry2 and
growth factor receptor-bound protein 2 (Grb2), an adaptor
between RTK and the ERK pathway.
50
These results suggest that
DYRK1A kinase activity could have a positive function in both
EGFR and FGFR signaling, at least in the context of the adult
CNS. In agreement with this, activation of the AKT pathway, a
known transducer of RTK activation, has been observed n several
transgenic Dyrk1A mouse models, concomitant with an increase
in expression of CCND1 (the gene coding for cyclin D1) in post-
natal and adult stages.
50,51
Together, these data suggest that
Dyrk1A might act mainly as a negative regulator of cell cycle dur-
ing neural development, but also as a positive regulator of cell pro-
liferation in the adult CNS, probably due to differential expression
of downstream substrates or regulatory molecules.
Regulation of Quiescence through the DREAM
Complex
An important group of proteins that negatively regulate the cell
cycle are the retinoblastoma (RB) tumor suppressor protein and the
related family members p107 (RBL1) and p130 (RBL2).
52
p130
has been shown to accumulate in G0, when it interacts with E2F4
to repress E2F-dependent gene transcription.
53-55
P130 and E2F4
are part of a larger multisubunit protein complex, the mammalian
DREAM (dimerization partner, RB-like, E2F, and multivulval
class B) complex.
56,57
In a recent study, phosphorylation of the
DREAM component LIN52 at S28 by DYRK1A was shown to reg-
ulate complex formation in G0 and E2F repression.
58
Inhibition of
DYRK1A activity or a point mutation in LIN52 disrupted
DREAM assembly and reduced the ability of tumor cells to enter
quiescence and the capacity of fibroblasts to undergo Ras-induced
senescence
58
(Fig. 1). These authors have shown that overexpres-
sion of DYRK1A (but not a kinase-deficient mutant) can inhibit
proliferation and colony formation of a panel of tumor cell lines
from different tissues.
58
Moreover, by phosphorylating DYRK1A,
LATS2, a component of the Hippo pathway, could enhance the
kinase activity of DYRK1A toward the DREAM subunit LIN52.
Intriguingly, the LATS2 locus is physically linked with RB1 on
13q, and this region frequently displays loss of heterozygosity in
human cancers.
59
Together, these results provide a possible expla-
nation for the proposed tumor suppressor role of DYRK1A, espe-
cially at the tumor initiation stages. However, the same mechanism
could have a protective role later on in the response to chemother-
apy. In fact, a recent report indicates that inhibition of DYRK1A
could enhance imatinib-induced cell death in gastrointestinal
tumors by favoring apoptosis at the expense of cell quiescence.
60
Documented Roles of DYRK1A in Cancer
Epidemiologic studies suggest that individuals with DS have
an increased risk of acute megakaryoblastic leukemia (AMLK)
and acute lymphoblastic leukemia (ALL) in combination with
acquired mutations of the transcription factor globin transcrip-
tion factor 1 (GATA1).
61-64
These GATA1 mutations are local-
ized in exon 2 and provoke the expression of a shorter isoform
named GATA1s.
61
However, non-DS individuals with germline
GATA1 mutations similar to those seen in DS-AMLK have no
predisposition to leukemia.
63
Moreover, trisomy for the DSCR
that includes DYRK1A markedly increases the proliferation of
megakaryocytes and is sufficient to cooperate with GATA1 muta-
tions in this mouse model, reinforcing the oncogenic function of
DYRK1A in this cell type.
65
GATA1 is essential for erythroid
and megakaryocytic development. GATA1s retains some of the
functions of the wild-type protein although it does not repress
oncogenic MYC and the proproliferative E2F.
66,67
This could
explain its oncogenic action but might also lead to a positive
feedback loop as E2F stimulates DYRK1A transcription, further
increasing DYRK1A expression.
68
In contrast to the increased risk of leukemia, individuals with
DS show a considerably reduced incidence of most solid
tumors.
69,70
It has been suggested that overexpression of
DYRK1A, in cooperation with DSCR1 (another gene located in
the DS critical region) could diminish angiogenesis through
attenuation of vascular endothelial growth factor (VEGF)-calci-
neurin- nuclear factor of activated T cells (NFAT) signaling in
www.tandfonline.com e970048-3Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
endothelial cells.
71,72
NFAT proteins are transcription factors
that are activated as a result of a calcium flux released from endo-
plasmic reticulum stores and from the activation of channels in
the plasma membrane. This increase in calcium provokes
dephosphorylation of NFAT by the phosphatase calcineurin and
its translocation to the nucleus, where it cooperates with other
factors and coactivators to promote gene transcription.
73
Since
the discovery of NFAT proteins 2 decades ago as promoters of
interleukin (IL)-2 during the activation of T cells,
74
it has
become increasingly clear that these proteins are not only
expressed in immune cells but are also overexpressed in human
solid tumors and hematologic malignancies. In general, activa-
tion of the NFAT pathway is considered to be a cancer-promot-
ing event by enhancing proliferation and survival (in T cell
lymphoid malignancies), by enhancing metastatic dissemination
(in epithelial cancers), and by promoting angiogenesis.
73,75
Phos-
phorylation of NFAT by DYRK1A
71,76
primes it for subsequent
phosphorylation by casein kinase 1 (CK1) and glycogen synthase
kinase 3 (GSK3), which drives the inactivation and nuclear
export of NFAT.
77-79
As VEGF is one of the known NFAT tar-
gets, DYRK1A overexpression could lead to decreased angiogene-
sis, thus contributing to the lower incidence of adult solid
tumors in DS individuals.
72
However, it is important to keep in
mind that DS is a multifactorial syndrome and the overexpres-
sion of other genes could be responsible for the reduced cancer
predisposition. Paradoxically, it has been proposed that
DYRK1A also contributes to megakaryocytic malignancies
through the inhibition of NFAT, although the possibility that
other substrates of the kinase are implicated in the oncogenic
response has not been excluded.
65
Therefore, it seems that while
DYRK1A and NFAT suppress the growth of epithelial and lym-
phoid tumors in the adult, they could act as megakaryocytic
oncogenes in children. This led us to hypothesize that DYRK1A
has opposite functions during normal development and carcino-
genesis of the NS and blood cells.
These context-dependent functions of NFAT have similarities
with other well-known substrates of DYRK1A and reinforce the
notion that this kinase can have different, and even antagonistic
functions, depending on the cellular context or even the cancer
stage. This idea has already been suggested by others, at least in
the DS context.
80
Figure 2 summarizes some of the DYRK1A
substrates, focusing on their pro- or antitumoral roles. For exam-
ple, NOTCH function is inhibited by DYRK1A phosphoryla-
tion, as previously mentioned.
29,39
Hyperactivation of the
NOTCH pathway has classically been viewed as oncogenic in
several cancers (solid tumors and blood cancers) although recent
studies have revealed tumor suppressor roles for NOTCH signal-
ing in myeloid malignancies.
81
Therefore, DYRK1A inhibition
in NOTCH-related cancers could have positive or negative con-
sequences for tumor growth and survival. A similar scenario
might exist for the protein REST, which has antitumor properties
in epithelial cancers but can function as an oncogene for neural
tumors.
40
REST was first identified as a cancer-related gene a
decade ago from studies in medulloblastoma (MD), a childhood
tumor of the cerebellum,
82,83
and was later also characterized as
an oncogene in neuroblastoma (NB), the most common
extracranial solid tumor in children
84
and glioblastoma (GBM),
a deadly adult brain tumor.
85,86
These data suggest that REST
controls the maintenance of embryonic and adult NPs and, when
deregulated, can be implicated in neural tumors. As we previ-
ously mentioned, REST appears to be downregulated in DS
brains. However, more recent observations indicate that REST
can activate DYRK1A transcription through a NRSE site in the
human DYRK1A promoter region. Moreover, REST and Dyrk1A
are coordinately expressed during neural development, and
DYRK1A imbalance can destabilize REST protein expression
and reduce its transcriptional activity.
35
Therefore, it seems that
in this case the regulation is even more complicated: DYRK1A
can work as a positive or a negative modulator of REST expres-
sion/activity, which in turn could have oncogenic or tumor sup-
pressor activity depending on the tissue.
Survival- or Apoptosis-Related Substrates
of DYRK1A
In contrast to the dual (pro- or antitumoral) function of some
of the genes mentioned above, there is a list of prosurvival or
antiapoptotic proteins that are activated upon DYRK1A phos-
phorylation and could be important for carcinogenesis in differ-
ent tissues (Fig. 3). Signal transducer and activator of
transcription (STAT) is a latent transcription factor that trans-
mits signals generated primarily by cell surface receptors into the
nucleus. STAT3 is transiently activated in normal cells but is
constitutively activated in a wide variety of hematologic malig-
nancies (leukemia, lymphomas, and multiple myelomas) and
solid tumors (such as head and neck, breast, lung, gastric, hepato-
cellular, colorectal, brain, and prostate cancers).
87
There is strong
evidence suggesting that aberrant STAT3 signaling promotes ini-
tiation and progression of cancer by either inhibiting apoptosis
or inducing proliferation, angiogenesis, invasion, and metasta-
sis.
88
Phosphorylation of STAT3 at position Y705 by Janus
kinase (JAK) induces its dimerization and nuclear transloca-
tion.
89
However, the STAT3 molecule contains a second phos-
phorylation site, S727, within its C terminus that can be
phosphorylated by DYRK1A
90
and is necessary to achieve maxi-
mal transcriptional activity.
89
In fact, recent data suggest that
phosphorylation at S727 plays a principal role in regulation of
cell survival activity and nuclear translocation of STAT3.
91
Nev-
ertheless, inhibition of DYRK1A has not been explored as a way
to modulate the growth of STAT3-related tumors. GLI1, a major
effector of Sonic hedgehog (SHH) signaling, is another onco-
genic transcription factor whose nuclear translocation and func-
tion seems to be mediated by DYRK1A phosphorylation, most
likely through the retention of GLI1 in the nucleus.
92
SHH-GLI
signaling regulates tumor growth and survival, as well as metasta-
sis, in a number of tumors.
93
However, although STAT3 is a
bona fide DYRK1A substrate, the relevance of GLI1 phosphoryla-
tion by the kinase and the possible applications of DYRK1A
inhibitors in SHH-dependent tumors is still under debate.
DYRK1A function has been linked to negative regulation of
the intrinsic apoptotic pathway through the phosphorylation of
e970048-4 Volume 2 Issue 1Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
caspase-9.
94,95
Caspase-9 is activated by a variety of apoptotic
stimuli that trigger the release of cytochrome c from mitochon-
dria. Once in the cytosol, cytochrome c induces the oligomeriza-
tion of apoptotic protease activating factor 1 (Apaf-1) and the
subsequent recruitment of procaspase-9. These events lead to
activation of procaspase-9 by autocatalytic processing and conse-
quently to the activation of effector caspase-3 and caspase-7.
96
Activation of caspase-9 is inhibited by phosphorylation at T125,
which is catalyzed, among others, by Dyrk1A.
94,95
This is impor-
tant during the development of retina progenitors,
95
and in the
response of cells to hyperosmotic stress.
97
Therefore, the inhibi-
tion of DYRK1A would lead to caspase-9 induction and could
be exploited to enhance the apoptotic response of cancer cells to
chemotherapy. In addition, DYRK1A might promote cell sur-
vival through phosphorylation and activation of sirtuin 1
(SIRT1; also known as silent mating type information regulation
2 homolog or NAD (C)-dependent protein deacetylase), which
participates in the stress response and cellular metabolism.
98
It
has been shown that DYRK1A and DYRK3 directly phosphory-
late SIRT1 at T522, promoting deacetylation of p53, and, more
importantly, that the knockdown of endogenous DYRK1A and
DYRK3 sensitizes cells to DNA damage-induced cell death.
99
These data, together with the impairment of quiescence mediated
by inhibition of the DREAM complex and inhibition of the
HIPPO pathway (which induces apoptosis) by DYRK1A, suggest
that DYRK1A could be a good therapeutic target to increase cell
death in response to both chemo- and radiotherapy in different
tumors, as has been suggested by others.
100
Despite these prosurvival functions of DYRK1A, and exclud-
ing its well-known link to DS-related leukemia, it was not until
recently that our group described a clear association of this kinase
with tumor growth. Our results suggest that DYRK1A is highly
expressed in a subset of GBMs, where it correlates with the
expression and genetic amplification of EGFR. In parallel to
events in normal NPs, downregulation of DYRK1A leads to an
increase in EGFR degradation and therefore to inhibition of the
self-renewal capacity of GBM cells.
101
Furthermore, overexpres-
sion of SPRY2 was able to compensate for the EGFR degradation
promoted by DYRK1A inhibition and rescue the self-renewal
effect. On top of this, we have shown that genetic or pharmaco-
logic blockade of DYRK1A severely impaired tumor growth in
vivo, thus opening the door to the use of DYRK1A inhibitors in
glioma therapy, at least for EGFR-dependent tumors.
101
EGFR
is a well-known oncogene in a variety of tumors and inhibitors of
Figure 2. Double-edged regulation of tumorigenesis by DYRK1A. DYRK1A has been associated with protumoral activity (green boxes) by activating
(green arrows) known oncogenic proteins (red boxes) or by inhibiting (red lines) tumor suppressors (red boxes). However, an antitumoral capacity of
DYRK1A has been also described through its activation of tumor suppressors or its inhibition of oncogenic proteins. To add complexity to DYRK1A func-
tion, some of the known substrates of this kinase can have both oncogenic and tumor suppressor activities (green and red boxes), depending on the cel-
lular context and the developmental stage.
www.tandfonline.com e970048-5Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
its tyrosine kinase activity are being tested in many of these,
including GBM, although the results have not always been always
optimal.
102
Impaired endocytic downregulation of this receptor
is frequently associated with cancer. Indeed, dominant-negative
forms of CBL have been identified as oncogenes in human mye-
loid neoplasms
103
and SPRY2 has tumor-promoting activity in
colon cancer.
104
Therefore, the induction of EGFR degradation
mediated by inhibition of DYRK1A could represent an alterna-
tive strategy for EGFR-related cancers such as colon, lung, and
head and neck tumors.
To summarize this section, there is not a simple answer to the
question of whether DYRK1A functions as an oncogene or a
tumor suppressor (Fig. 2). In addition, it is important to note
that the relevance of some of the targets described above has not
been confirmed in a tumor context, or even in cancer cell lines.
Moreover, in the most conceivable situations several targets
would act in a combinatorial way. Therefore, the final response
of a cell to DYRK1A overexpression or inhibition probably
depends on the molecular and cellular context of each cancer.
DYRK1A Inhibitors and their Possible Use in Cancer
Therapeutics
In recent years there has been increased interest in inhibition
of DYRK1A activity for the treatment of the mental impairment
associated with several neurodegenerative diseases (including
Alzheimer’s Disease) and DS.
12,105
The fact that this kinase is
extremely dosage dependent increases its therapeutic potential as
a target. The goal then would be not to inhibit DYRK1A
completely but rather to a level comparable with that of healthy
conditions, which could reduce the potential side effects of tar-
geting this kinase. Most DYRK1A inhibitors are ATP competi-
tors and act by binding within the DYRK1A kinase domain
(direct competitors) or by preventing the functionality of the
ATP binding site (indirect competitors).
6
They can be classified
based on their structure (see
100
for a comprehensive review) or
based on their origin: i.e., natural compounds and their deriva-
tives, synthetic inhibitors, and broad spectrum kinase inhibitors
that show activity against DYRK1A. Table 1 reviews the most
potent DYRK1A inhibitors and their possible implications in
cancer. Many of them can inhibit other members of the DYRK
family, as well as structurally related kinases such as CDKs
(important regulators of cell cycle progression) and CDK-like
kinases (CLKs), which participate in pre-mRNA splicing.
106
Dif-
ferent modifications of these compounds have been envisioned in
order to increase specificity and avoid side effects.
Among DYRK1A inhibitors, the b-carboline alkaloid known
as harmine is currently the most selective and effective, although
it also blocks other DYRKs, especially DYRK1B. It functions as
an ATP-competitive inhibitor to specifically block the S/T kinase
activity.
6,107
Harmine is isolated from divergent plant species,
including the South American vine Banisteriopsis caapi and the
mideastern shrub Peganum harmala. It is a component of ayahua-
sca, a hallucinogenic brew of plant extracts that has been used for
centuries in healing and to cure illnesses, including some types of
cancer. However, several active principles could be involved in
this action and there is no direct evidence that targeting
DYRK1A is the relevant anticancer action of ayahuasca.
108
Har-
mine has been shown to inhibit neovessel formation in vitro and
in vivo through the regulation of several angiogenic factors and
inflammatory cytokines.
109
Moreover, harmine and related
b-carboniles display cytostatic and/or cytotoxic activity toward
cancer cells, including leukemia, colon, liver, gastric, and glioma
cells.
65,101
,
110-114
However, harmine has long been known to be
a potent inhibitor of monoamine oxidase-A
115
and to have hallu-
cinogenic properties as a result of its affinity for the tryptamine
and serotonin receptor binding sites.
116
These properties seri-
ously limit the in vivo therapeutic applications of this compound.
Nonetheless, molecular docking analysis showed that harmine
has many degrees of freedom in the ATP-binding pocket of
DYRK1A and this could be exploited to more selectively inhibit
the kinase.
117
Glioma cells are also sensitive in vitro to INDY, a
benzothiazol derivative.
101
The in vitro biological activity of
INDY has been confirmed to involve blockage of Tau phosphor-
ylation and restoration of NFAT activity, and it can rescue the
developmental defects of Dyrk1A overexpressing tadpoles in
vivo.
105,118
However, nothing is currently known about the phar-
macokinetic properties of this compound in whole animals or
humans, which hampers its therapeutic use. Another 2 synthetic
compounds, lamerallins and meriolins, have shown some anti-
cancer and proapoptotic effects in cancer cells and mouse glioma
models, although other kinases (especially CDKs) could be
implicated in this activity.
119-123
Epigallocatechin gallate (EGCG) is a polyphenol and a major
catechin component of green tea that shows selective inhibition
of DYRK1A compared to other structurally and functionally
related kinases by functioning as a noncompetitive inhibitor of
ATP.
124
Interestingly, a diet rich in green tea clearly improved
the brain structure defects and cognitive impairments of
DYRK1A-overexpressing mice.
51
EGCG has been shown to have
anticancer activity by inhibiting topoisomerases I/II, the antia-
poptotic enzyme Bcl-xl, and cancer promoting proteases. More-
over, it has additional health properties as it shows potent
antibacterial and antiviral activity and elicits antioxidative and
anti-inflammatory capacity by suppressing the nitric oxide syn-
thase pathway.
105
Although it seems clear that the anticancer
activities of green tea and EGCG are not limited to DYRK1A
inhibition, their safety for human consumption make them good
candidates for use in DYRK1A-related tumors and neurodegen-
erative diseases.
Most of the inhibitors mentioned in Table 1 have the capacity
to block other DYRK proteins, especially the closely related
DYRK1B (also known as MIRK). MIRK has been characterized
as a negative regulator of cell proliferation that modulates the
protein stability of several cell cycle-related molecules.
125,126
However, upregulation of MIRK expression and/or constitutive
activation of this kinase has been observed in several different
types of cancer, where it has been associated with the survival of
tumor cells in response to stress.
127
For example, depletion of
Mirk has been associated with apoptosis in lung cancer
128
and
also in pancreatic and ovarian cancer cell lines, where it seems to
e970048-6 Volume 2 Issue 1Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
modulate the levels of reactive oxygen species (ROS).
129-131
Therefore, the effect of some DYRK1A inhibitors in cancer could
also be due to MIRK inhibition, especially if they are associated
with changes in the response of tumor cells to hypoxia, nutrient
deprivation, and/or cytotoxic stimuli.
Conclusions and Future Perspectives
The results of several in vitro and in vivo studies link
DYRK1A activity with cell cycle exit, oncogene-induced senes-
cence, and cell differentiation, especially in the embryonic NS.
This notion is reinforced by the nature of many known DYRK1A
substrates, and also by the decreased cancer susceptibility of indi-
viduals with DS. Therefore one would expect this gene to behave
as a tumor suppressor, at least for the initial cancer stages. How-
ever, recent evidence indicates that DYRK1A can induce clono-
genic and prosurvival properties in certain types of cell or in
certain developmental conditions, and that this kinase can be
considered as an oncogene for at least 2 types of cancer: myeloid
leukemias and gliomas. As a proof of principle, harmine, a potent
DYRK1A inhibitor, can block cell growth and tumorigenesis in
those tumors. Moreover, DYRK1A might confer chemo- and
radioresistance capacities in several tumors by controlling the bal-
ance between quiescence and apoptosis. We propose that newly
identified DYRK1A inhibitors, which are being developed pri-
marily for their use in mental diseases, should also be considered
as anticancer agents, at least for AMKL and GBM. Moreover,
considering the relevance of DYRK1A in the NS, it would be
important to test the efficacy of such inhibitors in other neural
tumors such as oligodendrogliomas, which express high levels of
DYRK1A, and MB, given the important cerebellar expression of
this kinase. In such brain tumors, the regulation of EGFR turn-
over by DYRK1A might participate in the oncogenic action of
this kinase, in addition to modulation of REST, which is highly
expressed in cerebellar NPs, and SHH and/or NOTCH signal-
ing, which are associated with NS development. Special attention
should be given to secondary hits of DYRK1A inhibitors and to
the fact that DYRK1A blockade could affect several pathways.
However, in the case of cancer some promiscuity may be
Table 1. DYRK1A inhibitors. The table shows the IC
50
of different classes of DYRK1A inhibitors against DYRK1A, other DYRK proteins, and different kinases
(only targets with submicromolar IC
50
values are included). Cancer-related studies performed with those inhibitors are also indicated
Compound name
IC
50
(nM)
Evidence related to cancer
(Chemical class) DYRK1A Other targets therapy Refs.
Natural inhibitors Harmine (b-carboline) 22400 DYRK1B (166300), DYRK2 (900
1,900), DYRK3 (8001000), DYRK4
(80,000), MAO-A (5), CLK1 (27)
Angiogenesis inhibition;
Cytotoxic activity in tumor cell lines
in vitro (glioma, leukemia, colon,
gastric, liver), and in vivo (glioma)
101,107,111115,117
EGCG (Polyphenol) 40330 Vimentin (3), COMT (70) Clinical trials on cancer: urothelial,
bladder, prostate, myeloma, breast,
lung
124,132,133
Staurosporine (glycosilated
indolocarbazole)
20 Aurora A (7.2), Aurora B (20), Chk1
(1), Ftl3 (3), HGK (1), Ikkb (0.5), Jak2
(1), KDR (10), SYK (4)
134
Synthetic inhibitors Lamerallins (Chromenoindole) 405,000 CDK5 (720->10,000), GSK3 (310-
>10,000), CDK1/CyclinB, PIM1, CK1
(708,000)
Apoptosis induction and multidrug
resistance phenotype reversion
119121,135
INDY (Benzothiazol) 200 DYRK1B (240) Antiproliferative in gliomas
101,118
Meriolins (Pyrimidinylindol/
azaindol)
30 CDK1 (7170), CDK2 (318), CDK5
(3170), CDK9 (5.618), GSK3 (21
400), CK1 (50200)
Antiproliferative and proapoptotic
effects in tumor cells and glioma
xenografts
122,123
Variolin B (Pyrimidinylindol/
azaindol)
80 CDK1 (60), CDK2 (80) CDK5 (90)
CDK9 (26) GSK3 (70) CK1 (5)
122
Meridianins (Pyrimidinylindol/
azaindol)
34900 CDK5 (680->10,000), CK1 (490-
>10,000), CLK1 (3070)
136,137
KHCB19 (dichloroindol) 55 CLK1 (20), CLK3 (500)
138
Imidazolone (Leucettamine) 701,000 CLK1 (1571), GSK3(2138)
139
Promiscuous
kinase inhibitors
Purlavanol A (Purine) 300 CDK2 (30100), PAK4 (100), SRC
(<100 ), CDK2/CyclinA (100)
107,124
A-443654 (Pyridine) <10 DYRK2, DYRK3 (<100 ), ERK8, RSK1,
RSK2, PKBa, PKBb, S6K1, PKA,
ROCK2, PRK2, PKCa,PKD1,MSK1,
SmMLCK, SK3b, CDK2-CyclinA,
PIM1,2,3,MST2,HIPK2 (<100 )
Initially identied as a potent Akt
inhibitor for use in anticancer
research eld
140
TBB, TBI, DMAT, TDI (Tetrahalo-
bicycles)
10010,000 DYRK2 (30035,000), DYRK3 (2,000
5,000), CK2 (100600), PIM1 (100
1,000), PIM2 (2004,000), PIM3 (70
1,000)
117,141143
www.tandfonline.com e970048-7Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
acceptable and might even represent an advantage, further
encouraging the preclinical and clinical analysis of such com-
pounds in cancer therapeutics.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Funding
This study was supported by grants from Ministerio de Econ-
omıa y Competitividad, Fondo de Investigacion Sanitaria, PI12/
00775 and from Ministerio de Economıa y Competitividad, Red
Tematica de Investigacion Cooperativa en Cancer (RD12/0036/
0027) to PSG.
References
1. Becker W, Joost HG. Structural and functional char-
acteristics of Dyrk, a novel subfamily of protein kin-
ases with dual specificity. Prog Nucleic Acid Res Mol
Biol 1999; 62:1-17; PMID:9932450; http://dx.doi.
org/10.1016/S0079-6603(08)60503-6
2. Aranda S, Laguna A, de la LS. DYRK family of pro-
tein kinases: evolutionary relationships, biochemical
properties, and functional roles. FASEB J 2011; 25
(2):449-62; PMID:21048044; http://dx.doi.org/
10.1096/fj.10-165837
3. Kentrup H, Becker W, Heukelbach J, Wilmes A,
Schurmann A, Huppertz C, Kainulainen H, Joost
HG. Dyrk, a dual specificity protein kinase with
unique structural features whose activity is dependent
on tyrosine residues between subdomains VII and
VIII. J Biol Chem 1996; 271(7):3488-95;
PMID:8631952; http://dx.doi.org/10.1074/jbc.271.
7.3488
4. Himpel S, Panzer P, Eirmbter K, Czajkowska H,
Sayed M, Packman LC, Blundell T, Kentrup H,
Grotzinger J, Joost HG, et al. Identification of the
autophosphorylation sites and characterization of their
effects in the protein kinase DYRK1A. Biochem J
2001; 359(Pt 3):497-505; PMID:11672423; http://
dx.doi.org/10.1042/0264-6021:3590497
5. Lochhead PA, Sibbet G, Morrice N, Cleghon V. Acti-
vation-loop autophosphorylation is mediated by a
novel transitional intermediate form of DYRKs. Cell
2005; 121(6):925-36; PMID:15960979; http://dx.
doi.org/10.1016/j.cell.2005.03.034
6. Becker W, Sippl W. Activation, regulation, and inhi-
bition of DYRK1A. FEBS J 2011; 278(2):246-56;
PMID:21126318; http://dx.doi.org/10.1111/j.1742-
4658.2010.07956.x
7. Guimera J, Casas C, Pucharcos C, Solans A, Dome-
nech A, Planas AM, Ashley J, Lovett M, Estivill X,
Pritchard MA, et al. A human homologue of Dro-
sophila minibrain (MNB) is expressed in the neuronal
regions affected in Down syndrome and maps to the
critical region. Hum Mol Genet 1996; 5(9):1305-10;
PMID:8872470; http://dx.doi.org/10.1093/hmg/
5.9.1305
8. Song WJ, Sternberg LR, Kasten-Sportes C, Keuren
ML, Chung SH, Slack AC, Miller DE, Glover TW,
Chiang PW, Lou L, et al. Isolation of human and
murine homologues of the Drosophila minibrain
gene: human homologue maps to 21q22.2 in the
Down syndrome "critical region". Genomics 1996;
38(3):331-9; PMID:8975710; http://dx.doi.org/
10.1006/geno.1996.0636
9. Shindoh N, Kudoh J, Maeda H, Yamaki A, Minosh-
ima S, Shimizu Y, Shimizu N. Cloning of a human
homolog of the Drosophila minibrain/rat Dyrk gene
from "the Down syndrome critical region" of chromo-
some 21. Biochem Biophys Res Commun 1996; 225
(1):92-9; PMID:8769099; http://dx.doi.org/10.1006/
bbrc.1996.1135
10. Hammerle B, Elizalde C, Galceran J, Becker W, Teje-
dor FJ. The MNB/DYRK1A protein kinase: neurobi-
ological functions and Down syndrome implications.
J Neural Transm Suppl 2003;(67):129-37;
PMID:15068245; http://dx.doi.org/10.1007/978-3-
7091-6721-2_11
11. Park J, Song WJ, Chung KC. Function and regulation
of Dyrk1A: towards understanding Down syndrome.
Cell Mol Life Sci 2009; 66(20):3235-40;
PMID:19685005; http://dx.doi.org/10.1007/s00018-
009-0123-2
12. Becker W, Soppa U, Tejedor FJ. DYRK1A: a poten-
tial drug target for multiple Down syndrome neuropa-
thologies. CNS Neurol Disord Drug Targets 2014; 13
(1):26-33; PMID:24152332; http://dx.doi.org/
10.2174/18715273113126660186
13. Okui M, Ide T, Morita K, Funakoshi E, Ito F, Ogita
K, Yoneda Y, Kudoh J, Shimizu N. High-level expres-
sion of the Mnb/Dyrk1A gene in brain and heart dur-
ing rat early development. Genomics 1999; 62
(2):165-71; PMID:10610708; http://dx.doi.org/
10.1006/geno.1999.5998
14. Fotaki V, Dierssen M, Alcantara S, Martinez S, Marti
E, Casas C, Visa J, Soriano E, Estivill X, Arbones ML.
Dyrk1A haploinsufficiency affects viability and causes
developmental delay and abnormal brain morphology
in mice. Mol Cell Biol 2002; 22(18):6636-47;
PMID:12192061; http://dx.doi.org/10.1128/MCB.
22.18.6636-6647.2002
15. Fotaki V, Martinez dL, Estivill X, Arbones M, Dier-
ssen M. Haploinsufficiency of Dyrk1A in mice leads
to specific alterations in the development and regula-
tion of motor activity. Behav Neurosci 2004; 118
(4):815-21; PMID:15301607; http://dx.doi.org/
10.1037/0735-7044.118.4.815
16. Smith DJ, Stevens ME, Sudanagunta SP, Bronson
RT, Makhinson M, Watabe AM, O’Dell TJ, Fung J,
Weier HU, Cheng JF, et al. Functional screening of 2
Mb of human chromosome 21q22.2 in transgenic
mice implicates minibrain in learning defects associ-
ated with Down syndrome. Nat Genet 1997; 16
(1):28-36; PMID:9140392; http://dx.doi.org/
10.1038/ng0597-28
17. Ahn KJ, Jeong HK, Choi HS, Ryoo SR, Kim YJ, Goo
JS, Choi SY, Han JS, Ha I, Song WJ. DYRK1A BAC
transgenic mice show altered synaptic plasticity with
learning and memory defects. Neurobiol Dis 2006;
22(3):463-72; PMID:16455265; http://dx.doi.org/
10.1016/j.nbd.2005.12.006
18. Altafaj X, Dierssen M, Baamonde C, Marti E, Visa J,
Guimera J, Oset M, Gonzalez JR, Florez J, Fillat C,
et al. Neurodevelopmental delay, motor abnormalities
and cognitive deficits in transgenic mice overexpress-
ing Dyrk1A (minibrain), a murine model of Down’s
syndrome. Hum Mol Genet 2001; 10(18):1915-23;
PMID:11555628; http://dx.doi.org/10.1093/hmg/
10.18.1915
19. Tejedor FJ, Hammerle B. MNB/DYRK1A as a multi-
ple regulator of neuronal development. FEBS J 2011;
278(2):223-35; PMID:21156027; http://dx.doi.org/
10.1111/j.1742-4658.2010.07954.x
20. Dierssen M, de Lagran MM. DYRK1A (dual-specific-
ity tyrosine-phosphorylated and -regulated kinase
1A): a gene with dosage effect during development
and neurogenesis. ScientificWorldJournal 2006;
6:1911-22; PMID:17205196; http://dx.doi.org/
10.1100/tsw.2006.319
21. Tejedor F, Zhu XR, Kaltenbach E, Ackermann A,
Baumann A, Canal I, Heisenberg M, Fischbach KF,
Pongs O. minibrain: a new protein kinase family
involved in postembryonic neurogenesis in Drosoph-
ila. Neuron 1995; 14(2):287-301; PMID:7857639;
http://dx.doi.org/10.1016/0896-6273(95)90286-4
22. Degoutin JL, Milton CC, Yu E, Tipping M, Bosveld
F, Yang L, Bellaiche Y, Veraksa A, Harvey KF. Riqui-
qui and minibrain are regulators of the hippo pathway
downstream of Dachsous. Nat Cell Biol 2013; 15
(10):1176-85; PMID:23955303; http://dx.doi.org/
10.1038/ncb2829
23. Moller RS, Kubart S, Hoeltzenbein M, Heye B, Vogel
I, Hansen CP, Menzel C, Ullmann R, Tommerup N,
Ropers HH, et al. Truncation of the Down syndrome
candidate gene DYRK1A in two unrelated patients
with microcephaly. Am J Hum Genet 2008; 82
(5):1165-70; PMID:18405873; http://dx.doi.org/
10.1016/j.ajhg.2008.03.001
24. Hammerle B, Vera-Samper E, Speicher S, Arencibia R,
Martinez S, Tejedor FJ. Mnb/Dyrk1A is transiently
expressed and asymmetrically segregated in neural pro-
genitor cells at the transition to neurogenic divisions.
Dev Biol 2002; 246(2):259-73; PMID:12051815;
http://dx.doi.org/10.1006/dbio.2002.0675
25. Hammerle B, Carnicero A, Elizalde C, Ceron J, Mar-
tinez S, Tejedor FJ. Expression patterns and subcellu-
lar localization of the Down syndrome candidate
protein MNB/DYRK1A suggest a role in late neuro-
nal differentiation. Eur J Neurosci 2003; 17
(11):2277-86; PMID:12814361; http://dx.doi.org/
10.1046/j.1460-9568.2003.02665.x
26. Hammerle B, Elizalde C, Tejedor FJ. The spatio-tem-
poral and subcellular expression of the candidate
Down syndrome gene Mnb/Dyrk1A in the develop-
ing mouse brain suggests distinct sequential roles in
neuronal development. Eur J Neurosci 2008; 27
(5):1061-74; PMID:18364031; http://dx.doi.org/
10.1111/j.1460-9568.2008.06092.x
27. Yabut O, Domogauer J, D’Arcangelo G. Dyrk1A over-
expression inhibits proliferation and induces prema-
ture neuronal differentiation of neural progenitor cells.
J Neurosci 2010; 30(11):4004-14; PMID:20237271;
http://dx.doi.org/10.1523/JNEUROSCI.4711-09.2010
28. Soppa U, Schumacher J, Florencio Ortiz, V, Pasqua-
lon T, Tejedor FJ, Becker W. The Down syndrome-
related protein kinase DYRK1A phosphorylates p27
(Kip1) and Cyclin D1 and induces cell cycle exit and
neuronal differentiation. Cell Cycle 2014; 13
(13):2084-100; PMID:24806449; http://dx.doi.org/
10.4161/cc.29104
29. Hammerle B, Ulin E, Guimera J, Becker W, Guille-
mot F, Tejedor FJ. Transient expression of Mnb/
Dyrk1a couples cell cycle exit and differentiation of
neuronal precursors by inducing p27KIP1 expression
and suppressing NOTCH signaling. Development
2011; 138(12):2543-54; PMID:21610031; http://dx.
doi.org/10.1242/dev.066167
30. Park J, Oh Y, Yoo L, Jung MS, Song WJ, Lee SH, Seo
H, Chung KC. Dyrk1A phosphorylates p53 and inhib-
its proliferation of embryonic neuronal cells. J Biol
Chem 2010; 285(41):31895-906; PMID:20696760;
http://dx.doi.org/10.1074/jbc.M110.147520
31. Harvey K, Tapon N. The Salvador-Warts-Hippo path-
way - an emerging tumour-suppressor network. Nat
Rev Cancer 2007; 7(3):182-91; PMID:17318211;
http://dx.doi.org/10.1038/nrc2070
32. Badouel C, Garg A, McNeill H. Herding Hippos:
regulating growth in flies and man. Curr Opin Cell
Biol 2009; 21(6):837-43; PMID:19846288; http://
dx.doi.org/10.1016/j.ceb.2009.09.010
33. Reddy BV, Irvine KD. The Fat and Warts signaling
pathways: new insights into their regulation, mecha-
nism and conservation. Development 2008; 135
(17):2827-38; PMID:18697904; http://dx.doi.org/
10.1242/dev.020974
e970048-8 Volume 2 Issue 1Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
34. Halder G, Johnson RL. Hippo signaling: growth con-
trol and beyond. Development 2011; 138(1):9-22;
PMID:21138973; http://dx.doi.org/10.1242/dev.
045500
35. Lu M, Zheng L, Han B, Wang L, Wang P, Liu H,
Sun X. REST regulates DYRK1A transcription in a
negative feedback loop. J Biol Chem 2011; 286
(12):10755-63; PMID:21252229; http://dx.doi.org/
10.1074/jbc.M110.174540
36. Chen JY, Lin JR, Tsai FC, Meyer T. Dosage of
Dyrk1a shifts cells within a p21-cyclin D1 signaling
map to control the decision to enter the cell cycle.
Mol Cell 2013; 52(1):87-100; PMID:24119401;
http://dx.doi.org/10.1016/j.molcel.2013.09.009
37. Yang EJ, Ahn YS, Chung KC. Protein kinase Dyrk1
activates cAMP response element-binding protein
during neuronal differentiation in hippocampal pro-
genitor cells. J Biol Chem 2001; 276(43):39819-24;
PMID:11518709; http://dx.doi.org/10.1074/jbc.
M104091200
38. Kelly PA, Rahmani Z. DYRK1A enhances the mito-
gen-activated protein kinase cascade in PC12 cells by
forming a complex with Ras, B-Raf, and MEK1. Mol
Biol Cell 2005; 16(8):3562-73; PMID:15917294;
http://dx.doi.org/10.1091/mbc.E04-12-1085
39. Fernandez-Martinez J, Vela EM, Tora-Ponsioen M,
Ocana OH, Nieto MA, Galceran J. Attenuation of
Notch signalling by the Down-syndrome-associated
kinase DYRK1A. J Cell Sci 2009; 122(Pt 10):1574-
83; PMID:19383720; http://dx.doi.org/10.1242/
jcs.044354
40. Negrini S, Prada I, D’Alessandro R, Meldolesi J.
REST: an oncogene or a tumor suppressor? Trends
Cell Biol 2013; 23(6):289-95; PMID:23414932;
http://dx.doi.org/10.1016/j.tcb.2013.01.006
41. Ballas N, Grunseich C, Lu DD, Speh JC, Mandel G.
REST and its corepressors mediate plasticity of neuro-
nal gene chromatin throughout neurogenesis. Cell
2005; 121(4):645-57; PMID:15907476; http://dx.
doi.org/10.1016/j.cell.2005.03.013
42. Guardavaccaro D, Frescas D, Dorrello NV, Peschiar-
oli A, Multani AS, Cardozo T, Lasorella A, Iavarone
A, Chang S, Hernando E, et al. Control of chromo-
some stability by the beta-TrCP-REST-Mad2 axis.
Nature 2008; 452(7185):365-9; PMID:18354482;
http://dx.doi.org/10.1038/nature06641
43. Bahn S, Mimmack M, Ryan M, Caldwell MA, Jau-
niaux E, Starkey M, Svendsen CN, Emson P. Neuro-
nal target genes of the neuron-restrictive silencer
factor in neurospheres derived from fetuses with
Down’s syndrome: a gene expression study. Lancet
2002; 359(9303):310-5; PMID:11830198; http://dx.
doi.org/10.1016/S0140-6736(02)07497-4
44. Canzonetta C, Mulligan C, Deutsch S, Ruf S,
O’Doherty A, Lyle R, Borel C, Lin-Marq N, Delom
F, Groet J, et al. DYRK1A-dosage imbalance perturbs
NRSF/REST levels, deregulating pluripotency and
embryonic stem cell fate in Down syndrome. Am J
Hum Genet 2008; 83(3):388-400; PMID:18771760;
http://dx.doi.org/10.1016/j.ajhg.2008.08.012
45. Lepagnol-Bestel AM, Zvara A, Maussion G, Quignon
F, Ngimbous B, Ramoz N, Imbeaud S, Loe-Mie Y,
Benihoud K, Agier N et al. DYRK1A interacts with
the REST/NRSF-SWI/SNF chromatin remodelling
complex to deregulate gene clusters involved in the
neuronal phenotypic traits of Down syndrome. Hum
Mol Genet 2009; 18(8):1405-14; PMID:19218269;
http://dx.doi.org/10.1093/hmg/ddp047
46. Ferron SR, Pozo N, Laguna A, Aranda S, Porlan E,
Moreno M, Fillat C, de la Luna S, Sanchez P, Arbones
ML et al. Regulated segregation of kinase Dyrk1A
during asymmetric neural stem cell division is critical
for EGFR-mediated biased signaling. Cell Stem Cell
2010; 7(3):367-79; PMID:20804972; http://dx.doi.
org/10.1016/j.stem.2010.06.021
47. Aranda S, Alvarez M, Turro S, Laguna A, de la LS.
Sprouty2-mediated inhibition of fibroblast growth
factor signaling is modulated by the protein kinase
DYRK1A. Mol Cell Biol 2008; 28(19):5899-911;
PMID:18678649; http://dx.doi.org/10.1128/MCB.
00394-08
48. Cabrita MA, Christofori G. Sprouty proteins, master-
minds of receptor tyrosine kinase signaling. Angiogen-
esis 2008; 11(1):53-62; PMID:18219583; http://dx.
doi.org/10.1007/s10456-008-9089-1
49. Rubin C, Zwang Y, Vaisman N, Ron D, Yarden Y.
Phosphorylation of carboxyl-terminal tyrosines modu-
lates the specificity of Sprouty-2 inhibition of different
signaling pathways. J Biol Chem 2005; 280(10):9735-
44; PMID:15637081; http://dx.doi.org/10.1074/jbc.
M408308200
50. Abekhoukh S, Planque C, Ripoll C, Urbaniak P, Paul
JL, Delabar JM, Janel N. Dyrk1A, a serine/threonine
kinase, is involved in ERK and Akt activation in the
brain of hyperhomocysteinemic mice. Mol Neurobiol
2013; 47(1):105-16; PMID:22923366; http://dx.doi.
org/10.1007/s12035-012-8326-1
51. Guedj F, Pereira PL, Najas S, Barallobre MJ, Chabert
C, Souchet B, Sebrie C, Verney C, Herault Y,
Arbones M et al. DYRK1A: a master regulatory pro-
tein controlling brain growth. Neurobiol Dis 2012;
46(1):190-203 PMID:22293606; http://dx.doi.org/
10.1016/j.nbd.2012.01.007
52. Weinberg RA. The retinoblastoma protein and cell
cycle control. Cell 1995; 81(3):323-30;
PMID:7736585; http://dx.doi.org/10.1016/0092-
8674(95)90385-2
53. Cobrinik D, Whyte P, Peeper DS, Jacks T, Weinberg
RA. Cell cycle-specific association of E2F with the
p130 E1A-binding protein. Genes Dev 1993; 7
(12A):2392-404; PMID:8253385; http://dx.doi.org/
10.1101/gad.7.12a.2392
54. Vairo G, Livingston DM, Ginsberg D. Functional
interaction between E2F-4 and p130: evidence for dis-
tinct mechanisms underlying growth suppression by
different retinoblastoma protein family members.
Genes Dev 1995; 9(7):869-81; PMID:7705662;
http://dx.doi.org/10.1101/gad.9.7.869
55. Smith EJ, Leone G, DeGregori J, Jakoi L, Nevins JR.
The accumulation of an E2F-p130 transcriptional
repressor distinguishes a G0 cell state from a G1 cell
state. Mol Cell Biol 1996; 16(12):6965-76;
PMID:8943352
56. Litovchick L, Sadasivam S, Florens L, Zhu X, Swanson
SK, Velmurugan S, Chen R, Washburn MP, Liu XS,
DeCaprio JA. Evolutionarily conserved multisubunit
RBL2/p130 and E2F4 protein complex represses
human cell cycle-dependent genes in quiescence. Mol
Cell 2007; 26(4):539-51; PMID:17531812; http://dx.
doi.org/10.1016/j.molcel.2007.04.015
57. Sadasivam S, Decaprio JA. The DREAM complex:
master coordinator of cell cycle-dependent gene
expression. Nat Rev Cancer 2013; 13(8):585-95;
PMID:23842645; http://dx.doi.org/10.1038/
nrc3556
58. Litovchick L, Florens LA, Swanson SK, Washburn
MP, Decaprio JA. DYRK1A protein kinase promotes
quiescence and senescence through DREAM complex
assembly. Genes Dev 2011; 25(8):801-13;
PMID:21498570; http://dx.doi.org/10.1101/gad.
2034211
59. Tschop K, Conery AR, Litovchick L, Decaprio JA,
Settleman J, Harlow E, Dyson N. A kinase shRNA
screen links LATS2 and the pRB tumor suppressor.
Genes Dev 2011; 25(8):814-30; PMID:21498571;
http://dx.doi.org/10.1101/gad.2000211
60. Boichuk S, Parry JA, Makielski KR, Litovchick L,
Baron JL, Zewe JP, Wozniak A, Mehalek KR, Korze-
niewski N, Seneviratne DS, et al. The DREAM com-
plex mediates GIST cell quiescence and is a novel
therapeutic target to enhance imatinib-induced apo-
ptosis. Cancer Res 2013; 73(16):5120-9;
PMID:23786773; http://dx.doi.org/10.1158/0008-
5472.CAN-13-0579
61. Wechsler J, Greene M, McDevitt MA, Anastasi J,
Karp JE, Le Beau MM, Crispino JD. Acquired
mutations in GATA1 in the megakaryoblastic leuke-
mia of Down syndrome. Nat Genet 2002; 32(1):148-
52; PMID:12172547; http://dx.doi.org/10.1038/
ng955
62. Hitzler JK, Cheung J, Li Y, Scherer SW, Zipursky A.
GATA1 mutations in transient leukemia and acute
megakaryoblastic leukemia of Down syndrome. Blood
2003; 101(11):4301-4; PMID:12586620; http://dx.
doi.org/10.1182/blood-2003-01-0013
63. Hollanda LM, Lima CS, Cunha AF, Albuquerque
DM, Vassallo J, Ozelo MC, Joazeiro PP, Saad ST,
Costa FF. An inherited mutation leading to produc-
tion of only the short isoform of GATA-1 is associated
with impaired erythropoiesis. Nat Genet 2006; 38
(7):807-12; PMID:16783379; http://dx.doi.org/
10.1038/ng1825
64. Taub JW, Mundschau G, Ge Y, Poulik JM, Qureshi
F, Jensen T, James SJ, Matherly LH, Wechsler J,
Crispino JD. Prenatal origin of GATA1 mutations
may be an initiating step in the development of mega-
karyocytic leukemia in Down syndrome. Blood 2004;
104(5):1588-9; PMID:15317736; http://dx.doi.org/
10.1182/blood-2004-04-1563
65. Malinge S, Bliss-Moreau M, Kirsammer G, Diebold
L, Chlon T, Gurbuxani S, Crispino JD. Increased
dosage of the chromosome 21 ortholog Dyrk1a pro-
motes megakaryoblastic leukemia in a murine model
of Down syndrome. J Clin Invest 2012; 122(3):948-
62; PMID:22354171; http://dx.doi.org/10.1172/
JCI60455
66. Klusmann JH, Godinho FJ, Heitmann K, Maroz A,
Koch ML, Reinhardt D, Orkin SH, Li Z. Develop-
mental stage-specific interplay of GATA1 and IGF sig-
naling in fetal megakaryopoiesis and leukemogenesis.
Genes Dev 2010; 24(15):1659-72; PMID:20679399;
http://dx.doi.org/10.1101/gad.1903410
67. Maroz A, Stachorski L, Emmrich S, Reinhardt K, Xu J,
Shao Z, Kabler S, Dertmann T, Hitzler J, Roberts I,
et al. GATA1s induces hyperproliferation of eosino-
phil precursors in Down syndrome transient leukemia.
Leukemia 2014; 28(6):1259-70; PMID:24336126;
http://dx.doi.org/10.1038/leu.2013.373
68. Maenz B, Hekerman P, Vela EM, Galceran J, Becker
W. Characterization of the human DYRK1A pro-
moter and its regulation by the transcription factor
E2F1. BMC Mol Biol 2008; 9:30; PMID:18366763;
http://dx.doi.org/10.1186/1471-2199-9-30
69. Hasle H, Clemmensen IH, Mikkelsen M. Incidence
of cancer in individuals with Down syndrome. Tidsskr
Nor Laegeforen 2000; 120(24):2878-81; PMID:
11143409
70. Nizetic D, Groet J. Tumorigenesis in Down’s syn-
drome: big lessons from a small chromosome. Nat
Rev Cancer 2012; 12(10):721-32; PMID:22996602;
http://dx.doi.org/10.1038/nrc3355
71. Arron JR, Winslow MM, Polleri A, Chang CP, Wu
H, Gao X, Neilson JR, Chen L, Heit JJ, Kim SK,
et al. NFAT dysregulation by increased dosage of
DSCR1 and DYRK1A on chromosome 21. Nature
2006; 441(7093):595-600; PMID:16554754; http://
dx.doi.org/10.1038/nature04678
72. Baek KH, Zaslavsky A, Lynch RC, Britt C, Okada Y,
Siarey RJ, Lensch MW, Park IH, Yoon SS, Minami T,
et al. Down’s syndrome suppression of tumour growth
and the role of the calcineurin inhibitor DSCR1.
Nature 2009; 459(7250):1126-30; PMID:19458618;
http://dx.doi.org/10.1038/nature08062
73. Mancini M, Toker A. NFAT proteins: emerging roles
in cancer progression. Nat Rev Cancer 2009; 9
(11):810-20; PMID:19851316; http://dx.doi.org/
10.1038/nrc2735
74. Shaw JP, Utz PJ, Durand DB, Toole JJ, Emmel EA,
Crabtree GR. Identification of a putative regulator of
early T cell activation genes. Science 1988; 241
(4862):202-5; PMID:3260404; http://dx.doi.org/
10.1126/science.3260404
75. Muller MR, Rao A. NFAT, immunity and cancer: a
transcription factor comes of age. Nat Rev Immunol
www.tandfonline.com e970048-9Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
2010; 10(9):645-56; PMID:20725108; http://dx.doi.
org/10.1038/nri2818
76. Gwack Y, Sharma S, Nardone J, Tanasa B, Iuga A, Sri-
kanth S, Okamura H, Bolton D, Feske S, Hogan PG,
et al. A genome-wide Drosophila RNAi screen identi-
fies DYRK-family kinases as regulators of NFAT.
Nature 2006; 441(7093):646-50; PMID:16511445;
http://dx.doi.org/10.1038/nature04631
77. Beals CR, Sheridan CM, Turck CW, Gardner P,
Crabtree GR. Nuclear export of NF-ATc enhanced by
glycogen synthase kinase-3. Science 1997; 275
(5308):1930-4; PMID:9072970; http://dx.doi.org/
10.1126/science.275.5308.1930
78. Zhu J, Shibasaki F, Price R, Guillemot JC, Yano T,
Dotsch V, Wagner G, Ferrara P, McKeon F. Intramo-
lecular masking of nuclear import signal on NF-AT4
by casein kinase I and MEKK1. Cell 1998; 93
(5):851-61; PMID:9630228; http://dx.doi.org/
10.1016/S0092-8674(00)81445-2
79. Okamura H, Garcia-Rodriguez C, Martinson H, Qin
J, Virshup DM, Rao A. A conserved docking motif
for CK1 binding controls the nuclear localization of
NFAT1. Mol Cell Biol 2004; 24(10):4184-95;
PMID:15121840; http://dx.doi.org/10.1128/MCB.
24.10.4184-4195.2004
80. Birger Y, Izraeli S. DYRK1A in Down syndrome: an
oncogene or tumor suppressor? J Clin Invest 2012;
122(3):807-10; PMID:22354166; http://dx.doi.org/
10.1172/JCI62372
81. Lobry C, Oh P, Mansour MR, Look AT, Aifantis I.
Notch signaling: switching an oncogene to a tumor
suppressor. Blood 2014; 123(16):2451-9;
PMID:24608975; http://dx.doi.org/10.1182/blood-
2013-08-355818
82. Lawinger P, Venugopal R, Guo ZS, Immaneni A,
Sengupta D, Lu W, Rastelli L, Marin Dias Carneiro
A, Levin V, Fuller GN, et al. The neuronal repressor
REST/NRSF is an essential regulator in medulloblas-
toma cells. Nat Med 2000; 6(7):826-31;
PMID:10888935; http://dx.doi.org/10.1038/77565
83. Su X, Gopalakrishnan V, Stearns D, Aldape K, Lang
FF, Fuller G, Snyder E, Eberhart CG, Majumder S.
Abnormal expression of REST/NRSF and Myc in
neural stem/progenitor cells causes cerebellar tumors
by blocking neuronal differentiation. Mol Cell Biol
2006; 26(5):1666-78; PMID:16478988; http://dx.
doi.org/10.1128/MCB.26.5.1666-1678.2006
84. Palm K, Metsis M, Timmusk T. Neuron-specific
splicing of zinc finger transcription factor REST/
NRSF/XBR is frequent in neuroblastomas and con-
served in human, mouse and rat. Brain Res Mol Brain
Res 1999; 72(1):30-9; PMID:10521596; http://dx.
doi.org/10.1016/S0169-328X(99)00196-5
85. Conti L, Crisafulli L, Caldera V, Tortoreto M, Brilli
E, Conforti P, Zunino F, Magrassi L, Schiffer D, Cat-
taneo E. REST controls self-renewal and tumorigenic
competence of human glioblastoma cells. PLoS One
2012; 7(6):e38486; PMID:22701651; http://dx.doi.
org/10.1371/journal.pone.0038486
86. Kamal MM, Sathyan P, Singh SK, Zinn PO, Mari-
setty AL, Liang S, Gumin J, El-Mesallamy HO, Suki
D, Colman H, et al. REST Regulates Oncogenic
Properties of Glioblastoma Stem Cells. Stem Cells
2012; 30(3):405-14; PMID:22228704; http://dx.doi.
org/10.1002/stem.1020
87. Siveen KS, Sikka S, Surana R, Dai X, Zhang J, Kumar
AP, Tan BK, Sethi G, Bishayee A. Targeting the
STAT3 signaling pathway in cancer: role of synthetic
and natural inhibitors. Biochim Biophys Acta 2014;
1845(2):136-54; PMID:24388873; http://dx.doi.org/
10.1016/j.bbcan.2013.12.005
88. Kamran MZ, Patil P, Gude RP. Role of STAT3 in
cancer metastasis and translational advances. Biomed
Res Int 2013; 2013:421821; PMID:24199193;
http://dx.doi.org/10.1155/2013/421821
89. Wen Z, Zhong Z, Darnell JE, Jr. Maximal activation
of transcription by Stat1 and Stat3 requires both tyro-
sine and serine phosphorylation. Cell 1995; 82
(2):241-50; PMID:7543024; http://dx.doi.org/
10.1016/0092-8674(95)90311-9
90. Matsuo R, Ochiai W, Nakashima K, Taga T. A new
expression cloning strategy for isolation of substrate-
specific kinases by using phosphorylation site-specific
antibody. J Immunol Methods 2001; 247(1-2):141-
51; PMID:11150545; http://dx.doi.org/10.1016/
S0022-1759(00)00313-6
91. Sakaguchi M, Oka M, Iwasaki T, Fukami Y, Nishigori
C. Role and regulation of STAT3 phosphorylation at
Ser727 in melanocytes and melanoma cells. J Invest
Dermatol 2012; 132(7):1877-85; PMID:22418867;
http://dx.doi.org/10.1038/jid.2012.45
92. Mao J, Maye P, Kogerman P, Tejedor FJ, Toftgard R,
Xie W, Wu G, Wu D. Regulation of Gli1 transcrip-
tional activity in the nucleus by Dyrk1. J Biol Chem
2002; 277(38):35156-61; PMID:12138125; http://
dx.doi.org/10.1074/jbc.M206743200
93. Altaba A. Hedgehog signaling and the Gli code in
stem cells, cancer, and metastases. Sci Signal 2011; 4
(200):t9; PMID:22114144; http://dx.doi.org/
10.1126/scisignal.2002540
94. Seifert A, Allan LA, Clarke PR. DYRK1A phosphory-
lates caspase 9 at an inhibitory site and is potently
inhibited in human cells by harmine. FEBS J 2008;
275(24):6268-80; PMID:19016842; http://dx.doi.
org/10.1111/j.1742-4658.2008.06751.x
95. Laguna A, Aranda S, Barallobre MJ, Barhoum R, Fer-
nandez E, Fotaki V, Delabar JM, de la Luna S, de la
Villa P, Arbones ML. The protein kinase DYRK1A
regulates caspase-9-mediated apoptosis during retina
development. Dev Cell 2008; 15(6):841-53;
PMID:19081073; http://dx.doi.org/10.1016/j.devcel.
2008.10.014
96. Pop C, Timmer J, Sperandio S, Salvesen GS. The
apoptosome activates caspase-9 by dimerization. Mol
Cell 2006; 22(2):269-75; PMID:16630894; http://
dx.doi.org/10.1016/j.molcel.2006.03.009
97. Seifert A, Clarke PR. p38alpha- and DYRK1A-depen-
dent phosphorylation of caspase-9 at an inhibitory site
in response to hyperosmotic stress. Cell Signal 2009;
21(11):1626-33; PMID:19586613; http://dx.doi.org/
10.1016/j.cellsig.2009.06.009
98. Imai SI, Guarente L. NAD and sirtuins in aging and
disease. Trends Cell Biol 2014; 24(8):464-71;
PMID:24786309; http://dx.doi.org/10.1016/j.tcb.
2014.04.002
99. Guo X, Williams JG, Schug TT, Li X. DYRK1A and
DYRK3 promote cell survival through phosphoryla-
tion and activation of SIRT1. J Biol Chem 2010; 285
(17):13223-32; PMID:20167603; http://dx.doi.org/
10.1074/jbc.M110.102574
100. Ionescu A, Dufrasne F, Gelbcke M, Jabin I, Kiss R,
Lamoral-Theys D. DYRK1A kinase inhibitors with
emphasis on cancer. Mini Rev Med Chem 2012; 12
(13):1315-29; PMID:23016545
101. Pozo N, Zahonero C, Fernandez P, Linares JM,
Ayuso A, Hagiwara M, Perez A, Ricoy JR, Hernan-
dez-Laın A, Sepulveda JM, et al. Inhibition of
DYRK1A destabilizes EGFR and reduces EGFR-
dependent glioblastoma growth. J Clin Invest 2013;
123(6):2475-87; PMID:23635774; http://dx.doi.org/
10.1172/JCI63623
102. Zahonero C, Sanchez-Gomez P. EGFR-dependent
mechanisms in glioblastoma: towards a better thera-
peutic strategy. Cell Mol Life Sci 2014;
PMID:24671641; http://dx.doi.org/10.1007/s00018-
014-1608-1
103. Kales SC, Ryan PE, Nau MM, Lipkowitz S. Cbl and
human myeloid neoplasms: the Cbl oncogene comes
of age. Cancer Res 2010; 70(12):4789-94;
PMID:20501843; http://dx.doi.org/10.1158/0008-
5472.CAN-10-0610
104. Barbachano A, Ordonez-Moran P, Garcia JM, San-
chez A, Pereira F, Larriba MJ, Martınez N, Hernandez
J, Landolfi S, Bonilla F, et al. SPROUTY-2 and E-
cadherin regulate reciprocally and dictate colon cancer
cell tumourigenicity. Oncogene 2010; 29(34):4800-
13; PMID:20543868; http://dx.doi.org/10.1038/
onc.2010.225
105. Smith B, Medda F, Gokhale V, Dunckley T, Hulme
C. Recent advances in the design, synthesis, and bio-
logical evaluation of selective DYRK1A inhibitors: a
new avenue for a disease modifying treatment of
Alzheimer’s? ACS Chem Neurosci 2012; 3(11):857-
72; PMID:23173067; http://dx.doi.org/10.1021/
cn300094k
106. Hagiwara M. Alternative splicing: a new drug target of
the post-genome era. Biochim Biophys Acta 2005;
1754(1-2):324-31; PMID:16260193; http://dx.doi.
org/10.1016/j.bbapap.2005.09.010
107. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ,
McLauchlan H, Klevernic I, Arthur JS, Alessi DR,
Cohen P. The selectivity of protein kinase inhibitors:
a further update. Biochem J 2007; 408(3):297-315;
PMID:17850214; http://dx.doi.org/10.1042/
BJ20070797
108. Schenberg EE. Ayahuasca and cancer treatment.
SAGE Open Med 2013; 1; http://dx.doi.org/
10.1177/2050312113508389
109. Hamsa TP, Kuttan G. Harmine inhibits tumour spe-
cific neo-vessel formation by regulating VEGF,
MMP, TIMP and pro-inflammatory mediators both
in vivo and in vitro. Eur J Pharmacol 2010; 649(1-
3):64-73; PMID:20858484; http://dx.doi.org/
10.1016/j.ejphar.2010.09.010
110. Zaker F, Oody A, Arjmand A. A study on the antitu-
moral and differentiation effects of peganum harmala
derivatives in combination with ATRA on leukaemic
cells. Arch Pharm Res 2007; 30(7):844-9; PMID:
17703736; http://dx.doi.org/10.1007/BF02978835
111. Dai F, Chen Y, Song Y, Huang L, Zhai D, Dong Y,
Lai L, Zhang T, Li D, Pang X, et al. A natural small
molecule harmine inhibits angiogenesis and suppresses
tumour growth through activation of p53 in endothe-
lial cells. PLoS One 2012; 7(12):e52162;
PMID:23300602; http://dx.doi.org/10.1371/journal.
pone.0052162
112. Eid SY, El Readi MZ, Eldin EE, Fatani SH, Wink M.
Influence of combinations of digitonin with selected
phenolics, terpenoids, and alkaloids on the expression
and activity of P-glycoprotein in leukaemia and colon
cancer cells. Phytomedicine 2013; 21(1):47-61;
PMID:23999162; http://dx.doi.org/10.1016/j.
phymed.2013.07.019
113. Bei YY, Yuan ZQ, Zhang L, Zhou XF, Chen WL, Xia
P, Liu Y, You BG, Hu XJ, Zhu QL, et al. Novel self-
assembled micelles based on palmitoyl-trimethyl-chi-
tosan for efficient delivery of harmine to liver cancer.
Expert Opin Drug Deliv 2014; 11(6):843-54;
PMID:24655139; http://dx.doi.org/10.1517/
17425247.2014.893292
114. Zhang H, Sun K, Ding J, Xu H, Zhu L, Zhang K, Li
X, Sun W, et al. Harmine induces apoptosis and
inhibits tumor cell proliferation, migration and inva-
sion through down-regulation of cyclooxygenase-2
expression in gastric cancer. Phytomedicine 2014; 21
(3):348-55; PMID:24176842; http://dx.doi.org/
10.1016/j.phymed.2013.09.007
115. Kim H, Sablin SO, Ramsay RR. Inhibition of mono-
amine oxidase A by beta-carboline derivatives. Arch Bio-
chem Biophys 1997; 337(1):137-42; PMID:8990278;
http://dx.doi.org/10.1006/abbi.1996.9771
116. Airaksinen MM, Lecklin A, Saano V, Tuomisto L,
Gynther J. Tremorigenic effect and inhibition of
tryptamine and serotonin receptor binding by beta-
carbolines. Pharmacol Toxicol 1987; 60(1):5-8;
PMID:3562389; http://dx.doi.org/10.1111/j.1600-
0773.1987.tb01711.x
117. Adayev T, Wegiel J, Hwang YW. Harmine is an ATP-
competitive inhibitor for dual-specificity tyrosine
phosphorylation-regulated kinase 1A (Dyrk1A). Arch
Biochem Biophys 2011; 507(2):212-8;
PMID:21185805; http://dx.doi.org/10.1016/j.abb.
2010.12.024
e970048-10 Volume 2 Issue 1Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
118. Ogawa Y, Nonaka Y, Goto T, Ohnishi E, Hiramatsu
T, Kii I, Yoshida M, Ikura T, Onogi H, Shibuya H,
et al. Development of a novel selective inhibitor of
the Down syndrome-related kinase Dyrk1A. Nat
Commun 2010; 1:86; PMID:20981014; http://dx.
doi.org/10.1038/ncomms1090
119. Gallego MA, Ballot C, Kluza J, Hajji N, Martoriati A,
Castera L, Cuevas C, Formstecher P, Joseph B,
Kroemer G, et al. Overcoming chemoresistance of
non-small cell lung carcinoma through restoration of
an AIF-dependent apoptotic pathway. Oncogene
2008; 27(14):1981-92; PMID:17906690; http://dx.
doi.org/10.1038/sj.onc.1210833
120. Baunbaek D, Trinkler N, Ferandin Y, Lozach O,
Ploypradith P, Rucirawat S, Ishibashi F, Iwao M,
Meijer L. Anticancer alkaloid lamellarins inhibit pro-
tein kinases. Mar Drugs 2008; 6(4):514-27;
PMID:19172192; http://dx.doi.org/10.3390/
md20080026
121. Ballot C, Kluza J, Lancel S, Martoriati A, Hassoun
SM, Mortier L, Vienne JC, Briand G, Formstecher P,
Bailly C, et al. Inhibition of mitochondrial respiration
mediates apoptosis induced by the anti-tumoral alka-
loid lamellarin D. Apoptosis 2010; 15(7):769-81;
PMID:20151196; http://dx.doi.org/10.1007/s10495-
010-0471-2
122. Echalier A, Bettayeb K, Ferandin Y, Lozach O, Clem-
ent M, Valette A, Liger F, Marquet B, Morris JC,
Endicott JA et al. Meriolins (3-(pyrimidin-4-yl)-7-
azaindoles): synthesis, kinase inhibitory activity, cellu-
lar effects, and structure of a CDK2/cyclin A/meriolin
complex. J Med Chem 2008; 51(4):737-751;
PMID:18232649; http://dx.doi.org/10.1021/
jm700940h
123. Jarry M, Lecointre C, Malleval C, Desrues L, Schouft
MT, Lejoncour V, Liger F, Lyvinec G, Joseph B,
Loaec N, et al. Impact of meriolins, a new class of
cyclin-dependent kinase inhibitors, on malignant gli-
oma proliferation and neo-angiogenesis. Neuro Oncol
2014; 16(11):1484-98 PMID:24891448; http://dx.
doi.org/10.1093/neuonc/nou102
124. Bain J, McLauchlan H, Elliott M, Cohen P. The spe-
cificities of protein kinase inhibitors: an update. Bio-
chem J 2003; 371(Pt 1):199-204; PMID:12534346;
http://dx.doi.org/10.1042/BJ20021535
125. Mercer SE, Friedman E. Mirk/Dyrk1B: a multifunc-
tional dual-specificity kinase involved in growth arrest,
differentiation, and cell survival. Cell Biochem Bio-
phys 2006; 45(3):303-15; PMID:16845176; http://
dx.doi.org/10.1385/CBB:45:3:303
126. Becker W. Emerging role of DYRK family protein
kinases as regulators of protein stability in cell cycle
control. Cell Cycle 2012; 11(18):3389-94;
PMID:22918246; http://dx.doi.org/10.4161/cc.21404
127. Friedman E. Mirk/Dyrk1B in cancer. J Cell Biochem
2007; 102(2):274-9; PMID:17583556; http://dx.doi.
org/10.1002/jcb.21451
128. Gao J, Zheng Z, Rawal B, Schell MJ, Bepler G, Haura
EB. Mirk/Dyrk1B, a novel therapeutic target, mediates
cell survival in non-small cell lung cancer cells. Cancer
Biol Ther 2009; 8(17):1671-9; PMID:19633423;
http://dx.doi.org/10.4161/cbt.8.17.9322
129. Friedman E. Mirk/dyrk1B Kinase in Ovarian Cancer.
Int J Mol Sci 2013; 14(3):5560-75; PMID:23528858;
http://dx.doi.org/10.3390/ijms14035560
130. Deng X, Ewton DZ, Friedman E. Mirk/Dyrk1B
maintains the viability of quiescent pancreatic cancer
cells by reducing levels of reactive oxygen species.
Cancer Res 2009; 69(8):3317-24; PMID:19351855;
http://dx.doi.org/10.1158/0008-5472.CAN-08-2903
131. Deng X, Mercer SE, Sun CY, Friedman E. The nor-
mal function of the cancer kinase Mirk/dyrk1B is to
reduce reactive oxygen species. Genes Cancer 2014; 5
(1-2):22-30; PMID:24955215
132. Yang CS, Wang X, Lu G, Picinich SC. Cancer pre-
vention by tea: animal studies, molecular mechanisms
and human relevance. Nat Rev Cancer 2009; 9
(6):429-39; PMID:19472429; http://dx.doi.org/
10.1038/nrc2641
133. Chen D, Wang CY, Lambert JD, Ai N, Welsh WJ,
Yang CS. Inhibition of human liver catechol-O-meth-
yltransferase by tea catechins and their metabolites:
structure-activity relationship and molecular-model-
ing studies. Biochem Pharmacol 2005; 69(10):1523-
31; PMID:15857617; http://dx.doi.org/10.1016/j.
bcp.2005.01.024
134. Sanchez C, Salas AP, Brana AF, Palomino M, Pineda-
Lucena A, Carbajo RJ, Mendez C, Moris F, Salas JA.
Generation of potent and selective kinase inhibitors
by combinatorial biosynthesis of glycosylated indolo-
carbazoles. Chem Commun (Camb) 2009;(27):4118-
20; PMID:19568652; http://dx.doi.org/10.1039/
b905068j
135. Neagoie C, Vedrenne E, Buron F, Merour JY, Rosca
S, Bourg S, Lozach O, Meijer L, Baldeyrou B, Lan-
siaux A, et al. Synthesis of chromeno[3,4-b]indoles as
Lamellarin D analogues: a novel DYRK1A inhibitor
class. Eur J Med Chem 2012; 49:379-96;
PMID:22305342; http://dx.doi.org/10.1016/j.
ejmech.2012.01.040
136. Akue-Gedu R, Debiton E, Ferandin Y, Meijer L,
Prudhomme M, Anizon F, Moreau P. Synthesis and
biological activities of aminopyrimidyl-indoles struc-
turally related to meridianins. Bioorg Med Chem
2009; 17(13):4420-4; PMID:19477650; http://dx.
doi.org/10.1016/j.bmc.2009.05.017
137. Giraud F, Alves G, Debiton E, Nauton L, Thery V,
Durieu E, Ferandin Y, Lozach O, Meijer L, Anizon F,
et al. Synthesis, protein kinase inhibitory potencies,
and in vitro antiproliferative activities of meridianin
derivatives. J Med Chem 2011; 54(13):4474-89;
PMID:21623630; http://dx.doi.org/10.1021/
jm200464w
138. Fedorov O, Huber K, Eisenreich A, Filippakopoulos
P, King O, Bullock AN, Szklarczyk D, Jensen LJ, Fab-
bro D, Trappe J, et al. Specific CLK inhibitors from a
novel chemotype for regulation of alternative splicing.
Chem Biol 2011; 18(1):67-76; PMID:21276940;
http://dx.doi.org/10.1016/j.chembiol.2010.11.009
139. Debdab M, Renault S, Lozach O, Meijer L, Paquin L,
Carreaux F, Bazureau JP. Synthesis and preliminary
biological evaluation of new derivatives of the marine
alkaloid leucettamine B as kinase inhibitors. Eur J
Med Chem 2010; 45(2):805-10; PMID:19879673;
http://dx.doi.org/10.1016/j.ejmech.2009.10.009
140. Fala F, Blalock WL, Tazzari PL, Cappellini A, Chiar-
ini F, Martinelli G, Tafuri A, McCubrey JA, Cocco L,
Martelli AM et al. Proapoptotic activity and chemo-
sensitizing effect of the novel Akt inhibitor (2S)-1-
(1H-Indol-3-yl)-3-[5-(3-methyl-2H-indazol-5-yl)pyr-
idin-3-yl]oxypropan2-ami ne (A443654) in T-cell
acute lymphoblastic leukemia. Mol Pharmacol 2008;
74(3):884-95; PMID:18577685; http://dx.doi.org/
10.1124/mol.108.047639
141. Pagano MA, Bain J, Kazimierczuk Z, Sarno S, Ruz-
zene M, Di Maira G, Elliott M, Orzeszko A, Cozza
G, Meggio F, et al. The selectivity of inhibitors of
protein kinase CK2: an update. Biochem J 2008; 415
(3):353-65; PMID:18588507; http://dx.doi.org/
10.1042/BJ20080309
142. Pagano MA, Andrzejewska M, Ruzzene M, Sarno S,
Cesaro L, Bain J, Elliott M, Meggio F, Kazimierczuk
Z, Pinna LA. Optimization of protein kinase CK2
inhibitors derived from 4,5,6,7-tetrabromobenzimida-
zole. J Med Chem 2004; 47(25):6239-47;
PMID:15566294; http://dx.doi.org/10.1021/
jm049854a
143. Sarno S, de Moliner E, Ruzzene M, Pagano MA, Bat-
tistutta R, Bain J, Fabbro D, Schoepfer J, Elliott M,
Furet P et al. Biochemical and three-dimensional-
structural study of the specific inhibition of protein
kinase CK2 by [5-oxo-5,6-dihydroindolo-(1,2-a)qui-
nazolin-7-yl]acetic acid (IQA). Biochem J 2003; 374
(Pt 3):639-46; PMID:12816539; http://dx.doi.org/
10.1042/BJ20030674
www.tandfonline.com e970048-11Molecular & Cellular Oncology
Downloaded by [78.25.98.238] at 13:47 20 August 2015
... DYRK1A (Dual-specificity Tyrosine (Y) Regulated Kinase) is a multifunctional kinase that phosphorylates protein substrates involved in a wide range of cellular process such as cell cycle, transcription, signaling, and apoptosis (reviewed in (Ananthapadmanabhan et al., 2023;Atas-Ozcan et al., 2021;Fernandez-Martinez et al., 2015)). Therefore, it is not surprising that proper dosage of this protein is critical for a variety of developmental processes. ...
... DYRK1A could also regulate cell numbers and growth in the face via its roles in the cell cycle. This protein has actually been shown to suppress cell cycle by modulating several cell cycle regulators such as D cyclins, CDK inhibitors, p53, the LIN52 subunit of DREAM complex as well as the spindle microtubules (Fernandez-Martinez et al., 2015;Iness et al., 2019;Litovchick et al., 2011;Liu et al., 2014) (Ananthapadmanabhan et al., 2023). Therefore, Dyrk1a deficiency might then be expected to increase proliferation in the developing embryo. ...
Preprint
Loss of function mutations in the dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) gene are associated with craniofacial malformations in humans. Here we characterized the effects of deficient DYRK1A in craniofacial development using a developmental model, Xenopus laevis. Dyrk1a mRNA and protein was expressed throughout the developing head and was enriched in the branchial arches which contribute to the face and jaw. Consistently, reduced Dyrk1a function, using dyrk1a morpholinos and pharmacological inhibitors, resulted in orofacial malformations including hypotelorism, altered mouth shape, slanted eyes, and narrower face accompanied by smaller jaw cartilage and muscle. Inhibition of Dyrk1a function resulted in misexpression of key craniofacial regulators including transcription factors and members of the retinoic acid signaling pathway. Two such regulators, sox9 and pax3 are required for neural crest development and their decreased expression corresponds with smaller neural crest domains within the branchial arches. Finally, we determined that the smaller size of the faces, jaw elements and neural crest domains in embryos deficient in Dyrk1a could be explained by increased cell death and decreased proliferation. This study is the first to provide insight into why craniofacial birth defects might arise in humans with DYRK1A mutations.
... Due to its location at DSCR, overexpression of DYRK1A may result in cerebellar malformation, reduction in cortical neurons, and synaptic abnormality [9]. Overactivation of DYRK1A may cause a range of diseases, such as neurodegenerative disorders [10,11], diabetes mellitus [12,13], and cancers [14][15][16]. As DYRK1A inhibition may reverse the aforementioned diseases, DYRK1A is regarded as a potential target. ...
Article
Full-text available
Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is implicated in accumulation of amyloid β-protein (Aβ) and phosphorylation of Tau proteins, and thus represents an important therapeutic target for neurodegenerative diseases. Though many DYRK1A inhibitors have been discovered, there is still no marketed drug targeting DYRK1A. This is partly due to the lack of effective and safe chemotypes. Therefore, it is still necessary to identify new classes of DYRK1A inhibitors. By performing virtual screening with the workflow mainly composed of pharmacophore modeling and molecular docking as well as the following DYRK1A inhibition assay, we identified compound L9, ((Z)-1-(((5-phenyl-1H-pyrazol-4-yl)methylene)-amino)-1H-tetrazol-5-amine), as a moderately active DYRK1A inhibitor (IC50: 1.67 μM). This compound was structurally different from the known DYRK1A inhibitors, showed a unique binding mode to DYRK1A. Furthermore, compound L9 showed neuroprotective activity against okadaic acid (OA)-induced injury in the human neuroblastoma cell line SH-SY5Y by regulating the expression of Aβ and phosphorylation of Tau protein. This compound was neither toxic to the SH-SY5Y cells nor to the human normal liver cell line HL-7702 (IC50: >100 μM). In conclusion, we have identified a novel DYRK1A inhibitor with neuroprotective activity through virtual screening and in vitro biological evaluation, which holds the promise for further study. Graphical abstract
... DYRK1A has also been implicated in the control of the cell cycle and cell proliferation. Depending on the cellular context, DYRK1A can either promote or inhibit cell proliferation, and has both oncogenic and tumor suppressor characteristics (15,16). ...
Preprint
The human dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) is implicated in the pathology of Down syndrome, microcephaly, and cancer, however the exact mechanism through which it functions is unknown. Here, we have studied the role of the Drosophila ortholog of DYRK1A, Minibrain (Mnb), in brain development. The neuroblasts (neural stem cells) that eventually give rise to differentiated neurons in the adult brain are formed from a specialized tissue in the larval optic lobe called the neuroepithelium, in a tightly regulated process. Molecular marker analysis of mnb mutants revealed alterations in the neuroepithelium and neuroblast regions of developing larval brains. Using affinity purification-mass spectrometry (AP-MS), we identified the novel Mnb binding partners Ral interacting protein (Rlip) and RALBP1 associated Eps domain containing (Reps). Rlip and Reps physically and genetically interact with Mnb, and the three proteins may form a ternary complex. Mnb phosphorylates Reps, and human DYRK1A binds to the Reps orthologs REPS1 and REPS2. Furthermore, Mnb engages the small GTPase Ras-like protein A (Rala) to regulate brain and wing development. This work uncovers a previously unrecognized early role of Mnb in the neuroepithelium and defines the functions of the Mnb/Reps/Rlip/Rala signaling network in brain development. Significance statement The kinase Minibrain(Mnb)/DYRK1A regulates the development of the brain and other tissues across many organisms. Here we show the critical importance of Mnb within the developing neuroepithelium. Advancing our understanding of Mnb function, we identified novel protein interactors of Mnb, Reps and Rlip, which function together with Mnb to regulate growth in Drosophila melanogaster . We also identify and characterize a role for the small GTPase Rala in Mnb-regulated growth and nervous system development. This work reveals an early role of Mnb in brain development and identifies a new Mnb/Reps/Rlip/Rala signaling axis.
... Dyrk1a gene dosage affects many stages of neural development. In human, DYRK1A is located on chromosome 21, and it has been associated with a range of conditions including Alzheimer's disease, Down syndrome, microcephaly, autism, and intellectual disability (21)(22)(23)(24)(25)(26). Patients harboring the same point mutation in the DYR-K1A gene display different phenotypes ranging from severe autism to relatively minor neurological impairment (25), strongly suggesting that the genetic background modifies the penetrance of the mutation, similar to many neurodevelopmental disorders. ...
Article
The power and scope of disease modeling can be markedly enhanced through the incorporation of broad genetic diversity. The introduction of pathogenic mutations into a single inbred mouse strain sometimes fails to mimic human disease. We describe a cross-species precision disease modeling platform that exploits mouse genetic diversity to bridge cell-based modeling with whole organism analysis. We developed a universal protocol that permitted robust and reproducible neural differentiation of genetically diverse human and mouse pluripotent stem cell lines and then carried out a proof-of-concept study of the neurodevelopmental gene DYRK1A . Results in vitro reliably predicted the effects of genetic background on Dyrk1a loss-of-function phenotypes in vivo. Transcriptomic comparison of responsive and unresponsive strains identified molecular pathways conferring sensitivity or resilience to Dyrk1a1A loss and highlighted differential messenger RNA isoform usage as an important determinant of response. This cross-species strategy provides a powerful tool in the functional analysis of candidate disease variants identified through human genetic studies.
... Since its discovery, DYRK1A has been considered a molecule associated with the pathogenesis of Down syndrome (23). With further research into the function of DYRK1A, it has been found to play important roles in the cell cycle (24), cell differentiation (25), apoptosis (26,27), splicing regulation (28), and tumorigenesis (29). Many recent studies have identified it as a potential target for the treatment of neurodegenerative diseases and cancer (30,31). ...
Article
Full-text available
Coronaviruses (CoVs) pose a major threat to human and animal health worldwide, which complete viral replication by hijacking host factors. Identifying host factors essential for the viral life cycle can deepen our understanding of the mechanisms of virus–host interactions. Based on our previous genome-wide CRISPR screen of α-CoV transmissible gastroenteritis virus (TGEV), we identified the host factor dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), but not DYRK1B, as a critical factor in TGEV replication. Rescue assays and kinase inhibitor experiments revealed that the effect of DYRK1A on viral replication is independent of its kinase activity. Nuclear localization signal modification experiments showed that nuclear DYRK1A facilitated virus replication. Furthermore, DYRK1A knockout significantly downregulated the expression of the TGEV receptor aminopeptidase N (ANPEP) and inhibited viral entry. Notably, we also demonstrated that DYRK1A is essential for the early stage of TGEV replication. Transmission electron microscopy results indicated that DYRK1A contributes to the formation of double-membrane vesicles in a kinase-independent manner. Finally, we validated that DYRK1A is also a proviral factor for mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. In conclusion, our work demonstrated that DYRK1A is an essential host factor for the replication of multiple viruses, providing new insights into the mechanism of virus–host interactions and facilitating the development of new broad-spectrum antiviral drugs. IMPORTANCE Coronaviruses, like other positive-sense RNA viruses, can remodel the host membrane to form double-membrane vesicles (DMVs) as their replication organelles. Currently, host factors involved in DMV formation are not well defined. In this study, we used transmissible gastroenteritis virus (TGEV) as a virus model to investigate the regulatory mechanism of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) on coronavirus. Results showed that DYRK1A significantly inhibited TGEV replication in a kinase-independent manner. DYRK1A knockout (KO) can regulate the expression of receptor aminopeptidase N (ANPEP) and endocytic-related genes to inhibit virus entry. More importantly, our results revealed that DYRK1A KO notably inhibited the formation of DMV to regulate the virus replication. Further data proved that DYRK1A is also essential in the replication of mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. Taken together, our findings demonstrated that DYRK1A is a conserved factor for positive-sense RNA viruses and provided new insights into its transcriptional regulation activity, revealing its potential as a candidate target for therapeutic design.
Article
Full-text available
Metastatic cancer remains incurable as patients eventually loose sensitivity to targeted therapies and chemotherapies, further leading to poor clinical outcome. Thus, there is a clear medical gap and urgent need to develop efficient and improved targeted therapies for cancer patients. In this study, we investigated the role of DYRK1A kinase in regulating cancer progression and evaluated the therapeutic potential of DYRK1A inhibition in invasive solid tumors, including colon and triple-negative breast cancers. We uncovered new roles played by the DYRK1A kinase. We found that blocking DYRK1A gene expression or pharmacological inhibition of its kinase activity via harmine efficiently blocked primary tumor formation and the metastatic tumor spread in preclinical models of breast and colon cancers. Further assessing the underlying molecular mechanisms, we found that DYRK1A inhibition resulted in increased expression of the G1/S cell cycle regulators while decreasing expression of the G2/M regulators. Combined, these effects release cancer cells from quiescence, leading to their accumulation in G1/S and further delaying/preventing their progression toward G2/M, ultimately leading to growth arrest and tumor growth inhibition. Furthermore, we show that accumulation of cancer cells in G1/S upon DYRK1A inhibition led to significant potentiation of G1/S targeting chemotherapy drug responses in vitro and in vivo. This study underscores the potential for developing novel DYRK1A-targeting therapies in colon and breast cancers and, at the same time, further defines DYRK1A pharmacological inhibition as a viable and powerful combinatorial treatment approach for improving G1/S targeting chemotherapy drugs treatments in solid tumors. Cell cycle is one of the main pathways to be dysregulated in tumorigenesis and cancer progression. As such targeting cell cycle remains a favored approach for developing anti-cancer therapeutics 1. Protein phosphorylation has been implicated in carcinogenesis by regulating many cellular processes such as proliferation, apoptosis, differentiation, and metabolism 2. Because protein kinases are easily druggable, extensive efforts have been spent to explore their potential as targeted therapy in various types of cancer and in fact, many current anti-cancer drugs and treatments rely upon protein kinase inhibition 3. The dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) can auto-phosphorylate their activation loop on tyrosine residue while phosphorylating their specific substrates on threonine and serine residues 4. Of all the 7 DYRK family members, DYRK1A has been the most extensively studied 5. The DYRK1A gene maps to human chromosome 21 within the Down syndrome critical region (DSCR) and its overexpression has been implicated in neuronal development deficits and brain abnormalities in Down syndrome 6. DYRK1A regulates cell cycle and differentiation of neuronal cells by inducing G0/G1 arrest through phosphorylation and subsequent degradation of cyclin D as well as through stabilization of the
Article
Full-text available
Metastatic cancer remains incurable as patients eventually loose sensitivity to targeted therapies and chemotherapies, further leading to poor clinical outcome. Thus, there is a clear medical gap and urgent need to develop efficient and improved targeted therapies for cancer patients. In this study, we investigated the role of DYRK1A kinase in regulating cancer progression and evaluated the therapeutic potential of DYRK1A inhibition in invasive solid tumors, including colon and triple-negative breast cancers. We uncovered new roles played by the DYRK1A kinase. We found that blocking DYRK1A gene expression or pharmacological inhibition of its kinase activity via harmine efficiently blocked primary tumor formation and the metastatic tumor spread in preclinical models of breast and colon cancers. Further assessing the underlying molecular mechanisms, we found that DYRK1A inhibition resulted in increased expression of the G1/S cell cycle regulators while decreasing expression of the G2/M regulators. Combined, these effects release cancer cells from quiescence, leading to their accumulation in G1/S and further delaying/preventing their progression toward G2/M, ultimately leading to growth arrest and tumor growth inhibition. Furthermore, we show that accumulation of cancer cells in G1/S upon DYRK1A inhibition led to significant potentiation of G1/S targeting chemotherapy drug responses in vitro and in vivo. This study underscores the potential for developing novel DYRK1A-targeting therapies in colon and breast cancers and, at the same time, further defines DYRK1A pharmacological inhibition as a viable and powerful combinatorial treatment approach for improving G1/S targeting chemotherapy drugs treatments in solid tumors.
Article
Full-text available
Triple-negative breast cancer (TNBC) is known for its heterogeneous complexity and is often difficult to treat. TNBC lacks the expression of major hormonal receptors like estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 and is further subdivided into androgen receptor (AR) positive and AR negative. In contrast, AR negative is also known as quadruple-negative breast cancer (QNBC). Compared to AR-positive TNBC, QNBC has a great scarcity of prognostic biomarkers and therapeutic targets. QNBC shows excessive cellular growth and proliferation of tumor cells due to increased expression of growth factors like EGF and various surface proteins. This study briefly reviews the limited data available as protein biomarkers that can be used as molecular targets in treating TNBC as well as QNBC. Targeted therapy and immune checkpoint inhibitors have recently changed cancer treatment. Many studies in medicinal chemistry continue to focus on the synthesis of novel compounds to discover new antiproliferative medicines capable of treating TNBC despite the abundance of treatments currently on the market. Drug repurposing is one of the therapeutic methods for TNBC that has been examined. Moreover, some additional micronutrients, nutraceuticals, and functional foods may be able to lower cancer risk or slow the spread of malignant diseases that have already been diagnosed with cancer. Finally, nanomedicines, or applications of nanotechnology in medicine, introduce nanoparticles with variable chemistry and architecture for the treatment of cancer. This review emphasizes the most recent research on nutraceuticals, medication repositioning, and novel therapeutic strategies for the treatment of TNBC.
Article
Full-text available
Objectives: Comprehensively review the evidence regarding the use of ayahuasca, an Amerindian medicine traditionally used to treat many different illnesses and diseases, to treat some types of cancer. Methods: An in-depth review of the literature was conducted using PubMed, books, institutional magazines, conferences and online texts in nonprofessional sources regarding the biomedical knowledge about ayahuasca in general with a specific focus in its possible relations to the treatment of cancer. Results: At least nine case reports regarding the use of ayahuasca in the treatment of prostate, brain, ovarian, uterine, stomach, breast, and colon cancers were found. Several of these were considered improvements, one case was considered worse, and one case was rated as difficult to evaluate. A theoretical model is presented which explains these effects at the cellular, molecular, and psychosocial levels. Particular attention is given to ayahuasca’s pharmacological effects through the activity of N,N-dimethyltryptamine at intracellular sigma-1 receptors. The effects of other components of ayahuasca, such as harmine, tetrahydroharmine, and harmaline, are also considered. Conclusion: The proposed model, based on the molecular and cellular biology of ayahuasca’s known active components and the available clinical reports, suggests that these accounts may have consistent biological underpinnings. Further study of ayahuasca’s possible antitumor effects is important because cancer patients continue to seek out this traditional medicine. Consequently, based on the social and anthropological observations of the use of this brew, suggestions are provided for further research into the safety and efficacy of ayahuasca as a possible medicinal aid in the treatment of cancer.
Article
Full-text available
Mirk kinase is a gene upregulated and sometimes amplified in pancreatic cancers and in ovarian cancers, but expressed at very low levels in most normal diploid cells except for skeletal muscle. The muscle cell function of Mirk kinase selected for by cancer cells is unknown. It is now shown that Mirk protein is expressed at low levels and is largely nuclear in cycling skeletal muscle C2C12 myoblasts, but is translocated to the cytoplasm and upregulated when myoblasts initiate differentiation, as shown by immunofluorescence staining and by cell fractionation. Either Mirk depletion or Mirk kinase inhibition increased ROS levels in cycling C2C12 myoblasts. However, Mirk protein is localized in the cytoplasm of mature muscle fibers, specifically in the fast twitch fibers of human skeletal muscle where toxic ROS levels are generated by muscle contraction. C2C12 myoblasts at high density in differentiation media fuse to form differentiated postmitotic myotubes that can contract. A Mirk kinase inhibitor induced a dose-dependent increase in ROS in this model for fast twitch fibers of human skeletal muscle. Efficient Mirk depletion in SU86.86 pancreatic cancer cells by an inducible shRNA decreased expression of eight antioxidant genes. Thus both cancer cells and differentiated myotubes utilize Mirk kinase to relieve oxidative stress.
Article
Full-text available
Background Glioblastomas are the most frequent and most aggressive primary brain tumors in adults. The median overall survival is limited to a few months despite surgery, radiotherapy, and chemotherapy. It is now clearly established that hyperactivity of cyclin-dependent kinases (CDKs) is one of the processes underlying hyperproliferation and tumoral growth. The marine natural products meridianins and variolins, characterized as CDK inhibitors, display a kinase-inhibitory activity associated with cytotoxic effects. In order to improve selectivity and efficiency of these CDK inhibitors, a series of hybrid compounds called meriolins have been synthesized.
Article
Full-text available
A fundamental question in neurobiology is how the balance between proliferation and differentiation of neuronal precursors is maintained to ensure that the proper number of brain neurons is generated. Substantial evidence implicates DYRK1A (dual specificity tyrosine-phosphorylation-regulated kinase 1A) as a candidate gene responsible for altered neuronal development and brain abnormalities in Down syndrome. Recent findings support the hypothesis that DYRK1A is involved in cell cycle control. Nonetheless, how DYRK1A contributes to neuronal cell cycle regulation and thereby affects neurogenesis remains poorly understood. In the present study we have investigated the mechanisms by which DYRK1A affects cell cycle regulation and neuronal differentiation in a human cell model, mouse neurons, and mouse brain. Dependent on its kinase activity and correlated with the dosage of overexpression, DYRK1A blocked proliferation of SH-SY5Y neuroblastoma cells within 24 h and arrested the cells in G 1 phase. Sustained overexpression of DYRK1A induced G 0 cell cycle exit and neuronal differentiation. Furthermore, we provide evidence that DYRK1A modulated protein stability of cell cycle-regulatory proteins. DYRK1A reduced cellular Cyclin D1 levels by phosphorylation on Thr286, which is known to induce proteasomal degradation. In addition, DYRK1A phosphorylated p27(Kip1) on Ser10, resulting in protein stabilization. Inhibition of DYRK1A kinase activity reduced p27(Kip1) Ser10 phosphorylation in cultured hippocampal neurons and in embryonic mouse brain. In aggregate, these results suggest a novel mechanism by which overexpression of DYRK1A may promote premature neuronal differentiation and contribute to altered brain development in Down syndrome.
Article
Full-text available
Glioblastoma is a particularly resilient cancer, and while therapies may be able to reach the brain by crossing the blood-brain barrier, they then have to deal with a highly invasive tumor that is very resistant to DNA damage. It seems clear that in order to kill aggressive glioma cells more efficiently and with fewer side effects on normal tissue, there must be a shift from classical cytotoxic chemotherapy to more targeted therapies. Since the epidermal growth factor receptor (EGFR) is altered in almost 50 % of glioblastomas, it currently represents one of the most promising therapeutic targets. In fact, it has been associated with several distinct steps in tumorigenesis, from tumor initiation to tumor growth and survival, and also with the regulation of cell migration and angiogenesis. However, inhibitors of the EGFR kinase have produced poor results with this type of cancer in clinical trials, with no clear explanation for the tumor resistance observed. Here we will review what we know about the expression and function of EGFR in cancer and in particular in gliomas. We will also evaluate which are the possible molecular and cellular escape mechanisms. As a result, we hope that this review will help improve the design of future EGFR-targeted therapies for glioblastomas.
Article
The transcription factor NF-AT responds to Ca²⁺-calcineurin signals by translocating to the nucleus, where it participates in the activation of early immune response genes. Calcineurin dephosphorylates conserved serine residues in the amino terminus of NF-AT, resulting in nuclear import. Purification of the NF-AT kinase revealed that it is composed of a priming kinase activity and glycogen synthase kinase-3 (GSK-3). GSK-3 phosphorylates conserved serines necessary for nuclear export, promotes nuclear exit, and thereby opposes Ca²⁺-calcineurin signaling. Because GSK-3 responds to signals initiated by Wnt and other ligands, NF-AT family members could be effectors of these pathways.
Article
Nicotinamide adenine dinucleotide (NAD(+)) is a classical coenzyme mediating many redox reactions. NAD(+) also plays an important role in the regulation of NAD(+)-consuming enzymes, including sirtuins, poly-ADP-ribose polymerases (PARPs), and CD38/157 ectoenzymes. NAD(+) biosynthesis, particularly mediated by nicotinamide phosphoribosyltransferase (NAMPT), and SIRT1 function together to regulate metabolism and circadian rhythm. NAD(+) levels decline during the aging process and may be an Achilles' heel, causing defects in nuclear and mitochondrial functions and resulting in many age-associated pathologies. Restoring NAD(+) by supplementing NAD(+) intermediates can dramatically ameliorate these age-associated functional defects, counteracting many diseases of aging, including neurodegenerative diseases. Thus, the combination of sirtuin activation and NAD(+) intermediate supplementation may be an effective antiaging intervention, providing hope to aging societies worldwide.