Sie sind auf Seite 1von 14

Human Molecular Genetics, 2015, Vol. 24, No.

21 60666079

doi: 10.1093/hmg/ddv318
Advance Access Publication Date: 11 August 2015
Original Article

ORIGINAL ARTICLE

Elucidating the role of the A2A adenosine receptor


in neurodegeneration using neurons derived from
Huntingtons disease iPSCs
Feng-Lan Chiu1,, Jun-Tasi Lin2,, Ching-Yu Chuang1,3,, Ting Chien2,,
Chiung-Mei Chen4, Kai-Hsiang Chen5, Han-Yun Hsiao2, Yow-Sien Lin2,
Yijuang Chern2, * and Hung-Chih Kuo1,3, *
1
Institute of Cellular and Organismic Biology, Academia Sinica, 2Institute of Biomedical Sciences, Academia
Sinica and 3Genomics Research Center, Academia Sinica, Taipei, Taiwan, 4Department of Neurology, Chang Gung
Memorial Hospital, Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
and 5Department of Neurology, National Taiwan University Hospital (Hsinchu Branch), Hsinchu, Taiwan
*To whom correspondence should be addressed at: Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2 Academia Rd., Nankang,
Taipei 115, Taiwan. Tel: +886 227899580; Fax: +886 227899587; Email: kuohuch@gate.sinica.edu.tw or Institute of Biomedical Sciences, Academia Sinica,
No.128, Sec. 2 Academia Rd., Nankang, Taipei 115, Taiwan. Tel: +886E 227893913; Fax: +886 227829143; Email: bmychern@ibms.sinica.edu.tw

Abstract
Huntingtons disease (HD) is an autosomal-dominant degenerative disease caused by a cytosine-adenine-guanine trinucleotide
expansion in the Huntingtin (htt) gene. The most vulnerable brain areas to mutant HTT-evoked toxicity are the striatum and
cortex. In spite of the extensive efforts that have been devoted to the characterization of HD pathogenesis, no disease-modifying
therapy for HD is currently available. The A2A adenosine receptor (A2AR) is widely distributed in the brain, with the highest level
observed in the striatum. We previously reported that stimulation of the A2AR triggers an anti-apoptotic effect in a rat neuron-
like cell line (PC12). Using a transgenic mouse model (R6/2) of HD, we demonstrated that A2AR-selective agonists effectively
ameliorate several major symptoms of HD. In the present study, we show that human iPSCs can be successfully induced to
differentiate into DARPP32-positive, GABAergic neurons which express the A2AR in a similar manner to striatal medium spiny
neurons. When compared with those derived from control subjects (CON-iPSCs), these HD-iPSC-derived neurons exhibited a
higher DNA damage response, based on the observed expression of H2AX and elevated oxidative stress. This is a critical
observation, because oxidative damage and abnormal DNA damage/repair have been reported in HD patients. Most
importantly, stimulation of the A2AR using selective agonists reduced DNA damage and oxidative stress-induced apoptosis in
HD-iPSC-derived neurons through a cAMP/PKA-dependent pathway. These ndings support our hypothesis that human
neurons derived from diseased iPSCs might serve as an important platform to investigate the benecial effects and underlying
mechanisms of A2AR drugs.

Equal contribution.
Received: May 12, 2015. Revised and Accepted: July 31, 2015
The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com

6066
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6067

Introduction cell line-derived neurotrophic factor (2628). In genetic mouse


models of HD, accumulating evidence indicates that stimulation
Huntingtons disease (HD) is an inherited, autosomal-dominant,
of the A2AR may promote neuronal survival. For example, it has
progressive neurodegenerative disease, caused by abnormal ex-
been shown that an A2AR agonist (CGS21680) can (i) signicantly
pansion of cytosine-adenine-guanine (CAG) repeats in exon1 of
delay deterioration of motor coordination (23), (ii) increase prote-
the huntingtin (htt) gene. CAG repeats in excess of 36 result in
asome activity and reduce the accumulation of mutant HTT ag-
the production of mutated Huntingtin (HTT) proteins with an ab-
gregates (23,29) and (iii) attenuate NMDA-induced toxicity in
normally long polyglutamine stretch; these proteins form aggre-
corticostriatal slices of a transgenic mouse model of HD (R6/2)
gates in the cytoplasm and nucleus, which cause degradation of
(30). Accordingly, A2AR-related drugs have been implicated as
neurons, particularly GABAergic medium-sized spiny projection
possible treatments for human HD.
neurons (MSNs) in the striatum. MSNs are involved in the regula-
In this study, we generated and characterized human HD-
tion of movement, emotion and cognitive ability (1,2), and thus
iPSCs with a CAG expansion mutation in the htt gene, and
HD patients gradually suffer from increased irritability and de-
induced their differentiation into GABAergic and DARPP-32-
pression, involuntary movements (chorea), rigidity and eventually
positive neurons. Consistent with previous reports (8,9,31,32),
dementia. Although the mutation which causes HD is known, our
we did not observe mutant HTT aggregation in neurons derived
understanding of HD pathogenesis remains poor. Moreover, there
from HD-iPSCs. Nevertheless, we observed selective vulnerability
is currently no effective strategy to delay the onset or slow down
of HD-iPSC-derived neurons to oxidative stress, which is an
the progression of HD.
important feature of HD pathology (3335). Furthermore, we de-
Various models have been developed to facilitate investiga-
monstrated that activation of the A2AR by selective agonists
tion into disease pathogenesis and to develop therapeutics. How-
(CGS21680 and APEC) reduced oxidative stress-induced DNA
ever, most studies of HD have utilized transgenic mice or
damage and caspase 3 activation in the HD-iPSC-derived GA-
immortalized neurons carrying the mutant htt gene (3), and
BAergic neurons. Our data imply that the human HD-iPSC
these models may not fully recapitulate human HD. Recent
model recapitulates, at least in part, the HD disease phenotype
advances in human-induced pluripotent stem cell (iPSC) tech-
in vitro, and thus may be a potential platform for drug screening.
nology have enabled the generation of patient-specic iPSC
lines for studying disease pathogenesis and for use as a drug
screening platform (4,5). Multiple research groups have isolated Results
several human HD-iPSCs, and induced their differentiation into
Derivation and characterization of HD iPSCs
a disease-relevant cell type [DARPP-32-positive cells, which are
expressed in the majority of MSNs (6)]; however, the differenti- To generate iPSCs, broblasts or peripheral blood mononuclear
ation efciency was low (510%) (79). Increasing such efciency cells obtained from four non-HD controls and two patients diag-
is important for the effective study of disease pathogenesis. nosed with HD were transduced with ve reprograming factors,
Cell-autonomous events contribute to mutant HTT-induced namely OCT4, SOX2, KLF4, c-MYC and NANOG (OSKMN) (Supple-
toxicity in affected neurons in mammalian cell culture systems mentary Material, Table S1). Following 23 weeks under human
and transgenic mouse models of HD. Several studies have sug- embryonic stem cell (hESC) culture conditions, several compact,
gested that multiple mechanisms may underlie the neurotoxic refractive, ESC-like colonies began to emerge. Four to six iPSC
effect of mutant HTT, including excitotoxicity (10,11), oxidative lines were isolated for each subject and characterized by various
stress and mitochondrial abnormalities (10,12), defects in cal- assays. Specically, the integration of exogenous OSKMN trans-
cium homeostasis (13,14), ubiquitin-proteasome system impair- genes into the genomes of the HD-iPSC clones was conrmed
ments (15,16) and transcriptional dysregulation (12,17). However, by PCR analysis at passage six (Fig. 1A). RT-PCR analysis of the
such evidence is insufcient to account for the selective vulner- HD-iPSC lines revealed reactivation and expression of the en-
ability of neurons, because mutant HTT is ubiquitously ex- dogenous pluripotency-associated markers, including OCT4,
pressed in the brain and body. Consequently, these studies SOX2, NANOG, KLF4 and c-MYC, whereas the exogenous OSKMN
imply that additional components are involved in neuronal cell transgenes were found to be predominantly silenced (Fig. 1B).
death in HD. PCR analysis of genomic DNA was also used to show that HD-
Adenosine is a well-characterized regulator of neuronal sur- iPSC-A1 and HD-iPSC-B4 contained mutant htt with CAG expan-
vival, which exerts its function through binding to specic cell sion (both with 43 repeats), as also observed for their parental HD
surface adenosine receptors. To date, four adenosine receptor broblasts (Fig. 1A and C). In contrast, non-HD control iPSCs
subtypes, A1, A2A, A2B and A3, have been identied. The A2A ad- (CON-iPSCs) contained only normal htt alleles with shorter CAG
enosine receptor (A2AR) is a Gs-coupled receptor. Upon stimula- triple repeats. Immunocytochemical (ICC) analysis with anti-
tion, the A2AR activates multiple classical signaling pathways, bodies against ESC antigens was used to demonstrate that
including the production of cAMP that activates PKA (18,19). HD-iPSCs express pluripotency-associated genes and markers
The A2AR exhibits differential distribution within the brain, without losing their pluripotency characteristics after extended
with the highest expression in the encephalin-expressing MSN culture in vitro (Fig. 1D). The in vivo developmental propensity of
of the striatum (20,21). Expression of this receptor is affected by HD-iPSCs was examined using the teratoma formation assay.
mutant HTT, and is gradually down-regulated at the transcrip- Following intramuscular injection of undifferentiated HD-iPSCs
tional level during the progression of HD (2224). Nonetheless, into immunocompromised mice, teratomas consisting of cell types
the ability of the A2AR to enhance the cAMP level in the brain of representing all three embryonic germ layers were formed (Fig. 1E).
HD mice is similar to that of WT mice (23), suggesting that A2AR
signaling in HD is amplied. Thus, the A2AR remains a potential
Differentiation of HD-iPSCs into disease-relevant
drug target of HD (25). This possibility is of great interest because
neuronal subtypes
it has been shown that tonic activation of the A2AR is required for
the function of several neurotrophic factors, including brain- We proceeded to investigate the neuronal differentiation poten-
derived neurotrophic factor, broblast growth factor and glial tial of the HD-iPSCs. As shown in Figure 2A, HD-iPSCs were
6068 | Human Molecular Genetics, 2015, Vol. 24, No. 21

Figure 1. Characterization of human HD-iPSC cell lines. (A) Genomic PCR was used to show that all HD-iPSC lines in this study possessed integrated viral OCT4, SOX2, KLF4,
c-MYC and NANOG (OSKMN) genes. (B) Semi-quantitative PCR analysis was used to show that the viral OSKMN exogenous genes (exo) were silenced and endogenous loci
(endo) of OSKMN were reactivated in these HD-iPSC lines. (C) Genomic PCR and sequencing conrmed the CAG expansion mutation. The H9 hESC line (hESC), normal
human foreskin broblasts (CON-HF) and HD dermal broblasts from HD patients (HD-A/B-HF) were used as controls. -actin and GAPDH were used as loading
controls. (D) Phase contrast and ICC analysis of pluripotency markers (OCT4, SOX2, Nanog and SSEA4) in HD-iPSC lines (A1, A7, B4 and B16). (E) Hematoxylin and
eosin staining of tissue sections of teratomas derived from two HD-iPSC lines (A1 and B4) grafted into immunodecient mice. Scale bar: 50 m.

subjected to embryoid body (EB) formation followed by step-wise RT-PCR was used to show that HD-iPSC neurons contained
in vitro differentiation. Following replating of EBs onto a cell cul- mutant htt with CAG expansion, while CON-iPSC neurons con-
ture surface in N2 media containing 20 ng/ml bFGF, rosette-like tained only normal htt with shorter CAG triple repeats (Fig. 2D).
structures resembling early neuroepithelium and neural pro- Since GABAergic projection neurons are the primary neuronal
genitors began to emerge from EB outgrowths. ICC staining and subtype affected in HD brains, we proceeded to examine the
RT-PCR analysis were used to demonstrate that these cells ex- proportion of GABAergic neurons in our model. To enhance
press neural progenitor markers, such as Nestin, SOX1 and neuronal differentiation, we cultured the differentiated neural
PAX6, but not the ESC markers OCT4 or Nanog (Fig. 2B and C). Fur- progenitors in B27 media for an additional 46 weeks after
ther culture of the neural progenitors in N2/B27 media with 4 weeks of culture in B27/N2 media. Next, we interrogated the
10 ng/ml bFGF directed their differentiation into various neural differentiated cells at various developmental stages, using
populations, including TUJ1-expressing or MAP2-expressing GABAergic neuron markers, including - aminobutyric acid
neurons, and Olig2-expressing or GFAP-expressing glia cells (GABA), Glutamate decarboxylase (GAD) 65, Calbindin and dopa-
(Fig. 2B and C). In addition to expression of neural markers, we mine- and cAMP-regulated neuronal phosphoprotein (DARPP-
also detected the mHTT transcript in iPSC-derived neurons. 32). We observed that cells at the early stages of differentiation
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6069

Figure 2. Human HD-iPSCs differentiate into neural lineages. (A) Schematic procedure of GABAergic neuron differentiation. (B) Immunostaining and (C) semi-quantitative
PCR of HD-iPSC neural derivatives at 48 weeks with the indicated antibodies and primers. Nuclei were stained with DAPI. Scale bar: 50 m. (D) RT-PCR was used to detect
htt transcripts with expanded CAG repeats (upper band). GAPDH was used as an internal control.

already expressed GABA, while some even expressed the striatal are GABAergic projection neurons, and that the neural differenti-
neuron markers GAD65 and Calbindin; cells expressing DARPP- ation efciencies are similar between CON-iPSC- and HD-iPSC-
32, a MSN marker, were observed rarely under this condition derived NPCs.
(data not shown). At later stages of differentiation, the majority
of cells were process-bearing neurons that expressed TUJ1 and
HD-iPSC neurons are more susceptible to H2O2-induced
MAP2; in addition, a high proportion of cells at this stage (over
DNA damage than controls
70%) were positive for GABAergic projection neuron markers:
GABA, GAD65, DARPP-32, or Calbindin (Fig. 3AD, Supplementary Increasing evidence indicates that oxidative stress plays an im-
Material, Fig. S1B and Supplementary Material, Table S5). We as- portant role in HD pathogenesis (3335). To examine whether
sessed the homogeneity of the neuronal populations derived HD-iPSC-derived neurons are vulnerable to oxidative stress-
from CON- or HD-iPSCs. First, we examined viable cells by Trypan induced DNA damage, we treated neurons derived from both
blue exclusion assay at weeks 4, 8 and 12 of neural differentiation; HD- and CON-iPSCs with H2O2. ICC analysis and immunoblotting
this revealed that the expansion rates are similar between CON- with an antibody against H2AX, as a DNA damage marker (36),
and HD-iPSC-derived NPCs (Supplementary Material, Fig. S1A). In were used to show that H2O2 induced H2AX activation in a
addition, the neuronal populations derived from CON- and HD- time- and dose-dependent manner in neurons derived from
iPSCs exhibit similar homogeneity, as revealed by ICC, Western both HD- and CON-iPSCs (Fig. 4 and Supplementary Material,
blotting and RT-qPCR analysis (Fig. 3D and E, Supplementary Ma- S3); this suggests that oxidative insult promotes DNA double-
terial, Fig. S1BC and S2). Together, these results indicate that the strand breaks (DSBs) in the iPSC-derived neurons. However, H2O2
majority of neuronal cells derived from HD-iPSCs or CON-iPSCs induced a signicantly greater level of H2AX in HD-iPSC-derived
6070 | Human Molecular Genetics, 2015, Vol. 24, No. 21

Figure 3. Human HD-iPSCs differentiate into GABAergic neurons. ICC staining of GABAergic projection neuron markers in representative HD-iPSC-A1 (A), -B4 (B) and CON-
iPSC-1 (C) neural derivatives at 14 weeks. Nuclei were stained with DAPI. Scale bar: 50 m. (D) The percentage of DARPP-32 and GABA double-positive cells among DAPI-
labeled nuclei of neurons derived from HD-iPSC-A1 and HD-iPSC-B4, and CON-iPSC-1. At least 200 cells were counted for each experiment. (E) Western blotting with two
primary DARPP-32 antibodies ( purchased from Abcam and SCBT) in neurons derived from CON- and HD-iPSCs. Human brain tissue (hBT) and HD- and CON-iPSCs were
used as positive and negative controls, respectively. An antibody against GAPDH was used for the protein loading control.

neurons (Fig. 4CE). We also compared the global gene expression the responses of iPSC-derived neurons during elevation of
proles of HD- and CON-iPSC-derived neurons by microarray ana- reactive oxygen species (ROS). We hypothesized that activation
lysis (Supplementary Material, Table S6), which revealed that of the A2AR may prevent DNA damage and cell death arising
genes encoding proteins involved in the repair of DNA DSBs from elevated ROS in HD-iPSC-derived neurons. To test this hy-
(RAD51) and protection against oxidative stress (GSST1, GSST2) pothesis, we treated HD- and CON-iPSC-derived neurons with
were down-regulated in HD-iPSC-derived neurons. Collectively, either CGS or APEC, followed by H2O2 insult. As demonstrated
these data indicate that HD-iPSC neurons are more susceptible by ICC analysis and immunoblotting, both CGS (Fig. 6A, C, E
to H2O2-induced DNA damage. and Supplementary Material, Fig. S4A) and APEC (Fig. 6B and
D, and Supplementary Material, Fig. S4B) signicantly reduced
the H2O2-induced activation of H2AX in HD-iPSC-derived neu-
Activation of the A2AR protects HD-iPSC neurons
rons in a dosage-dependent manner. We also conrmed that
from H2O2-induced DNA damage and apoptosis
CGS has the same protective effect in CON-iPSC-derived neu-
through a PKA-dependent pathway
rons (Supplementary Material, Fig. S4C and D). To investigate
Activation of the A2AR has been previously demonstrated to have whether the HD-iPSC-derived neurons are susceptible to cell
several benecial effects in genetic mouse models of HD death upon H2O2 exposure, we treated HD- and CON-iPSC-
(23,29,37). To gain a better understanding of how the A2AR exerts derived neurons with H2O2, and performed TUNEL assays and
its neuroprotective effect, we rst examined the expression of the measured active caspase 3 to quantify the percentage of
A2AR in HD-iPSC-derived neurons. ICC analysis, RT-qPCR and cells with apoptotic traits. We observed that H 2O2 treatment
immunoblotting were used to demonstrate that iPSC-derived resulted in a greater enhancement of active caspase 3 in HD-
neurons express the A2AR (Fig. 5), and that HD-iPSC-derived neu- iPSC-derived neurons than in their counterparts derived
rons express a lower level of the A2AR than control iPSC-derived from CON-iPSCs (Fig. 7C, D and F). Consistent with the
neurons (Fig. 5B and C). We subsequently investigated the effect effects on caspase-3 activation, H 2 O 2 treatment resulted in a
of the A2AR on H2O2-induced DNA damage by treating HD-iPSC- signicantly higher proportion of TUNEL-positive HD-iPSC-
derived neurons with two selective A2AR agonists (CGS-21680, derived neurons than TUNEL-positive CON-iPSC-derived
CGS; 2-[(2-aminoethylamino) carbonylethylphenylethylamino]- neurons. Addition of CGS reduced the TUNEL-positive cells to
5-N- ethylcarboxamidoadenosine, APEC), and then determining 10% (Fig. 7G).
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6071

Figure 4. H2O2 stimulation induced greater H2AX activation in HD-iPSCs than in controls. CON-iPSC-1- and HD-iPSC-B16-derived neurons were treated with (A and C)
100 M H2O2 for the indicated times or (B and D) H2O2 at the indicated concentrations for 60 min. (A and B) Immunouorescence analysis of neurons with H2AX and
TUJ1. Nuclei were stained with DAPI. Scale bar: 20 m. Quantitative analysis of H2AX intensity from (C) Figure 4A and Supplementary Material, Figure S3A and (D)
Figure 4B and Supplementary Material, Figure S3B. The total intensity of H2AX was quantied and normalized to cell numbers by counting DAPI-labeled nuclei. At
least 500 cells were quantied for each condition of each experiment. Data are presented as the mean SEM of three independent experiments. *, compared with
CON-iPSC; P < 0.05 (by one-way ANOVA). (E) Western blot analyses of H2AX in the indicated CON-iPSC and HD-iPSC neuron derivatives following treatment with 0, 50,
or 100 M H2O2 for 60 min. GAPDH was used as a protein loading control. Histograms correspond with the lanes at the top of the adjacent panel (from left to right).
Data are representative of three independent experiments. *, P < 0.05 (by Students t test).

To determine the basis for the protective effect of A2AR agon- As it has been previously shown that A 2AR activation pre-
ist on H2O2-induced DNA damage and apoptosis, we used im- vents toxic stimuli-induced apoptosis through PKA activation
munoblotting to examine induction of H2AX and cleaved (38,39), we hypothesized that PKA activation may be involved
capase-3 in CON-iPSC neurons transfected with A2AR-shRNA or in the A2AR-mediated protection of HD-iPSC-derived neurons.
control-shRNA. A2AR protein was readily detected in cells trans- Hence, we pre-treated HD-iPSC-derived neurons with an A2AR
fected with a scrambled control shRNA, while the level of expres- antagonist (SCH58261) and a selective PKA inhibitor (H-89)
sion was signicantly reduced in cells transfected with the A2AR prior to H2O2 and CGS21680 exposure, and subsequently exam-
shRNA (Supplementary Material, Fig. S5A). As shown in Supple- ined the extent of DNA damage (H2AX) and apoptosis (TUNEL
mentary Material, Figure S5B and S5C, treatment with CGS de- and caspase 3 activation). As demonstrated by ICC analysis and
creased the H2O2-mediated induction of H2AX and cleaved immunoblotting, we found that the protective effect of CGS
caspase-3 in control-shRNA transfectants. In contrast, the pro- against both H2O2-induced DNA damage (Fig. 7A, B and E) and
tective effect was blocked in A2AR-shRNA transfectants. These cell death (Fig. 7C, D, F and G) can be abolished by treatment
ndings support the conclusion that A2AR activation plays an im- with SCH58261 or H-89. Together, these data indicate that acti-
portant role in H2O2-induced DNA damage and apoptosis. vation of the A2AR exerts a benecial effect on HD-iPSC-derived
6072 | Human Molecular Genetics, 2015, Vol. 24, No. 21

Figure 5. HD-iPSC-derived neurons express the A2AR. (A) Immunouorescence analysis of CON-iPSC and HD-iPSC neuron derivatives stained with A2AR and DARPP-32
antibodies. Nuclei were stained with DAPI. Scale bar: 20 m. (B) Relative expression values of A2AR mRNA. Columns represent the average fold-change values obtained
from CON- and HD-iPSC-derived neurons at week 10 of neuron differentiation. Black, control neuron combination (CON-1 and CON-2); Gray, HD neuron combination (HD-
A1, A7, B4 and B16). Error bars represent SEM. (C) Western blot analyses of A2AR protein in CON-iPSC and HD-iPSC neuron derivatives. GAPDH was used as a protein loading
control. Histograms correspond with the lanes in the panel on the right (from left to right). Data are representative of three independent experiments. *, P < 0.05 (by
Students t test).

neurons, protecting them from DNA damage and cell death via iPSC counterparts (32). The HD-iPSC consortium and Carter
activation of PKA. et al. (8,40) reported that HD-iPSC differentiated cultures were
highly susceptible to stressors, including BDNF withdrawal,
glutamate pulsing, or the addition of cellular stressors (H2O2
Discussion and 3-MA). Our results demonstrate that oxidative stress is in-
Here, we describe the generation of human HD-iPSCs with a CAG volved in the pathogenesis of HD; HD-iPSC-derived neurons
expansion mutation in the endogenous htt gene, and their were more susceptible to H2O2-induced DNA damage and cell
differentiation into HD-relevant, DARPP-32-positive, GABAergic death than their counterparts derived from normal iPSCs.
neurons. We conrmed that these cells exhibit selective vulner- These results are consistent with previous reports that H2O2
ability to oxidative stress, which is an important feature of HD treatment resulted in a strong DNA damage response in bro-
pathology. Using these HD-iPSC-derived neurons as an in vitro blasts derived from HD patients (41), and increased cell death in
model, we have demonstrated that activation of the A2AR by human HD iPSCs (8,40). It has also been suggested that the asso-
two A2AR-selective agonists (CGS and APEC) can reduce oxidative ciation between HD and increased ROS and oxidative stimuli
stress-induced DNA damage and neuronal apoptosis in HD iPSC- (8,42) underlies mutant htt-dependent cell death (8,43).
derived neurons, through a PKA-dependent pathway. This study Recent evidence indicates that oxidative stress-induced de-
demonstrates the feasibility of using human-iPSC-derived MSN fects in DNA damage repair can lead to neuronal cell death
neurons as a drug screening platform for HD. Importantly, the (33,44,45). Our microarray analysis data (Supplementary Mater-
CAG repeat number in the htt gene of our HD-iPSCs is 43, which ial, Table S6) revealed that expression of RAD51, which is in-
is in the range for most HD patients, and is much lower than volved in recombination and repair of damaged DNA, is
that of previously described HD-iPSCs (8,9,31). Thus, the results reduced in HD-iPSC neuron derivatives as compared with coun-
obtained from our HD-iPSC cells are likely to reect HD pathogen- terparts from CON-iPSCs. We also observed that expression of
esis observed in the majority of HD patients. Collectively, our genes encoding cellular glutathione antioxidant enzymes [gluta-
ndings suggest that the human HD-iPSC model recapitulates thione S-transferase theta 1 and 2 (GSTT1, GSST2) and Glutathi-
HD-relevant pathogenesis, and may thus be suitable for further one peroxidase 8 (GPX-8)] was slightly decreased in HD-iPSC-
development into a platform for screening novel therapeutic derived neurons (Supplementary Material, Table S6). These
strategies. results are consistent with the reported increase in oxidative
Human iPSC-based models for HD have been developed to stress biomarkers and reduction in antioxidant systems in HD
replicate specic molecular and biological phenotypes. However, patients (42). Despite the presence of such defects, we did not ob-
consistent with previous reports (8,9,31,32), we did not observe serve increased mortality under basal neuronal culture condi-
mutant HTT aggregation, an overt HD phenotype, in neurons de- tions in the long term. Nevertheless, these changes may
rived from HD-iPSCs. Recently, human HD iPSC lines were used to increase the sensitivity of HD-iPSC-derived neurons to oxidative
derive neuronal precursors; these precursors exhibited a mild in- stress, thereby resulting in the accumulation of ROS, and conse-
crease in caspase activity upon growth factor deprivation (9,31), quent DNA damage and cell death. Moreover, our results revealed
and lysomal activity was signicantly increased in HD-iPSCs that one of the protective functions of the A2AR in human MSN
and HD-iPSC-derived neurons as compared with their CON- neurons derived from HD-iPSCs is the prevention of DNA damage
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6073

Figure 6. Stimulation of the A2AR decreased H2O2-induced H2AX activation in HD-iPSCs. HD-iPSC-A1 and -B16-derived neurons were treated with (A) CGS or (B) APEC at
the indicated concentrations for 1 h, and then exposed to 100 M H2O2 for 1 h. (A and B) Immunouorescence analysis of neurons stained with H2AX and TUJ1. Nuclei
were stained with DAPI (blue). Scale bar: 20 m. (C and D) Quantitative analysis of the intensity of H2AX from Figure 6A and B, respectively. The total intensity of H2AX
was quantied and normalized to total cell number by counting DAPI-positive nuclei. At least 500 cells were quantied for each condition of each experiment. Data are
presented as the mean SEM of three independent experiments. *, P < 0.05 (by one-way ANOVA). (E) Immunoblotting against H2AX in iPSC-derived neurons, following
treatment with CGS at the indicated concentrations for 1 h and subsequent exposure to 100 M H2O2 for 1 h. GAPDH was used as a protein loading control. Histograms
correspond with the lanes at the top of the adjacent panel (from left to right). Data are representative of three independent experiments. *, P < 0.05 (by Students t test).

evoked by elevated oxidative stress. This new nding provides by hypoxia (46) and serum-deprivation (39,47) in PC12 cells, and
important insights into the function of the A2AR as a potential NGF withdrawal in sympathetic neurons (48). Our current nd-
therapeutic target for HD in human neurons. ings demonstrate that CGS and APEC prevent DNA damage and
Although the gene causing HD is known, the mechanism apoptosis induced by H2O2 in HD-iPSC-derived neurons (Figs 6
underlying the increased vulnerability of a specic neuronal and 7), suggesting that the A2AR exerts a positive effect on the
population in HD remains elusive. Previous studies have sug- survival of human HD-iPSC-derived neurons. Additionally, we
gested a role of the A2AR in cell survival. For example, stimulation conrmed the protective effect of an A2AR agonist on H2O2-
of the A2AR has been shown to protect against cell death induced induced DNA damage and apoptosis by shRNA knockdown of
6074 | Human Molecular Genetics, 2015, Vol. 24, No. 21

Figure 7. The protective effects of A2AR agonists are mediated through activation of PKA. The HD-iPSC-derived neurons were pre-treated with SCH58261 (SCH, 10 M) or
H-89 (H89, 10 M) and then treated with CGS21680 (CGS, 10 M) and 100 M H2O2 for 1 h (DNA damage) or 6 h (cell death), respectively. Immunouorescence analysis of
neurons stained with (A) H2AX and TUJ1 or (C) active caspase-3 and TUJ1. Nuclei were stained with DAPI. Scale bar: 20 m. Quantitative analysis of the intensity of (B)
H2AX and (D) active caspase-3, as normalized to total cell number by counting DAPI-labeled nuclei. At least 500 cells were quantied for each condition of each
experiment. Immunoblotting of neurons with antibodies against (E) H2AX and (F) active caspase 3. GAPDH was used as a protein loading control. Histograms
correspond with the lanes at the top of the adjacent panel (from left to right). Data are representative of three independent experiments. (G) Percentage of TUNEL-
positive cells within total cells. Data are presented as the mean SEM of three independent experiments. At least 200 cells were quantied for each condition of each
experiment. *, P < 0.05 [by one-way ANOVA in (B) and (D); by Students t test in (E), (F) and (G)].

the A2AR. Our results demonstrated that CGS treatment de- Fig. S5B and S5C). These ndings further support the conclusion
creased DNA damage and apoptosis induced by H2O2 insult in that A2AR activation plays an important role in H2O2-induced
control-shRNA transfectants, whereas the protective effect was DNA damage and apoptosis. Our ndings are consistent with pre-
blocked in A2AR-shRNA transfectants (Supplementary Material, vious reports, which showed that A2AR agonists are benecial in a
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6075

transgenic mouse model of HD (R6/2) and mouse striatal progeni- Isolation of broblasts from patients with HD
tor cell lines (Hdh 7Q and Hdh 109Q) (23,37,4951). In line with the and generation of HD-iPSCs
aforementioned observations supportive of a protective role for
Dermal broblasts from two patients (A and B) with HD were ob-
the A2AR in human and rodent models of HD, genetic removal
tained through skin biopsy; written informed consent was ob-
of the A2AR signicantly exacerbates the progression of HD in a
tained in accordance with the protocol approved by the Internal
HD mouse model (N171-82Q) (52). Together, these ndings
Research Board of CHANG GUNG Memorial Hospital. Four human
strongly suggest that the A2AR is a worthwhile drug target for HD.
non-HD control iPSC lines (CON-iPSC-1, 2, 3 and 5) were derived
Stimulation of the A2AR activates at least two major cellular
from adult foreskin broblasts (CFB-50 iPSC) (64), newborn fore-
signaling cascades: adenylyl cyclase/cAMP/PKA and PKC-
skin broblasts (ATCC, CRL-2429), adult dermal broblasts and
mediated pathways (19). Several reports have indicated that com-
adult peripheral blood mononuclear cells (PBMCs), respectively
ponents involved in regulating the PKA pathway are disrupted
(Supplementary Material, Table S1). Derivation of iPSCs was per-
during HD progression (22,53,54). Therefore, the role of PKA in
formed as described previously (64).
the protective function of the A2AR is worthy of further evalu-
ation. A2AR-mediated protection against H2O2-induced DNA
damage and cell death in HD-iPSC-derived neurons appears to Cultivation and neural differentiation of hiPSCs
be mediated by PKA, because such protection was blocked by a Undifferentiated human HD-iPSCs (HD-iPSC-A1, A7, B4 and B16)
PKA inhibitor, H-89 (Fig. 7). Our results are consistent with earlier and human CON-iPSCs (CON-iPSC-1, 2, 3 and 5) were routinely
reports, which demonstrated that A2AR activation protects cells maintained and subcultivated as previously described (64). For
against hypoxia and growth factor withdrawal via PKA signaling neural differentiation, iPSCs were dissociated with 1 mg/ml dis-
(39,46,55). pase (Sigma), and then left to form embryoid bodies (EBs) in
The ability of iPSCs to differentiate into GABAergic MSNs, the ultra-low attachment dishes (Corning) in hESC media [Dulbeccos
most susceptible cell type in HD pathogenesis, will be the crux to modied Eagles medium (DMEM)/F12, 20% knockout serum re-
successfully model HD in culture. Procedures for inducing differ- placement, 1% non-essential amino acids (NEAA), 2 m L-glu-
entiation of hESCs and iPSCs into GABAergic neurons have been tamine, 100 m 2-mercaptoethanol and 5 ng/ml basic broblast
described previously (79,56,57). These protocols often involved growth factor (bFGF)] containing 10 M SB431542 and 100 nM
treatment with various morphogens, such as SHH and WNT LDN193189 for 3 days; EBs were then transferred to N2 media
inhibitor (DKK1), to direct hESC differentiation toward the GA- [DMEM/F12, 1X N2 supplement, 1% NEAA, 1 m sodium pyru-
BAergic neuronal lineage (79,57). By using a step-wise in vitro dif- vate, 2 m L-glutamine and 20 ng/ml bFGF (all purchased from
ferentiation protocol combining EB formation, neural induction Invitrogen)] containing 10 M SB431542 and 100 nM LDN193189
by small molecules, treatment with inhibitors of the TGF path- for another 2 days. Next, EBs were plated on tissue culture dishes
way (SB431542) and the BMP pathway (LDN193189) (58), and (Nunc) in N2 media, and media were replaced every 2 days. After
mechanical isolation/purication of neural progenitors and 34 weeks of differentiation, neural rosette structures (termed
neurons, we induced 6070% of control iPSCs or HD-iPSCs to neural stem cells; NSCs) were manually picked and transferred
differentiate into GABA- and DARPP-32-double positive neu- to Matrigel-coated culture dishes in N2 medium. The iPSC-
rons (Fig. 3D). The high yield of GABA- and DARPP-32-double derived NSCs were expanded as spherical aggregates.
positive neurons obtained may be attributed to effective neur- For striatal differentiation, the iPSC-derived neurospheres
al induction by the BMP and TGF inhibitors and the mechan- were picked and cut into small pieces (12 mm in diameter)
ical isolation/purication of neural progenitors and early using a needle on a weekly basis, and then plated onto new Ma-
neural cells. This is of particular interest because the default trigel-coated dishes in N2/B27 medium [(DMEM/F12: Neurobasal
neuronal differentiation of human expanded neuron precur- medium (1 : 1), 0.5X N2, 0.5X B27, 1% NEAA, 0.5 m sodium pyru-
sors typically produces GABAergic neurons, irrespective of vate, 2 m L-glutamine and 10 ng/ml bFGF)] for 4 weeks; the
the region from which these cells were harvested from the media were subsequently replaced with B27 medium [Neuroba-
embryo (59,60). sal medium, 1X B27, 1% NEAA, 2 m L-glutamine] for another
In summary, we have elucidated the role of the A2AR in HD 46 weeks. At weeks 4, 8 and 12 of neural differentiation, viable
using HD-iPSC-derived neurons as a disease modeling system. cells were dissociated with TrypLE (Invitrogen, Carlsbad, CA,
Our results imply that A2AR activation may protect MSN against USA) and counted using the Trypan blue exclusion assay in a
ROS-induced damage, and suggest that the A2AR may be a hemocytometer.
novel therapeutic target for HD.

Cell stress, DNA damage and apoptosis assays


Materials and Methods Neurons derived from iPSCs were dissociated into single cells
using TrypLE, seeded onto Matrigel-coated coverslips or appro-
Reagents
priate culture dishes and then cultured for an additional 3 days
SB431542, 2-[ p-(2-carboxyethyl) phenylethylamino]-5-N-ethyl- to enhance cell attachment. Cells were pre-incubated with ad-
carboxamidoadenosine [CGS21680, (61)], and 5-amino-7-(2-phe- enosine deaminase (ADA) (1 unit/ml) for 4 h to eliminate en-
nylethyl)-2-(2-furyl)- pyrazolo [4,3-epsilon]-1,2,4-triazolo[1,5-c) dogenous adenosine, and then treated with an A2AR agonist
pyrimidine [SCH58261, (62)] were purchased from Sigma Aldrich (CGS21680 or APEC) of the indicated concentration for 1 h, prior
(St Louis, Mom USA). LDN193189 was purchased from STEM- to the addition of H2O2 (100 M). Cells were then incubated with
GENT (San Diego, CA, USA). H-89 was purchased from Bio- H2O2 for 1 and 6 h for the measurement of H2AX and caspase-
Mol (Plymouth Meeting, PA, USA). Adenosine deaminase 3 activation, respectively. To characterize the underlying mech-
was purchased from Roche (Nutley, NJ, USA). 2-[(2-aminoethy- anism, an A2AR antagonist (SCH58261) or a PKA inhibitor (H-89)
lamino) carbonylethylphenylethylamino]-5-N- ethylcarboxami- was applied after ADA treatment (as described above) for 10
doadenosine [APEC, (63)] was a generous gift from Dr Kenneth or 30 min, respectively, followed by treatment with the indi-
A. Jacobson. cated A 2A R agonist. Cells were xed and permeabilized for
6076 | Human Molecular Genetics, 2015, Vol. 24, No. 21

immunocytochemical analysis and the terminal deoxynucleo- Knockdown of the A2AR


tidyl transferase-mediated dUTP-biotin nick end-labeling
The A2AR knockdown constructs were produced by subcloning
(TUNEL) assay. The TUNEL assay was performed using the
the 21-nucleotide shRNA sequences of the human A2AR gene
DeadEnd Fluorometric TUNEL System (Promega, Wisconsin,
(Adora2a) (Adora2a shRNA-1, 5-AGACCTTCCGCAAGATCATTC-3;
USA), according to the manufacturers instructions. The per-
Adora2a shRNA-2, 5- CAGAACGTCACCAACTACTTT-3) into the
centage of TUNEL-positive cells was calculated from the total
pLKO.1-AS1010 vector. Preparation and transfection of lentiviral
number of cells from three independent experiments. At
vectors were performed as previously described (67). CON-iPSC-
least 200 cells were quantied in each case.
derived neurons were transduced with lentiviral vectors producing
shRNA targeting the A2AR at a multiplicity of three in the presence
of polybrene (5 g/mL). Proteins were extracted from the trans-
Genomic DNA and RNA extraction and PCR analysis
duced cells at 4 days post-transduction.
of gene expression
Genomic DNA and total RNA were extracted with Trizol reagent
(Invitrogen, Carlsbad, CA, USA), according to the manufacturers
Teratoma generation
recommendations. RNA (1 g) was treated with DNase I (Qiagen,
Germantown, MD, USA) and puried using the RNeasy kit (Qia- For teratoma generation, 5 106 undifferentiated iPSCs were sub-
gen, Germantown, MD, USA), before being reverse-transcribed cutaneously injected into NOD-SCID mice. After 1012 weeks,
using Superscript II (Invitrogen). The expression of total, en- mice were sacriced and teratomas were isolated. All animal ex-
dogenous and viral transgenes of OCT4, SOX2, KLF4 and MYC, periments were approved by the Animal Care and Use Committee
as well as neural-related genes, were examined using semi- of Academia Sinica, and performed in accordance with the Insti-
quantitative PCR, with previously described primers (65,66) tutional Animal Care and Use Committee guidelines of Academia
and those in Supplementary Material, Tables S2 and S3. Quan- Sinica.
titative PCR (RT-qPCR) was performed with SYBR Green Master
Mix and an Applied Biosystems 7900HT Fast Real-Time PCR Sys-
tem using gene-specic primers (Supplementary Material, Microarray analysis
Table S3).
Total RNA was extracted from CON-iPSC-1, CON-iPSC-5, HD-iPSC-
A1, HD-iPSC-A7 and HD-iPSC-B16-derived neurons at week 10 of
neural differentiation. All gene expression data were obtained
Immunocytochemistry (ICC), immunohistochemistry
using the Affymetrix microarray platform by the Affymetrix
and microscopy
Gene Expression Service Laboratory at Academia Sinica, Taiwan.
Immunostaining was performed as previously described (29). Chips were scanned with an Affymetrix GeneChip Scanner 7G,
Cells were xed in 4% paraformaldehyde/DPBS for 20 min, per- and data were analyzed with GeneSpring X software (Agilent,
meabilized with 0.05% NP-40 in PBS for 20 min and then blocked Santa Clara, CA, USA). Raw data were normalized independently
in 3% normal goat serum/DPBS for 1 h. Cells were incubated with for each cell line using Robust Multichip Average. The NCBI acces-
primary antibodies overnight at 4C, washed and incubated with sion number for the microarray data reported in this article is
the corresponding secondary antibody for 60 min. Cells were GSE59051. Changes in gene expression with a fold change >1
counterstained with 1 g/ml DAPI/DPBS for 3 min. The primary and an adjusted P < 0.05 were considered to be signicantly
and secondary antibodies are listed in Supplementary Material, different.
Table S4. Images were captured on an Olympus Ix71 microscope
or a laser-scanning confocal microscope LSM700-meta (Carl
Zeiss). The intensity of uorescence was quantied using Statistical analyses
Image J software (National Institutes of Health, USA), and the
relative intensity of uorescence was normalized to the cell num- Unless stated otherwise, all experiments were performed in trip-
ber. At least three randomly selected elds in each coverslip with licate and the results are shown as the mean SEM. One-way
a total of over 500 cells were counted for each case. For histo- analysis of variance (ANOVA) and Students t test were used for
chemical analysis, H&E staining was performed using an Auto- statistical analysis; P < 0.05 was considered signicant.
stainer XL Leica ST5010 (Leica Microsystem).

Supplementary Material
Immunoblotting
Supplementary Material is available at HMG online.
Total proteins were extracted using RIPA lysis buffer fortied
with protease inhibitors (Complete Mini; Roche-Boehringer). For
detection of the A2AR, protein lysate was extracted using RIPA
lysis buffer containing 4 M urea. Protein concentration was deter- Acknowledgements
mined using the BCA Protein Assay Kit (Pierce). Samples (1025 g We thank Dr Kenneth A. Jacobson (National Institute of Diabetes
each) were then subjected to SDS-PAGE, before being electro- and Digestive and Kidney Diseases, National Institutes of Health,
blotted onto a PVDF membrane (Millipore). Membranes were USA) and the Chemical Synthesis Program of the National Insti-
blocked in 1% BSA in PBS prior to sequential probing with primary tute of Mental Health (Rockville, MD, USA) for the generous gift of
antibody and HRP-conjugated secondary antibody in blocking APEC. We thank Lee Stone and Chun-Ying Yu for technical
solution. Target proteins were visualized by ECL (Amersham- support.
Pharmacia) with an LAS-4000 mini device (FUJI Photo Film).
Primary and secondary antibodies used in immunoblotting are Conict of Interest statement. None of the co-authors have any con-
listed in Supplementary Material, Table S4. icts of interest to declare.
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6077

Funding Early mitochondrial calcium defects in Huntingtons disease


are a direct effect of polyglutamines. Nat. Neurosci., 5, 731
This work was supported by grants from the National Science
736.
Council (102-2321-B-001 -067 -MY3 to H.-C.K.; 100-2320-B-001-
15. Seo, H., Sonntag, K.C. and Isacson, O. (2004) Generalized brain
0110MY3 to Y.C.) and Academia Sinica (AS-103-TP-B10 and Sum-
and skin proteasome inhibition in Huntingtons disease. Ann.
mit project grant to H.-C.K.; AS-103-TP-B10 and 103-Academia
Neurol., 56, 319328.
Sinica Investigation Award-06 to Y.C.).
16. Wang, J., Wang, C.E., Orr, A., Tydlacka, S., Li, S.H. and Li, X.J.
(2008) Impaired ubiquitin-proteasome system activity in the
synapses of Huntingtons disease mice. J. Cell. Biol., 180, 1177
References 1189.
1. Kelly, C.M., Dunnett, S.B. and Rosser, A.E. (2009) Medium 17. Brown, T.B., Bogush, A.I. and Ehrlich, M.E. (2008) Neocortical
spiny neurons for transplantation in Huntingtons disease. expression of mutant huntingtin is not required for altera-
Biochem. Soc. Trans., 37, 323328. tions in striatal gene expression or motor dysfunction in a
2. Reiner, A., Albin, R.L., Anderson, K.D., DAmato, C.J., Penney, transgenic mouse. Hum. Mol. Genet., 17, 30953104.
J.B. and Young, A.B. (1988) Differential loss of striatal projec- 18. Chern, Y., King, K., Lai, H.L. and Lai, H.T. (1992) Molecular
tion neurons in Huntington disease. Proc. Natl. Acad. Sci cloning of a novel adenosine receptor gene from rat brain. Bio-
USA., 85, 57335737. chem. Biophys. Res. Commun., 185, 304309.
3. Pouladi, M.A., Morton, A.J. and Hayden, M.R. (2013) Choosing 19. Chern, Y., Lai, H.L., Fong, J.C. and Liang, Y. (1993) Multiple me-
an animal model for the study of Huntingtons disease. Nat. chanisms for desensitization of A2a adenosine receptor-
Re. Neurosci., 14, 708721. mediated cAMP elevation in rat pheochromocytoma PC12
4. Dimos, J.T., Griswold-Prenner, I., Grskovic, M., Irion, S., cells. Mol. Pharmacol., 44, 950958.
Johnson, C. and Vaisberg, E. (2011) Induced pluripotent 20. Rosin, D.L., Robeva, A., Woodard, R.L., Guyenet, P.G. and
stem cells as human disease models. Ann. Rep. Med. Chem., Linden, J. (1998) Immunohistochemical localization of ad-
46, 369383. enosine A2A receptors in the rat central nervous system.
5. Soldner, F. and Jaenisch, R. (2012) Medicine. iPSC disease J. Comp. Neurol., 401, 163186.
modeling. Science, 338, 11551156. 21. Shearman, L.P. and Weaver, D.R. (1997) [125I]4-aminobenzyl-
6. Ivkovic, S. and Ehrlich, M.E. (1999) Expression of the striatal 5-N-methylcarboxamidoadenosine (125I)AB-MECA) labels
DARPP-32/ARPP-21 phenotype in GABAergic neurons re- multiple adenosine receptor subtypes in rat brain. Brain
quires neurotrophins in vivo and in vitro. J. Neurosci., 19, Res., 745, 1020.
54095419. 22. Chiang, M.C., Lee, Y.C., Huang, C.L. and Chern, Y. (2005) cAMP-
7. Aubry, L., Bugi, A., Lefort, N., Rousseau, F., Peschanski, M. and response element-binding protein contributes to sup-
Perrier, A.L. (2008) Striatal progenitors derived from human pression of the A2A adenosine receptor promoter by mutant
ES cells mature into DARPP32 neurons in vitro and in quino- Huntingtin with expanded polyglutamine residues. J. Biol.
linic acid-lesioned rats. Proc. Natl. Acad. Sci. USA, 105, 16707 Chem., 280, 1433114340.
16712. 23. Chou, S.Y., Lee, Y.C., Chen, H.M., Chiang, M.C., Lai, H.L.,
8. HD iPSC Consortium (2012) Induced pluripotent stem cells Chang, H.H., Wu, Y.C., Sun, C.N., Chien, C.L., Lin, Y.S. et al.
from patients with Huntingtons disease show CAG-repeat- (2005) CGS21680 attenuates symptoms of Huntingtons dis-
expansion-associated phenotypes. Cell Stem Cell, 11, 264278. ease in a transgenic mouse model. J. Neurochem., 93, 310320.
9. Zhang, N., An, M.C., Montoro, D. and Ellerby, L.M. (2010) Char- 24. Martinez-Mir, M.I., Probst, A. and Palacios, J.M. (1991) Adeno-
acterization of human Huntingtons disease cell model from sine A2 receptors: selective localization in the human basal
induced pluripotent stem cells. PLoS Curr., 2, Rrn1193. ganglia and alterations with disease. Neuroscience, 42,
10. Kim, S.H., Thomas, C.A., Andre, V.M., Cummings, D.M., 697706.
Cepeda, C., Levine, M.S. and Ehrlich, M.E. (2011) Forebrain 25. Popoli, P., Blum, D., Domenici, M.R., Burnouf, S. and Chern, Y.
striatal-specic expression of mutant huntingtin protein (2008) A critical evaluation of adenosine A2A receptors as po-
in vivo induces cell-autonomous age-dependent alterations tentially druggable targets in Huntingtons disease. Curr.
in sensitivity to excitotoxicity and mitochondrial function. Pharm. Des., 14, 15001511.
ASN Neuro., 3, e0060. 26. Canals, M., Marcellino, D., Fanelli, F., Ciruela, F., de Benedetti,
11. Graham, R.K., Pouladi, M.A., Joshi, P., Lu, G., Deng, Y., Wu, N.P., P., Goldberg, S.R., Neve, K., Fuxe, K., Agnati, L.F., Woods, A.S.
Figueroa, B.E., Metzler, M., Andre, V.M., Slow, E.J. et al. (2009) et al. (2003) Adenosine A2A-dopamine D2 receptor-receptor
Differential susceptibility to excitotoxic stress in YAC128 heteromerization: qualitative and quantitative assessment
mouse models of Huntington disease between initiation by uorescence and bioluminescence energy transfer. J. Biol.
and progression of disease. J. Neurosci., 29, 21932204. Chem., 278, 4674146749.
12. Thomas, E.A., Coppola, G., Tang, B., Kuhn, A., Kim, S., Gesch- 27. Flajolet, M., Wang, Z., Futter, M., Shen, W., Nuangchamnong,
wind, D.H., Brown, T.B., Luthi-Carter, R. and Ehrlich, M.E. N., Bendor, J., Wallach, I., Nairn, A.C., Surmeier, D.J. and
(2011) In vivo cell-autonomous transcriptional abnormalities Greengard, P. (2008) FGF acts as a co-transmitter through ad-
revealed in mice expressing mutant huntingtin in striatal but enosine A(2A) receptor to regulate synaptic plasticity. Nat.
not cortical neurons. Hum. Mol. Genet., 20, 10491060. Neurosci., 11, 14021409.
13. Rosenstock, T.R., Bertoncini, C.R., Teles, A.V., Hirata, H., Fer- 28. Gomes, C.A., Vaz, S.H., Ribeiro, J.A. and Sebastiao, A.M. (2006)
nandes, M.J. and Smaili, S.S. (2010) Glutamate-induced al- Glial cell line-derived neurotrophic factor (GDNF) enhances
terations in Ca2+ signaling are modulated by mitochondrial dopamine release from striatal nerve endings in an adeno-
Ca2+ handling capacity in brain slices of R6/1 transgenic sine A2A receptor-dependent manner. Brain Res., 1113, 129
mice. Eur. J. Neurosci., 32, 6070. 136.
14. Panov, A.V., Gutekunst, C.A., Leavitt, B.R., Hayden, M.R., 29. Lin, J.T., Chang, W.C., Chen, H.M., Lai, H.L., Chen, C.Y., Tao, M.
Burke, J.R., Strittmatter, W.J. and Greenamyre, J.T. (2002) H. and Chern, Y. (2013) Regulation of feedback between
6078 | Human Molecular Genetics, 2015, Vol. 24, No. 21

protein kinase A and the proteasome system worsens Hun- transgenic mouse model of Huntingtons disease. J. Neuro-
tingtons disease. Mol. Cell Biol., 33, 10731084. chem., 79, 12461249.
30. Ferrante, A., Martire, A., Armida, M., Chiodi, V., Pezzola, A., 45. Schilling, G., Cooneld, M.L., Ross, C.A. and Borchelt, D.R.
Potenza, R.L., Domenici, M.R. and Popoli, P. (2010) Inuence (2001) Coenzyme Q10 and remacemide hydrochloride ameli-
of CGS 21680, a selective adenosine A(2A) receptor agonist, orate motor decits in a Huntingtons disease transgenic
on NMDA receptor function and expression in the brain of mouse model. Neurosci. Lett., 315, 149153.
Huntingtons disease mice. Brain Res., 1323, 184191. 46. Kobayashi, S. and Millhorn, D.E. (1999) Stimulation of expres-
31. An, M.C., Zhang, N., Scott, G., Montoro, D., Wittkop, T., Moon- sion for the adenosine A2A receptor gene by hypoxia in PC12
ey, S., Melov, S. and Ellerby, L.M. (2012) Genetic correction of cells. A potential role in cell protection. J. Biol. Chem., 274,
Huntingtons disease phenotypes in induced pluripotent 2035820365.
stem cells. Cell Stem Cell, 11, 253263. 47. Lee, Y.C., Yang, Y.C., Huang, C.L., Kuo, T.Y., Lin, J.H., Yang, D.
32. Camnasio, S., Delli Carri, A., Lombardo, A., Grad, I., Mariotti, M. and Huang, N.K. (2012) When cytokinin, a plant hormone,
C., Castucci, A., Rozell, B., Lo Riso, P., Castiglioni, V., Zuccato, meets the adenosine A2A receptor: a novel neuroprotectant
C. et al. (2012) The rst reported generation of several induced and lead for treating neurodegenerative disorders? PLoS
pluripotent stem cell lines from homozygous and heterozy- One, 7, e38865.
gous Huntingtons disease patients demonstrates mutation 48. Ramirez, S.H., Fan, S., Maguire, C.A., Perry, S., Hardiek, K.,
related enhanced lysosomal activity. Neurobiol. Dis., 46, 4151. Ramkumar, V., Gelbard, H.A., Dewhurst, S. and Maggirwar,
33. Browne, S.E., Bowling, A.C., MacGarvey, U., Baik, M.J., Berger, S.B. (2004) Activation of adenosine A2A receptor protects
S.C., Muqit, M.M., Bird, E.D. and Beal, M.F. (1997) Oxidative sympathetic neurons against nerve growth factor withdraw-
damage and metabolic dysfunction in Huntingtons disease: al. J. Neurosci. Res., 77, 258269.
selective vulnerability of the basal ganglia. Ann. Neurol., 41, 49. Chiang, M.C., Chen, H.M., Lai, H.L., Chen, H.W., Chou, S.Y.,
646653. Chen, C.M., Tsai, F.J. and Chern, Y. (2009) The A2A adenosine
34. Browne, S.E., Ferrante, R.J. and Beal, M.F. (1999) Oxidative receptor rescues the urea cycle deciency of Huntingtons
stress in Huntingtons disease. Brain Pathol., 9, 147163. disease by enhancing the activity of the ubiquitin-prote-
35. Chen, C.M. (2011) Mitochondrial dysfunction, metabolic de- asome system. Hum. Mol. Genet., 18, 29292942.
cits, and increased oxidative stress in Huntingtons disease. 50. Huang, N.K., Lin, J.H., Lin, J.T., Lin, C.I., Liu, E.M., Lin, C.J.,
Chang Gung Med. J., 34, 135152. Chen, W.P., Shen, Y.C., Chen, H.M., Chen, J.B. et al. (2011) A
36. Rogakou, E.P., Pilch, D.R., Orr, A.H., Ivanova, V.S. and Bonner, new drug design targeting the adenosinergic system for Hun-
W.M. (1998) DNA double-stranded breaks induce histone tingtons disease. PLoS One, 6, e20934.
H2AX phosphorylation on serine 139. J. Biol. Chem., 273, 51. Martire, A., Pepponi, R., Domenici, M.R., Ferrante, A., Chiodi,
58585868. V. and Popoli, P. (2013) BDNF prevents NMDA-induced toxicity
37. Ju, T.C., Chen, H.M., Lin, J.T., Chang, C.P., Chang, W.C., Kang, J. in models of Huntingtons disease: the effects are genotype
J., Sun, C.P., Tao, M.H., Tu, P.H., Chang, C. et al. (2011) Nuclear specic and adenosine A(2A) receptor is involved.
translocation of AMPK-alpha1 potentiates striatal neurode- J. Neurochem., 125, 225235.
generation in Huntingtons disease. J. Cell. Biol., 194, 209227. 52. Mievis, S., Blum, D. and Ledent, C. (2011) A2A receptor knock-
38. Bshesh, K., Zhao, B., Spight, D., Biaggioni, I., Feokistov, I., De- out worsens survival and motor behaviour in a transgenic
nenberg, A., Wong, H.R. and Shanley, T.P. (2002) The A2A re- mouse model of Huntingtons disease. Neurobiol. Dis., 41,
ceptor mediates an endogenous regulatory pathway of 570576.
cytokine expression in THP-1 cells. J. Leukoc. Biol., 72, 1027 53. Giampa, C., Laurenti, D., Anzilotti, S., Bernardi, G., Menniti, F.
1036. S. and Fusco, F.R. (2010) Inhibition of the striatal specic
39. Huang, N.K., Lin, Y.W., Huang, C.L., Messing, R.O. and Chern, Y. phosphodiesterase PDE10A ameliorates striatal and cortical
(2001) Activation of protein kinase A and atypical protein kinase pathology in R6/2 mouse model of Huntingtons disease.
C by A(2A) adenosine receptors antagonizes apoptosis due to PLoS One, 5, e13417.
serum deprivation in PC12 cells. J. Biol. Chem., 276, 1383813846. 54. Hebb, A.L., Robertson, H.A. and Denovan-Wright, E.M. (2004)
40. Carter, R.L., Chen, Y., Kunkanjanawan, T., Xu, Y., Moran, S.P., Striatal phosphodiesterase mRNA and protein levels are re-
Putkhao, K., Yang, J., Huang, A.H., Parnpai, R. and Chan, A.W. duced in Huntingtons disease transgenic mice prior to the
(2014) Reversal of cellular phenotypes in neural cells derived onset of motor symptoms. Neuroscience, 123, 967981.
from Huntingtons disease monkey-induced pluripotent 55. Kobayashi, S., Conforti, L., Pun, R.Y. and Millhorn, D.E.
stem cells. Stem Cell Reports, 3, 585593. (1998) Adenosine modulates hypoxia-induced responses
41. Giuliano, P., De Cristofaro, T., Affaitati, A., Pizzulo, G.M., Feli- in rat PC12 cells via the A2A receptor. J. Physiol., 508 (Pt 1),
ciello, A., Criscuolo, C., De Michele, G., Filla, A., Avvedimento, 95107.
E.V. and Varrone, S. (2003) DNA damage induced by polyglu- 56. Jeon, I., Lee, N., Li, J.Y., Park, I.H., Park, K.S., Moon, J., Shim, S.
tamine-expanded proteins. Hum. Mol. Genet., 12, 23012309. H., Choi, C., Chang, D.J., Kwon, J. et al. (2012) Neuronal proper-
42. Tunez, I., Sanchez-Lopez, F., Aguera, E., Fernandez-Bolanos, ties, in vivo effects, and pathology of a Huntingtons disease
R., Sanchez, F.M. and Tasset-Cuevas, I. (2011) Important role patient-derived induced pluripotent stem cells. Stem Cells, 30,
of oxidative stress biomarkers in Huntingtons disease. J. Med. 20542062.
Chem., 54, 56025606. 57. Delli Carri, A., Onorati, M., Lelos, M.J., Castiglioni, V., Faedo, A.,
43. Goswami, A., Dikshit, P., Mishra, A., Mulherkar, S., Nukina, N. Menon, R., Camnasio, S., Vuono, R., Spaiardi, P., Talpo, F. et al.
and Jana, N.R. (2006) Oxidative stress promotes mutant hun- (2013) Developmentally coordinated extrinsic signals drive
tingtin aggregation and mutant huntingtin-dependent cell human pluripotent stem cell differentiation toward authen-
death by mimicking proteasomal malfunction. Biochem. Bio- tic DARPP-32+ medium-sized spiny neurons. Development,
phys. Res. Commun., 342, 184190. 140, 301312.
44. Bogdanov, M.B., Andreassen, O.A., Dedeoglu, A., Ferrante, R.J. 58. Chambers, S.M., Fasano, C.A., Papapetrou, E.P., Tomishima,
and Beal, M.F. (2001) Increased oxidative damage to DNA in a M., Sadelain, M. and Studer, L. (2009) Highly efcient neural
Human Molecular Genetics, 2015, Vol. 24, No. 21 | 6079

conversion of human ES and iPS cells by dual inhibition of receptor binding subunit by photoafnity crosslinking. Proc.
SMAD signaling. Nat. Biotechnol., 27, 275280. Natl. Acad. Sci. USA, 86, 65726576.
59. Carpenter, M.K., Cui, X., Hu, Z.Y., Jackson, J., Sherman, S., Sei- 64. Huang, H.P., Yu, C.Y., Chen, H.F., Chen, P.H., Chuang, C.Y., Lin,
ger, A. and Wahlberg, L.U. (1999) In vitro expansion of a multi- S.J., Huang, S.T., Chan, W.H., Ueng, T.H., Ho, H.N. et al. (2010)
potent population of human neural progenitor cells. Exp. Factors from human embryonic stem cell-derived broblast-
Neurol., 158, 265278. like cells promote topology-dependent hepatic differenti-
60. Jain, M., Armstrong, R.J., Tyers, P., Barker, R.A. and Rosser, A.E. ation in primate embryonic and induced pluripotent stem
(2003) GABAergic immunoreactivity is predominant in neu- cells. J. Biol. Chem., 285, 3351033519.
rons derived from expanded human neural precursor cells 65. Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T.,
in vitro. Exp. Neurol., 182, 113123. Tomoda, K. and Yamanaka, S. (2007) Induction of pluripotent
61. Zhang, G., Franklin, P.H. and Murray, T.F. (1994) Activation of stem cells from adult human broblasts by dened factors.
adenosine A1 receptors underlies anticonvulsant effect of Cell, 131, 861872.
CGS21680. Eur. J. Pharmacol., 255, 239243. 66. Takahashi, K. and Yamanaka, S. (2006) Induction of pluripo-
62. Belardinelli, L., Shryock, J.C., Ruble, J., Monopoli, A., Dionisot- tent stem cells from mouse embryonic and adult broblast
ti, S., Ongini, E., Dennis, D.M. and Baker, S.P. (1996) Binding of cultures by dened factors. Cell, 126, 663676.
the novel nonxanthine A2A adenosine receptor antagonist 67. Lin, I.Y., Chiu, F.L., Yeang, C.H., Chen, H.F., Chuang, C.Y.,
[3H]SCH58261 to coronary artery membranes. Circ. Res., 79, Yang, S.Y., Hou, P.S., Sintupisut, N., Ho, H.N., Kuo, H.C. et al.
11531160. (2014) Suppression of the SOX2 neural effector gene by
63. Barrington, W.W., Jacobson, K.A., Hutchison, A.J., Williams, PRDM1 promotes human germ cell fate in embryonic stem
M. and Stiles, G.L. (1989) Identication of the A2 adenosine cells. Stem Cell Reports, 2, 189204.

Das könnte Ihnen auch gefallen