ArticlePDF Available

A calcineurin- and NFAT-dependent pathway is involved in -synuclein-induced degeneration of midbrain dopaminergic neurons

Authors:

Abstract and Figures

Parkinson's disease (PD), the most common degenerative movement disorder, is caused by a preferential loss of midbrain dopaminergic (mDA) neurons. Both α-synuclein (α-syn) missense and multiplication mutations have been linked to PD. However, the underlying intracellular signalling transduction pathways of α-syn-mediated mDA neurodegeneration remain elusive. Here, we show that transgenic expression of PD-related human α-syn A53T missense mutation promoted calcineurin (CN) activity and the subsequent nuclear translocation of nuclear factor of activated T cells (NFATs) in mDA neurons. α-syn enhanced the phosphatase activity of CN in both cell-free assays and cell lines transfected with either human wild-type or A53T α-syn. Furthermore, overexpression of α-syn A53T mutation significantly increased the CN-dependent nuclear import of NFATc3 in the mDA neurons of transgenic mice. More importantly, a pharmacological inhibition of CN by cyclosporine A (CsA) ameliorated the α-syn-induced loss of mDA neurons. These findings demonstrate an active involvement of CN- and NFAT-mediated signalling pathway in α-syn-mediated degeneration of mDA neurons in PD.
A53T transgenic mice show significant neuron loss accompanied by NFATc3 translocation in midbrain dopaminergic neurons. ( A ) Western blot was used to determine the level of A53T a -syn overexpression in the midbrain homogenate from 1-month-old A53T transgenic mice compared with littermate nTg mice using an antiserum (C20) recognizing both human and mouse a -syn. b -actin and TH were used as the loading control. The bar graph estimates the level of a -syn overexpression (normalized against the TH expression) in the midbrain of 1-month-old A53T mice compared with that of age-matched littermate nTg mice ( n 1⁄4 4 per genotype). ( B ) Western blot detection of NFATc3 in total, cytoplasmic and nuclear homogenates from the midbrains of nTg and A53T mice ( n 1⁄4 4 per group). The expression of b -actin (total and cytoplasmic) or nucleoporin 62 (nuclear) was used as the loading control. The histograms represent the quantification of total, cytoplasmic and nuclear NFATc3 corrected by the loading control. Data were presented as mean + SEM. ∗∗ P , 0.01 compared with the nTg group. ( C ) Representative images of NFATc3 (green) and TH (red) co-staining in the midbrain sections of 1-month-old nTg and A53T mice. Topro3 (blue) staining marked the nucleus. The arrowhead points to the cytoplasmic localization of NFATc3. The asterisks label the nuclear localization of NFATc3. Scale bar: 10 m m. ( D ) The percentage of total cellular NFATc3 staining of TH-positive neurons ( n 1⁄4 4 animals per genotype, and n 1⁄4 16 neurons per animals) and the nuclear/cytoplasmic ratio of NFATc3 staining. All histograms represent A53T mice values as a percentage compared with the nTg group. Data were presented as mean + SEM. ∗∗ P , 0.01.
… 
Content may be subject to copyright.
A calcineurin- and NFAT-dependent pathway is
involved in a-synuclein-induced degeneration
of midbrain dopaminergic neurons
Jing Luo1,2, Lixin Sun2, Xian Lin2,
{
, Guoxiang Liu2, Jia Yu2, Loukia Parisiadou2,
Chengsong Xie2, Jinhui Ding3and Huaibin Cai2,
1
Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology
Beijing Key Laboratory, Beijing 100875, China,
2
Transgenics Section and and
3
Bioinformatics Core, Laboratory of
Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
Received May 29, 2014; Revised and Accepted July 14, 2014
Parkinson’s disease (PD), the most common degenerative movement disorder, is caused by a preferential loss of
midbrain dopaminergic (mDA) neurons. Both a-synuclein (a-syn) missense and multiplication mutations have
been linked to PD. However, the underlying intracellular signalling transduction pathways of a-syn-mediated
mDA neurodegeneration remain elusive. Here, we show that transgenic expression of PD-related human
a-syn A53T missense mutation promoted calcineurin (CN) activity and the subsequent nuclear translocation
of nuclear factor of activated T cells (NFATs) in mDA neurons. a-syn enhanced the phosphatase activity of CN
in both cell-free assays and cell lines transfected with either human wild-type or A53T a-syn. Furthermore, over-
expression of a-syn A53T mutation significantly increased the CN-dependent nuclear import of NFATc3 in the
mDA neurons of transgenic mice. More importantly, a pharmacological inhibition of CN by cyclosporine A
(CsA) ameliorated the a-syn-induced loss of mDA neurons. These findings demonstrate an active involvement
of CN- and NFAT-mediated signalling pathway in a-syn-mediated degeneration of mDA neurons in PD.
INTRODUCTION
Parkinson’s disease (PD) is pathologically characterized by a
preferential loss of midbrain dopaminergic (mDA) neurons in
the substantia nigra pas compacta (SNpc) and the presence
of a-synuclein (a-syn)-containing cytoplasmic inclusions,
termed Lewy bodies and Lewy neurites (1). Both missense mu-
tation and gene multiplication in a-syn cause autosomal domin-
ant forms of familial PD (2). In addition, the a-syn gene locus
is also associated with the more common sporadic PD (3).
Together, these genetic and neuropathological studies clearly
indicate a prominent role of a-syn in the pathogenesis of PD.
A variety of in vitro and in vivo experiments have been con-
ducted to determine the underlying pathogenic mechanisms of
the a-syn-induced degeneration of mDA neurons (48). For
example, a-syn has been shown to interact with and affect the ac-
tivity of the enzymes phospholipase D (9), protein kinase C,
extracellular regulated kinases (10) and protein phosphatase
2A (11). In addition, a-syn binds to Ca
2+
through a novel
C-terminal domain, which affects the functional properties of
a-syn (12). However, only a few of these studies have been
carried out in SNpc DA neurons. The signalling pathways of
a-syn-mediated mDA neuron loss remain to be established.
Calcineurin (CN) is a Ca
2+
/calmodulin-dependent serine/
threonine-specific protein phosphatase enriched in neurons
(13). The nuclear factor of activated T-cell (NFAT) family of
transcription factors, including NFATc1, NFATc2, NFATc3
and NFATc4, are the main downstream targets of CN (14).
In the resting cells, NFAT proteins are hyperphosphorylated
and mainly reside in the cytoplasm. Upon activation, NFAT pro-
teins undergo rapid dephosphorylation by CN and translocate
into the nucleus, where they regulate gene transcription, in
many cases via associations with other transcription factors
(15). While the NFAT family of transcription factors was
Present address: Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
To whom correspondence should be addressed at: Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes
of Health, Building 35, Room 1A116, MSC 3707, 35 Convent Drive, Bethesda, MD 20892-3707, USA. Tel: +1 3014028087; Fax: +1 3014802830;
Email: caih@mail.nih.gov
Published by Oxford University Press 2014. This work is written by (a) US Government employee(s) and is in the public domain
in the US.
Human Molecular Genetics, 2014 1–8
doi:10.1093/hmg/ddu377
HMG Advance Access published July 28, 2014
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
initially characterized in the immune system, recent studies have
highlighted the importance of this family of proteins in neurons,
where they are involved in the regulation of synaptic plasticity,
axonal growth and neuronal survival (16,17). However, the in-
volvement of CN and NFAT in the a-syn-mediated degeneration
of mDA neurons is unclear.
In our present study, we investigated whether the presence of
pathogenic a-syn affected the CN/NFAT signalling pathway in
Human Embryonic Kidney 293 (HEK293) cells transfected with
either wild-type (WT) or PD-related A53T mutant a-syn and in
mDA neurons of a-syn A53T transgenic mice (18). We found
that the overexpression of a-syn activated the CN and NFAT
pathway in cell lines and mDA neurons, whereas the inhibition
of CN/NFAT activity protected mDA neurons against
a-syn-mediated cytotoxicity.
RESULTS
a-syn activates the phosphatase activity of calcineurin
in cell-free assays
The CN phosphatase activity can be determined using the
chromogenic substrate para-nitrophenyl phosphate (p-NPP)
(19). We found that recombinant human a-syn proteins sig-
nificantly enhanced the CN-mediated dephosphorylation of
p-NPP in a cell-free assay (Fig. 1A). We then further examined
the effect of a-syn on the CN activity by using RII peptide as
the specific substrate in additional cell-free assays (20). Com-
pared with the conveniently measurable p-NPP assay, the RII
peptide assay is more sensitive and only a small amount of CN
is needed (21). The sequence of phospho-RII peptide represents
the phosphorylation site of the regulatory subunit of cAMP-
dependent protein kinase, a well characterized and more
physiological phosphopeptide substrate (20). We found that
a-syn significantly enhanced the CN-induced dephosphoryla-
tion of phospho-RII in a dose-dependent manner (Fig. 1B).
Overexpression of WT or A53T a-syn enhances
CN enzymatic activity and induces the translocation
of NFATc1 and NFATc3 in HEK293 cells
CN normally consists of one catalytic subunit of calcineurin A
(CnA) and one regulatory subunit of calcineurin B (CnB).
The phosphatase activity of CN is fully activated upon the
calcium-dependent binding of calmodulin to the CnA CnB
complex in response to the elevation of intracellular calcium
(21). To explore the potential regulatory role of a-syn on CN,
we examined the CN phosphatase activity and the CnA expres-
sion level in HEK293 cells transiently transfected with WT or
PD-linked mutant A53T a-syn. We found that the phosphatase
activity of CN was significantly increased by 28 and 35% in
cells transfected with either WT or A53T a-syn compared
with cells transfected with empty vectors (Fig. 2A). Moreover,
the expression levels of the CnA subunit protein were not signifi-
cantly altered in the a-syn-expressing cells (Fig. 2B). These
results suggest that the overexpression of a-syn enhanced the ac-
tivity of CN without affecting its protein expression levels.
We next examined the activation of NFAT family proteins in
a-syn-expressing cells. Antibodies against four different NFAT
family members, including NFATc1, NFATc2, NFATc3 and
NFATc4, were used to measure the expression of endogenous
NFAT in the HEK293 cells. Consistent with a previous report
(22), HEK293 cells predominantly expressed NFATc1 and
NFATc3. To investigate whether the a-syn-induced activation
of CN is sufficient to trigger the nuclear translocation of
NFAT proteins, we examined the cytoplasmic and nuclear distri-
bution of NFATc1 and NFATc3 in HEK293 cells transfected
with either empty vector or a-syn expression constructs.
Forty-eight hours after transfection, the cells were harvested
for western blot analysis. The overexpression of both WT and
A53T a-syn substantially altered the cytosolic and nuclear distri-
bution of NFATc1 and NFATc3; a significant decrease in the
cytoplasmic fraction and an increase in the nuclear fraction
were observed (Fig. 2C). Accordingly, the nucleus/cytoplasm
ratios of NFATc1 and NFATc3 proteins were significantly
increased in the a-syn-transfected cells (Fig. 2D and E). More-
over, the total NFATc1 and NFATc3 levels did not differ in
the whole cell extracts prepared from a-syn and empty vector-
transfected cells (data not shown). Together, these data demon-
strate that the overexpression of a-syn may lead to the enhanced
CN activity and subsequent nuclear translocation of the NFAT
family of transcription factors in cultured cells.
Stimulation of calcium ionophore ionomycin leads to nuclear
translocation of NFATc3 in cultured mDA neurons
We investigated the expression levels of Nfatc1, Nfatc2, Nfatc3
and Nfatc4 mRNA in the SNpc DA neurons of 12-month-old
mice by sequencing the total RNA prepared from the SNpc
DA neurons isolated by laser capture microdissection. RNA
sequencing was also used to compare the expression of Nfat
family mRNA in the whole brain of 12-month-old mice. The
RNA sequencing analyses showed that Nfatc3 mRNA was pre-
dominantly expressed in the mouse SNpc DA neurons and
whole brain (Fig. 3A). To investigate the activation of CN/
NFAT pathway in mDA neurons, we treated the cultured
neurons with ionomycin and then stained them with antibodies
specific for NFATc1-c4. Ionomycin can induce the release of
calcium from the intracellular storage place (23). The mDA
neurons did not show obvious staining for NFATc1, NFATc2
and NFATc4 (data not shown). In the resting cells, NFATc3
signals were mainly detected in the cytoplasm, whereas ionomy-
cin treatment led to an almost complete nuclear translocation of
Figure 1. a-syn activates the phosphatase activity of purified calcineurin. (A–B)
The effects of recombinant a-syn proteins on the purified CN activity were deter-
mined in vitro using p-NPP (A) or RII peptide (B) as the substrate. The CN activ-
ity assayed in the absence of a-syn represented 100% activity. Data were
presented as mean +SEM (n¼3). ∗∗∗p,0.001 compared with the control
group.
2Human Molecular Genetics, 2014
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
NFATc3 (Fig. 3B). Here, tyrosine hydroxylase (TH), a cytosolic
protein(24), served not onlyas a marker for dopaminergic neurons
but also as an indicator of the cytosol of these neurons (Fig. 3B).
These results suggest that NFATc3 is abundantly expressed in
mDA neurons and can be regulated by Ca
2+
stimulation.
The nuclear translocation of NFATc3 is significantly
increased in the mDA neurons of A53T a-syn
transgenic mice
To further examine whether NFATc3 is activated in the mDA
neurons of A53T a-syn transgenic mice, we examined the
expression levels of NFATc3 in the nuclear and cytoplasmic
fractions of midbrain homogenates from 1-month-old non-
transgenic (nTg) and A53T a-syn transgenic mice by western
blot analysis. The total levels of NFATc3 were not significantly
changed in the midbrain homogenates of A53T a-syn mice com-
pared with controls (Fig. 4A). In contrast, the transgenic mouse
midbrain homogenates showed a marked increase of NFATc3
immunoreactivity in the nuclear fraction and a trend towards
the decrease of NFATc3 in the cytoplasmic fraction compared
with the controls (Fig. 4B, top panel). Moreover, a significant in-
crease in the nuclear/cytoplasmic ratio of NFATc3 was found in
the mDA neurons of A53T a-syn mice (Fig. 4B, bottom panel).
To further confirm the increased nuclear translocation of
NFATc3 in the mDA neurons of A53T a-syn transgenic mice,
we checked the subcellular distribution of NFATc1, NFATc2,
Figure 2. The overexpression of WT or A53T a-syn enhances the calcineurin en-
zymatic activity and induces the translocation of NFATc1 and NFATc3 in
HEK293 cells. (A) The CN activity was measured using a synthetic peptide,
RII, as the substrate and presented in form of millimoles of phosphate
released/mg of protein/min at 308C(n¼4). Data were presented as mean +
SEM. P,0.05, ∗∗P,0.01 compared with the empty vector group. (B) Repre-
sentative western blot analyses of CN expression levels in three groups (n¼4).
Data were presented as mean +SEM. (C) Western blot detection of NFATc1 and
NFATc3 in the cytoplasmic and nuclear fractions from the control vector-,
WT- and A53T a-syn-transfected HEK293 cells (n¼4). Forty-eight hours
after transfection, the cells were harvested for western blot analysis. The
b-actin (cytoplasmic) and HDAC1 (nuclear) expression levels were used as
the loading control. The histograms represent the quantification of cytoplasmic
and nuclear NFATc3 corrected by the loading control. (D,E) Bar graph
depicts the nuclear/cytoplasmic ratios of NFATc1 and NFATc3. All histograms
in D and E represent values as a percentagecompared with the control group. Data
were presented as mean+SEM. P,0.05, ∗∗P,0.01.
Figure 3. Stimulation of calcium ionophore ionomycin leads to the nuclear trans-
location of NFATc3 in cultured midbrain DA neurons. (A) RNA sequencing
reveals the expression of Nfatc1, Nfatc2, Nfatc3 and Nfatc4 in SNpc DA
neurons and the whole brain of 12-month-old control mice. Two independent
SNpc and whole brain RNA samples were analysed. (B) Representative
images show co-staining of endogenous NFATc3 (green) and TH (red) in
primary mDA neurons with (+) or without ( ) ionomycin stimulation. Scale
bar: 20 mm.
Human Molecular Genetics, 2014 3
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
NFATc3 and NFATc4 in the mDA neurons of 1-month-old
A53T a-syn transgenic and littermate nTg mice by immunos-
taining. The mDA neurons of A53T a-syn transgenic mice did
not show obvious staining for NFATc1, NFATc2 and NFATc4
antibodies (data not shown). In contrast, NFATc3 staining was
detected in the mDA neurons of both nTg and A53T a-syn trans-
genic mice (Fig. 4C). While the NFATc3 signals were mainly
detected in the cytosol of nTg neurons, they were predominantly
distributed in the nuclei of mDA neurons in A53T a-syn trans-
genic mice (Fig. 4C). An additional image analysis revealed a
significant increase in the nuclear distribution of NFATc3 in
the mDA neurons of A53T a-syn transgenic mice compared
with the controls (Fig. 4D). Therefore, both western blot and im-
munocytochemistry analyses demonstrate that overexpression
of PD-related A53T a-syn leads to the nuclear translocation of
NFATc3 in mDA neurons.
Pharmacological inhibition of CN activity ameliorated
a-syn-induced mDA neuron loss in primary culture
The immunosuppressant cyclosporine A (CsA) is a specific in-
hibitor of CN (21). We found that pre-treatment with CsA
blocked the WT a-syn-induced dephosphorylation of NFATc3
in transfected HEK293 cells (Fig. 5A). We then investigated
whether CN/NFATc3 activation was involved in the a-syn-
mediated loss of TH-positive mDA neurons (18). We treated
mDA neuronal cultures from neonatal A53T a-syn and litter-
mate control pups with the CN inhibitor CsA or vehicle
(DMSO) after 5 days in vitro and then counted the numbers of
surviving TH-positive neurons 2 days after the treatment. We
observed an 47% loss of TH-positive mDA neurons in the
vehicle-treated A53T a-syn cultures compared with the controls
(Fig. 5B). In contrast, treatment with 1 mMCsA, which did not
affect the survival of control TH-positive neurons, significantly
increased the survival of TH-positive neurons in the A53T a-syn
cultures compared with the vehicle-treated ones (Fig. 5B). Fur-
thermore, we found that CsA treatment blocked the nuclear
translocation of NFATc3 in the TH-positive dopaminergic
neurons of A53T a-syn cultures (Fig. 5C). These observations
provide direct evidence that the CN/NFATc3 pathway is
involved in a-syn-induced mDA neuron loss.
DISCUSSION
In this study, we employed a new line of a-syn A53T conditional
transgenic mice to investigate the CN/NFAT signalling pathway
in mDA neurons (18). We found that a-syn promoted the CN
phosphatase activity, leading to NFATc3 nuclear import in cell
cultures and mDA neurons of transgenic mice expressing
PD-related A53T a-syn. Moreover, the pharmacological inhib-
ition of CN activity ameliorated a-syn-induced loss of mDA
neurons. These findings suggest that the CN/NFATc3 signalling
pathway may contribute to a-syn-mediated mDA neuron loss
in PD.
a-syn overexpression has been used to generate cellular
and animal models of PD. The overexpression of WT or
mutant a-syn induces cell death in dopaminergic cell lines and
primary dopaminergic neuron cultures (8,25). Transgenic mice
expressing WT or mutant a-syn show motor deficits and
Figure 4. A53T transgenic mice show significant neuron loss accompanied by NFATc3 translocation in midbrain dopaminergic neurons. (A) Western blot was used to
determine the level of A53T a-syn overexpression in the midbrain homogenate from 1-month-old A53T transgenic mice compared with littermate nTg mice using an
antiserum (C20) recognizing both human and mouse a-syn. b-actin and TH were used as the loading control. The bar graph estimates the level of a-syn overexpression
(normalized against the TH expression) in the midbrain of 1-month-old A53T mice compared with that of age-matched littermate nTg mice (n¼4 per genotype). (B)
Western blot detection of NFATc3 in total, cytoplasmic and nuclear homogenates from the midbrains of nTg and A53T mice (n¼4 per group). The expression of
b-actin (total and cytoplasmic) or nucleoporin 62 (nuclear) was used as the loading control. The histograms represent the quantification of total, cytoplasmic and
nuclear NFATc3 corrected by the loading control. Data were presented as mean+SEM. ∗∗P,0.01 compared with the nTg group. (C) Representative images of
NFATc3 (green) and TH (red) co-staining in the midbrain sections of 1-month-old nTg and A53T mice. Topro3 (blue) staining marked the nucleus. The arrowhead
points to the cytoplasmic localization of NFATc3. The asterisks label the nuclear localization of NFATc3. Scale bar: 10 mm. (D) The percentage of total cellular
NFATc3 staining of TH-positive neurons (n¼4 animals per genotype, and n¼16 neurons per animals) and the nuclear/cytoplasmic ratio of NFATc3 staining.
All histograms represent A53T mice values as a percentage compared with the nTg group. Data were presented as mean +SEM. ∗∗P,0.01.
4Human Molecular Genetics, 2014
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
changes in dopamine levels (5). Although the excessive aggrega-
tion of a-syn has been associated with neurodegeneration, the
mechanism by which a-syn injures dopaminergic neurons
remains to be fully established. Several hypotheses have been
proposed, including a-syn-induced Ca
2+
dyshomeostasis.
More recently, oligomeric forms of a-syn have been proposed
to be the most neurotoxic form of this protein. a-syn oligomers
trigger Ca
2+
influx and the subsequent caspase activation in cul-
tured neurons and neuroblastoma cells (26,27). a-syn has been
suggested to be able to form Ca
2+
permeable pores in the
plasma membrane, much like other aggregating proteins, such
as amyloid bpeptides and prion proteins (28). Notably, the
SNpc DA neurons are pace-making neurons that keep firing
through an L-type calcium channel (29). As the result, the alter-
ation of calcium homeostasis may make the SNpc DA neurons
more vulnerable to PD-related degeneration (29). In line with
this notion, the analyses of central nervous system tissues from
patients with PD suggest a role for cellular Ca
2+
overload in
the death of vulnerable neurons in this disease (29). The hypoth-
esis that a-syn may trigger Ca
2+
influx, together with the more
general concept that perturbed Ca
2+
homeostasis is of central
importance to neurodegenerative processes (30), prompted us
to determine the potential effects of increased a-syn levels on
processes downstream of the Ca
2+
-signalling pathway.
In our study, we identified a new calcium-dependent pathway
in dopaminergic neuron loss. We found that a-syn directly acti-
vated CN activity on the small phosphorylated compound p-NPP
as well as the 19 amino acid phosphopeptide RII. In addition, we
also observed a similar increase in the CN activity in WT or
A53T a-syn-transfected HEK293 cells. The activation of CN
leads to the dephosphorylation of key signal transduction mole-
cules, including the NFAT family of transcription factors (31).
The dephosphorylated NFAT is transferred from the cytosol
into the nucleus, where it induces the expression of target
genes, such as cytokine genes, in human T cells in cooperation
with other transcription factors, such as AP-1 (32). Here, we
found a typical nuclear translocation of NFATc3 in mDA
neurons in response to ionomycin stimulation. More import-
antly, we observed a significant translocation of NFATc3 from
the cytosol to the nucleus in the mDA neurons of A53T a-syn
transgenic mice. However, the function and downstream
targets of NFAT in mDA neurons remain to be determined.
Although originally described in T cells, NFATs are now
known to participate in the regulation of CN-mediated transcrip-
tional activity in axonal growth, dendritic branching and pre-
synaptic differentiation (17,33,34). A combined deletion of
either NFATc3/NFATc4 or NFATc2/NFATc3/NFATc4 iso-
forms leads to a marked deficiency in axonal development
(16). Moreover, NFATc3 and NFATc4 have also been impli-
cated in the regulation of neuronal survival (35). For example,
NFATc4 activation has also been recently proposed to mediate
deafferentation-induced neuronal loss in the cochlear nucleus
(36). Furthermore, increased CN levels and the associated shut-
tling of NFATc3 and NFATc4 from the cytosol to the nucleus are
indicated in methamphetamine-induced neuron death (37).
NFAT translocation also induced FasL protein expression in stri-
atal GABAergic neurons, which may be related to neuronal
apoptosis and cognitive defects in patients who abuse metham-
phetamine (38). Given that NFAT activation contributes to
the loss of neurons, the a-syn-mediated translocation of
NFATc3 may contribute to the mDA neurodegeneration in
PD. In support of this hypothesis, we found that treatment with
the CN inhibitor CsA rescued the a-syn-induced loss of
primary mDA neuron cultures. Notably, CsA not only inhibits
CN-dependent NFAT transcriptional activation but also blocks
the mitochondrial Ca
2+
fluxes by binding to the mitochondrial
receptor cyclophylin D (CypD) (39,40). However, whether
the mitochondrial calcium homeostasis is altered in the
mDA neurons of A53T a-syn transgenic mice remains to be
determined. In addition, it will be interesting to identify the
downstream targets of NFATc3 in mDA neurons, which
may provide new molecular targets for potential therapeutic
interventions.
MATERIALS AND METHODS
Cell line culture and transfection
HEK293 cells were cultured in 100-mm dishes with Dulbecco’s
Modified Eagle’s Medium supplemented with 10% heat-
inactivated foetal bovine serum (FBS) (Invitrogen) and
penicillin/streptomycin (Sigma Aldrich). Six micrograms of
Figure 5. CsA treatment ameliorated A53T a-syn-induced loss of midbrain
dopaminergic neurons. (A) CsA induces NFATc3 phosphorylation in HEK293
cells transfected with WT a-syn. (B) Survival of TH-positive neurons treated
with DMSO or 1 mMCsA. Y-axis represents the survival rate (%) of TH-positive
neurons. Six pairs of control and A53T cultures were analysed for DMSO or CsA
treatment, respectively. For each culture, 200 500 TH-positive neurons were
counted. Data were presented as mean +SEM. ∗∗P,0.01, ∗∗∗P,0.001 com-
pared with the A53T DMSO group. (C) Representative images show co-staining
of NFATc3 (green) and TH (red) in primary mDA neurons from neonatal A53T
a-syn pups treated with CsA or DMSO. Scale bar: 20 mm.
Human Molecular Genetics, 2014 5
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
WT and A53T
a
-syn cDNAs in the pcDNA3.1expression vector
(Invitrogen) were used for each transfection via the Fugene 6
Transfection Reagent (Roche Applied Science) according to
the manufacturer’s instructions. The cells were allowed to
grow for 48 h after transfection before being harvested for the
following experiments. The cell suspension was washed with
three volumes of ice-cold phosphate-buffered saline by repeated
centrifugation at 500×gfor 2 min at 48C. The cells were lysed in
appropriate buffers to determine the CN enzyme activity and for
the western blot analysis.
Nuclear and cytoplasmic fractionation
The NE-PER nuclear and cytoplasmic extraction kit (Thermo
Fisher Scientific, Inc.) was used to separate the cytoplasmic
and nuclear fractions of HEK293 cells and mouse midbrain
tissues according to the instruction of the manufacturer.
b-actin was used as the loading control for the cytosolic proteins,
and histone deacetylase-1 (HDAC1) was used as the loading
control of nuclear proteins.
Western blot analysis
The HEK293 cells or midbrain tissues were homogenized in
SDS buffer (50 mMTris HCl, 150 mMNaCl, 2 mMEDTA, pH
7.6, and 2% SDS) supplemented with protease inhibitors
(Roche Applied). Following 15-min incubation on ice, the
protein extracts were clarified by centrifugation at 15 000 ×g
for 30 min at 48C. The protein contents of the supernatants
were quantified using an assay kit based on bicinchoninic
acid (Thermo Scientific), and the supernatants were then sepa-
rated by 4 12% NuPage Bis-Tris polyacrylamide gel electro-
phoresis (Invitrogen) using MES or MOPS running buffer
(Invitrogen). After transferring to nitrocellulose membranes,
the membranes were immunoblotted with the appropriate dilu-
tions of primary antibodies: a-syn (1 : 500, Santa Cruz),
b-actin (1 : 1000, Sigma), tyrosine hydroxylase (anti-TH anti-
body, 1 : 1000, Santa Cruz), NFATc3 (F-1 monoclonal antibody,
1 : 500, Santa Cruz), NFATc1 (7A6 monoclonal antibody, 1 :
500, Santa Cruz) or calcineurin (pan-calcineurin A antibody,
1 : 1000, Cell Signaling). The signals were visualized by
enhanced chemiluminescence development (Thermo Scientific)
and quantified by a Scion Image System (Frederick, MD).
Calcineurin activity assay
The CnA and CnB subunits for the activity assay were expressed
and purified according to the description (40). The protein purity
was analysed by SDS PAGE. The purified CnA was concen-
trated with an Amicon Ultra Filter Unit and diluted in 50 mM
Tris HCl, 0.5 mMdithiothreitol, 0.1 mg/ml BSA and 50% gly-
cerol. A colorimetric assay was used to determine the activities
of CN with 20 mMp-NPP or RII peptide as the substrate. The
reaction was terminated after reacting at 308C for 10 min.
The same vehicle without recombinant human a-syn (ProSpec,
Israel) wasused as a control. Therecombinant humana-synuclein
produced in Escherichia Coli is purified by proprietary chromato-
graphic techniques, and the purity is .95.0% as determined by
SDS– PAGE. The CN activity of each sample was determined
in triplicate. The phosphatase activities are presented as % of
the control.
The CN activity in HEK293 cells was determined with a Cal-
cineurin Cellular Assay Kit (PLUS-AK-816, Enzo Life
Sciences) according to manufacturer’s instructions. Briefly,
the CN activity was measured as the dephosphorylation rate of
the RII peptide. The amount of PO
4
released was calorimetri-
cally determined with the classic malachite green reagent. The
activity was calculated as the difference in protein phosphatase
activity in 2×assay buffer and 2 ×EGTA buffer. The CN activ-
ity of each sample was determined in triplicate. The phosphatase
activities are presented as millimoles of phosphate released/mg
of protein/min at 308C.
Human A53T
a
-syn transgenic mice
The PITX3-IRES-tTA/tetO-A53T double transgenic mice were
generated as previously described (18). Mice were housed in a
12-h light/dark cycle and fed a regular diet ad libitum. All
mouse work followed the guidelines approved by the Institution-
al Animal Care and Use Committees of the National Institute of
Child Health and Human Development, NIH.
Genotyping
The genomic DNA was extracted from a tail biopsy using the
DirectPCR Lysis Reagent (Viagen Biotech, Inc., Los Angeles,
CA, USA) and subjected to PCR amplification using specific
sets of PCR primers for each genotype, including PITX3-
IRES2-tTA transgenic mice (PITX3-F: GACTGGCTTGCC
CTCGTCCCA and PITX3-R: GTGCACCGAGGCCCCAGA
TCA), a-syn A53T transgenic mice (PrpEx2-F: TACTGCTC
CATTTTGCGTGA and SNCA-R: TCCAGAATTCCTTCCTG
TGG) and tetO-H2Bj-GFP mice (H2BGFP0872F, AAGTTC
ATCTGCACCACCG and H2BGFP1416R, TCCTTGAAGAA
GATGGTGCG).
Immunohistochemistry and light microscopy
To immune-stain the mouse midbrain sections, the mice were
sacrificed and then perfused via cardiac infusion with 4% paraf-
ormaldehyde in cold PBS. To obtain frozen sections, the brain
tissues were removed, cryo-protected in 30% sucrose for 24 h
and sectioned at 40 mm thickness using a cryostat (Leica
CM1950). Antibodies specific to NFATc3 (1 : 300, Sigma
Aldrich USA, St. Louis, MO, USA), a-syn and tyrosine hydro-
xylase (TH) (1 : 1000, Pel-Freez Biologicals, Rogers, AR,
USA) were used as suggested by the manufacturers. Alexa
488- or Alexa 568-conjugated secondary antibody (1 : 500, Invi-
trogen) was used to visualize the staining. The fluorescence
images were captured using a laser scanning confocal micro-
scope (LSM 510; Zeiss, Thornwood, NJ, USA). Sections from
Bregma-2.92 to -3.16 mm of control and A53T transgenic
mice were used for immunostaining and the following image
analyses. Sixteen TH-positive neurons were selected from
each brain that showed intact nuclear structure based on
Topro3 staining. The paired images in all figures were collected
at the same gain and offset settings. Post-collection processing
was uniformly applied to all paired images. The images are pre-
sented as either a single optic layer after acquisition in z-series
stack scans at 0.8-mm intervals from individual fields or as
maximum intensity projections to represent confocal stacks.
6Human Molecular Genetics, 2014
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
Laser capture microdissection and RNA-sequencing
analysis
We adopted whole-genome gene expression analyses of DA
neurons isolated from the SNpc of 12-month-old a-syn A53T
transgenic mice and age-matched control mice. To facilitate
the identification of mDA neurons, we generated the
Pitx3-tTA::tetO-H2Bj-GFP::tetO-A53T triple transgenic mice
and control Pitx3-tTA::tetO-H2Bj-GFP double transgenic
mice, in which the histone-GFP fusion proteins (H2Bj-GFP)
are restricted to the nucleus of TH-positive DA neurons in
both the SNpc and ventral tegmental area (VTA) (18). The mid-
brain DA neurons were isolated directly without any staining
owing to the strong GFP signals, and the integrity of RNA was
preserved for the later RNAseq experiments. The strong GFP
signals allowed directly isolate the midbrain DA neurons
without any staining and help to preserve the integrity of RNA
for the later RNAseq experiments. The total RNA was extracted
with the PicoPure Isolation kit (Applied Biosystems). The
genomic DNA was eliminated during the RNA isolation
process. The RNA quantity was measured with NanoDrop-
Spectrophotometer, and the quality was evaluated with BioAna-
lyzer. The libraries for TruSequencing were set up from 100-ng
total RNA fragmented to 200-base pair length. The cDNA li-
braries were amplified by PCR and validated by the BioAnaly-
zer. The deep RNA sequencing was performed on the Illumina
HiSeq 2500 on 2 ×100 bp type for 200 cycles with the Illumina
TruSeq SBS kit. After sequencing instrument generates the se-
quencing images, both image analysis step and base call steps
were run using standard Illumina pipeline. Raw sequences
were filtered and trimmed based upon quality scores over read
cycles. Then, we aligned the paired-end sequencing reads to
mouse reference genome (mm10) using Bowtie2 (2.1.0)
package and Samtools (0.1.14) toolkit. We then utilized Cuf-
flinks (2.1.1) to annotate sequencing reads and estimate tran-
scripts abundances. The 10-mm transcript sequences from
NCBI Reference Sequence Database were used as the annotation
reference. We used DEGSeq, a Bioconductor R package, in
downstream count-based analysis for differential expression
among samples with different genotypes.
Primary neuronal culture and treatment
Primary midbrain neuronal cultures were prepared from P0 pups
of breeding pairs fed with doxycycline (DOX). Briefly, individ-
ual midbrain containing SNpc and VTA was subjected to papain
digestion (5 U/ml, Worthington) for 40 min at 378C. The
digested tissue was carefully triturated into single cells using in-
creasingly smaller pipette tips. The cells were then centrifuged at
250×gfor 5 min and re-suspended in warm Basal Medium
Eagle supplemented with 5% heat-inactivated FBS, 1×B27
(Gibco), 1×N2 (Gibco), 1×GlutaMAX, 0.45% D-glucose
(Sigma), 10 U/ml penicillin (Gibco) and 10 g/ml streptomycin
(Gibco). The dissociated cells from each midbrain were
equally divided and plated onto four 12-mm round coverslips
pre-coated with poly-D-lysine and laminin (BD Bioscience),
and the slips were maintained at 378C in a 95% O
2
- and 5% CO
2
-
humidified incubator. Twenty-four hours after seeding, the
cultures were switched to serum-free medium supplemented
with 5 Mcytosine-D-arabino-furanoside (Sigma), which was
used to suppress the proliferation of glia. The cells in two
sister coverslips were maintained in the presence of 1 mg/ml
DOX after plating. After 5 days in vitro, DOX-treated or non-
treated cells were exposed to 1 mMCsA (Sigma) or DMSO
vehicle control for another 48 h. The cells were then fixed with
4% paraformaldehyde and 4% sucrose in PBS for 15 min, per-
meabilized by 0.1% Triton X-100 for 5 min and blocked in
10% non-immune donkey serum for 1 h at room temperature
(RT). The cells were then double-labelled with primary anti-
bodies against TH (1 : 1000, Santa Cruz) and NFATc3 (1 :
1000, Sigma) overnight at 48C in a humidified chamber. After
three washes with PBS, donkey-derived secondary antibodies
conjugated to Alexa Fluor 488 and Alexa Fluor 546 (1 : 1000,
Invitrogen) were applied and incubated for 1 h at RT in the
dark. After extensive washes, the nuclei were stained with
Topro3 iodide (1 : 1000, Invitrogen). Finally, the coverslips
were mounted on glass slides with Prolong Gold antifade
reagent (Invitrogen), and the fluorescence signals were detected
using a laser scanning confocal microscope (LSM 510; Zeiss).
The total number of all TH-positive neurons on each of the
four sister coverslips was counted under a 25×objective.
Owing to the different amounts of midbrain neurons in each
mouse, we used the relative survival rate of TH-positive
neurons in each midbrain. The rate was calculated by dividing
the number of TH-positive neurons on the non-DOX-treatment
coverslip with the number of TH-positive neurons on the DOX-
treated coverslip from the same midbrain preparation.
Image analysis
To quantitatively assess the marker protein distributions, images
were taken using identical settings and exported to ImageJ (NIH)
for imaging analyses. The images were converted to an 8-bit
colour scale (fluorescence intensity from 0 to 255) using
ImageJ. The areas of interest were first selected with Polygon
or Freehand selection tools and then subjected to measurement
by mean optical intensities or area fractions. The mean intensity
for the background area was subtracted from the selected area to
determine the net mean intensity.
Statistical analysis
A statistical analysis was performed using GraphPad Prism 5
(GraphPad Software, Inc., La Jolla, CA). The data are presented
as the mean +SEM. Statistical significances were determined
by comparing means of different groups and conditions
using t-test, one-way ANOVA with post hoc test. P,0.05,
∗∗P,0.01, ∗∗∗P,0.001.
ACKNOWLEDGEMENTS
The authors thank members of Cai lab for providing various
supports, Mr Christopher Letson and Dr J. Raphael Gibbs for
helping with RNAseq experiments, and China Scholarship
Council (CSC) for its international exchange programs.
Conflict of Interest statement. None declared.
FUNDING
This work was supported in part by the intramural research pro-
grams of National Institute on Aging (AG000928, AG000929)
Human Molecular Genetics, 2014 7
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
and by the National Natural Science Foundation of China
(Project 81072648 and 81373389).
REFERENCES
1. Forno, L.S. (1996) Neuropathology of Parkinson’s disease. J. Neuropathol.
Exp. Neurol.,55, 259– 272.
2. Hardy, J., Cai, H., Cookson, M.R., Gwinn-Hardy, K. and Singleton, A.
(2006) Genetics of Parkinson’s disease and parkinsonism. Ann. Neurol.,60,
389398.
3. Singleton,A.B., Farrer, M., Johnson, J., Singleton, A., Hague, S., Kachergus,
J., Hulihan, M., Peuralinna, T., Dutra, A., Nussbaum, R. et al. (2003)
a-Synuclein locus triplication causes Parkinson’s Disease. Science,
302, 841.
4. Feany, M.B. and Bender, W.W. (2000) A drosophila model of Parkinson’s
disease. Nature,404, 394– 398.
5. Giasson, B.I., Duda, J.E., Quinn, S.M., Zhang, B., Trojanowski, J.Q. and
Lee, V.M. (2002) Neuronal alpha-synucleinopathy with severe movement
disorder in mice expressing A53T human alpha-synuclein. Neuron,34,
521533.
6. Lakso, M., Vartiainen,S., Moilanen, A.M., Sirvio, J., Thomas, J.H., Nass, R.,
Blakely, R.D. and Wong, G. (2003) Dopaminergic neuronal loss and motor
deficits in Caenorhabditis elegans overexpressing human alpha-synuclein.
J. Neurochem.,86, 165– 172.
7. Thiruchelvam, M.J., Powers, J.M., Cory-Slechta, D.A. and Richfield, E.K.
(2004) Risk factors for dopaminergic neuron loss in human alpha-synuclein
transgenic mice. Eur. J. Neurosci.,19, 845854.
8. Zhou, W., Schaack, J., Zawada, W.M. and Freed, C.R. (2002)
Overexpression of human alpha-synuclein causes dopamine neuron death in
primary human mesencephalic culture. Brain Res.,926, 4250.
9. Ahn, B.H., Rhim, H., Kim, S.Y., Sung, Y.M., Lee, M.Y., Choi, J.Y.,
Wolozin, B., Chang, J.S., Lee, Y.H., Kwon, T.K. et al. (2002)
Alpha-synuclein interacts with phospholipase D isozymes and inhibits
pervanadate-induced phospholipase D activation in human embryonic
kidney-293 cells. J. Biol. Chem.,277, 12334– 12342.
10. Ostrerova, N., Petrucelli, L., Farrer, M., Mehta, N., Choi, P., Hardy, J. and
Wolozin, B. (1999) Alpha-synuclein shares physical and functional
homology with 143-3 proteins. J. Neurosci.,19, 57825791.
11. Peng, X.M., Tehranian, R., Dietrich, P., Stefanis, L. and Perez, R.J. (2005)
Alpha-synuclein activation of protein phosphatase 2A reduces tyrosine
hydroxylase phosphorylation in dopaminergic cells. J. Cell Sci.,118,
3523– 3530.
12. Nielsen, M.S., Vorum, H., Lindersson, E. and Henning Jensen, P. (2001)
Ca
2+
binding to a-synuclein regulates ligand binding and oligomerization.
J. Biol. Chem.,276, 2268022684.
13. Klee, C.B., Crouch, T.H. and Krinks, M.H. (1979) Calcineurin: a calcium-
and calmodulin-binding protein of the nervous system. Proc. Natl. Acad. Sci.
USA,76, 6270– 6273.
14. Crabtree, G.R. and Olson, E.N. (2002) NFAT signaling: choreographing the
social lives of cells. Cell,109, S67S79.
15. Macian, F. (9999) NFAT proteins: key regulators of T-cell development and
function. Nat. Rev. Immunol.,5, 472– 484.
16. Graef, I.A., Wang, F., Charron, F., Chen, L., Neilson, J., Tessier-Lavigne, M.
and Crabtree, G.R. (2003) Neurotrophins and netrins require calcineurin/
NFAT signaling to stimulate outgrowth of embryonic axons. Cell,113,
657670.
17. Nguyen, T. and Giovanni, S.Di. (2008) NFAT signaling in neural
development and axon growth. Int. J. Dev. Neurosci.,26, 141145.
18. Lin, X., Parisiadou, L., Yu, J., Liu, G., Sun, L., Sgobio, C., Shim, H., Gu,
X.L., Luo, J., Long, C.X. et al. (2012) Heterologous expression of
Parkinson’s disease-related mutant alpha-synuclein causes the dysfunction
of Nurr1 and degeneration of midbrain dopaminergic neurons in transgenic
mice. J. Neurosci.,32, 9248– 9264.
19. Pallen, C.J. andWang, J.H. (1983) Calmodulin-stimulateddephosphorylation
of p-nitrophenyl phosphate and free phosphotyrosine by calcineurin. J. Biol.
Chem.,258, 8550 –8553.
20. Blumenthal, D., Takio, K., Hansen, R.S. and Krebs, E.G. (1986)
Dephosphorylation of cAMP-dependent protein kinase regulatory subunit
(Type II) by calmodulin-dependent protein phosphatase. J. Biol. Chem.,262,
8140– 8145.
21. Rusnak, F. and Mertz, P. (2000) Calcineurin: form and function. Physiol.
Rev.,80, 1483– 1521.
22. Li, G.D., Zhang, X., Li, R., Wang, X.D., Wang, Y.L., Han, K.J., Qian, X.P.,
Yang, C.G., Liu, P., Wei, Q. et al. (2008) CHP2 activates the calcineurin/
nuclear factor of activated T cells signaling pathway and enhances the
oncogenic potential of HEK293 cells. J. Biol. Chem.,283, 32660– 32668.
23. Liu, C. and Hermann, T.E. (1978) Characterization of ionomycin as a
calcium ionophore. J. Biol. Chem.,253, 5892–5894.
24. Matsuoka, Y., Vila, M., Lincoln, S., McCormack, A., Picciano, M.,
LaFrancois, J., Yu, X., Dickson, D., Langston, W.J., McGowan, E. et al.
(2001) Lack of nigral pathology in transgenic mice expressing human
alpha-synuclein driven by the tyrosine hydroxylase promoter. Neurobiol.
Dis.,8, 535– 539.
25. Zhou, W., Hurlbert, M.S., Schaack, J., Prasad, K.N. and Freed, C.R. (2000)
Overexpression of human alpha-synuclein causes dopamine neuron death in
rat primary culture and immortalized mesencephalon-derived cells. Brain
Res.,866, 33 43.
26. Danzer, K.M., Haasen, D., Karow, A.R., Moussaud, S., Habeck, M., Giese,
A., Kretzschmar, H., Hengerer, B. and Kostka, M. (2007) Different species
of a-synuclein oligomers induce calcium influx and seeding. J. Neurosci.,
27, 9220– 9232.
27. Martin, Z.S., Neugebauer, V., Dineley, K.T., Kayed, R., Zhang, W., Reese,
L.C. and Taglialatela, G. (2012) Alpha-synuclein oligomers oppose
long-term potentiation and impair memory through a calcineurin-dependent
mechanism: relevance to human synucleopathic diseases. J. Neurochem.,
120, 440– 452.
28. Hettiarachchi, N.T., Parker, A., Dallas, M.L., Pennington, K., Hung, C.C.,
Pearson, H.A., Boyle, J.P., Robinson, P. and Peers, C. (2009) a-Synuclein
modulation of Ca
2+
signaling in human neuroblastoma (SH-SY5Y) cells.
J. Neurochem.,111, 1192– 1201.
29. Surmeier, D.J., Guzman, J.N. and Sanchez-Padilla, J. (2010) Calcium,
cellular aging, and selective vulnerability in Parkinson’s disease. Cell
Calcium,47, 175– 182.
30. Mattson, M.P. (2007) Calcium and neurodegeneration. Aging Cell,6,
337–350.
31. Loh, C., Shaw, K.T., Carew, J., Viola, J., Luo, C., Perrino, B.A. and Rao, A.
(1996) Calcineurin binds the transcription factor NFAT1 and reversibly
regulates its activity. J. Biol. Chem.,271, 10884–10891.
32. Rao, A. (2009) Signaling to gene expression: calcium, calcineurin and
NFAT. Nat. Immunol.,10,35.
33. Yoshida, T. and Mishina, M. (2005) Distinct roles of calcineurin-nuclear
factor of activated T-cells and protein kinase A-cAMP response
element-binding protein signaling in presynaptic differentiation.
J. Neurosci.,25, 3067– 3079.
34. Schwartz, N., Schohl, A. and Ruthazer, E.S. (2009) Neural activity regulates
synaptic properties and dendritic structure in vivo through calcineurin/
NFAT signaling. Neuron,62, 655– 669.
35. Ulrich, J.D., Kim, M-S., Houlihan, P.R., Shutov, L.P., Mohapatra, D.P.,
Strack, S. and Usachev, Y.M. (2012) Distinct activation properties of the
nuclear factor of activated T-cells (NFAT) isoforms NFATc3 and NFATc4
in neurons. J. Biol. Chem.,287, 3759437609.
36. Luoma, J.I. and Zirpel, L. (2008) Deafferentation-induced activation of
NFAT (nuclear factor of activated T-cells) in cochlear nucleus neurons
during a developmental critical period: a role for NFATc4-dependent
apoptosis in the CNS. J. Neurosci.,28, 3159– 3169.
37. Jayanthi,S., Deng, X.L., Ladenheim, B., McCoy, M.T., Cluster, A., Cai, N.S.
and Cadet, J.L. (2005) Calcineurin/NFAT-induced up-regulation of the Fas
ligand/Fas death pathway is involved in methamphetamine-induced
neuronal apoptosis. Proc. Natl. Acad. Sci. USA,102, 868– 873.
38. Liu, J., Farmer,, J.D. Jr., Lane, W.S., Friedman, J., Weissman, I. and
Schreiber, S.L. (1991) Calcineurin is a common target of cyclophilin-
cyclosporin A and FKBP-FK506 complexes. Cell,66, 807 815.
39. Fournier, N., Ducet, G. and Crevat, A. (1987) Action of cyclosporine on
mitochondrial calcium fluxes. J. Bioenerg. Biomembr.,19, 297– 303.
40. Wang, H., Yao, S., Lin, W., Du, Y., Xiang, B., He, S., Huang, C. and Wei, Q.
(2007) Different roles of Loop 7 in inhibition of calcineurin. Biochem.
Biophys. Res. Commun.,362, 925– 929.
8Human Molecular Genetics, 2014
at Galter Health Sciences Library on September 11, 2014http://hmg.oxfordjournals.org/Downloaded from
... Strikingly, elevated levels of the active form of CaN have been reported in post-mortem brain tissue from patients with confirmed ␣-synuclein pathology [4]. A growing body of evidence underscores the centrality of CaN in the pathogenesis and progression of PD, particularly through its regulation of downstream transcription factors [4,[7][8][9][10][11][12][13]. Notably, CaN inhibition may extend dopamine action by preventing dephosphorylation of Dopamine-and cAMP-regulated phosphoprotein (DARPP-32) [13]. ...
... Notably, CaN inhibition may extend dopamine action by preventing dephosphorylation of Dopamine-and cAMP-regulated phosphoprotein (DARPP-32) [13]. Furthermore, CaN inhibition counteracts ␣-synuclein-induced toxicity, including neurodegeneration and motor impairments, in preclinical models of PD pathology [4,8,12]. However, the therapeutic manipulation of CaN demands precision, as both its deletion and overexpression have been linked to amplified ␣-synuclein toxicity [10]. ...
... CaN dephosphorylates nuclear factor of activated T-cells (NFAT), triggering its relocation to the nucleus and subsequent activation of immune response genes [14][15][16]. Alongside its well-recognized role in peripheral immune modulation, NFAT activation within the CNS contributes to neuroinflammation and neuronal death [12,17]. Consequently, immunomodulatory strategies, particularly CaN inhibition, emerge as a promising avenue for therapeutic intervention in PD. ...
Article
Full-text available
Background: Preclinical evidence suggests calcineurin inhibitors (CNIs) combat α-synuclein-induced neuronal dysfunction and motor impairments. However, whether CNIs prevent or treat Parkinson’s disease (PD) in humans has never been investigated. Objective: We seek to ascertain if prescription of CNIs is linked to a decreased prevalence of PD in a varied patient population and to glimpse into the mechanism(s) and target site through which CNIs might decrease PD prevalence. Methods: We analyzed electronic health records (EHRs) from patients prescribed the brain penetrant CNI tacrolimus (TAC), the peripherally restricted CNI cyclosporine (CySp), or the non-CNI sirolimus (SIR). For comparison, EHRs from a diverse population from the same network served as a general population-like control. After propensity-score matching, prevalence, odds, and hazards of PD diagnoses among these cohorts were compared. Results: Patients prescribed CNIs have decreased odds of PD diagnosis compared to the general population-like control, while patients prescribed SIR do not. Notably, patients prescribed TAC have a decreased prevalence of PD compared to patients prescribed SIR or CySp. Conclusions: Our results suggest CNIs, especially those acting within the brain, may prevent PD. The reduced prevalence of PD in patients prescribed TAC, compared to patients prescribed SIR, suggests that mechanisms of calcineurin inhibition— other than immunosuppression, which is common to both drugs— are driving the reduction. Therefore, CNIs may provide a promising therapeutic approach for PD.
... One instance is proven inside the necessity of the nuclear issue of activated T cells (NFATs) for regulating both gene expression and immune responses. Within the nuclear surroundings, α-synuclein's capability to interact with NFAT has a long way-attaining effects [90,122]. Its ability to either activate or inhibit NFAT-dependent genes results in immune responses; in particular its potential to avert T cell activation [25]. ...
... Through binding with calcineurin -a regulatory protein for NFATα-synuclein can negatively affect signaling activity associated with this factor. Ultimately such impact could lead not only to changes in gene expression but also impairments such as DNA damage and complications within the nucleocytoplasmic transport efficacy [90]. Mitochondrial dysfunction is another consequential hallmark associated with PD through autophagy lysosomal pathway (ALP) and ubiquitin-proteasome system all under α-synuclein influence (Fig. 3) [28]. ...
Article
Parkinson's disease (PD) is a progressive disorder that belongs to a class of neurodegenerative disorders (NDs) called Synucleinopathies. It has characterized by the misfolding and aggregation of a-synuclein. Our understanding of PD continues to evolve, and so does our approach to treatment. including therapies aimed at delaying pathology, quitting neuronal loss, and shortening the course of the disease by selectively targeting essential proteins suspected to play a role in PD pathogenesis. One emerging approach that is generating significant interest is Targeted Protein Degradation (TPD). TPD is an innovative method that allows us to specifically break down certain proteins using specially designed molecules or peptides, like PROteolysis-TArgeting-Chimera (PROTACs). This approach holds great promise, particularly in the context of NDs. In this review, we will briefly explain PD and its pathogenesis, followed by discussing protein degradation systems and TPD strategy in PD by reviewing synthesized small molecules and peptides. Finally, future perspectives and challenges in the field are discussed.
... So, these physical interactions within a Cav1.2 channel complex could support rapid short-term modulation of Ca 2+ entry under the control of G-protein-coupled receptors. Interestingly, a calcineurin-dependent pathway is involved in α-synuclein-induced degeneration of midbrain dopaminergic neurons [45]. ...
Article
Full-text available
Pacemaking activity in substantia nigra dopaminergic neurons is generated by the coordinated activity of a variety of distinct somatodendritic voltage- and calcium-gated ion channels. We investigated whether these functional interactions could arise from a common localization in macromolecular complexes where physical proximity would allow for efficient interaction and co-regulations. For that purpose, we immunopurified six ion channel proteins involved in substantia nigra neuron autonomous firing to identify their molecular interactions. The ion channels chosen as bait were Cav1.2, Cav1.3, HCN2, HCN4, Kv4.3, and SK3 channel proteins, and the methods chosen to determine interactions were co-immunoprecipitation analyzed through immunoblot and mass spectrometry as well as proximity ligation assay. A macromolecular complex composed of Cav1.3, HCN, and SK3 channels was unraveled. In addition, novel potential interactions between SK3 channels and sclerosis tuberous complex (Tsc) proteins, inhibitors of mTOR, and between HCN4 channels and the pro-degenerative protein Sarm1 were uncovered. In order to demonstrate the presence of these molecular interactions in situ, we used proximity ligation assay (PLA) imaging on midbrain slices containing the substantia nigra, and we could ascertain the presence of these protein complexes specifically in substantia nigra dopaminergic neurons. Based on the complementary functional role of the ion channels in the macromolecular complex identified, these results suggest that such tight interactions could partly underly the robustness of pacemaking in dopaminergic neurons.
... Moreover, we showed that inhibition of CnA with CsA and with VIVIT, a specific inhibitor of the CnA-NFAT pathway, can revert this transcription factor activation and block aSyn-O neurotoxicity, as characterized by the almost complete inhibition of cell death, reversal of neural ramification deterioration, and loss of pre-synaptic synapsin 1 positive puncta formation (Figs. 1, 2 and 3). Consistently, the NFAT transcription factor has already been demonstrated to be important in neural and axonal growth and development (Graef et al. 2003(Graef et al. , 2001Nguyen and Giovanni 2008), survival (Ulrich et al. 2012), and death (Luoma and Zirpel 2008;Jayanthi et al. 2005;Liu et al. 1991), being localized in the nucleus of neurons in aSyn transgenic mice in postmortem brain tissue from PD patients (Caraveo et al. 2014), it can be activated by aSyn in mice neurons and its modulation by the non-NFAT-specific inhibitor (CsA) can ameliorated the aSyn-induced loss of mDA neurons (Luo et al. 2014). Further, Ca + /CnA/NFAT signaling pathway is linked to several nervous system diseases as Down Syndrome, Alzheimer's, Huntington's, traumatic brain injuries, amyotrophic lateral sclerosis, and others (Kipanyula et al. 2016;Park et al. 2009;Minami 2014;Sompol and Norris 2018) suggesting a relevant role for this transcription factor pathway in nervous system physiological maintenance and development, including Parkinson's disease. ...
Article
Full-text available
Background: Soluble oligomeric forms of alpha-synuclein (aSyn-O) are believed to be one of the main toxic species in Parkinson's disease (PD) leading to degeneration. aSyn-O can induce Ca2+ influx, over activating downstream pathways leading to PD phenotype. Calcineurin (CN), a phosphatase regulated by Ca2+ levels, activates NFAT transcription factors that are involved in the regulation of neuronal plasticity, growth, and survival. Methods: Here, using a combination of cell toxicity and gene regulation assays performed in the presence of classical inhibitors of the NFAT/CN pathway, we investigate NFAT's role in neuronal degeneration induced by aSyn-O. Results: aSyn-O are toxic to neurons leading to cell death, loss of neuron ramification and reduction of synaptic proteins which are reversed by CN inhibition with ciclosporin-A or VIVIT, a NFAT specific inhibitor. aSyn-O induce NFAT nuclear translocation and transactivation. We found that aSyn-O modulates the gene involved in the maintenance of synapses, synapsin 1 (Syn 1). Syn1 mRNA and protein and synaptic puncta are drastically reduced in cells treated with aSyn-O which are reversed by NFAT inhibition. Conclusions: For the first time a direct role of NFAT in aSyn-O-induced toxicity and Syn1 gene regulation was demonstrated, enlarging our understanding of the pathways underpinnings synucleinopathies.
... The interaction between calcineurin and α-synuclein is facilitated by calciumcalmodulin [50]. Calcineurin triggers neuroinflammation by activating nuclear factor of activated cells (NFAT) transcription, leading to inflammation and neuron death [51]. Calmodulin regulates small conductance calcium-activated K+ (SK) channels, which prevent the release of neurotransmitter by increasing the channel's potential above polarization [52]. ...
Article
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta, leading to motor symptoms such as tremors, rigidity, and bradykinesia. Current therapeutic strategies for PD are limited and mainly involve symptomatic relief, with no available treatment for the underlying causes of the disease. Therefore, there is a need for new therapeutic approaches that target the underlying pathophysiological mechanisms of PD. Calcium homeostasis is an essential process for maintaining proper cellular function and survival, including neuronal cells. Calcium dysregulation is also observed in various organelles, including the endoplasmic reticulum (ER), mitochondria, and lysosomes, resulting in organelle dysfunction and impaired inter-organelle communication. The ER, as the primary calcium reservoir, is responsible for folding proteins and maintaining calcium homeostasis, and its dysregulation can lead to protein misfolding and neurodegeneration. The crosstalk between ER and mitochondrial calcium signaling is disrupted in PD, leading to neuronal dysfunction and death. In addition, a lethal network of calcium cytotoxicity utilizes mitochondria, ER and lysosome to destroy neurons. This review article focused on the complex role of calcium dysregulation and its role in aggravating functioning of organelles in PD so as to provide new insight into therapeutic strategies for treating this disease. Targeting dysfunctional organelles, such as the ER and mitochondria and lysosomes and whole network of calcium dyshomeostasis can restore proper calcium homeostasis and improve neuronal function. Additionally targeting calcium dyshomeostasis that arises from miscommunication between several organelles can be targeted so that therapeutic effects of calcium are realised in whole cellular territory.
... Strikingly, the limited neuroprotective effect of VIVIT peptide disappeared when LD is co-treated, resembling retrograde DAergic neurodegeneration. As CaN substrates with PxIxIT docking site include NFAT family proteins [39], we speculate that neuroinflammation might play a role in the current context [69,70] and thus VIVIT treatment can protect neurons by blocking NFAT activation. As VIVIT is ineffective for CaN-DRP1 axis, it cannot offer the same effect any further when LD is administered in the PD model. ...
Article
Full-text available
Background Parkinson’s disease (PD), a highly prevalent neuro-motor disorder is caused due to progressive loss of dopaminergic (DAergic) neurons at substantia nigra region of brain. This leads to depleted dopamine (DA) content at striatum, thus affecting the fine tuning of basal ganglia. In patients, this imbalance is manifested by akinesia, catalepsy and tremor. PD associated behavioral dysfunctions are frequently mitigated by l-DOPA (LD) therapy, a precursor for DA synthesis. Due to progressive neurodegeneration, LD eventually loses applicability in PD. Although DA is cytotoxic, it is unclear whether LD therapy can accelerate PD progression or not. LD itself does not lead to neurodegeneration in vivo, but previous reports demonstrate that LD treatment mediated excess DA can potentiate neurotoxicity when PD associated genetic or epigenetic aberrations are involved. So, minimizing DA toxicity during the therapy is an absolute necessity to halt or slowdown PD progression. The two major contributing factors associated with DA toxicity are: degradation by Monoamine oxidase and DAquinone (DAQ) formation. Results Here, we report that apoptotic mitochondrial fragmentation via Calcineurin (CaN)-DRP1 axis is a common downstream event for both these initial cues, inhibiting which can protect cells from DA toxicity comprehensively. No protective effect is observed, in terms of cell survival when only PxIxIT domain of CaN is obstructed, demonstrating the importance to block DRP1-CaN axis specifically. Further, evaluation of the impact of DA exposure on PD progression in a mice model reveal that LD mediated behavioral recovery diminishes with time, mostly because of continued DAergic cell death and dendritic spine loss at striatum. CaN inhibition, alone or in combination with LD, offer long term behavioral protection. This protective effect is mediated specifically by hindering CaN-DRP1 axis, whereas inhibiting interaction between CaN and other substrates, including proteins involved in neuro-inflammation, remained ineffective when LD is co-administered. Conclusions In this study, we conclude that DA toxicity can be circumvented by CaN inhibition and it can mitigate PD related behavioral aberrations by protecting neuronal architecture at striatum. We propose that CaN inhibitors might extend the therapeutic efficacy of LD treatment.
... Another study has found that the overexpression of pathogenic α-Syn can induce the nuclear translocation of NFATc1 and NFATc3, which is relative to α-Syn-induced degeneration of midbrain dopaminergic neurons (Luo et al., 2014). Nevertheless, little evidence is available to illuminate the role of microglial NFATc1 in α-Syn pathology. ...
Article
Full-text available
Synucleinopathies refer to a range of neurodegenerative diseases caused by abnormal α-synuclein (α-Syn) deposition, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Their pathogenesis is strongly linked to microglial dysfunction and neuroinflammation, which involves the leucine-rich-repeat kinase 2 (LRRK2)-regulated nuclear factor of activated T-cells (NFAT). Of the NFAT family, NFATc1 has been found to be increasingly translocated into the nucleus in α-syn stimulation. However, the specific role of NFATc1-mediated intracellular signaling in PD remains elusive in regulating microglial functions. In the current study, we crossbred LRRK2 or NFATc1 conditional knockout mice with Lyz2Cre mice to generate mice with microglia-specific deletion of LRRK2 or NFATc1, and by stereotactic injection of fibrillary α-Syn, we generated PD models in these mice. We found that LRRK2 deficiency enhanced microglial phagocytosis in the mice after α-Syn exposure and that genetic inhibition of NFATc1 markedly diminished phagocytosis and α-Syn elimination. We further demonstrated that LRRK2 negatively regulated NFATc1 in α-Syn-treated microglia, in which microglial LRRK2-deficiency facilitated NFATc1 nuclear translocation, CX3CR1 upregulation, and microglia migration. Additionally, NFATc1 translocation upregulated the expression of Rab7 and promoted the formation of late lysosomes, resulting in α-Syn degradation. In contrast, the microglial NFATc1 deficiency impaired CX3CR1 upregulation and the formation of Rab7-mediated late lysosomes. These findings highlight the critical role of NFATc1 in modulating microglial migration and phagocytosis, in which the LRRK2-NFATc1 signaling pathway regulates the expression of microglial CX3CR1 and endocytic degradative Rab7 to attenuate α-synuclein immunotoxicity.
... Both of these two regions are vital for the function of TRIM proteins. Moreover, TRIM17 directly resisted the degradation of another transcription factor NFATc3, which is linked to the degeneration of dopaminergic neurons in the midbrain induced by α-Syn in PD patients (Mojsa et al. 2015;Luo et al. 2014;Caraveo et al. 2014). ...
Article
Full-text available
Tripartite motif (TRIM) protein superfamily is a group of E3 ubiquitin ligases characterized by the conserved RING domain, the B-box domain, and the coiled-coil domain (RBCC). It is widely involved in various physiological and pathological processes, such as intracellular signal transduction, cell cycle regulation, oncogenesis, and innate immune response. Central nervous system (CNS) diseases are composed of encephalopathy and spinal cord diseases, which have a high disability and mortality rate. Patients are often unable to take care of themselves and their life quality can be seriously declined. Initially, the function research of TRIM proteins mainly focused on cancer. However, in recent years, accumulating attention is paid to the roles they play in CNS diseases. In this review, we integrate the reported roles of TRIM proteins in the pathological process of CNS diseases and related signaling pathways, hoping to provide theoretical bases for further research in treating CNS diseases targeting TRIM proteins. Graphical Abstract TRIM proteins participated in CNS diseases. TRIM protein family is characterized by a highly conserved RBCC domain, referring to the RING domain, the B-box domain, and the coiled-coil domain. Recent research has discovered the relations between TRIM proteins and various CNS diseases, especially Alzheimer’s disease, Parkinson’s disease, and ischemic stroke.
Article
Calcium (Ca2+) and Ca2+-regulated enzymes calpain and calcineurin are the key molecules of signaling mechanisms in neurons and ensure the normal course of intracellular neurochemical and neurophysiological processes. The imbalance and increase in the intracellular level of Ca2+ correlates with the activation of calpain and calcineurin. Inactivation of endogenous inhibitors and/or absence of exogenous pharmacological inhibitors of these enzymes may induce a cascade of intracellular mechanisms that are detrimental to the structural integrity and functional activity of neurons. The interrelated processes of Ca2+ imbalance, dysregulation of calpain and calcineurin are directly related to the development of intracellular pathophysiological reactions leading to the degeneration and death of selective neuronal populations in neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The review briefly presents the characteristics of calpain and calcineurin, their interrelated role in the neurodegeneration processes. Data on the efficiency of the exogenous inhibitors (in vivo, in vitro) point out the potential role of pharmacological regulation of calpain and calcineurin for neuroprotection.
Article
Full-text available
NFAT1 (previously termed NFATp) is a cytoplasmic transcription factor involved in the induction of cytokine genes. We have previously shown that the dephosphorylation of NFAT1, accompanied by its nuclear translocation and increased DNA binding activity, is regulated by calcium- and calcineurin-dependent mechanisms, as each of these hallmarks of NFAT1 activation is elicited by ionomycin and blocked by the immunosuppressive drugs cyclosporin A and FK506 (Shaw, K. T.-Y., Ho, A. M., Raghavan, A., Kim, J., Jain, J., Park, J., Sharma, S., Rao, A., and Hogan, P. G.(1995) Proc. Natl. Acad. Sci. U. S. A. 92, 11205-11209). Here we show that the activation state of NFAT1 in T cells is remarkably sensitive to the level of calcineurin activity. Addition of cyclosporin A, even in the presence of ongoing ionomycin stimulation, results in rephosphorylation of NFAT1, its reappearance in the cytoplasm, and a return of its DNA binding activity to low levels. Similar effects are observed upon removal of ionomycin or addition of EGTA. We also demonstrate a direct interaction between calcineurin and NFAT1 that is consistent with a direct enzyme-substrate relation between these two proteins and that may underlie the sensitivity of NFAT1 activation to the level of calcineurin activity. The NFAT1-calcineurin interaction, which involves an N-terminal region of NFAT1 conserved in other NFAT family proteins, may provide a target for the design of novel immunosuppressive drugs.
Article
Full-text available
The Ca2+/calcineurin(CaN)-dependent transcription factor NFAT (nuclear factor of activated T-cells) is implicated in regulating dendritic and axonal development, synaptogenesis and neuronal survival. Despite the increasing appreciation for the importance of NFAT-dependent transcription in the nervous system, the regulation and function of specific NFAT isoforms in neurons is poorly understood. Here, we compare the activation of NFATc3 and NFATc4 in hippocampal and dorsal root ganglion (DRG) neurons following electrically-evoked elevations of intracellular Ca2+ concentration ([Ca2+]i). We find that NFATc3 undergoes rapid dephosphorylation and nuclear translocation that is essentially complete within 20 min, while NFATc4 remains phosphorylated and localized to the cytosol, only exhibiting nuclear localization following prolonged (1-3 hrs) depolarization. Knocking down NFATc3, but not NFATc4, strongly diminished NFAT-mediated transcription induced by mild depolarization in neurons. By analyzing NFATc3/NFATc4 chimeras we find that the region containing the serine rich region-1 (SRR1) mildly affects initial NFAT translocation, while the region containing the SP repeats is critical for determining the magnitude of NFAT activation and nuclear localization upon depolarization. Knockdown of glycogen synthase kinase 3β (GSK3β) significantly increased the depolarization-induced nuclear localization of NFATc4. In contrast, inhibition of p38 or mTOR kinases had no significant effect on nuclear import of NFATc4. Thus, electrically-evoked [Ca2+]i elevation in neurons rapidly and strongly activates NFATc3, whereas activation of NFATc4 requires a coincident increase in [Ca2+]i and suppression of GSK3β with differences in the SP-containing region giving rise to these distinct activation properties of NFATc3 and NFATc4.
Article
Full-text available
α-Synuclein (α-syn) plays a prominent role in the degeneration of midbrain dopaminergic (mDA) neurons in Parkinson's disease (PD). However, only a few studies on α-syn have been performed in the mDA neurons in vivo, which may be attributed to a lack of α-syn transgenic mice that develop PD-like severe degeneration of mDA neurons. To gain mechanistic insights into the α-syn-induced mDA neurodegeneration, we generated a new line of tetracycline-regulated inducible transgenic mice that overexpressed the PD-related α-syn A53T missense mutation in the mDA neurons. Here we show that the mutant mice developed profound motor disabilities and robust mDA neurodegeneration, resembling some key motor and pathological phenotypes of PD. We also systematically examined the subcellular abnormalities that appeared in the mDA neurons of mutant mice and observed a profound decrease of dopamine release, the fragmentation of Golgi apparatus, and the impairments of autophagy/lysosome degradation pathways in these neurons. To further understand the specific molecular events leading to the α-syn-dependent degeneration of mDA neurons, we found that overexpression of α-syn promoted a proteasome-dependent degradation of nuclear receptor-related 1 protein (Nurr1), whereas inhibition of Nurr1 degradation ameliorated the α-syn-induced loss of mDA neurons. Given that Nurr1 plays an essential role in maintaining the normal function and survival of mDA neurons, our studies suggest that the α-syn-mediated suppression of Nurr1 protein expression may contribute to the preferential vulnerability of mDA neurons in the pathogenesis of PD.
Article
Full-text available
Calcineurin, a calmodulin-binding protein from brain, has been shown to possess a metal ion-dependent and calmodulin-stimulated phosphatase activity towards phosphorylase kinase and inhibitor-1 (Stewart, A. A., Ingebritsen, T. S., Manalan, A., Klee, C. B., and Cohen, P. (1982) FEBS Lett. 137, 80-84). In this report, we show that calcineurin can also dephosphorylate p-nitrophenyl phosphate and free phosphotyrosine. However, calcineurin does not show significant activity towards phosphothreonine, phosphoserine, or several other low molecular weight phosphocompounds tested. As we have found with phosphorylase kinase and phosphocasein, the dephosphorylation of p-nitrophenyl phosphate and free phosphotyrosine is stimulated by calmodulin and is metal ion-dependent with the order of efficiency being Mn2+ much greater than Co2+ greater than Ca2+. The dephosphorylation of these substrates appears to be an intrinsic property of calcineurin and is not due to contamination by alkaline phosphatases since the pH optimum for calcineurin activity occurs at a neutral rather than an alkaline pH. The dephosphorylation of p-nitrophenyl phosphate provides an easy, rapid, and accurate method for the quantification of calcineurin activity as well as permitting insight into reaction kinetics. The dephosphorylation of free phosphotyrosine by calcineurin suggests that this compound may be a physiological substrate of calcineurin.
Article
Calcineurin is a eukaryotic Ca ²⁺ - and calmodulin-dependent serine/threonine protein phosphatase. It is a heterodimeric protein consisting of a catalytic subunit calcineurin A, which contains an active site dinuclear metal center, and a tightly associated, myristoylated, Ca ²⁺ -binding subunit, calcineurin B. The primary sequence of both subunits and heterodimeric quaternary structure is highly conserved from yeast to mammals. As a serine/threonine protein phosphatase, calcineurin participates in a number of cellular processes and Ca ²⁺ -dependent signal transduction pathways. Calcineurin is potently inhibited by immunosuppressant drugs, cyclosporin A and FK506, in the presence of their respective cytoplasmic immunophilin proteins, cyclophilin and FK506-binding protein. Many studies have used these immunosuppressant drugs and/or modern genetic techniques to disrupt calcineurin in model organisms such as yeast, filamentous fungi, plants, vertebrates, and mammals to explore its biological function. Recent advances regarding calcineurin structure include the determination of its three-dimensional structure. In addition, biochemical and spectroscopic studies are beginning to unravel aspects of the mechanism of phosphate ester hydrolysis including the importance of the dinuclear metal ion cofactor and metal ion redox chemistry, studies which may lead to new calcineurin inhibitors. This review provides a comprehensive examination of the biological roles of calcineurin and reviews aspects related to its structure and catalytic mechanism.
Article
Calcium signaling activates the phosphatase calcineurin and induces movement of NFATc proteins into the nucleus, where they cooperate with other proteins to form complexes on DNA. Nuclear import is opposed by kinases such as GSK3, thereby rendering transcription continuously responsive to receptor occupancy. Disruptions of the genes involved in NFAT signaling are implicating this pathway as a regulator of developmental cell–cell interactions.
Article
α-Synucleinopathies are neurodegenerative disorders that range pathologically from the demise of select groups of nuclei to pervasive degeneration throughout the neuraxis. Although mounting evidence suggests that α-synuclein lesions lead to neurodegeneration, this remains controversial. To explore this issue, we generated transgenic mice expressing wild-type and A53T human α-synuclein in CNS neurons. Mice expressing mutant, but not wild-type, α-synuclein developed a severe and complex motor impairment leading to paralysis and death. These animals developed age-dependent intracytoplasmic neuronal α-synuclein inclusions paralleling disease onset, and the α-synuclein inclusions recapitulated features of human counterparts. Moreover, immunoelectron microscopy revealed that the α-synuclein inclusions contained 10–16 nm wide fibrils similar to human pathological inclusions. These mice demonstrate that A53T α-synuclein leads to the formation of toxic filamentous α-synuclein neuronal inclusions that cause neurodegeneration.
Article
The etiology and pathogenesis of Parkinson’s disease have remained a mystery since the first description of the disease, but the neuropathology of the disorder, as we now know it, is fairly simple and straightforward. The most important lesion is in the substantia nigra pars compacta, where nerve cells degenerate together with their nigro-striatal fiber tract, causing a severe depletion of dopamine in the basal ganglia.
Article
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by the appearance of intracytoplasmic inclusions called Lewy bodies (LB) in dopamine neurons in the substantia nigra and the progressive loss of these neurons. Recently, mutations in the α-synuclein gene have been identified in early-onset familial PD, and α-synuclein has been shown to be a major component of LB in all patients. Yet, the pathophysiological function of α-synuclein remains unknown. In this report, we have investigated the toxic effects of adenovirus-mediated α-synuclein overexpression on dopamine neurons in rat primary mesencephalic cultures and in a rat dopaminergic cell line – the large T-antigen immortalized, mesencephalon-derived 1RB3AN27 (N27). Adenovirus-transduced cultures showed high-level expression of α-synuclein within the cells. Overexpression of human mutant α-synuclein (Ala53Thr) selectively induced apoptotic programmed cell death of primary dopamine neurons as well as N27 cells. The mutant protein also potentiated the neurotoxicity of 6-hydroxydopamine (6-OHDA). By contrast, overexpression of wild-type human α-synuclein was not directly neurotoxic but did increase cell death after 6-OHDA. Overexpression of wild-type rat α-synuclein had no effect on dopamine cell survival or 6-OHDA neurotoxicity. These results indicate that overexpression of human mutant α-synuclein directly leads to dopamine neuron death, and overexpression of either human mutant or human wild-type α-synuclein renders dopamine neurons more vulnerable to neurotoxic insults.
Article
J. Neurochem. (2012) 120 , 440–452. Abstract Intracellular deposition of fibrillar aggregates of α‐synuclein (αSyn) characterizes neurodegenerative diseases such as Parkinson’s disease (PD) and dementia with Lewy bodies. However, recent evidence indicates that small αSyn oligomeric aggregates that precede fibril formation may be the most neurotoxic species and can be found extracellularly. This new evidence has changed the view of pathological αSyn aggregation from a self‐contained cellular phenomenon to an extracellular event and prompted investigation of the putative effects of extracellular αSyn oligomers. In this study, we report that extracellular application of αSyn oligomers detrimentally impacts neuronal welfare and memory function. We found that oligomeric αSyn increased intracellular Ca ²⁺ levels, induced calcineurin (CaN) activity, decreased cAMP response element‐binding protein (CREB) transcriptional activity and resulted in calcineurin‐dependent death of human neuroblastoma cells. Similarly, CaN induction and CREB inhibition were observed when αSyn oligomers were applied to organotypic brain slices, which opposed hippocampal long‐term potentiation. Furthermore, αSyn oligomers induced CaN, inhibited CREB and evoked memory impairments in mice that received acute intracerebroventricular injections. Notably, all these events were reversed by pharmacological inhibition of CaN. Moreover, we found decreased active CaN and reduced levels of phosphorylated CREB in autopsy brain tissue from patients affected by dementia with Lewy bodies, which is characterized by deposition of αSyn aggregates and progressive cognitive decline. These results indicate that exogenously applied αSyn oligomers impact neuronal function and produce memory deficits through mechanisms that involve CaN activation.