Mend 1743

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

ORIGINAL RESEARCH

Oxytocin-Stimulated NFAT Transcriptional Activation


in Human Myometrial Cells

Jason N. A. Pont, Craig A. McArdle, and Andrs Lpez Bernal


Bristol University, School of Clinical Sciences, Bristol BS1 3NY, United Kingdom

Oxytocin (OXT) is a peptide hormone that binds the OXT receptor on myometrial cells, initiating
an intracellular signaling cascade, resulting in accumulation of intracellular calcium and smooth
muscle contraction. In other systems, an elevation of intracellular Ca2! stimulates nuclear trans-
location of the transcription factor, nuclear factor of activated T cells (NFAT), which is transcrip-
tionally active in arterial and ileal smooth muscle. Here we have investigated the role of NFAT in
the mechanism of action of OXT. Human myometrial cells expressed all five NFAT isoforms
(NFATC1C4 and -5). Myometrial cells were transduced with a recombinant adenovirus expressing
a NFATC1-EFP reporter, and a semi-automated imaging system was used to monitor effects of OXT
on reporter localization in live cells. OXT induced a concentration-dependent nuclear transloca-
tion of NFATC1-EFP in a reversible manner, which was inhibited by OXT antagonists and calcineu-
rin inhibitors. Pulsatile stimulation with OXT caused intermittent, pulse-frequency-dependent,
nuclear translocation of NFATC1-EFP, which was more efficient than sustained stimulation. OXT
induced nuclear translocation of endogenous NFAT that was transcriptionally active, because OXT
stimulated activity of a NFAT-response element-luciferase reporter and induced calcineurin-NFAT
dependent expression of RGS2, RCAN1, and PTGS2 (COX2) mRNA. Furthermore, OXT-dependent
transcription was dependent on protein neosynthesis; cycloheximide abolished RGS2 transcription
but augmented RCAN1 and COX2 transcriptional readouts. This study identifies a novel signaling
mechanism within the myometrium, whereby calcineurin-NFAT signaling mediates OXT-induced
transcriptional activity. Furthermore, we show NFATC1-EFP is responsive to pulses of OXT, a
mechanism by which myometrial cells could decode OXT pulse frequency. (Molecular
Endocrinology 26: 17431756, 2012)

T he uterus primarily functions as a quiescent environ-


ment for the developing fetus during pregnancy, be-
fore switching from a relaxed to a contractile state at term
elucidated, making intervention and treatment of preterm
labor problematic (5, 6).
Oxytocin (OXT) is a potent uterotonic hormone, re-
resulting in parturition. Complications arise when partu- leased from the posterior pituitary in a pulsatile manner.
rition occurs too early or too late, but more often when it As parturition progresses, OXT pulse frequency and du-
is premature. Ten percent of births worldwide are pre- ration increase (7), thus enhancing the frequency and
term (defined as delivery preceding 37 wk gestation) (1) force of myometrial contractions. Although the clinical
with 85% of infant morbidity and mortality attributed to use of OXT in the induction and augmentation of labor is
prematurity (2). Interestingly, nearly 50% of preterm well established (8, 9), its importance in initiating partu-
births are idiopathic, with no obvious etiology (1), and rition physiologically is debatable. Animal experiments
are therefore potentially preventable (3, 4). However, the have shown that OXT-deficient mice exhibit normal la-
mechanisms by which labor is initiated have yet to be fully bor and delivery, although the pups do not survive due to

ISSN Print 0888-8809 ISSN Online 1944-9917 Abbreviations: AM, Acetoxymethyl ester; BAPTA, 1,2-bis(o-aminophenoxy)ethane-
Printed in U.S.A. N,N,N",N"-tetraacetic acid; [Ca2!]i, intracellular calcium; COX2, cyclooxygenase 2; CX,
Copyright 2012 by The Endocrine Society cycloheximide; CyclA, cyclosporin A; FCS, fetal calf serum; !-Gal, !-galactosidase; GPCR,
doi: 10.1210/me.2012-1057 Received February 20, 2012. Accepted July 12, 2012. G protein-coupled receptor; N:C, nuclear to cytoplasmic ratio; NFAT, nuclear factor of
First Published Online August 17, 2012 activated T cells; NF"B, nuclear factor "-light-chain-enhancer of activated B cells; OXT,
oxytocin; OXTR, OXT receptor; PFA, paraformaldehyde; pfu, plaque-forming units; PSS,
physiological salt solution; qPCR, real-time PCR; RCAN1, regulator of calcineurin 1; RGS2,
regulator of G protein signaling 2.

Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1743


1744 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

loss of the milk ejection reflex (10, 11). This suggests that the NFAT-binding motif in DNA promoter regions and
OXT may play an essential role in the establishment of cooperates with cofactors, i.e. activator protein 1, globin
lactation, but only a permissive role during parturition. In transcription factor 1, and myocyte enhancer factor 2 to
women, there is an increase in OXT receptor (OXTR) enhance gene expression (44, 45). As [Ca2!]i declines,
density in the myometrium during late pregnancy; how- NFAT is rephosphorylated by kinases such as c-JUN N-
ever, there is no further increase near the onset of labor terminal kinase and glycogen synthase kinase 3 (46 48),
(12, 13), indicating that OXTR density is not a defining promoting nuclear export of NFAT via a chromosome
factor in initiating parturition. Maternal plasma OXT region maintenance protein 1/exportin mechanism (49).
levels are not elevated before the onset of parturition in Pulsatile release of hormones is important for regulat-
women (14); however, in addition to being released into ing physiological processes and coordinating Ca2! signal-
the circulation by the posterior pituitary, OXT is also ing and thus transcription within a cell; i.e. pulses of OXT
synthesized and secreted by the decidua, with OXT determine luteolysis and maintenance of pregnancy in the
mRNA and protein expression in this tissue increasing sheep (50), and pulses of GnRH regulate gene expression
prepartum (1517). This suggests a possible paracrine in the gonadotrophs (51, 52). A pulsatile system is phys-
mechanism by which locally synthesized OXT may be iologically advantageous in comparison with sustained
involved in the initiation of parturition at term. release of a hormone for several reasons: intermittent
OXT binds to the OXTR, a G protein-coupled recep- stimulation of a receptor does not result in desensitization
tor (GPCR), on myometrial cells to initiate an intracellu- and internalization of the receptor, as with sustained
lar signaling pathway culminating in intracellular calcium stimulation (53), and allows for recycling of the receptor
([Ca2!]i) accumulation (18, 19), and thus uterine smooth to the surface between stimulations for a maximal re-
muscle contraction through activation of myosin light sponse. Moreover, the energy cost to the cell of intermit-
chain kinase (20 22). In conjunction with contraction, tently elevating [Ca2!]i is far less than the energy required
Ca2! signaling also mediates activation of certain tran- to maintain elevated [Ca2!]i levels for a sustained period
scription factors, i.e. nuclear factor "-light-chain-en- of time. Furthermore, pulse frequency is an important
hancer of activated B cells (NF"B), c-JUN N-terminal mechanism for encoding transcriptional responses; i.e.
kinases and nuclear factor of activated T cells (NFAT). GnRH pulse frequency is decoded within gonadotrophs
NFAT was first identified in T cells, being essential for to stimulate differential transcriptional readouts (51),
induction of cytokine gene expression during the immune and a similar system may be involved in GH-regulated
response (23). Outside the immune system, NFAT has gene transcription (54). NFAT nuclear shuttling has been
been physiologically implicated in skeletal (24) and car- shown to be dependent on Ca2! oscillation and hormonal
diac (25, 26) muscle hypertrophy, skeletal muscle growth pulse frequency, with the greater the frequency, the
and development (27, 28), heart valve formation (29), greater the amplitude of NFAT nuclear localization (51,
and vascular development (30 32). To date, there have 55, 56). This suggests that NFAT is an important mech-
been no studies on the function of NFAT in the myome- anism in decoding pulse frequency to differentially regu-
trium; however, experiments in mice have shown that late gene transcription.
inhibition of NFAT signaling at term delays delivery by In this report, we demonstrate for the first time that
several hours (33), suggesting that the NFAT pathway NFAT is involved in the mechanism of action of OXT in
may be involved in the initiation of parturition. human myometrial cells. OXT induces translocation of
Five members of the NFAT family of transcription fac- NFAT to the nucleus via a calcineurin-mediated mecha-
tors have been isolated: NFATC2 (NFAT1/p) (34), nism, with NFAT nuclear localization mirroring pulses of
NFATC1 (NFAT2) (35), NFATC4 (NFAT3) (36), OXT in a frequency-dependent manner. We show that
NFATC3 (NFAT4/x) (36 38), and NFAT5 (TonEBP)
pulsatile OXT can provide a more efficient signal for
(39, 40). NFATC1 4 signaling is mediated by the Ca2!/
NFAT activation than a sustained stimulus and that
calmodulin-dependent phosphatase calcineurin (41),
OXT-induced gene expression is dependent on calcineu-
whereas NFAT5 activity is activated by osmotic stress
rin-NFAT activity.
(40). In resting cells, the four calcineurin-sensitive
NFATC1 4 transcription factors are predominantly lo-
calized to the cytosol in a phosphorylated state. On ele-
Materials and Methods
vation of [Ca2!]i, calcineurin induces NFAT dephosphor-
ylation, unmasking nuclear localization signals (42) and Reagents
stimulating cotranslocation of the calcineurin-NFAT DMEM, fetal calf serum (FCS), collagenase type II, dispase,
complex to the nucleus (43). In the nucleus, NFAT binds penicillin, streptomycin Hoechst nuclear stain, fluo4 Ca2! in-
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1745

dicator, !-mercaptoethanol, and primers were ordered from In- NFATC1-EFP nuclear translocation
vitrogen (Paisley, UK). Deoxyribonuclease, elastase, atosiban, Myometrial cells were seeded at 5000 cells per well in black-
coenzyme A, and ATP was purchased from Sigma (Dorset, UK). walled 96-well plates. Next day, cells were transduced with a
L368899, cyclosporin A (CyclA), FK506, and cycloheximide NFATC1-EFP adenovirus (GE Healthcare, Buckinghamshire,
(CX) were purchased from Tocris (Bristol, UK). OXT was from UK), at 2 # 104 plaque-forming units (pfu)/#l in DMEM (2%
Calbiochem (Darmstadt, Germany), ionomycin from Ascent FCS) for 16 24 h. On the day of experiment, cells were loaded
Scientific (Abcam, Cambridge, UK), and luciferin from Promega with 0.5 #M Hoechst nuclear stain in physiological salt solution
(Southampton, UK). All basic chemicals were from Sigma unless (PSS) [127 mM NaCl, 0.5 mM NaH2PO4, 1.8 mM CaCl2, 2 mM
otherwise stated. MgCl2, 5 mM KCl2, 5 mM NaHCO3, 10 mM HEPES (pH 7.5),
0.1% BSA, 10 mM glucose] and incubated at 37 C for 30 min.
Antibodies For live cell experiments, cell populations were imaged before
Primary antibodies were mouse monoclonal anti-NFATC1 stimulation with OXT, with subsequent images being acquired
at the relevant time points after stimulation (see figure legends).
(7A6) antibody (SC7294) from Santa Cruz Biotechnology
For repeated stimulation experiments, cells were washed five
(Santa Cruz, CA) and rabbit polyclonal antidesmin (ab15200),
times with PSS between 5-min stimulations with OXT (the fre-
mouse monoclonal anti-actin (ab18147), and rabbit monoclo-
quency and amplitude of stimulation is described in the figure
nal anticalponin (ab46794) from Abcam (Cambridge, UK). Sec-
legends). For fixed cell imaging, cells were pretreated with the
ondary antibodies were Alexa fluor 488 goat antirabbit IgG
relevant compound (as described in figure legends) and stimu-
(heavy and light chains) from Invitrogen, and DyLight 488 Af-
lated with OXT or ionomycin before terminating the reaction
finiPure donkey antimouse IgG (heavy and light chains) from with ice-cold PBS, fixation in 4% paraformaldehyde (PFA) for
Jackson ImmunoResearch Laboratories (Suffolk, UK). 20 min, and imaging.

Myometrial cell dispersion and culture IN Cell Analyzer image analysis


This study was approved by the South West Research Ethics Image analysis and fluorescence quantification was per-
Committee. Human myometrial biopsies were obtained, with formed using the IN Cell Analyzer Work station version 3.5
informed consent from premenopausal women at hysterectomy. software (IN Cell Investigator; GE Healthcare). Cytoplasmic
Myometrial cell isolation and culture was adapted from the and nuclear regions were defined by green channel (fluo4 and
protocol described by Phaneuf et al. (57). Briefly, dissected tis- NFATC1-EFP) and blue channel (Hoechst nuclear stain) im-
sue was incubated at 37 C, with gentle shaking, in DMEM (4.5 ages, respectively. The fluorescence readouts for each region
g/liter glucose, glutamate), with 300 U/ml collagenase type II, were used to calculate cell population and individual cell fluo-
0.3 U/ml dispase, 30 U/ml deoxyribonuclease, and 0.09 U/ml rescence. For graphical representation and statistical analysis,
elastase for 1 h. The supernatant was spun for 10 min at 378 # fluo4 nuclear intensity (background subtracted) was standard-
g, and the cell pellet was resuspended in DMEM supplemented ized to resting cell nuclear intensity, and NFATC1-EFP cyto-
with 10% FCS and 1% penicillin-streptomycin. After two ad- plasmic and nuclear intensity were used to calculate the nuclear
ditional incubations, collected cells were pooled into T75 flasks to cytoplasmic ratio (N:C) as previously described (51).
for culture. Cultured cells were used experimentally from pas-
sages 310. Immunocytochemistry
Myometrial cells were seeded at 5000 cells per well in black-
IN Cell Analyzer 1000 image acquisition walled 96-well plates. Next day, cells were washed with PBS,
Imaging experiments were performed using the IN Cell An- fixed in 4% PFA for 20 min, and permeabilized with 0.1%
alyzer 1000 (GE Healthcare, Buckinghamshire, UK) semiauto- Triton X-100 for 10 min. Nonspecific primary antibody binding
mated imaging platform as previously described (51). Images was blocked with normal goat serum (Invitrogen) (desmin and
were acquired in a single field of view for live cell imaging and calponin) or normal donkey serum (Sigma) (NFATC1 and ac-
three fields for fixed cell imaging. Experiments were performed tin) for 2 h. Cells were treated with primary antibody for 24 h at
in triplicate wells with 50 200 cells per field. 4 C. Primary antibodies used were NFATC1 (1:100), desmin
(1:150), actin (1:400), and calponin (1:150). Next day, cells
were washed in PBS six times before incubation with goat anti-
Measurement of [Ca2!]i
rabbit (1:200) (desmin and calponin) and donkey antimouse
Myometrial cells were seeded at 5000 cells per well in black- (1:200) (NFATC1 and actin) secondary antibody. Cells were
walled 96-well sample plates (Appleton Woods, Birmingham, again washed with PBS six times before staining with Hoechst
UK). Next day, cells were washed before loading with 2.5 #M nuclear stain for 30 min and image acquisition by the IN Cell
fluo4 Ca2! indicator and 0.5 #M Hoechst nuclear stain, along Analyzer 1000 as previously described. Image analysis was by
with other compounds if required (as described in figure leg- IN Cell Analyzer work station version 3.5 software as previ-
ends), in phenol red-free DMEM (10% FCS, 25 mM HEPES) ously described. Primary antibody cytoplasmic fluorescence was
and incubated at 37 C for 30 min. After incubation, sample defined by green channel images. A filter was set for cytoplasmic
plates were loaded into the IN Cell Analyzer 1000 for imaging. intensity at 260, 250, and 300 arbitrary fluorescent units for
Live cell populations were imaged before stimulation with OXT desmin, actin, and calponin images, respectively, to calculate the
(as described in figure legends), using a robotic needle, with percentage of positive cells for each primary antibody. For
subsequent images acquired every 12 sec for 60 sec. graphical representation and statistical analyzes, a NFATC1
1746 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

N:C was calculated as previously described and normalized to 4-fold on reaction completion. Gene expression was quantified
the untreated control. by real-time PCR (qPCR) with Power SYBR Green Master Mix
(Applied Biosystems, Foster City, CA), using a 7500 real-time
Luciferase assay PCR system (Applied Biosystems, Foster City, CA). A template
Myometrial cells were seeded at 5000 cells per well in black- of 2 #l cDNA was run per reaction in a final reaction volume of
walled 96-well plates. Next day, cells were transduced with a 20 #l (75 nM forward and reverse primers, Table 1) for 45 cycles
NFAT response element luciferase reporter (NFAT-RE-Luc) of 95 C for 15 sec and 60 C for 60 sec. A dissociation stage
(Addgene, Cambridge, UK; plasmid 10959) and !-galactosidase followed to ensure the amplification of a single product. Rela-
(!-Gal) adenovirus as previously described (51, 58) at 1 # 104 tive mRNA expression levels were quantified by the Pfaffl
pfu/#l and 5 # 103 pfu/#l, respectively, in DMEM (2% FCS) for method of relative quantification (59), as used by Peeters et al.
16 24 h. Cells were treated with inhibitors where necessary and (60), and standardized to the housekeeping genes 18S and RNA
stimulated with OXT in PSS at the desired concentration and polymerase II. Expression levels were normalized to control val-
time (see figure legends) before the reaction was terminated with ues for graphical representation and statistical analysis.
ice-cold PBS, treated with lysis buffer, and frozen then thawed to
aid lysis. Luciferase reagent [33.3 mM dithiothreitol, 270 #M Promoter sequence analysis for NFAT-binding
coenzyme A, 530 #M ATP, 470 #M luciferin in assay buffer motifs
containing 20 mM Tricene, 0.1 mM EDTA, 1.07 mM Regulator of G protein signaling 2 (RGS2), regulator of cal-
(MgCO3)4Mg(OH)2, and 2.67 mM MgSO4] was added to cell cineurin 1 (RCAN1), and cyclooxygenase 2 (COX2) promoter
lysates to assess luminescence with a Lumat LB 9507 luminom- sequences were derived using the UCSC genome browser (UCSC
eter (Berthold Technologies, Hertfordshire, UK). !-Gal activity Genome Bioinformatics, University of California, Santa Cruz,
was used to correct for adenovirus transduction efficiency, be- CA). The promoter sequences were entered into the Meme Suite
ing measured by incubation of the remaining lysate with chlo- program (61) and using the Fimo Algorithm (62) scanned for
rophenol-red-galactopyranoside (Roche Applied Sciences, West NFAT-binding motifs. Statistical significance was taken as P $
Sussex, UK) mix (878 #M chlorophenol-red-galactopyranoside, 0.001.
175 #M !-mercaptoethanol, 60 mM Na2HPO4, 40 mM
NaH2PO4, 10 mM KCl, and 1 mM MgSO4) at 37 C for 48 h, with
Statistical analysis
!-Gal activity measured using a microplate reader (Revelation
Statistical analysis was performed with GraphPad Prism ver-
version 4.22; Dynex Technologies, West Sussex, UK). Light
sion 5 (GraphPad Software, La Jolla, CA) using one-way or
units were standardized to !-Gal activity and normalized to
two-way ANOVA with Dunnetts or Bonferroni post hoc test as
control for graphical representation and statistical analysis.
indicated. Results shown are mean % SEM, with P $ 0.05 con-
sidered statistically significant. All experiments were repeated at
Quantitative real-time PCR least three times using cells from three or more separate donors.
Myometrial cells were seeded at 100,000 cells per well in
six-well plates and grown to 60 80% confluency. Cells were
treated with OXT, plus or minus inhibitors, for the appropriate
times (as described in figure legends). The stimulations were Results
terminated with ice-cold PBS (ribonuclease free) before addition
of lysis buffer (50 #M mercaptoethanol). RNA was isolated Characterization of myometrial cell phenotype
using an RNeasy mini kit (QIAGEN, West Sussex, UK), accord- The phenotype of myometrial cells in our culture sys-
ing to the manufacturers protocol. Quantity and quality of tem were validated using primary antibodies for the
RNA was assessed by the NanoDrop 1000 full spectrum 220/
smooth muscle cell markers desmin, actin, and calponin
750-nm spectrophotometer (Labtech International, East Sussex,
UK). cDNA was synthesized using the High Capacity cDNA (Supplemental Fig. 1, published on The Endocrine Soci-
Reverse Transcription Kit (Applied Biosystems, Foster City, etys Journals Online web site at http://mend.endojour-
CA) following the manufacturers instructions and diluted nals.org). From a sample size of 1200 2400 cells, 95.4 %

TABLE 1. Primer pairs for qPCR


Gene nomenclature Forward primer (5!3!) Reverse primer (5!3!)
RNA18S1 (18s) (1332) ATGGCCGTTCTTAGTTGGTG (1548) CGCTGAGCCAGTCAGTGTAG
POLR2A (RNAPOLII) (4453) GCACCACGTCCAATGACATTG (4719) GTGCGGCTGCTTCCATAAGC
NFATC2 (NFAT1) (2114) AAGAGCCAGCCCAACATGC (2220) CGTTTTCTCTTCCCATTGATGAC
NFATC1 (NFAT2) (1302) CTGTGCAAGCCGAATTCTCTGG (1379) ACTGACGTGAACGGGGCTGG
NFATC4 (NFAT3) (2189) GTCCTGATGGGAAGCTGCAATGG (2258) AGCGTCACCTCGTTGCTCTGC
NFATC3 (NFAT4) (303) GCGGCCTGCAGATCTTGAGC (404)TGATGTGGTAAGCAAAGTGGTGTGGT
NFAT5 (2458) GACACTGGCGGTGGACTGCG (2517) CTGGCTTCGACATCAGCATTCCT
RGS2 (236) CTTTCATCAAGCCTTCTCCTGA (299) GCTAGCAGCTCGTCAAATGC
RCAN1 (DSCR1) (493)GCTCAGACCTTACACATAGGAAGC (598) CCACTTGTTTCCATCCCACTG
PTGS2 (COX2) (590) CAAAGGTAAAAAGCAGCTTCCTG (671) CTGGGGATCAGGGATGAACT
The gene sequence position of each primers 5"-terminal nucleotide is indicated in parentheses preceding each primer sequence.
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1747

1.3% of cells were positive for desmin, 94 % 0.28% pos- fluo4-AM intensity returning to near baseline within 1
itive for actin, and 99.1 % 0.33% positive for calponin. min (Fig. 2A).
Within the no-primary-antibody control group, 96.3 % To assess the effect of OXT on NFAT activity, myo-
0.54% of cells were negative for desmin, 99 % 0.003% metrial cells were transduced with a recombinant adeno-
negative for actin, and 98.4 % 0.9% negative for virus expressing a NFATC1-EFP imaging reporter. In un-
calponin. stimulated cells, NFATC1-EFP was largely cytoplasmic;
however, on treatment with OXT, NFATC1-EFP trans-
Expression of NFAT isoforms in human located to the nucleus (Fig. 2 and Supplemental Fig. 1).
myometrium This effect was quantified by an automated imaging sys-
Because there has been no previous description of tem, which defined the NFATC1-EFP fluorescence inten-
NFAT expression in myometrial smooth muscle, we de- sity in the cytoplasm and nucleus to calculate the
signed probes to identify NFAT isoforms by real-time NFATC1-EFP (N:C) intensity ratio in cell populations
PCR and found that NFATC1, NFATC2, NFATC3, and individual cells (Supplemental Fig. 2). Sustained
NFATC4, and NFAT5 were expressed in cultured myo- treatment with OXT induced a concentration-dependent
metrial cells. The calcineurin-insensitive NFAT5 was ex- increase in NFATC1-EFP N:C ratio (EC50 & 1.14 % 0.15
pressed in greatest abundance in comparison with the nM) (Fig. 2B) within the cell population, which paralleled
calcineurin-dependent NFATC1 4 (P $ 0.001). The levels the effect of OXT on fluo4-AM intensity. The effect was
of expression of the calcineurin-sensitive isoforms were slower in onset than [Ca2!]i accumulation, with
NFATC4 more than NFATC1, and NFATC3 (P $ 0.01) NFATC1-EFP translocating to the nucleus within 5 min.
and NFATC4 more than NFATC2 (P $ 0.001) (Fig. 1). The NFATC1-EFP N:C ratio was maximal from 30 40
min and remained sustained for at least 70 min (Fig. 2B).
OXT-induced NFATC1-EFP nuclear translocation in OXT-induced translocation of NFATC1-EFP to the
human myometrial cells nucleus was inhibited by the OXTR antagonists atosiban
The NFAT signaling pathway is activated by an eleva- and L368899 (Fig. 3A) and by the calcineurin inhibitors
tion of [Ca2!]i in arterial and ileal smooth muscle cells CyclA and FK506 (Fig. 3B). Pretreatment with the intra-
(63, 64). To confirm OXT induction of Ca2! signaling in cellular Ca2! chelator BAPTA [1,2-bis(o-aminophe-
myometrial cells, a fluo4-acetoxymethyl ester (AM) Ca2! noxy)ethane-N,N,N",N"-tetraacetic acid]-AM or the use
indicator was used. Treatment of myometrial cells with of Ca2!-free PSS blocked NFATC1-EFP nuclear translo-
OXT caused a concentration-dependent (EC50 & 1.18 % cation in response to OXT (Fig. 3C). The Ca2! ionophore
0.18 nM) and time-dependent increase in [Ca2!]i (Fig. ionomycin also induced NFATC1-EFP nuclear localiza-
2A). The response was maximal 12 sec after stimulus at tion, the effect being suppressed by CyclA, FK506,
the higher concentrations (10'6 and 10'7 M) and 24 sec BAPTA-AM, and Ca2!-free media (Fig. 3, B and C) but
for the lower concentrations (10'8-10'10 M), with not inhibited by L36899 or atosiban.

Effect of pulsatile OXT on NFATC1-EFP nuclear


translocation
To investigate whether NFATC1-EFP nuclear translo-
cation is dependent on sustained OXTR occupancy or
triggered by the initial binding of OXT to its receptor, we
measured responses in the presence of brief or sustained
OXT exposure. Myometrial cells were treated with OXT
for 5 min followed by a repeated wash period to remove
the agonist (Fig. 4). Within the initial 5-min stimulation,
NFATC1-EFP translocated to the nucleus (35% of max-
imal response seen with sustained receptor occupancy).
FIG. 1. NFAT mRNA expression in human myometrial cells. Real-time After the wash, there was no additional increase in the
PCR was performed on cDNA collected from cultured human
NFATC1-EFP N:C ratio, with the signal gradually declin-
myometrial cells. Data shown are relative mRNA copy number,
mean % SE, from five independent donors, calculated by the Pfaffl ing toward baseline within 40 min. This shows that OXT-
method of analysis and normalized to the housekeeping genes 18S induced nuclear translocation of NFATC1-EFP is revers-
and RNA polymerase II. Statistical analysis was performed by one-way ible and requires constant receptor occupancy for a
ANOVA, with Bonferroni multiple-comparison post hoc test. Statistics
shown are in comparison with relative NFATC4 expression. *, P $ sustained signal. This reversibility raised the possibility
0.01; **, P $ 0.001. that pulsatile OXT treatment would, in turn, cause pul-
1748 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

FIG. 2. OXT induces accumulation of [Ca2!]i and NFATC1-EFP nuclear translocation. Human myometrial cells were loaded with fluo4-AM (A) or
transduced with a NFATC1-EFP adenovirus (B) and stained with Hoechst nuclear stain before imaging. Cells were stimulated with OXT, in a
concentration-dependent manner at the concentration (molar) and time indicated. The concentration of OXT used (molar) is indicated by the
symbols, which apply to both A and B. The control (Ctrl) group was not stimulated with OXT. Live cell image acquisition and analysis was
performed as described in Materials and Methods. Representative images shown are before and after treatment with OXT (10'7 M). Scale bar, 60
#m. Data shown are fluo4 intensity (background subtracted) normalized to time zero (A) and NFATC1-EFP intensity (background subtracted) N:C
ratio (B), mean % SE, from three separate experiments and independent donors. Statistical analyses were performed by one-way ANOVA, with
Dunnetts post hoc test, which show statistical difference compared with the untreated control in terms of area under the curve in A and B, as
follows: for a concentration of OXT of 10'9 M, P $ 0.01, and for 10'8-10'6 M, P $ 0.001.

satile NFATC1-EFP translocation responses, so we exam- by five washes, over a time course of 3 h, at varying
ined the effect of repeated 5-min pulses of OXT on concentrations and pulse frequency. Each exposure to
NFATC1-EFP nuclear localization. Myometrial cells OXT caused a concentration-dependent translocation of
were subjected to 5-min stimulations with OXT, followed NFATC1-EFP to the nucleus, with similar kinetics, i.e.

FIG. 3. OXT-induced NFATC1-EFP nuclear translocation is inhibited by OXT antagonists, calcineurin inhibitors, and inhibition of Ca2! signaling. Human
myometrial cells were transduced with a NFATC1-EFP adenovirus before pretreatment with OXT antagonists atosiban (10'5 M) and L368899 (10'5 M)
(A), calcineurin inhibitors CyclA (10'6 M) and FK506 (10'6 M) (B), and the [Ca2!]i chelator BAPTA-AM (10'6 M) and replacement of PSS with Ca2!-free
solution (C) for 30 min. Cells were stimulated with OXT (10'7 M) or ionomycin (2 #M) for 60 min before terminating the reaction with ice-cold PBS, fixing
in 4% PFA, and staining with Hoechst. Fixed cell image acquisition and analysis were performed as described in Materials and Methods. Data shown are
NFATC1-EFP intensity (background subtracted) N:C ratio, mean % SE, from three separate experiments and independent donors. Statistical analyses were
performed by one-way ANOVA with Dunnetts post hoc test compared with the untreated control. *, P $ 0.01; **, P $ 0.001.
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1749

dent increase in NFAT-RE-Luc activity (EC50 & 84.6 %


0.24 nM) (Fig. 7, A and B). OXT-stimulated (10'7 M)
NFAT-RE luciferase activity was abolished after pretreat-
ment with the OXTR antagonist L368899 (10'5 M) and
with the calcineurin inhibitor CyclA (10'6 M), [luciferase
activity (fold induction): control, 1 % 0; OXT, 2 % 0.11;
L368889, 0.65 % 0.15; CyclA, 0.86 % 0.14; data not
shown].
The increase in NFAT-RE-Luc activity in response to
OXT led us to investigate the effect of OXT on transcrip-
tional activity. For this, we chose genes whose expression
was known to be either OXT dependent in the myome-
trium or NFAT dependent in other smooth muscle cells.
FIG. 4. OXT induction of NFATC1-EFP nuclear translocation is The genes selected for analysis were RGS2 (65) and pros-
reversible. Human myometrial cells were transduced with a NFATC1- taglandin-endoperoxide synthase 2 (PTGS2/COX2) (66)
EFP adenovirus and stained with Hoechst nuclear stain before imaging.
(OXT dependent) and RCAN1 (67), coagulation factor II
Cells were stimulated with either sustained OXT (10'7 M) for 70 min,
or briefly, 5 min, followed by five washes (gray rectangle). The control (thrombin) receptor (F2R/PAR1) (68), and transient re-
(Ctrl) group was not stimulated with OXT. Images were acquired at the ceptor potential cation channel, subfamily C, member 1
time points indicated. Data shown are NFATC1-EFP intensity (TRPC1) (69) (NFAT dependent). COX2 expression has
(background subtracted) N:C ratio, mean % S.E. from three separate
experiments and independent donors. Statistical analysis was by two also been shown to be NFAT dependent (67). Gene acti-
way ANOVA which demonstrated that brief vs. sustained stimulation vation was measured by real-time PCR.
with OXT was a significant source of variation, P $ 0.01. Exposure of myometrial cells to OXT increased
mRNA levels of RGS2, RCAN1, and COX2 (Fig. 8A).
maximal response at 5 min, followed by a gradual decline The response was significant after 4 6 h of stimulation
of NFATC1-EFP N:C after washing, despite the variance with OXT, with a 2.5- to 4-fold increase in mRNA. Pre-
in amplitude (Fig. 5A) and pulse frequency (Fig. 5, B and treatment with L368899 or CyclA prevented the increase
C). Treatment of cells with a pulse of OXT every 30 min in mRNA (Fig. 8B), indicating that OXT-induced gene
resulted in the NFATC1-EFP signal not returning to base- expression is mediated through the OXTR and dependent
line between pulses, resulting in a cumulative effect (Fig. on the calcineurin-NFAT signaling pathway. OXT did
5B). Furthermore, the areas under the curve of the not influence expression of PAR1 or TRPC1 (data not
NFATC1-EFP N:C traces in response to sustained treat- shown).
ment with OXT and pulsatile stimulation (30-min wash
periods) over a 3-h time course were of similar magnitude The effect of OXT on gene expression requires
(Fig. 5D). both transcription and translation
Despite induction of RGS2 gene expression being me-
OXT stimulates endogenous NFAT nuclear diated by the calcineurin-NFAT pathway, it has previ-
translocation and transcriptional activity ously been reported that the RGS2 promoter does not
Because OXT stimulated translocation of a NFATC1- contain the NFAT consensus-binding motif (67), raising
EFP reporter construct to the nucleus of myometrial cells, the question whether OXT-induced activation of gene
we investigated the effect of OXT on endogenous NFAT expression may involve the synthesis of intermediary pro-
nuclear localization using a NFATC1 primary antibody. teins. To search for NFAT-binding motif expression in
A 30-min treatment with OXT (10'6 M) stimulated trans- the promoter regions of RGS2, RCAN1, and COX2, we
location of NFATC1 to the nucleus with CyclA inhibiting used the Meme Suite program as described in Materials
the response (Fig. 6). NFATC1 nuclear translocation was and Methods. The search identified NFAT-binding motifs
also induced by ionomycin [NFATC1 N:C ratio (fold within the promoter regions of RCAN1 and COX2 but
change): control, 1 % 0; ionomycin, 1.62 % 0.19; data not did not identify the sequence within %2.5 kb of the RGS2
shown]. promoter. Therefore, we investigated the role protein
To assess endogenous NFAT transcriptional activity, neosynthesis may play in OXT-induced gene expression.
myometrial cells were transduced with a NFAT response Human myometrial cells were pretreated with the protein
element luciferase reporter (NFAT-RE-Luc). Cells were synthesis inhibitor CX before stimulation with OXT. CX
then stimulated with a sustained concentration of OXT alone did not influence RGS2 transcription but had a
for 6 h, which elicited a concentration- and time-depen- tendency to increase transcription of RCAN1 and COX2.
1750 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

phospholipase C-coupled GPCR, to in-


crease [Ca2!]i and activate Ca2! effec-
tor proteins, such as myosin light chain
kinase, to initiate myometrial contrac-
tions. In this study, we are interested in
the potential for other Ca2! effector
proteins (i.e. transcription factors such
as NFAT) to mediate longer-time ef-
fects of OXT on gene expression. We
focused on NFAT for two reasons:
first, NFAT is a Ca2!-dependent tran-
scription factor that regulates gene ex-
pression in other smooth muscle cells
(6770); second, NFAT activation is
known to be dependent on stimulus
pulse frequency in other systems (51,
55, 56), and therefore, NFAT may de-
code OXT pulse frequency. In this re-
port, we have defined NFAT isoform
expression in myometrial cells by
qPCR, monitored NFAT activation by
the OXTR with a NFATC1-EFP re-
porter, assessed endogenous NFAT ac-
tivity with an NFATC1 primary anti-
body and NFAT-RE-Luc construct,
FIG. 5. NFATC1-EFP nuclear translocation during pulsatile OXT treatment. Human and finally, used qPCR to investigate
myometrial cells were transduced with a NFATC1-EFP adenovirus and stained with Hoechst putative NFAT and OXT target genes
nuclear stain before imaging. AC, Cells were stimulated with either sustained or 5-min within the myometrium.
pulses of OXT at the indicated concentration (molar) (A) or at 10'7 M (B and C) and indicated
time points, followed by washes (five times) (gray rectangle). D, Histogram of area under the Myometrial cells in our cell culture
curves of NFATC1-EFP traces in B and C. Ctrl, Control; PF, pulse frequency. Live cell image system were characterized using pri-
acquisition and analysis were performed as described in Materials and Methods. Data shown mary antibodies raised toward the
are NFATC1-EFP intensity (background subtracted) N:C ratio, mean % SE, from at least three
separate experiments and independent donors. In A, the y-axis is offset for clarity so the
smooth muscle cell markers desmin,
traces are clearly distinguishable, the amplitude of the offset is as follows: 10'9, !0.5 N:C; actin, and calponin. Over 90% of all
10'8, !1 N:C; 10'7, !1.5 N:C. Statistical analysis was by two-way ANOVA with Bonferroni myometrial cells were positive for
post hoc test, which demonstrated that OXT concentration was a significant source of
desmin, actin, and calponin, indicating
variation at P $ 0.05 (A), and one-way ANOVA with Dunnetts post hoc test compared with
the untreated control: *, P $ 0.05; ** P $ 0.01 (D). that cell dispersion from myometrial
biopsies yielded a 90% pure myocyte
Joint stimulation with OXT and CX inhibited OXT-in- population. In addition, myometrial
duced RGS2 transcription but amplified the OXT-in- cells in our cell culture system have previously demon-
2!
duced transcriptional readouts for RCAN1 and COX2 strated robust phospholipase C and Ca signals in re-
(Fig. 9). These data indicate initiation of RGS2 transcrip- sponse to OXT (19, 57), thus indicating they retain their
tion is dependent on protein neosynthesis, whereas induc- functional responsiveness in culture and are a good model
tion of RCAN1 and COX2 transcription is protein neo- for investigating OXT-stimulated NFAT activation.
synthesis independent. This is the first description of NFAT isoforms in the
myometrium, defined by qPCR. mRNA for all known
NFAT isoforms (NFATC1C4 and -5) were expressed, in
Discussion contrast with arterial and ileal smooth muscle, which ex-
press only NFATC1, -C3, and -C4 (70) and NFATC3 and
OXT is a neuropeptide hormone that stimulates contrac- -C4 (64), respectively, indicating NFAT isoform expres-
tion of the pregnant uterus; it is involved in the augmen- sion is tissue specific.
tation and possibly the initiation of parturition and uter- Expression of NFAT isoforms in the myometrium led
otonic activity after delivery. OXT acts via the OXTR, a us to study the effect of Ca2! on NFAT activation by
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1751

stimulated NFATC1-EFP nuclear lo-


calization with similar response kinet-
ics (data not shown), demonstrating
that translocation of NFATC1-EFP is
dependent on Ca2! entry as opposed to
other aspects of OXTR signaling.
GPCR activation and ionomycin have
previously been shown to stimulate nu-
clear translocation of NFATC1-EFP in
gonadotrophs and HeLa cells with sim-
ilar kinetics (51). However, in arteries
stimulated with endothelin NFATC3,
nuclear translocation is slower in onset
than in the myometrium, and nuclear
localization is not sustained (70),
whereas in the ileum, ionomycin failed
to induce NFATC3 nuclear transloca-
tion (64), indicating that NFAT activa-
tion is likely to be isoform, tissue, and
stimulus dependent.
FIG. 6. OXT-induced nuclear translocation of endogenous NFATC1. Human myometrial cells OXT-induced nuclear translocation
were stimulated with OXT (10'6 M) for 30 min or pretreated with CyclA (10'6 M) for 30 min
before stimulation with OXT. After stimulation, the reaction was terminated with ice-cold PBS
of NFATC1-EFP is mediated through
and fixed in 4% PFA. Immunocytochemistry was performed as described in Materials and the OXTR and calcineurin, as demon-
Methods. Images show myometrial cells stained with NFATC1 primary antibody. The control strated by inhibition of NFAT activity
(Ctrl) group was not stimulated with OXT. Images were acquired and analyzed as described in
by the OXT antagonists atosiban and
Materials and Methods. Images shown are representative of 2500 3000 cells. Scale bar, 60
#m. Data shown are NFATC1 N:C fold change over control group, mean % SE, from at least L368899 and the calcineurin inhibitors
three separate experiments and independent donors. Statistical analysis was by one-way CyclA and FK506. Furthermore, re-
ANOVA with Dunnetts post hoc test compared with the untreated control. *, P $ 0.01 moval of extracellular Ca2! and the
use of the [Ca2!]i chelator BAPTA-AM
using NFATC1-EFP, because translocation of this re- both abolished the NFATC1-EFP response, indicating
porter from the cytoplasm to the nucleus can provide a that Ca2! influx into the cell and elevation of [Ca2!]i is
live cell readout for NFAT activation (51), as opposed to necessary for the initiation of NFATC1-EFP nuclear
immunocytochemistry for endogenous NFAT, which re- translocation.
quires cell fixation. First, we demonstrated that OXT Because prolonged exposure of OXT to myometrial
2! 2!
stimulates accumulation of [Ca ]i using a fluo4 Ca cells induced sustained nuclear localization of NFATC1-
indicator with an EC50 in the low nanomolar range as EFP, we investigated whether NFATC1-EFP nuclear
previously described (19). The fluo4 response peaked translocation is dependent on constant receptor occu-
faster with high concentrations of OXT than at lower pancy or whether the initial binding of OXT to its recep-
concentrations; this delay could simply reflect the fact tor is sufficient to generate a sustained NFATC1-EFP re-
that augmenting the concentration of OXT increases the sponse. We found that washing off OXT after stimulation
rate at which OXT-sensitive intracellular stores are de- resulted in nuclear export of NFATC1-EFP, demonstrat-
pleted and Ca2! mobilized. ing that nuclear localization of NFATC1-EFP is revers-
To assess whether mobilization of Ca2! by activation ible. This is in concordance with previous work in other
of the OXTR induces an NFAT response myometrial cells systems that found sustained GPCR occupancy, and
were transduced with a NFATC1-EFP and stimulated [Ca2!]i elevation is required to maintain NFATC1-EFP
with OXT. We found that OXT stimulated NFATC1- localization in the nucleus (51, 55, 56).
EFP translocation to the nucleus with an EC50, similar to To assess the physiological importance of stimulus
2!
OXT-induced accumulation of [Ca ]i. NFATC1-EFP pulse frequency on NFATC1-EFP nuclear translocation,
translocation is far slower in onset than Ca2! mobiliza- we studied the effect of pulses of OXT on the NFATC1-
tion (within minutes rather than seconds), suggesting EFP N:C. We found that NFATC1-EFP N:C mirrored
there is a rate-limiting step between Ca2! release and OXT pulse frequency in a concentration-dependent man-
nuclear translocation of NFATC1-EFP. Ionomycin also ner, demonstrating a cumulative, sawtooth effect. This is
1752 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

and has not yet been tested in regard to OXT-induced


gene expression). Moreover it costs the cell less energy to
maintain high [Ca2!]i levels for repeated short intervals
than for a constant time period. Interestingly, pulsatile
administration of OXT is more efficient than constant
infusion for the induction/augmentation of labor in
women (8, 71).
OXT-stimulated nuclear translocation of the
NFATC1-EFP reporter construct led us to investigate the
role of OXT in inducing endogenous NFAT nuclear
translocation and transcriptional activity using a
NFATC1 primary antibody and NFAT-RE-Luc reporter,
respectively. OXT stimulated endogenous NFATC1 nu-
clear translocation with the effect being mediated by cal-
cineurin, indicating NFATC1-EFP nuclear translocation
can be extended to an endogenous mechanism. OXT also
induced a time- and concentration-dependent increase in
NFAT-RE-Luc activity, demonstrating that activation of
NFAT by OXT in myometrial cells results in a transcrip-
tional output.
We have also investigated whether OXT induces gene
expression through the calcineurin-NFAT pathway. We
have shown for the first time that OXT stimulates the
expression of the calcineurin inhibitory factor RCAN1 in
FIG. 7. OXT induces transcriptional activity of endogenous NFAT. the myometrium. Moreover, we have confirmed that
Human myometrial cells were transduced with Ad-NFAT-RE-Luc and
Ad-!-Gal before sustained stimulation with OXT (10'6 M) for the
OXT induces expression of RGS2 (a regulator of G pro-
indicated time points (A) or in a concentration-dependent manner for tein signaling) and COX2 (an enzyme involved in pros-
6 h (B). The control (Ctrl) group was not stimulated with OXT. After taglandin synthesis) as previously shown in myometrial
stimulation, the reaction was terminated with ice-cold PBS before cell
cells (65, 66). The calcineurin inhibitor CyclA attenuated
lysis. Data shown are luciferase activity in luminescence units,
standardized to !-Gal activity, and normalized to control. Mean % SE is OXT-induced expression of all three genes, demonstrat-
displayed for at least three separate experiments and independent ing the importance of the calcineurin-NFAT signaling
donors. Statistical analyses were performed by one-way ANOVA with pathway in mediating OXT-induced gene expression in
Dunnetts post hoc test compared with time zero (A) or the untreated
control (B). *, P $ 0.05; **, P $ 0.01; ***, P $ 0.001. the myometrium. The physiological significance of OXT-
induced up-regulation of RGS2 and RCAN1 in myome-
in concordance with previous work that shows NFATC1- trial cells is not established, but its likely to be important
EFP nuclear translocation in response to pulsatile GnRH for the negative regulation of OXT signaling; RGS2 un-
(51). OXT is released in pulses in vivo; therefore, it is couples G protein-coupled receptor subunits (72), there-
important that NFATC1-EFP is responsive to a pulsatile fore controlling OXT signaling at the level of the OXTR.
OXT system, which highlights the physiological rele- RCAN1 inhibits calcineurin activity (73), thus inhibiting
vance of NFATC1-EFP N:C being dependent on OXT NFAT-induced gene transcription in response to OXT.
pulse frequency. Pulsatile systems are important for pre- On the other hand, the induction of COX2 by OXT
venting desensitization, signaling efficiency, and encod- would result in production of prostaglandins and ampli-
ing information. Within our system, there was no reduc- fication of the parturition cascade (66). Prostaglandins
tion in the amplitude of the NFATC1-EFP N:C ratio with are uterotonic agonists that could work in synergy with
each pulse, indicating that NFATC1-EFP nuclear trans- OXT to induce myometrial contractions at term; more-
location is not desensitized within the conditions studied. over, they participate in cervical ripening, which would
Furthermore, the amplitude of the NFATC1-EFP N:C facilitate a successful vaginal delivery (74).
ratio with pulsatile OXT stimulation over 4 h was similar Although the increase in RGS2 was found to be medi-
to that with sustained treatment, suggesting a pulsatile ated by calcineurin-NFAT, the absence of the NFAT con-
system is a more efficient mechanism of OXT-stimulated sensus-binding motif in the RGS2 promoter suggests that
NFATC1-EFP nuclear translocation than a sustained NFAT itself is not directly responsible for enhancing
stimulus (although this observation cannot be extended RGS2 gene expression, and there must be another inter-
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1753

FIG. 8. OXT induces gene expression via a calcineurin-NFAT-dependent pathway. Human myometrial cells were stimulated with sustained OXT
(10'7 M) for the indicated time points (A) or pretreated with L368899 (10'5 M) or CyclA (10'6 M) for 30 min before treatment with sustained OXT
(10'7 M) for 4 h (B). The control (Ctrl) group was not stimulated with OXT. After stimulation, the reaction was terminated with ice-cold PBS before
RNA extraction and cDNA conversion. Real-time PCR was performed on cDNA to quantify mRNA of RGS2, RCAN1, and COX2. Data shown are
relative mRNA expression calculated by the Pfaffl method of analysis, standardized to the housekeeping genes 18S and RNA polymerase II and
normalized to time zero (A) or the untreated control (B). Mean % SE is displayed for at least three separate experiments from independent donors.
Statistical analyses were performed by one-way ANOVA with Dunnetts post hoc test compared with time zero or untreated control. *, P $ 0.05;
**, P $ 0.01.

mediary factor. To test this, we pretreated cells with CX Because the calcineurin inhibitor CyclA inhibited Oxt in-
(an inhibitor of protein synthesis) before stimulation with duced RGS2 gene expression independently, it is possible
OXT. CX was found to completely inhibit OXT-induced that in OXT-stimulated cells NFAT participates in the tran-
RGS2 gene transcription, suggesting that protein neosyn- scription of another transcription factor that, when trans-
thesis is necessary for this mechanism of OXT action. lated into active protein, in turn induces RGS2 expression.

FIG. 9. The effect of inhibiting protein neosynthesis on OXT-induced gene expression. Human myometrial cells were stimulated with sustained
OXT (10'7 M) for 4 h, with or without pretreatment with CX (50 #M) for 30 min. After stimulation, the reaction was terminated with ice-cold PBS
before RNA extraction and cDNA conversion. Real-time PCR was performed on cDNA to quantify mRNA of RGS2, RCAN1, and COX2. Data shown
are relative mRNA expression calculated by the Pfaffl method of analysis, standardized to the housekeeping genes 18S and RNA polymerase II and
normalized to the untreated control. Mean % SE is displayed for at least three separate experiments from independent donors. Statistical analyses
by two-way ANOVA revealed OXT and CX to be significant sources of variation: RGS2, P $ 0.05 (OXT and CX); RCAN1, P $ 0.001 (OXT) and P $
0.01 (CX); COX2, P $ 0.001 (OXT and CX). The OXT-CX interaction was also found to be significant: RGS2, P $ 0.05; RCAN1, P $ 0.05; COX2,
P $ 0.01. Post hoc Bonferroni tests revealed statistical differences between treatment and control groups. *, P $ 0.05; **, P $ 0.01; ***, P $
0.001.
1754 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

In contrast, we have shown that CX does not inhibit preliminary evidence for translation-dependent negative
OXT-induced transcription of RCAN1 and COX2, indi- feedback regulation of OXTR-mediated NFAT signaling.
cating this mechanism of OXT action is protein synthesis The data provide novel insights into the role of OXT in
independent. Instead, CX amplifies the effect of OXT uterine function.
stimulation. A similar increase in transcription has also
been reported in fibroblasts pretreated with CX before
stimulation with TNF$ (75). Protein neosynthesis-depen- Acknowledgments
dent negative feedback loops are often found in signaling
cascades; for example, ERK-mediated expression of dual- We thank Alison Kirby and staff at St. Michaels Hospital, Bris-
tol, UK, for the collection of myometrial tissue, Professor James
specificity phosphatases (52, 76) and NF"B-mediated ex-
Uney for kindly donating the !-galactosidase adenovirus, and
pression of nuclear factor of " light polypeptide gene en-
Dr. Chris Helps for assistance in analysis of qPCR data.
hancer in B-cells inhibitor $ (75), where the phosphatases
and I"B$ negatively regulate ERK and NF"B activity, Address all correspondence and requests for reprints to: Ja-
respectively. The same could be true of NFAT-mediated son N. A. Pont, Bristol University, School of Clinical Sciences,
expression of RCAN1 and COX2, where CX blocks pro- Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY,
tein synthesis, removing putative negative regulators of United Kingdom. E-mail: jp5620@bris.ac.uk.
This work was supported by the BBSRC (Grant SG1525),
NFAT signaling, thus increasing the transcriptional re-
Wellcome Trust (Grant 083902/Z), and Action Medical Re-
sponse of NFAT target genes. These hypotheses need to search (Grant SP4612).
be tested in additional experiments. Disclosure Summary: The authors of this paper have nothing
We have identified a novel signaling pathway within to declare.
the myometrium, whereby OXT initiates NFAT signal-
ing, inducing transcriptional activity. These findings are
likely to be of physiological significance in the context of References
parturition. Pregnant mice administered with the cal-
1. Beck S, Wojdyla D, Say L, Betran AP, Merialdi M, Requejo JH,
cineurin inhibitor FK506 experienced delayed labor (33);
Rubens C, Menon R, Van Look PF 2010 The worldwide incidence
thus, it is possible that OXT, through induction of the of preterm birth: a systematic review of maternal mortality and
calcineurin-NFAT pathway, is responsible for the regula- morbidity. Bull World Health Organ 88:3138
tion of the initiation of labor. The transcriptional activity 2. Rush RW, Keirse MJ, Howat P, Baum JD, Anderson AB, Turnbull
AC 1976 Contribution of preterm delivery to perinatal mortality.
stimulated by OXT in the myometrium could produce Br Med J 2:965968
factors involved in the initiation and maintenance of uter- 3. Goldenberg RL, Culhane JF, Iams JD, Romero R 2008 Epidemiol-
ine contraction, i.e. COX2 through the synthesis of pros- ogy and causes of preterm birth. Lancet 371:75 84
4. Moutquin JM 2003 Classification and heterogeneity of preterm
taglandins, or regulatory factors such as RCAN1 and
birth. BJOG 110(Suppl 20):30 33
RGS2. Identification of a wider array of OXT- and cal- 5. Lpez Bernal A, Europe-Finner GN, Phaneuf S, Watson SP 1995
cineurin-NFAT-responsive genes in the myometrium will Preterm labour: a pharmacological challenge. Trends Pharmacol Sci
provide insight into how parturition is initiated, enabling 16:129 133
6. Muglia LJ, Katz M 2010 The enigma of spontaneous preterm birth.
development of therapies to prevent spontaneous prema- N Engl J Med 362:529 535
ture labor. 7. Fuchs AR, Romero R, Keefe D, Parra M, Oyarzun E, Behnke E
In summary, we have shown that human myometrial 1991 Oxytocin secretion and human parturition: pulse frequency
cells express the four Ca2!/calcineurin-sensitive NFAT and duration increase during spontaneous labor in women. Am J
Obstet Gynecol 165:15151523
isoforms (NFATC1C4) as well as the Ca2!/calcineurin- 8. Tribe RM, Crawshaw SE, Seed P, Shennan AH, Baker PN 2012
insensitive NFAT5. We demonstrate for the first time that Pulsatile versus continuous administration of oxytocin for induc-
the OXTR mediates translocation of a NFATC1-EFP re- tion and augmentation of labor: two randomized controlled trials.
Am J Obstetrics Gynecol 206:230 e231238
porter from the cytoplasm to the nucleus and that this
9. Turnbull AC, Anderson AB 1968 Induction of labour. II. Intrave-
mechanism is mediated by calcineurin. We also show how nous oxytocin infusion. J Obstetrics Gynaecol Brit Commonwealth
NFATC1-EFP translocation kinetics can make signaling 75:24 31
more efficient with a pulsatile OXT signal in contrast to 10. Nishimori K, Young LJ, Guo Q, Wang Z, Insel TR, Matzuk MM
1996 Oxytocin is required for nursing but is not essential for par-
sustained stimulation. Furthermore, we show that OXT turition or reproductive behavior. Proc Natl Acad Sci USA 93:
induces nuclear translocation of endogenous NFATC1 11699 11704
and stimulates endogenous NFAT-mediated activation of 11. Young 3rd WS, Shepard E, Amico J, Hennighausen L, Wagner KU,
LaMarca ME, McKinney C, Ginns EI 1996 Deficiency in mouse
a NFAT-Luc reporter. Moreover, we identify OXT tran-
oxytocin prevents milk ejection, but not fertility or parturition.
scriptional induction of three calcineurin-NFAT-sensitive J Neuroendocrinol 8:847 853
genes (RGS2, RCAN1, and COX2) as well as providing 12. Kimura T, Takemura M, Nomura S, Nobunaga T, Kubota Y, Inoue
Mol Endocrinol, October 2012, 26(10):17431756 mend.endojournals.org 1755

T, Hashimoto K, Kumazawa I, Ito Y, Ohashi K, Koyama M, Azuma 2009 Spatial and temporal regulation of coronary vessel formation
C, Kitamura Y, Saji F 1996 Expression of oxytocin receptor in by calcineurin-NFAT signaling. Development 136:33353345
human pregnant myometrium. Endocrinology 137:780 785 33. Tabata C, Ogita K, Sato K, Nakamura H, Qing Z, Negoro H,
13. Phaneuf S, Rodrguez Liares B, TambyRaja RL, MacKenzie IZ, Kumasawa K, Temma-Asano K, Tsutsui T, Nishimori K, Kimura T
Lpez Bernal A 2000 Loss of myometrial oxytocin receptors during 2009 Calcineurin/NFAT pathway: a novel regulator of parturition.
oxytocin-induced and oxytocin-augmented labour. J Reprod Fertil Am J Reprod Immunol 62:44 50
120:9197 34. McCaffrey PG, Luo C, Kerppola TK, Jain J, Badalian TM, Ho AM,
14. Kuwabara Y, Takeda S, Mizuno M, Sakamoto S 1987 Oxytocin Burgeon E, Lane WS, Lambert JN, Curran T, et al 1993 Isolation of
levels in maternal and fetal plasma, amniotic fluid, and neonatal the cyclosporin-sensitive T cell transcription factor NFATp. Science
plasma and urine. Arch Gynecol Obstet 241:1323 262:750 754
15. Blanks AM, Vatish M, Allen MJ, Ladds G, de Wit NC, Slater DM, 35. Northrop JP, Ho SN, Chen L, Thomas DJ, Timmerman LA, Nolan
Thornton S 2003 Paracrine oxytocin and estradiol demonstrate a GP, Admon A, Crabtree GR 1994 NF-AT components define a
spatial increase in human intrauterine tissues with labor. J Clin family of transcription factors targeted in T-cell activation. Nature
Endocrinol Metab 88:33923400 369:497502
16. Chibbar R, Miller FD, Mitchell BF 1993 Synthesis of oxytocin in 36. Hoey T, Sun YL, Williamson K, Xu X 1995 Isolation of two new
amnion, chorion, and decidua may influence the timing of human members of the NF-AT gene family and functional characterization
parturition. J Clin Invest 91:185192 of the NF-AT proteins. Immunity 2:461 472
17. Chibbar R, Wong S, Miller FD, Mitchell BF 1995 Estrogen stimu- 37. Ho SN, Thomas DJ, Timmerman LA, Li X, Francke U, Crabtree
lates oxytocin gene expression in human chorio-decidua. J Clin GR 1995 NFATc3, a lymphoid-specific NFATc family member that
Endocrinol Metab 80:567572 is calcium-regulated and exhibits distinct DNA binding specificity.
18. Phaneuf S, Europe-Finner GN, Carrasco MP, Hamilton CH, Lpez J Biol Chem 270:19898 19907
Bernal A 1995 Oxytocin signaling in human myometrium. Adv Exp 38. Masuda ES, Naito Y, Tokumitsu H, Campbell D, Saito F, Hannum
Med Biol 395:453 467 C, Arai K, Arai N 1995 NFATx, a novel member of the nuclear
19. Phaneuf S, Carrasco MP, Europe-Finner GN, Hamilton CH, Lpez factor of activated T cells family that is expressed predominantly in
Bernal A 1996 Multiple G proteins and phospholipase C isoforms the thymus. Mol Cell Biol 15:26972706
in human myometrial cells: implication for oxytocin action. J Clin 39. Lopez-Rodrguez C, Aramburu J, Rakeman AS, Rao A 1999
Endocrinol Metab 81:2098 2103
NFAT5, a constitutively nuclear NFAT protein that does not coop-
20. Arthur P, Taggart MJ, Mitchell BF 2007 Oxytocin and parturition:
erate with Fos and Jun. Proc Natl Acad Sci USA 96:7214 7219
a role for increased myometrial calcium and calcium sensitization?
40. Miyakawa H, Woo SK, Dahl SC, Handler JS, Kwon HM 1999
Front Biosci 12:619 633
Tonicity-responsive enhancer binding protein, a rel-like protein
21. Lpez Bernal A, Rivera J, Europe-Finner GN, Phaneuf S, Asbth G
that stimulates transcription in response to hypertonicity. Proc Natl
1995 Parturition: activation of stimulatory pathways or loss of
Acad Sci USA 96:2538 2542
uterine quiescence? Adv Exp Med Biol 395:435 451
41. Rao A, Luo C, Hogan PG 1997 Transcription factors of the NFAT
22. Word RA, Stull JT, Casey ML, Kamm KE 1993 Contractile ele-
family: regulation and function. Annu Rev Immunol 15:707747
ments and myosin light chain phosphorylation in myometrial tissue
42. Okamura H, Aramburu J, Garca-Rodrguez C, Viola JP, Raghavan
from nonpregnant and pregnant women. J Clin Invest 92:29 37
A, Tahiliani M, Zhang X, Qin J, Hogan PG, Rao A 2000 Concerted
23. Shaw JP, Utz PJ, Durand DB, Toole JJ, Emmel EA, Crabtree GR
dephosphorylation of the transcription factor NFAT1 induces a
1988 Identification of a putative regulator of early T cell activation
conformational switch that regulates transcriptional activity. Mo-
genes. Science 241:202205
lecular cell 6:539 550
24. Musar A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N 1999
IGF-1 induces skeletal myocyte hypertrophy through calcineurin in 43. Shibasaki F, Price ER, Milan D, McKeon F 1996 Role of kinases
association with GATA-2 and NF-ATc1. Nature 400:581585 and the phosphatase calcineurin in the nuclear shuttling of tran-
25. Bai S, Kerppola TK 2011 Opposing roles of FoxP1 and Nfat3 in scription factor NF-AT4. Nature 382:370 373
transcriptional control of cardiomyocyte hypertrophy. Mol Cell 44. Akazawa H, Komuro I 2003 Roles of cardiac transcription factors
Biol 31:3068 3080 in cardiac hypertrophy. Circ Res 92:1079 1088
26. Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Rob- 45. Macin F, Lpez-Rodrguez C, Rao A 2001 Partners in transcrip-
bins J, Grant SR, Olson EN 1998 A calcineurin-dependent tran- tion: NFAT and AP-1. Oncogene 20:2476 2489
scriptional pathway for cardiac hypertrophy. Cell 93:215228 46. Chow CW, Rincn M, Cavanagh J, Dickens M, Davis RJ 1997
27. Delling U, Tureckova J, Lim HW, De Windt LJ, Rotwein P, Molk- Nuclear accumulation of NFAT4 opposed by the JNK signal trans-
entin JD 2000 A calcineurin-NFATc3-dependent pathway regu- duction pathway. Science 278:1638 1641
lates skeletal muscle differentiation and slow myosin heavy-chain 47. Gomez MF, Bosc LV, Stevenson AS, Wilkerson MK, Hill-Eubanks
expression. Mol Cell Biol 20:6600 6611 DC, Nelson MT 2003 Constitutively elevated nuclear export activ-
28. Horsley V, Friday BB, Matteson S, Kegley KM, Gephart J, Pavlath ity opposes Ca2!-dependent NFATc3 nuclear accumulation in vas-
GK 2001 Regulation of the growth of multinucleated muscle cells cular smooth muscle: role of JNK2 and Crm-1. J Biol Chem 278:
by an NFATC2-dependent pathway. J Cell Biol 153:329 338 46847 46853
29. Ranger AM, Grusby MJ, Hodge MR, Gravallese EM, de la Brousse 48. Rinne A, Banach K, Blatter LA 2009 Regulation of nuclear factor of
FC, Hoey T, Mickanin C, Baldwin HS, Glimcher LH 1998 The activated T cells (NFAT) in vascular endothelial cells. J Mol Cell
transcription factor NF-ATc is essential for cardiac valve forma- Cardiol 47:400 410
tion. Nature 392:186 190 49. Kehlenbach RH, Dickmanns A, Gerace L 1998 Nucleocytoplasmic
30. Graef IA, Chen F, Chen L, Kuo A, Crabtree GR 2001 Signals trans- shuttling factors including Ran and CRM1 mediate nuclear export
duced by Ca2!/calcineurin and NFATc3/c4 pattern the developing of NFAT In vitro. J Cell Biol 141:863 874
vasculature. Cell 105:863 875 50. Wathes DC, Lamming GE 1995 The oxytocin receptor, luteolysis
31. Guo X, Zhou C, Sun N 2011 The neuropeptide catestatin promotes and the maintenance of pregnancy. J Reprod Fertil Supplement
vascular smooth muscle cell proliferation through the Ca2!-cal- 49:53 67
cineurin-NFAT signaling pathway. Biochem Biophys Res Commun 51. Armstrong SP, Caunt CJ, Fowkes RC, Tsaneva-Atanasova K,
407:807 812 McArdle CA 2009 Pulsatile and sustained gonadotropin-releasing
32. Zeini M, Hang CT, Lehrer-Graiwer J, Dao T, Zhou B, Chang CP hormone (GnRH) receptor signaling: does the Ca2!/NFAT signal-
1756 Pont et al. Oxytocin-Induced NFAT Activation Mol Endocrinol, October 2012, 26(10):17431756

ing pathway decode GnRH pulse frequency? J Biol Chem 284: NFAT4 movement in native smooth muscle. A role for differential
35746 35757 Ca2! signaling. J Biol Chem 276:15018 15024
52. Armstrong SP, Caunt CJ, Fowkes RC, Tsaneva-Atanasova K, 65. Park ES, Echetebu CO, Soloff S, Soloff MS 2002 Oxytocin stimu-
McArdle CA 2010 Pulsatile and sustained gonadotropin-releasing lation of RGS2 mRNA expression in cultured human myometrial
hormone (GnRH) receptor signaling: does the ERK signaling path- cells. Am J Physiol Endocrinol Metab 282:E580 E584
way decode GnRH pulse frequency? J Biol Chem 285:24360 66. Molnr M, Rig Jr J, Romero R, Hertelendy F 1999 Oxytocin
24371 activates mitogen-activated protein kinase and up-regulates cyclo-
53. Smith MP, Ayad VJ, Mundell SJ, McArdle CA, Kelly E, Lpez oxygenase-2 and prostaglandin production in human myometrial
Bernal A 2006 Internalization and desensitization of the oxytocin cells. Am J Obstet Gynecol 181:42 49
receptor is inhibited by Dynamin and clathrin mutants in human 67. Lee MY, Garvey SM, Baras AS, Lemmon JA, Gomez MF, Schoppee
embryonic kidney 293 cells. Mol Endocrinol 20:379 388 Bortz PD, Daum G, LeBoeuf RC, Wamhoff BR 2010 Integrative
54. Sundseth SS, Alberta JA, Waxman DJ 1992 Sex-specific, growth genomics identifies DSCR1 (RCAN1) as a novel NFAT-dependent
hormone-regulated transcription of the cytochrome P450 2C11 mediator of phenotypic modulation in vascular smooth muscle
and 2C12 genes. J Biol Chem 267:39073914 cells. Hum Mol Genet 19:468 479
55. Dolmetsch RE, Lewis RS, Goodnow CC, Healy JI 1997 Differential 68. Rosenkranz AC, Rauch BH, Freidel K, Schrr K 2009 Regulation of
activation of transcription factors induced by Ca2! response am- protease-activated receptor-1 by vasodilatory prostaglandins via
plitude and duration. Nature 386:855 858 NFAT. Cardiovasc Res 83:778 784
69. Morales S, Diez A, Puyet A, Camello PJ, Camello-Almaraz C, Bau-
56. Tomida T, Hirose K, Takizawa A, Shibasaki F, Iino M 2003 NFAT
tista JM, Pozo MJ 2007 Calcium controls smooth muscle TRPC
functions as a working memory of Ca2! signals in decoding Ca2!
gene transcription via the CaMK/calcineurin-dependent pathways.
oscillation. EMBO J 22:38253832
Am J Physiol Cell Physiol 292:C553C563
57. Phaneuf S, Europe-Finner GN, Varney M, MacKenzie IZ, Watson
70. Nilsson LM, Sun ZW, Nilsson J, Nordstrm I, Chen YW, Molken-
SP, Lpez Bernal A 1993 Oxytocin-stimulated phosphoinositide
tin JD, Wide-Swensson D, Hellstrand P, Lydrup ML, Gomez MF
hydrolysis in human myometrial cells: involvement of pertussis tox-
2007 Novel blocker of NFAT activation inhibits IL-6 production in
in-sensitive and -insensitive G-proteins. J Endocrinol 136:497509
human myometrial arteries and reduces vascular smooth muscle
58. Caunt CJ, Armstrong SP, Rivers CA, Norman MR, McArdle CA
cell proliferation. Am J Physiol Cell Physiol 292:C1167C1178
2008 Spatiotemporal regulation of ERK2 by dual specificity phos- 71. Dawood MY 1995 Novel approach to oxytocin induction-augmen-
phatases. J Biol Chem 283:2661226623 tation of labor. Application of oxytocin physiology during preg-
59. Pfaffl MW 2001 A new mathematical model for relative quantifi- nancy. Adv Exp Med Biol 395:585594
cation in real-time RT-PCR. Nucleic Acids Res 29:e45 72. Hepler JR 1999 Emerging roles for RGS proteins in cell signaling.
60. Peeters D, Peters IR, Helps CR, Gabriel A, Day MJ, Clercx C 2007 Trends Pharmacol Sci 20:376 382
Distinct tissue cytokine and chemokine mRNA expression in canine 73. Fuentes JJ, Genesc L, Kingsbury TJ, Cunningham KW, Prez-Riba
sino-nasal aspergillosis and idiopathic lymphoplasmacytic rhinitis. M, Estivill X, de la Luna S 2000 DSCR1, overexpressed in Down
Vet Immunol Immunopathol 117:95105 syndrome, is an inhibitor of calcineurin-mediated signaling path-
61. Bailey TL, Boden M, Buske FA, Frith M, Grant CE, Clementi L, ways. Hum Mol Genet 9:16811690
Ren J, Li WW, Noble WS 2009 MEME SUITE: tools for motif 74. Mackenzie IZ 2006 Induction of labour at the start of the new
discovery and searching. Nucleic Acids Res 37:W202W208 millennium. Reproduction 131:989 998
62. Grant CE, Bailey TL, Noble WS 2011 FIMO: scanning for occur- 75. Sung MH, Salvatore L, De Lorenzi R, Indrawan A, Pasparakis M,
rences of a given motif. Bioinformatics 27:10171018 Hager GL, Bianchi ME, Agresti A 2009 Sustained oscillations of
63. Gomez MF, Stevenson AS, Bonev AD, Hill-Eubanks DC, Nelson NF-"B produce distinct genome scanning and gene expression pro-
MT 2002 Opposing actions of inositol 1,4,5-trisphosphate and files. PloS one 4:e7163
ryanodine receptors on nuclear factor of activated T-cells regula- 76. Zhang T, Roberson MS 2006 Role of MAP kinase phosphatases in
tion in smooth muscle. J Biol Chem 277:37756 37764 GnRH-dependent activation of MAP kinases. J Mol Endocrinol
64. Stevenson AS, Gomez MF, Hill-Eubanks DC, Nelson MT 2001 36:4150

You might also like