You are on page 1of 13

Cell Biochem Biophys (2014) 68:497–509

DOI 10.1007/s12013-013-9728-z

ORIGINAL PAPER

Delphinidin Activates NFAT and Induces IL-2 Production


Through SOCE in T Cells
Evelyn Jara • Marı́a A. Hidalgo • Juan L. Hancke •
Alejandra I. Hidalgo • Sebastian Brauchi •
Luisa Nuñez • Carlos Villalobos • Rafael A. Burgos

Published online: 13 August 2013


Ó Springer Science+Business Media New York 2013

Abstract Delphinidin is an anthocyanidin that possesses increasing cytokine production through CRAC channel and
antioxidant and anti-inflammatory effects; however, some NFAT activation.
reports suggest that delphinidin has pro-inflammatory
properties. For this reason, we assessed the effect of del- Keywords Delphinidin  Calcium  T cells  NFAT 
phinidin on cytokine production in T cells. We demon- Cytokines
strated that delphinidin increased the cytosolic-free Ca2?
concentration by releasing Ca2? from intracellular stores Abbreviations
and increasing Ca2? entry. The putative Ca2? release SOCE Store-operated calcium entry
activated Ca2? (CRAC) channel inhibitors BTP2 and BTP2 N-(4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-
gadolinium reduced the calcium entry stimulated by the yl]phenyl)-4-methyl-1,2,3-thiadiazole-5-
anthocyanidin. Delphinidin induced nuclear factor of acti- carboxamide
vated T cells (NFAT) translocation and NFAT-Luc activity CRAC Ca2? release-activated Ca2?
in Jurkat cells and was dependent on the CRAC channel
and calcineurin pathway. Delphinidin increased the mRNA
expression and production of IL-2 in Jurkat cells and was
inhibited by BTP2 and cyclosporine A. Using peripheral
blood lymphocytes, we demonstrated that delphinidin Introduction
increased the production of IL-2 and IFN-c and was
inhibited by BTP2. Taken together, our results suggest that Previous studies have reported that anthocyanidins, which
delphinidin exerts immunostimulatory effects on T cells by are commonly found in pigmented fruits and vegetables,
stimulate an immune response by augmenting pro-inflam-
Electronic supplementary material The online version of this
matory cytokine expression [1–5]. Delphinidin is one of the
article (doi:10.1007/s12013-013-9728-z) contains supplementary most important anthocyanidins found in berries and dark
material, which is available to authorized users. grapes [6]. It has been suggested to be beneficial to human
health in many ways, such as providing antioxidant and
E. Jara  M. A. Hidalgo  J. L. Hancke  A. I. Hidalgo 
R. A. Burgos (&) anticancer effects [7, 8]. In RAW 264.7 macrophages,
Institute of Pharmacology and Morphophysiology, Universidad delphinidin increased the TNFa production induced by
Austral de Chile, P.O. Box 567, Valdivia, Chile LPS/IFN-c and was the most potent of all anthocyanidins
e-mail: rburgos1@uach.cl tested, which included malvidin, peonidin, and pelargoni-
S. Brauchi din [9]. Despite these previous studies, other authors have
Institute of Physiology, Faculty of Medicine, Universidad noted that delphinidin exerts anti-inflammatory effects on
Austral de Chile, Valdivia, Chile Jurkat cells by suppressing NF-jB acetylation and inhib-
iting TNFa and IL-6 production when the Jurkat cells were
L. Nuñez  C. Villalobos
Spanish Research Council, Institute of Molecular Biology induced by LPS [10]. Thus, the effect of delphinidin on T
and Genetics, University of Valladolid, Valladolid, Spain cell cytokine production is not clear.

123
498 Cell Biochem Biophys (2014) 68:497–509

In T cells, activation through the T cell receptor (TCR) 125 mM. Each stock aliquot of delphinidin was frozen
complex leads to a rapid increase in the cytoplasmic Ca2? at -80 °C, and a vial was dissolved for each experiment
concentration ([Ca2?]i), which triggers signals that are using HEPES buffer. Fura-2/acetoxymethyl ester (AM) and
essential for T cell activation, proliferation, gene expres- anti-mouse Alexa 488 (#A11017) were purchased from
sion, cytokine production and differentiation [11]. Calcium Molecular Probes (InvitrogenTM, CA, USA). N-(4-[3,5-bis
signaling mechanisms include the inositol 1,4,5-trisphos- (trifluoromethyl)-1H-pyrazol-1-yl]phenyl)-4-methyl-1,2,3-
phate (IP3)-induced release of Ca2? from the endoplasmic thiadiazole-5-carboxamide (BTP2) was obtained from
reticulum (ER) and store-operated Ca2? entry (SOCE) Calbiochem (Darmstadt, Germany). Gadolinium (Gd3?),
through CRAC channels [12]. cyclosporine A, and HistochoiceTM were obtained from
SOCE-mediated Ca2? signals regulate the expression of Sigma Chemical Co. (St. Louis, MO, USA). RPMI 1640
cytokines that are critical for immune responses. Deriva- medium, penicillin, and fetal bovine serum (FBS) were
tives of 3, 5-bistrifluoromethyl pyrazole (BTP), such as purchased from Hyclone (Logan, UT, USA). IL-2 ELISA
BTP2, inhibit T cell SOCE-dependent calcium signaling kits were purchased from Biosource Europe S.A. (Niv-
and the subsequent cytokine production at the transcrip- elles, Belgium). A monoclonal antibody against NFATc1
tional level [13]. The control of IL-2 gene expression in T was obtained from BD Transduction Laboratories (San
cells is predominately linked to the nuclear factor of acti- Diego, CA, USA). Anti-mouse IgG-HRP was obtained from
vated T cells (NFAT), which is regulated in a dynamic Cell Signaling (Beverly, MA, USA). Lymphocyte Separa-
manner by Ca2? levels [14]. In unstimulated cells, NFAT is tion Medium was obtained from Mediatech (Mediatech,
located in the cytoplasm in a highly phosphorylated form. Inc., VA, USA). The RNeasyÒ Plus Mini Kit was obtained
An increase in intracellular Ca2? activates calcineurin from Qiagen, Inc. (CA, USA). The IL-2 and IFN-c BD
(Cn), which is a serine-threonine phosphatase that dephos- OptEIA TM Kits were obtained from BD Biosciences. All
phorylates NFAT and thus induces NFAT’s translocation other reagents and chemicals were purchased from Merck
into the nucleus [15, 16]. Once in the nucleus, NFAT binds (Darmstadt, Germany).
to specific DNA sequences to activate the transcription of
NFAT-dependent genes [17–19].
Jurkat E6-1 Cell Culture
Impaired Ca2? signaling in T cells is linked to several
inherited immunodeficiency diseases, such as severe
The Jurkat E6-1 cell line (ATCCÒ CRL-1573TM) was
combined immunodeficiency (SCID) [20, 21]. While SCID
cultured in RPMI 1640 medium supplemented with 10 %
patients were originally identified by their susceptibility to
FBS, 1 mM sodium pyruvate, and 2 mM L-glutamine at
recurrent infections, it was later revealed that these patients
37 °C in a humidified incubator with 5 % CO2.
were completely deficient in CRAC channel function
[20–23]. The lack of SOCE in these patients is associated
with severely compromised T cell proliferation and a Isolation of Peripheral Blood Lymphocytes
decreased production of cytokines, such as IL-2, IL-4,
IL-10, IFN-c, and tumor necrosis factor (TNFa); patients Lymphocytes were separated from blood samples obtained
also exhibited markedly impaired antibody responses to T from healthy volunteer donors using protocols approved by
cell-dependent antigens [22–25]. In addition, T cells from the Ethical Committee; specifically, lymphocytes were
SCID patients show a pronounced attenuation of NFAT separated by flotation in Lymphocyte Separation Medium
dephosphorylation and nuclear translocation in response to using the manufacturer’s protocol and then utilized in
TCR stimulation or treatment with pharmacological agents, ELISA assays.
such as ionomycin or thapsigargin; however, these cells
maintain intact AP-1 and NF-jB activations [26]. In this Dye Loading
study, we demonstrate that delphinidin activates NFAT and
induces cytokine production through increased levels of Jurkat E6-1 cells were suspended in HBSS at a concen-
SOCE in T cells. tration of 20 9 106 cells/mL and incubated with 1 lM
Fura-2/AM for 30 min at 37 °C. The cells were then
washed twice, divided into 2 9 106 cells/mL aliquots and
Materials and Methods maintained at 4 °C in the dark until use. Cells were pelleted
by centrifugation at 1,200 rpm for 6 min and resuspended
Reagents in 2 mL of HEPES buffer (20 mM HEPES, pH 7.2;
140 mM NaCl; 10 mM glucose; 1 mM KCl; 1 mM Ca2?
Delphinidin was purchased from Extrasynthese (France) chloride (CaCl2) and 1 mM MgCl2). In some experiments,
and dissolved in DMSO at a stock concentration of the cells were suspended in Ca2?-free HEPES buffer with

123
Cell Biochem Biophys (2014) 68:497–509 499

0.3 mM Tris-EGTA that was added 20 s prior to the experiment. After 100 s of recording, 50 lM delphinidin
experiment. Experiments involving cationic inhibitors (i.e., was added; at 300 s, a pulse of 1 mM CaCl2 was given and
Gd3?) or an influx of strontium ions (Sr2?) were performed registered for 150 s. The area under the 150 s curve
in Ca2?-free HEPES medium without EGTA to avoid the (AUC150) was determined. The Ca2? influx was recorded
chelating effect of these compounds. as the excitation ratio (340/380) measured at a 509 nm
emission using an LS55 thermoregulated spectrofluorime-
Fluorescence Imaging of Cytosolic Calcium ter (Perkin-Elmer, USA).

Experiments with Fura-2/AM-loaded Jurkat cells were per- Immunofluorescence


formed using an inverted fluorescence microscope (Zeiss
Axiovert S100 TV). Jurkat cells were grown on coverslips The nuclear distribution of NFAT was measured using
coated with fibronectin (20 lg/mL) and incubated with immunofluorescence. Jurkat E6-1 cells were preincubated
1 lM Fura-2/AM for 90 min at room temperature in the with 10 lM BTP2, 1 lg/mL cyclosporine A or a vehicle
dark. Images were captured using an OrcaER camera for 15 min and then stimulated with 50 lM delphinidin
(Hamamatsu Photonics, Hamamatsu, Japan). Finally, the for 30 min. Cells were centrifuged using a Cytospin for
records and images were analyzed using Aquacosmos 2.0 10 min at 2009g onto 2 mg/mL poly-L-lysine-coated
image analysis software. slides. The cells were next fixed with HistochoiceTM for
10 min at room temperature and then permeabilized in
The Measurement of Calcium (Ca2?) and Strontium 0.3 % Triton-X 100/PBS for 15 min. Nonspecific binding
(Sr2?) Influx was blocked by incubating the cells with PBS containing
1 % BSA and 5 % nonfat milk for 1 h at room temperature.
To assess SOCE, Fura-2/AM-loaded Jurkat E6-1 cells Cells were incubated with the NFATc1-specific monoclo-
(2 9 106 cells/mL) were suspended in Ca2?-free HEPES nal antibody (1:200 dilution) overnight, followed by an
buffer, and 0.3 mM EGTA was added to each cuvette 20 s incubation with an anti-mouse Alexa 488 secondary anti-
prior to the experiment. At 100 s, 50 lM delphinidin was body. Covered slides were analyzed using confocal fluo-
added, and after 200 s of recording, one pulse of 1 mM rescence microscopy (LSM5 PASCAL microscope, Zeiss,
Ca2? or Sr2? was administered and recorded for 400 s. To Germany). The nuclear NFAT signal intensity was regis-
investigate delphinidin-mediated SOCE activation, Fura-2/ tered using Image Pro Plus v4.5 (Media Cybernetics, Inc.,
AM-loaded Jurkat E6-1 cells were preincubated with Gd3? USA).
or BTP2 for 15 min, and then, the influx of Ca2? and Sr2?
was recorded as the 340:380 excitation ratio measured at a Analysis of NFAT Dephosphorylation by Immunoblot
509 nm emission using a LS55 thermoregulated spectro-
fluorimeter (Perkin-Elmer, MA, USA). For the detection of NFAT phosphorylation, Jurkat E6-1
cells (5 9 106) were deprived of serum for 4 h, preincu-
The Assessment of Putative SOCE Inhibitors bated with cyclosporine A (1 lg/ml) or vehicle for 30 min,
on Ca2? Influx and then stimulated with 50 lM of delphinidin. Whole cell
lysates (60 lg of protein) were separated using SDS/PAGE
To assess the effect of putative SOCE inhibitors, different with 8 % gels and probed with an anti-NFATc1 mono-
approaches were used. First, Fura-2/AM-loaded Jurkat E6- clonal antibody (BD Transduction); labeled proteins were
1 cells (2 9 106 cells/mL) were suspended in HEPES detected with an enhanced chemiluminescence system
buffer without Ca2?, and 0.3 mM EGTA was added to (Perkin-Elmer Life Science, Boston, MA, USA).
each cuvette 20 s prior to experiment initiation. After 100 s
of recording, 50 lM delphinidin was added. In the second Bioluminescence Imaging of NFAT-Luciferase
approach, Fura-2/AM-loaded Jurkat E6-1 cells (2 9 106 Reporter Activity in Individual Living Cells
cells/mL) were suspended in HEPES with Ca2? buffer and
preincubated with the inhibitors for 15 min. After 100 s, Jurkat F6 cells that stably expressed a luciferase reporter
50 lM delphinidin was added; the dye fluorescence exci- gene under the control of triplet NFAT response elements
tation ratio was recorded for 200 s; and the area under the [27] were placed on fibronectin-coated glass coverslips and
200 s curve (AUC200) was estimated. For the final subjected to time-lapse bioluminescence imaging to visu-
approach, Fura-2/AM-loaded Jurkat E6-1 cells (2 9 106 alize the dynamics of NFAT transcriptional activity in
cells/mL) were suspended in Ca2?-free HEPES buffer and living cells. Cells were introduced into a Zeiss incubator,
preincubated with the inhibitors for 15 min; then, 0.3 mM which was attached over an inverted Zeiss S100 TV
EGTA was added to each cuvette 20 s prior to the microscope, and subjected to bioluminescence imaging for

123
500 Cell Biochem Biophys (2014) 68:497–509

16 h. Photonic emissions reflecting NFAT transcriptional MTT Assay


activity were taken every 15 min using a Hamamatsu
photon counting camera. Specific photonic emissions were Jurkat cells (5 9 104 cells/mL) were incubated with a
calculated as raw photonic emissions minus background vehicle or delphinidin (10, 50 or 100 lM) for 24 h. Cell
emissions (i.e., photonic emissions captured over 15 min in proliferation was assessed using the mitochondria-depen-
a field devoid of cells). Ionomycin (500 nM) plus PMA dent reduction of MTT to formazan. The reduction of MTT
(100 nM), delphinidin (50 lM) or a vehicle treatment was to formazan was estimated by measuring OD570 values
added at time 0. with a microplate reader (Varioskan Flash Multimode
Reader, Thermo Scientific, USA).
Flow Cytometric Analysis of NFAT Translocation
in Jurkat Nuclei Preparations Statistical Analysis

For the detection of NFAT translocation by flow cytome- Results were analyzed using one-way analysis of variance
try, we used the protocols described by Blaecke et al. In (ANOVA) and Dunnett’s multiple comparisons test. A
brief, Jurkat E6-1 cells (1 9 106 cells/mL) were preincu- significance level of 5 % was used.
bated with 10 lM BTP2 or 1 lg/mL cyclosporine A for
30 min and then stimulated with 50 lM delphinidin for
30 min. After the incubation period, the cells were washed Results
twice with PBS.
Nuclear preparations were achieved by incubating the cells Delphinidin Mobilizes Calcium in Jurkat Cells
in 200 ll of Pipes-Triton solution, as described previously
[28]. After two washes in PBS, the nuclei were stained with Following TCR activation, increase in intracellular calcium
mouse anti-NFATc1 antibodies (5 lg/mL) for 30 min at leads to the activation of PLCc1, increased levels of inositol-
4 °C. The nuclei were washed twice with PBS, incubated for 1,4,5-trisphosphate (InsP3), and the binding of InsP3 to the
30 min at 4 °C with anti-mouse Alexa 488 (1:200 dilution) InsP3 receptor (InsP3R), thereby inducing the release of Ca2?
and finally washed twice with PBS. The nuclei were assessed from the ER. The decreased Ca2? concentration in the ER
by flow cytometry on a FACSCanto II (Becton–Dickinson); triggers the activation of the ‘‘capacitative,’’ or SOCE, path-
the results were analyzed using the FlowJo v7.6 software. way, resulting in the opening of the store-operated Ca2?
channels in the plasma membrane, which are responsible for
qRT-PCR the inward calcium current (Icrac). Because the Ca2? from the
ER and the extracellular calcium influx are two sources of this
Total RNA was isolated from Jurkat E6-1 cells using the cation, which are associated with T cell activation [11], we
RNeasyÒ Plus Mini Kit according to the manufacturer’s assessed the effect of delphinidin on calcium mobilization by
instructions. Approximately 2 lg of total RNA, 0.5 lg of oligo measuring the calcium flux in Fura-2/AM-loaded Jurkat cells.
(dT)18 primer, 10 mM dNTPs, and 200 U of reverse trans- In situ calcium imaging in the presence of 1 mM CaCl2
criptase were used for each cDNA synthesis reaction. PCR revealed that during the perfusion of delphinidin a significant
amplifications were performed using the BrilliantÒ II SYBRÒ increase of intracellular Ca2? was observed (Fig. 1a, b).
Green QPCR Master Mix (Stratagene) according to the man- Treatment with varying concentrations of delphinidin (10, 50,
ufacturer’s instructions. All reactions were carried out using an and 100 lM) induced a slow and steady increase in [Ca2?]i.
Mx300p thermal cycler (Stratagene, CA, USA). IL-2-specific This effect was dose-dependent, as shown by representative
primers were used as follows: 50 -ACCAGGATGCTCACATT recordings and AUC150 measurements (Fig. 1c, d, respec-
TAAGTTTT-30 (sense) and 50 -GAGGTTTGAGTTCTTCTT tively), which increased following treatment with 50 or
CTAGACACTG-30 (antisense). 100 lM delphinidin, compared to vehicle-treated cells (i.e.,
0.2 % DMSO). All concentrations of delphinidin used in this
IL-2 and IFN-c Measurements experiment were not toxic to the Jurkat cells, as was demon-
strated in the MTT-based viability experiments (Fig. 1e).
Jurkat E6-1 cells (5 9 106 cells/mL) and peripheral blood The role of extracellular and intracellular calcium on the
lymphocytes (PBLs) (5 9 105 cells/mL) were cultured in calcium fluxes induced by 50 lM delphinidin was evalu-
24-well plates, preincubated with BTP2 (1, 5 or 10 lM), ated (Fig. 2). Fura-2/AM-loaded cells were incubated in
cyclosporine A (1 lg/mL) or a vehicle for 15 min and then Ca2?-free medium, and 0.3 mM EGTA was added 20 s
stimulated with 50 lM delphinidin. Supernatants were prior to delphinidin stimulation. Delphinidin treatment in
collected at 48 h post-stimulation and analyzed for IL-2 Ca2?-free medium induced a mild increase in intracellular
and IFN-c using the BD OptEIA TM Kit. calcium compared to delphinidin in the presence of Ca2?,

123
Cell Biochem Biophys (2014) 68:497–509 501

A C

B D

Fig. 1 Delphinidin induces calcium fluxes in Jurkat cells. Fura-2/ the fluorescence imaging of cytosolic calcium. c The effect of
AM-loaded Jurkat E6-1 cells were suspended in Ca2?-HEPES buffer delphinidin (10, 50, and 100 lM) on Fura-2/AM-loaded Jurkat cells
and then stimulated with delphinidin. The Ca2? influx was assessed in the presence of Ca2?. d The dose response effect of delphinidin on
by in situ calcium imaging and spectrofluorimetric assays. a Repre- the AUC150 of the Fura-2/AM 340/380 ratio. e The effect of
sentative fluorescence imaging of cytosolic calcium increases induced delphinidin on Jurkat cell viability using MTT assay is shown. Each
by treatment with 50 lM delphinidin (9400). The pseudocolor scale bar represents the mean of at least three independent experiments.
reflects low (0) and high ratios [1]. b Representative measurements of *p \ 0.05 and **p \ 0.01 compared with vehicle
the effects induced by 50 lM delphinidin on the 340/380 ratio during

suggesting the contribution of an intracellular compartment intracellular Ca2? concentrations and long-term functional
to this effect (Fig. 2a, b). In addition, we utilized BAPTA/ consequences [29].
AM to chelate the intracellular calcium and to assess the To evaluate the contribution of SOCE on the increase of
effect of delphinidin on ER Ca2? release. We observed an intracellular calcium induced by delphinidin, BTP2, which
almost complete inhibition of the calcium flux, which inhibits SOCE in Jurkat cells [13], was used. In the presence of
suggests the inhibition of intracellular calcium release; 1 mM CaCl2, BTP2 significantly reduced the delphinidin-
however, a mild increase in Ca2? was still recorded induced calcium flux in Fura-2/AM-loaded Jurkat cells
(Fig. 2c). The calcium flux induced by 50 lM delphinidin (Fig. 3a; Supplementary Data 1A). In addition, we tested the
was completely blocked in the presence of 50 lM BAPTA/ efficacy of Gd3?, a putative SOCE inhibitor, on delphinidin-
AM and 0.3 mM EGTA in the absence of CaCl2 (Fig. 2d). induced calcium flux in Fura-2/AM-loaded Jurkat cells
(Fig. 3d). It is widely known that lanthanides, such as Gd3?,
suppress CRAC channels in Drosophila S2 cells [30]. In the
Putative SOCE Inhibitors BTP2 and Gd3? Reduce presence of 1 mM CaCl2, the treatment of Fura-2/AM-loaded
the Delphinidin-Induced Ca2? Influx Jurkat cells with 1, 5, or 10 lM Gd3? significantly reduced the
AUC200 induced by 50 lM delphinidin (Supplementary Data
In T cells, the main mechanism for the influx of extracel- 1B), which suggests an interference with the Ca2? influx.
lular calcium across the plasma membrane is SOCE [11]. Next, we assessed the effect of BTP2 on the Ca2? influx
SOCE leads directly to a sustained increase in the induced by delphinidin in the absence of external calcium

123
502 Cell Biochem Biophys (2014) 68:497–509

Fig. 2 Ca2? chelation A B


interferes with delphinidin-
induced Ca2? flux. Jurkat cells
was loaded with Fura-2/AM,
resuspended in the presence or
absence of Ca2? and stimulated
with delphinidin. The Ca2?
influx was then measured using
spectrofluorimetric assays.
a The effect of treatment with
50 lM delphinidin on Fura-2/
AM-loaded Jurkat cells in the
presence of Ca2? is shown. The
effects of a b Ca2?-free solution
plus 0.3 mM EGTA, c 1 mM C D
Ca2? plus 50 lM BAPTA/AM,
and d Ca2?-free/0.3 mM EGTA
plus 50 lM BAPTA/AM on the
delphinidin-induced calcium
mobilization are shown

Fig. 3 Delphinidin-induced A D
Ca2? and Sr2? influxes are
inhibited by BTP2 and Gd3?.
The spectrofluorometric register
of the effect of BTP2 (10 lM)
(a) or Gd3? (10 lM) (d) on
delphinidin-induced Ca2?
movements in Fura-2/AM-
loaded Jurkat has been depicted.
In a second set of experiments,
Jurkat cells were stimulated
with 50 lM delphinidin in
Ca2?-free HEPES/0.3 mM B E
EGTA, and, then, a 1 mM
CaCl2 pulse was added. A
representative
spectrofluorimeter recording of
the effect of BTP2 (b) or Gd3?
(e) on the Ca2? influx induced
by delphinidin is shown. In a
third set of experiments, Fura-2/
AM-loaded Jurkat cells were
stimulated with 50 lM
delphinidin in Ca2?-free
HEPES. A pulse of 1 mM SrCl2 C F
was added, and the 340/380
ratio of fluorescence was
recorded. The effects of 10 lM
BTP2 or Gd3? on Sr2? influx
(c, f) are depicted

123
Cell Biochem Biophys (2014) 68:497–509 503

(Fig. 3b). For these experiments, we used Fura-2/AM-loa- phosphatase. Calcineurin activation is initiated by the
ded cells that were preincubated with varying concentrations binding of calcium to calmodulin; once activated, calci-
of BTP2 in the absence of external calcium and in the pres- neurin causes NFAT dephosphorylation, which is a key
ence of 0.3 mM EGTA. After 100 s of recording, 50 lM step in NFAT nuclear translocation [35]. Because the
delphinidin was added. We did not observe an inhibition of increase in calcium induced by delphinidin is able to
the intracellular calcium movements. However, after 200 s, activate the NFAT pathway, we analyzed the effect of
a pulse of 1 mM CaCl2 was added. Under these experimental delphinidin treatment on NFAT phosphorylation using
conditions, pretreatment with BTP2 (5 and 10 lM) signifi- western blot analysis. While NFAT migrated as a single
cantly reduced the delphinidin-induced Ca2? influx, as protein band with an apparent molecular mass of 140 kDa
measured by the AUC150 (Supplementary Data 2). in unstimulated cells, NFAT migrated as multiple bands in
Because delphinidin treatment induced intracellular cells stimulated with 50 lM of delphinidin (Fig. 4a).
Ca2? release, we assessed whether Gd3? interfered with Delphinidin induced a marked NFAT dephosphorylation at
this mechanism. To achieve this, we developed an exper- 15 min post-stimulation that was sustained for 4 h. In
imental protocol in the absence of external Ca2? and in the
presence of 0.3 mM EGTA. We added 50 lM delphinidin
to Fura-2/AM-loaded Jurkat cells either in the presence or A
absence of Gd3? and then measured the level of fluores-
cence emitted. After a period of 200 s, a pulse of 1 mM
CaCl2 was incorporated. In the absence of external cal-
cium, delphinidin induced a strong Ca2? influx (Fig. 3e).
Gd3? did modify the intracellular Ca2? release, as mea-
sured by the AUC200 (Supplementary Data 3A). The
addition of 1 mM CaCl2 increased the fluorescence, sug- B
gesting the existence of a Ca2? influx that was significantly
inhibited by Gd3? (Fig. 3e; Supplementary Data 3B).

BTP2 and Gd3? Reduce the Delphinidin-Induced


Sr2? Influx in Jurkat Cells

CRAC channels are known to be permeable to other cat-


ions, such as Sr2? [31]; therefore, in a manner similar to
Ca2?, delphinidin could induce the Sr2? influx in Jurkat
cells. To test this hypothesis, we investigated the impact of
delphinidin on Sr2? entry into cells. Although Sr2? has a
lower affinity for Fura-2/AM compared to Ca2? and its
binding causes less fluorescence, their isosbestic points and
340:380 ratio profiles are similar [32]. Furthermore, Sr2?
has been used as an indicator of SOCE in previous studies
[33, 34]. In the absence of external calcium, delphinidin
treatment increased the Sr2? influx when 1 mM Sr2? was
added into the medium at 300 s (Fig. 3c). The influx of Fig. 4 Delphinidin induces NFAT activation in Jurkat cells.
Sr2?, but not an intracellular Ca2? release, was inhibited by a Western blot analysis of cell extracts after various periods of
treatment with delphinidin. Jurkat cells were treated with vehicle or
10 lM BTP2 (Supplementary Data 4A, C). The inhibitor
delphinidin for 0, 5, 15, 30, 45, 60, 120, and 240 min. The
Gd3? (10 lM) also blocked the Sr2? influx (Fig. 3f). electrophoretic mobilities of the upper and lower bands, which
However, in contrast to BTP2, Gd3? affected both the correspond to the phosphorylated and dephosphorylated forms of
release of intracellular calcium and the influx of Sr2? NFAT1, respectively, are indicated by arrows. The data shown are
representative of at least three separate experiments. Jurkat F6 cells
(Fig. 3f; Supplementary Data 4B, D).
were placed on fibronectin-coated glass coverslips and subjected to
time-lapse bioluminescence imaging to visualize the dynamics of
Delphinidin Activates the NFAT Pathway NFAT transcriptional activity in living cells. Bioluminescent images
in Jurkat Cells of accumulated photonic emissions during a 15 min period in a
control medium or in medium containing 50 lM delphinidin (the
pseudocolor scale ranges from 0 to 50 photons per pixel). Data are
In T cells, sustained calcium influx is associated with the representative of at least three independent experiments for each
activation of calcineurin, which is a calmodulin-dependent condition

123
504 Cell Biochem Biophys (2014) 68:497–509

addition, we assessed the transcriptional activation of Next, cyclosporine A was used to study the role of
NFAT following delphinidin treatment using Jurkat cells calcineurin on NFAT nuclear translocation induced by
that express luciferase under the control of triplet NFAT delphinidin. Flow cytometric and immunofluorescence
response elements. These cells were subjected to time- analyses showed that the delphinidin-induced NFAT
lapse bioluminescence imaging to visualize the dynamics nuclear translocation was calcineurin-dependent, as the
of NFAT transcriptional activity in live Jurkat F6 cells. nuclear translocation was inhibited by cyclosporine A
Treatment with 50 lM delphinidin increased NFAT tran- treatment (Fig. 5d, e).
scriptional activity until 4 h post-stimulation, after which To assess whether NFAT participated in delphinidin-
NFAT transcriptional activity achieved a steady state that induced IL-2 production, Jurkat cells were incubated with
was sustained throughout the experimental time period cyclosporine A (1 lg/mL) for 30 min and then stimulated
(16 h) (Fig. 4b). As a positive control, treatment with with delphinidin. Treatment with 1 lg/mL cyclosporine A
ionomycin (500 nM) plus PMA (100 nM) also increased significantly reduced IL-2 mRNA expression (Fig. 5c) and
the NFAT transcriptional activity, and a maximal level of IL-2 production (Fig. 5f).
transcription was achieved at 6 h post-stimulation (data not In addition, we demonstrated that delphinidin induced
shown). the production of IL-2 and IFN-c in PBLs (Fig. 6a, b,
respectively). In both cases, this effect was significantly
blocked by BTP2, which suggests that cytokine production
Delphinidin-Induced NFAT Activation and IL-2 induced by delphinidin is also SOCE dependent (Fig. 6c,
Production are Dependent on SOCE and Calcineurin d). We demonstrated that the IFN-c production induced by
delphinidin in PBLs was reduced with 1 lg/mL of cyclo-
As SOCE and calcineurin are critical for NFAT activation, sporine A (Supplementary Data 5).
we analyzed the effect of delphinidin on NFAT translo-
cation via flow cytometric and immunofluorescence anal-
yses. The flow cytometric analysis of NFAT translocation Discussion
in nuclei purified from Jurkat E6-1 cells showed that del-
phinidin induced an increase in the amount of nuclear Polyphenols have been reported to exert a large number of
NFAT compared to control cells. This increase was beneficial physiological effects on humans [36]. Within the
blocked in Jurkat E6-1 cells that were preincubated with polyphenol group, anthocyanidins and their glycosylated
BTP2, and the translocation levels observed under these forms, anthocyanins, have gained significant attention due
experimental conditions were similar to the levels observed to the numerous studies that indicate their potential human
in controls (Fig. 5a). In addition, we corroborated the health benefits [37, 38]. Delphinidin has been reported to
nuclear translocation of NFAT using immunofluorescence possess antioxidant properties [39], as well as to exert
experiments. We observed that delphinidin increased the antimutagenic [40] and neuroprotective effects [41, 42]. In
presence of NFAT in the nuclei of Jurkat E6-1 cells addition, delphinidin displays a variety of effects on blood
compared to control cells. Furthermore, the preincubation vessels [43] and platelets [44] and has been shown to
of Jurkat E6-1 cells with BTP2 blocked the nuclear trans- reduce the risk of coronary artery disease [45].
location of NFAT (Fig. 5b). In the present work, we demonstrated that delphinidin
In T cells, calcium signals are a critical signal for the increases the intracellular calcium fluxes that induce
activation of effector functions, such as IL-2 production cytokine production in T cells.
[11]. To assess whether delphinidin could exert an immu- We demonstrated through spectrofluorimetric experi-
nostimulatory effect, we treated Jurkat cells with 50 lM ments that delphinidin induces a sustained calcium mobi-
delphinidin and measured the resulting IL-2 mRNA and lization in Jurkat E6-1 cells. Calcium imaging experiments
protein levels. Delphinidin increased the expression of IL-2 revealed that delphinidin treatment increased calcium
mRNA after 24 h of incubation (Fig. 5c) and the produc- movement, which decreased when it was withdrawn at
tion of IL-2 protein after 48 h of incubation (Fig. 5f). 6 min from the perfusion medium. In addition, in the
Higher concentrations of delphindin did not increase the absence of external calcium, delphinidin-induced calcium
expression of IL-2 mRNA or proteins more than observed fluxes were reduced, but not abolished. Calcium mobili-
with 50 lM. zation was abolished completely in the presence of EGTA
To assess whether CRAC channels and NFAT are involved plus 50 lM BAPTA/AM, indicating that intracellular Ca2?
in delphinidin-induced IL-2 production, Jurkat cells were stores play an important role in delphinidin-induced cal-
preincubated with BTP2 prior to delphinidin stimulation. cium mobilization.
BTP2 pretreatment significantly inhibited IL-2 mRNA Previously, treatment with 30 lM delphinidin was shown
expression (Fig. 5c) and IL-2 protein production (Fig. 5f). to increase calcium fluxes in bovine aortic endothelial cells.

123
Cell Biochem Biophys (2014) 68:497–509 505

A D

B E

C F

Fig. 5 Delphinidin-induced NFAT translocation and IL-2 mRNA graph shows the ratio of the fluorescence between nuclear NFAT and
expression and cytokine production are blocked by BTP2 and total cellular fluorescence. Jurkat cells were incubated with 10 lM
cyclosporine A. Jurkat cells were incubated with 10 lM BTP2 (a, BTP2 (c) or 1 lg/mL cyclosporine A (e, f) for 15 min and then
b) or 1 lg/mL cyclosporine A (e, f) for 15 min and then stimulated stimulated with 50 lM delphinidin. RNA and supernatants were
with 50 lM delphinidin for 15 min. Jurkat cell nuclei were then either extracted and subjected to analysis by qRT-PCR (c) or ELISA (f),
subjected to flow cytometric analysis to determine the amount of respectively. Each bar represents the mean ± SEM of at least three
NFAT present (a, d) or to a cytospin of cells before being fixed onto independent experiments. **p \ 0.01 compared to treatment with
glass coverslips for immunofluorescence analysis using propidium 50 lM delphinidin alone
iodide and the NFATc1 antibody-Alexa Fluor488 (b, e). The bar

In the absence of external calcium, the mean peak of [Ca2?]I, calcium, Gd3? and BTP2 inhibited calcium uptake when
as induced by delphinidin, was significantly reduced by 1 mM CaCl2 was incorporated into the medium. However,
70 ± 2 %, which suggests the involvement of both calcium while Gd3? reduced the delphinidin-induced calcium
influx and internal calcium release [46]. release from intracellular stores, BTP2 did not. This sug-
The main mechanism for the entry of extracellular Ca2? gests a more specific effect of BTP2 on the opening of
across the plasma membrane into T cells is via SOCE, CRAC channels in Jurkat cells [13].
which occurs when InsP3-induced depletion of the ER The inhibition of the calcium influx induced by BTP2
Ca2? stores triggers plasma membrane Ca2? entry through and Gd3? in Jurkat cells suggests that delphinidin activates
Ca2? release-activated calcium (CRAC) channels [47]. the uptake of extracellular calcium through CRAC chan-
We assessed the effects of two CRAC channel inhibi- nels. Consistent with these findings, Zitt et al. [13] dem-
tors, Gd3? and BTP2. Both inhibitors reduced delphinidin- onstrated that BTP2 specifically inhibited CRAC channels
induced calcium mobilization. In the absence of external in human peripheral blood CD4? and CD8? T cells and in

123
506 Cell Biochem Biophys (2014) 68:497–509

A B

C D

Fig. 6 BTP2 blocks IL-2 and IFN-c production induced by delphin- stimulated with 50 lM delphinidin for 48 h; at that point, the
idin in PBLs. Lymphocytes were separated from blood samples and supernatants were extracted and subjected to ELISA analysis for the
then incubated with delphinidin (10, 30, 50, or 100 lM) for 48 h. presence of IL-2 (c) or IFN-c (d). Each bar represents the
Next, the supernatants were extracted and subjected to ELISA mean ± SEM of at least three independent experiments. *p \ 0.05;
analysis for the presence of IL-2 (a) or IFN-c (b) production. PBL **p \ 0.01 compared to treatment with 50 lM delphinidin alone
cells were incubated with 1, 5, and 10 lM BTP2 for 15 min and then

Jurkat cells without affecting the other channels or trans- LoVo/ADR cells, a doxorubicin-resistant metastatic human
porters [13]. BTP2 has been shown to block SOCE in T colorectal adenocarcinoma cell line, and said to induce
lymphocytes by inhibiting some of the proteins putatively cytotoxicity and increase the production of radical oxygen
involved in this phenomenon, which include members of species [50]. Because H2O2 can be involved in the acti-
the transient receptor potential (TRP) superfamily, such as vation of CRAC channels in Jurkat cells [51], and antho-
TRPM4 [48]. cyanins have shown anti-proliferative effect in leukemia
In addition, we observed that delphinidin induced a cells [52], this could represent a potential mechanism
sustained Sr2? influx over time. This result corroborates implicated in the effect of delphinidin on T cells. However,
delphinidin-induced SOCE activation because the perme- this theory was discarded because the concentration of
ability to both Ca2? and Sr2? was increased. Furthermore, delphinidin used in T cells did not induce any toxic effects.
both BTP2 and Gd3? reduced delphinidin-induced Sr2? Here, using luminol, we were not able to not observe any
influx, suggesting that their entry was CRAC-dependent. increase in the production of radical oxygen species (data
However, the increase in fluorescence with Sr2? was not shown).
reduced compared to Ca2?. This result could be explained In T cells, the sustained influx of Ca2? is associated with
by the lower affinity of Sr2? for Fura-2/AM or a result of the activation of calcineurin, which is a Ca2?/calmodulin-
its binding, which might cause decreased fluorescence dependent Ser/Thr phosphatase that dephosphorylates
compared to Ca2? [32]. In addition, the relative perme- NFAT; the dephosphorylation of NFAT enables NFAT
ability for Sr2? was lower compared to Ca2? in the Jurkat nuclear translocation and transcriptional activity [53, 54].
cells, which exhibited a permeability ratio of 0.06 for Sr2? We assessed the NFAT phosphorylation status following
versus 1 for Ca2? [49]. Furthermore, we determined that delphinidin treatment. Treatment with 50 lM of delphini-
delphinidin was not able to induce Ba2? influx in Jurkat din induced NFAT dephosphorylation. Moreover, the
cells (data not shown). This result is consistent with the low strong NFAT dephosphorylation observed after 15 min of
permeability reported for this cation in Jurkat cells that delphinidin treatment was maintained through 240 min of
were stimulated with 1 lM thapsigargin in Ca2?-free stimulation.
K?-Ringer’s solution [49]. Moreover, in cytotoxic T cells, NFAT-h is a conserved module of *300 residues,
the store-operated calcium influx is seven times more which regulates NFAT nuclear/cytoplasmic trafficking in
selective for Ca2? over Sr2?, and 17 times more selective response to changes in intracellular Ca2? concentrations.
for Ca2? over Ba2? [49]. The mechanism of the delphini- The engagement of the TCR or treatment of cells with a
din-induced activation of the CRAC channels remains Ca2? ionophore activates calcineurin. Calcineurin then
unknown. The use of delphinidin has been described in dephosphorylates the NFAT-h domain, which leads to the

123
Cell Biochem Biophys (2014) 68:497–509 507

unmasking of NFAT’s nuclear localization sequences and 264.7 macrophages, delphinidin increased the TNFa pro-
subsequent NFAT nuclear translocation [55, 56]. In cells, duction induced by LPS/IFN-c [9]. The present finding on
NFAT remains in the nucleus while Ca2? is elevated, but it T cells supports a more complex immunomodulatory effect
is rapidly phosphorylated and exported to the cytoplasm of delphinidin on leukocytes. Due to the low bioavailability
following the termination of calcium signaling [17, 18]. of anthocyanins, a potential therapeutical effect, should be
Because delphinidin induced a strong and sustained Ca2? carefully considered to explain systemic effects. However,
flux in Jurkat cells, we assessed the extent of NFAT because anthocyanins are present in foods and several
translocation in response to delphinidin treatment. We antecedents have suggested potentially positive effects in
demonstrated that treatment with 50 lM delphinidin gastrointestinal diseases, we did not discard the potential
resulted in increased NFAT nuclear localization in Jurkat local immunological modulations induced by these com-
cells. In addition, BTP2 significantly inhibited this pounds. In support of this assumption, anthocyanins have
response, suggesting that NFAT nuclear translocation is been described as effective in animals models and humans
CRAC channel-dependent. Previously, BTP2 was demon- of colon cancer [61]. The IFN-c expressed by Th1 cells has
strated to inhibit NFAT activation in Jurkat cells and pri- been described as an effective antitumoral cytokine in
mary human T cells [13, 56, 57], which supports the colon adenocarcinoma and a possible autocrine enhancer of
critical importance of CRAC channels in NFAT activation. cytolytic T cell capacity [62, 63].
We also observed that cyclosporine A partially blocked the In conclusion, delphinidin treatment induced Ca2? and
2?
delphinidin-induced NFAT nuclear translocation. Consis- Sr influx through CRAC channels, as indicated by the
tent with these results, BTP2 has been shown to suppress ability of BTP2 and Gd3? to inhibit both events. Further-
calcineurin activation induced by PMA/ionomycin in Jur- more, BTP2 blocked delphinidin-induced NFAT activation,
kat cells, which further supports the role of CRAC channels which corroborates the involvement of SOCE in delphinidin-
in calcineurin activation [58]. mediated calcium signaling. Finally, delphinidin treatment
Finally, we demonstrated that delphinidin increased induced the expression and production of IL-2 mRNA and
NFAT transcriptional activity. Treatment with delphinidin protein, which is an effect that is mediated by SOCE and
induced a sustained increase in NFAT-luciferase reporter NFAT activation, as both processes were inhibited by BTP2
activity during the entire recording period. and cyclosporine A. However, as IL-2 production was not
These results suggest that delphinidin can induce the completely blocked by BTP2 and cyclosporine A, it is pos-
transcriptional activation of NFAT-dependent genes in T sible that other signaling pathways are involved in delphin-
cells, including genes that code for cytokines, such as IL-2 idin-mediated T cell activation. Taken together, these results
and IFN-c [11]. Consistent with this hypothesis, we suggest that delphinidin can increase the immune response
observed that delphinidin increased the expression and by stimulating cytokine production in T cells.
production of IL-2 mRNA and protein. Delphinidin treat-
ment enhanced IL-2 mRNA expression five times above Acknowledgments This work was supported by Grants from Con-
sorcio de Tecnologı́a e Innovación para la Salud CTI-Salud (CTE-06),
the levels observed in control cells while increasing IL-2 Chile (CONICYT 21090900 and CONICYT AT-24100037).
production by *250 pg/mL compared to control-treated
cells. BTP2 pretreatment significantly decreased delphini-
din-induced IL-2 mRNA expression and protein produc-
tion. In this context, the CRAC channels are fundamental
for the activation of T cell effector functions, particularly References
because BTP2 is able to specifically inhibit calcium-
dependent gene expression, including IL-2, IL-5, and IFN- 1. Barak, V., Birkenfeld, S., Halperin, T., & Kalickman, I. (2002).
c, in peripheral blood regulatory T lymphocytes and Jurkat The effect of herbal remedies on the production of human
inflammatory and anti-inflammatory cytokines. The Israel Med-
cells [56, 57]. Moreover, we observed that BTP2 can ical Association Journal, 4, 919–922.
reduce IL-2 and IFN-c production induced by delphinidin 2. McAnulty, L. S., Nieman, D. C., Dumke, C. L., Shooter, L. A.,
in peripheral blood T lymphocytes. In addition, cyclo- Henson, D. A., Utter, A. C., et al. (2011). Effect of blueberry
sporine A pretreatment resulted in a partial decrease of ingestion on natural killer cell counts, oxidative stress, and
inflammation prior to and after 2.5 h of running. Applied Physi-
delphinidin-induced IL-2 production. Because cyclospor- ology, Nutrition and Metabolism, 36, 976–984.
ine A completely inhibits IL-2 production induced by 3. Waknine-Grinberg, J. H., El-On, J., Barak, V., Barenholz, Y., &
PMA/ionomycin in Jurkat cells [59], our results suggest Golenser, J. (2009). The immunomodulatory effect of Sambucol on
that delphinidin-induced IL-2 production involves other leishmanial and malarial infections. Planta Medica, 75, 581–586.
4. Barak, V., Halperin, T., & Kalickman, I. (2001). The effect of
calcium-dependent signaling pathways. The anti-inflam- Sambucol, a black elderberry-based, natural product, on the
matory properties of delphinidin are well-known and have production of human cytokines: I. Inflammatory cytokines.
been demonstrated previously [60]. However, in RAW European cytokine network, 12, 290–296.

123
508 Cell Biochem Biophys (2014) 68:497–509

5. Wang, Y. P., Cheng, M. L., Zhang, B. F., Mu, M., Zhou, M. Y., primary immunodeficiency. Journal of Biological Chemistry,
Wu, J., et al. (2010). Effect of blueberry on hepatic and immu- 269, 32327–32335.
nological functions in mice. Hepatobiliary & Pancreatic Dis- 24. Weiss, A., Wiskocil, R. L., & Stobo, J. D. (1984). The role of T3
eases International, 9, 164–168. surface molecules in the activation of human T cells: A two-
6. Afaq, F., Syed, D. N., Malik, A., Hadi, N., Sarfaraz, S., Kweon, stimulus requirement for IL 2 production reflects events occurring
M. H., et al. (2007). Delphinidin, an anthocyanidin in pigmented at a pre-translational level. Journal of Immunology, 133,
fruits and vegetables, protects human HaCaT keratinocytes and 123–128.
mouse skin against UVB-mediated oxidative stress and apoptosis. 25. Feske, S., Gwack, Y., Prakriya, M., Srikanth, S., Puppel, S. H.,
The Journal of Investigative Dermatology, 127, 222–232. Tanasa, B., et al. (2006). A mutation in Orai1 causes immune
7. Kong, J. M., Chia, L. S., Goh, N. K., Chia, T. F., & Brouillard, R. deficiency by abrogating CRAC channel function. Nature, 441,
(2003). Analysis and biological activities of anthocyanins. Phy- 179–185.
tochemistry, 64, 923–933. 26. Feske, S., Draeger, R., Peter, H. H., Eichmann, K., & Rao, A.
8. Juranic, Z., & Zizak, Z. (2005). Biological activities of berries: (2000). The duration of nuclear residence of NFAT determines
From antioxidant capacity to anti-cancer effects. Biofactors, 23, the pattern of cytokine expression in human SCID T cells.
207–211. Journal of Immunology, 165, 297–305.
9. Wang, J., & Mazza, G. (2002). Effects of anthocyanins and other 27. Baldari, C. T., Di Somma, M. M., Majolini, M. B., Ulivieri, C.,
phenolic compounds on the production of tumor necrosis factor Milia, E., & Telford, J. L. (1998). NF-AT-luciferase reporter T
alpha in LPS/IFN-gamma-activated RAW 264.7 macrophages. cell lines as tools to screen immunosuppressive drugs. Biologi-
Journal of Agriculture and Food Chemistry, 50, 4183–4189. cals, 26, 1–5.
10. Seong, A. R., Yoo, J. Y., Choi, K., Lee, M. H., Lee, Y. H., Lee, J., 28. Blaecke, A., Delneste, Y., Herbault, N., Jeannin, P., Bonnefoy, J.
et al. (2011). Delphinidin, a specific inhibitor of histone acetyl- Y., Beck, A., et al. (2002). Measurement of nuclear factor-kappa
transferase, suppresses inflammatory signaling via prevention of B translocation on lipopolysaccharide-activated human dendritic
NF-kappaB acetylation in fibroblast-like synoviocyte MH7A cells by confocal microscopy and flow cytometry. Cytometry, 48,
cells. Biochemical and Biophysical Research Communications, 71–79.
410, 581–586. 29. Oh-hora, M., & Rao, A. (2008). Calcium signaling in lympho-
11. Feske, S. (2007). Calcium signalling in lymphocyte activation cytes. Current Opinion in Immunology, 20, 250–258.
and disease. Nature Reviews Immunology, 7, 690–702. 30. Zhang, S. L., Yeromin, A. V., Zhang, X. H., Yu, Y., Safrina, O.,
12. Putney, J. W, Jr. (1986). A model for receptor-regulated calcium Penna, A., et al. (2006). Genome-wide RNAi screen of Ca(2?)
entry. Cell Calcium, 7, 1–12. influx identifies genes that regulate Ca(2?) release-activated
13. Zitt, C., Strauss, B., Schwarz, E. C., Spaeth, N., Rast, G., Hat- Ca(2?) channel activity. Proceedings of the National Academy of
zelmann, A., et al. (2004). Potent inhibition of Ca2? release- Sciences of the United States of America, 103, 9357–9362.
activated Ca2? channels and T-lymphocyte activation by the 31. Zweifach, A., & Lewis, R. S. (1993). Mitogen-regulated Ca2?
pyrazole derivative BTP2. Journal of Biological Chemistry, 279, current of T lymphocytes is activated by depletion of intracellular
12427–12437. Ca2? stores. Proceedings of the National Academy of Sciences of
14. Shaw, J. P., Utz, P. J., Durand, D. B., Toole, J. J., Emmel, E. A., the United States of America, 90, 6295–6299.
& Crabtree, G. R. (1988). Identification of a putative regulator of 32. Xu-Friedman, M. A., & Regehr, W. G. (1999). Presynaptic
early T cell activation genes. Science, 241, 202–205. strontium dynamics and synaptic transmission. Biophysical
15. Rao, A., Luo, C., & Hogan, P. G. (1997). Transcription factors of Journal, 76, 2029–2042.
the NFAT family: Regulation and function. Annual Review of 33. Christian, E. P., Spence, K. T., Togo, J. A., Dargis, P. G., & Patel,
Immunology, 15, 707–747. J. (1996). Calcium-dependent enhancement of depletion-acti-
16. Crabtree, G. R., & Olson, E. N. (2002). NFAT signaling: Cho- vated calcium current in Jurkat T lymphocytes. Journal of
reographing the social lives of cells. Cell, 109(Suppl), S67–S79. Membrane Biology, 150, 63–71.
17. Zhu, J., & McKeon, F. (1999). NF-AT activation requires sup- 34. Zweifach, A., & Lewis, R. S. (1996). Calcium-dependent
pression of Crm1-dependent export by calcineurin. Nature, 398, potentiation of store-operated calcium channels in T lympho-
256–260. cytes. Journal of General Physiology, 107, 597–610.
18. Okamura, H., Aramburu, J., Garcia-Rodriguez, C., Viola, J. P., 35. Macian, F. (2005). NFAT proteins: Key regulators of T-cell
Raghavan, A., Tahiliani, M., et al. (2000). Concerted dephos- development and function. Nature Reviews Immunology, 5,
phorylation of the transcription factor NFAT1 induces a confor- 472–484.
mational switch that regulates transcriptional activity. Molecular 36. Harborne, J. B. (1988). Flavonoids in the environment: Structure-
Cell, 6, 539–550. activity relationships. Progress in Clinical and Biological
19. Shibasaki, F., Hallin, U., & Uchino, H. (2002). Calcineurin as a Research, 280, 17–27.
multifunctional regulator. Journal of Biochemistry, 131, 1–15. 37. Bell, D. R., & Gochenaur, K. (2006). Direct vasoactive and va-
20. Le Deist, F., Hivroz, C., Partiseti, M., Thomas, C., Buc, H. A., soprotective properties of anthocyanin-rich extracts. Journal of
Oleastro, M., et al. (1995). A primary T-cell immunodeficiency Applied Physiology, 100, 1164–1170.
associated with defective transmembrane calcium influx. Blood, 38. Vuorela, S., Salminen, H., Makela, M., Kivikari, R., Karonen, M.,
85, 1053–1062. & Heinonen, M. (2005). Effect of plant phenolics on protein and
21. Feske, S., Giltnane, J., Dolmetsch, R., Staudt, L. M., & Rao, A. lipid oxidation in cooked pork meat patties. Journal of Agricul-
(2001). Gene regulation mediated by calcium signals in T lym- ture and Food Chemistry, 53, 8492–8497.
phocytes. Nature Immunology, 2, 316–324. 39. Noda, Y., Kaneyuki, T., Mori, A., & Packer, L. (2002). Antiox-
22. Feske, S., Prakriya, M., Rao, A., & Lewis, R. S. (2005). A severe idant activities of pomegranate fruit extract and its anthocyani-
defect in CRAC Ca2? channel activation and altered K? channel dins: Delphinidin, cyanidin, and pelargonidin. Journal of
gating in T cells from immunodeficient patients. Journal of Agriculture and Food Chemistry, 50, 166–171.
Experimental Medicine, 202, 651–662. 40. Azevedo, L., Alves de Lima, P. L., Gomes, J. C., Stringheta, P.
23. Partiseti, M., Le Deist, F., Hivroz, C., Fischer, A., Korn, H., & C., Ribeiro, D. A., & Salvadori, D. M. (2007). Differential
Choquet, D. (1994). The calcium current activated by T cell response related to genotoxicity between eggplant (Solanum
receptor and store depletion in human lymphocytes is absent in a melanogena) skin aqueous extract and its main purified

123
Cell Biochem Biophys (2014) 68:497–509 509

anthocyanin (delphinidin) in vivo. Food and Chemical Toxicol- mechanisms involved. International Journal of Oncology, 23,
ogy, 45, 852–858. 705–712.
41. Galli, R. L., Shukitt-Hale, B., Youdim, K. A., & Joseph, J. A. 53. Aramburu, J., Garcia-Cozar, F., Raghavan, A., Okamura, H., Rao,
(2002). Fruit polyphenolics and brain aging: Nutritional inter- A., & Hogan, P. G. (1998). Selective inhibition of NFAT acti-
ventions targeting age-related neuronal and behavioral deficits. vation by a peptide spanning the calcineurin targeting site of
Annals of the New York Academy of Sciences, 959, 128–132. NFAT. Molecular Cell, 1, 627–637.
42. Youdim, K. A., & Deans, S. G. (2000). Effect of thyme oil and 54. Liu, Y., Cseresnyes, Z., Randall, W. R., & Schneider, M. F.
thymol dietary supplementation on the antioxidant status and (2001). Activity-dependent nuclear translocation and intranuclear
fatty acid composition of the ageing rat brain. British Journal of distribution of NFATc in adult skeletal muscle fibers. Journal of
Nutrition, 83, 87–93. Cell Biology, 155, 27–39.
43. Andriambeloson, E., Magnier, C., Haan-Archipoff, G., Lobstein, 55. Zhu, J., Shibasaki, F., Price, R., Guillemot, J. C., Yano, T.,
A., Anton, R., Beretz, A., et al. (1998). Natural dietary poly- Dotsch, V., et al. (1998). Intramolecular masking of nuclear
phenolic compounds cause endothelium-dependent vasorelaxa- import signal on NF-AT4 by casein kinase I and MEKK1. Cell,
tion in rat thoracic aorta. Journal of Nutrition, 128, 2324–2333. 93, 851–861.
44. Demrow, H. S., Slane, P. R., & Folts, J. D. (1995). Administra- 56. Djuric, S. W., BaMaung, N. Y., Basha, A., Liu, H., Luly, J. R.,
tion of wine and grape juice inhibits in vivo platelet activity and Madar, D. J., et al. (2000). 3,5-Bis(trifluoromethyl)pyrazoles: A
thrombosis in stenosed canine coronary arteries. Circulation, 91, novel class of NFAT transcription factor regulator. Journal of
1182–1188. Medicinal Chemistry, 43, 2975–2981.
45. Renaud, S., & de Lorgeril, M. (1992). Wine, alcohol, platelets, 57. Trevillyan, J. M., Chiou, X. G., Chen, Y. W., Ballaron, S. J.,
and the French paradox for coronary heart disease. Lancet, 339, Sheets, M. P., Smith, M. L., et al. (2001). Potent inhibition of
1523–1526. NFAT activation and T cell cytokine production by novel low
46. Martin, S., Andriambeloson, E., Takeda, K., & Andriantsitohaina, molecular weight pyrazole compounds. Journal of Biological
R. (2002). Red wine polyphenols increase calcium in bovine Chemistry, 276, 48118–48126.
aortic endothelial cells: A basis to elucidate signalling pathways 58. Sieber, M., & Baumgrass, R. (2009). Novel inhibitors of the
leading to nitric oxide production. British Journal of Pharma- calcineurin/NFATc hub: Alternatives to CsA and FK506? Cell
cology, 135, 1579–1587. Communication and Signaling, 7, 25.
47. Panyi, G., Vamosi, G., Bodnar, A., Gaspar, R., & Damjanovich, 59. Manger, B., Hardy, K. J., Weiss, A., & Stobo, J. D. (1986).
S. (2004). Looking through ion channels: Recharged concepts in Differential effect of cyclosporin A on activation signaling in
T-cell signaling. Trends in Immunology, 25, 565–569. human T cell lines. Journal of Clinical Investigation, 77,
48. Takezawa, R., Cheng, H., Beck, A., Ishikawa, J., Launay, P., 1501–1506.
Kubota, H., et al. (2006). A pyrazole derivative potently inhibits 60. Hou, D. X., Yanagita, T., Uto, T., Masuzaki, S., & Fujii, M.
lymphocyte Ca2? influx and cytokine production by facilitating (2005). Anthocyanidins inhibit cyclooxygenase-2 expression in
transient receptor potential melastatin 4 channel activity. LPS-evoked macrophages: Structure-activity relationship and
Molecular Pharmacology, 69, 1413–1420. molecular mechanisms involved. Biochemical Pharmacology, 70,
49. Zweifach, A. (2000). Target-cell contact activates a highly 417–425.
selective capacitative calcium entry pathway in cytotoxic T 61. Thomasset, S., Teller, N., Cai, H., Marko, D., Berry, D. P., Steward,
lymphocytes. Journal of Cell Biology, 148, 603–614. W. P., et al. (2009). Do anthocyanins and anthocyanidins, cancer
50. Cvorovic, J., Tramer, F., Granzotto, M., Candussio, L., Decorti, chemopreventive pigments in the diet, merit development as
G., & Passamonti, S. (2010). Oxidative stress-based cytotoxicity potential drugs? Cancer Chemotherapy and Pharmacology, 64,
of delphinidin and cyanidin in colon cancer cells. Archives of 201–211.
Biochemistry and Biophysics, 501, 151–157. 62. Rizzo, A., Pallone, F., Monteleone, G., & Fantini, M. C. (2011).
51. Grupe, M., Myers, G., Penner, R., & Fleig, A. (2010). Activation Intestinal inflammation and colorectal cancer: A double-edged
of store-operated I(CRAC) by hydrogen peroxide. Cell Calcium, sword? World Journal of Gastroenterology, 17, 3092–3100.
48, 1–9. 63. Gerber, S. A., Sedlacek, A. L., Cron, K. R., Murphy, S. P., Fre-
52. Hou, D. X., Ose, T., Lin, S., Harazoro, K., Imamura, I., Kubo, M., linger, J. G., & Lord, E. M. (2013). IFN-gamma mediates the
et al. (2003). Anthocyanidins induce apoptosis in human pro- antitumor effects of radiation therapy in a murine colon tumor.
myelocytic leukemia cells: Structure-activity relationship and American Journal of Pathology, 182, 2345–2354.

123

You might also like