You are on page 1of 7

PHYTOTHERAPY RESEARCH

Phytother. Res. 29: 1707–1713 (2015)


Published online 14 July 2015 in Wiley Online Library
(wileyonlinelibrary.com) DOI: 10.1002/ptr.5414

Differential Inhibition of T Lymphocyte


Proliferation and Cytokine Synthesis by
[6]-Gingerol, [8]-Gingerol, and [10]-Gingerol

Megan Bernard,1 Suzanne J. Furlong,1 Melanie R. Power Coombs1 and David W. Hoskin1,2,3*
1
Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
2
Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
3
Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada

[6]-Gingerol, [8]-gingerol, and [10]-gingerol are pungent components of fresh ginger, extracts of which inhibit various
components of the inflammatory response. Because little is known regarding the effect of gingerols with different
unbranched alkyl side chain lengths on the activation and effector function of T lymphocytes, we compared the effects
of [6]-gingerol, [8]-gingerol, and [10]-gingerol on murine T lymphocyte proliferation, expression of CD25 and CD69
activation markers, cytokine synthesis, and interleukin (IL)-2 receptor signaling. All three gingerols inhibited DNA
synthesis by T lymphocytes, as well as interferon-γ synthesis. In contrast, only [8]-gingerol and [10]-gingerol inhibited
CD25 and CD69 expression, and IL-2 synthesis. None of the gingerols affected IL-4 synthesis. Exogenous IL-2
enhanced T lymphocyte proliferation in the presence of [6]-gingerol but did not significantly increase T lymphocyte
proliferation in the presence of [8]-gingerol or [10]-gingerol. In line with this finding, [8]-gingerol and [10]-gingerol
impaired IL-2-induced proliferation of CTLL-2 cells, but constitutive CD25 expression was unaffected, indicating
inhibition of IL-2 receptor signaling. In general, [10]-gingerol and [8]-gingerol were more potent inhibitors of T
lymphocytes than [6]-gingerol. Suppression of T lymphocyte responses by gingerols suggests that these phytochemi-
cals may be beneficial in chronic inflammatory conditions associated with excessive or inappropriate T lymphocyte
activation. Copyright © 2015 John Wiley & Sons, Ltd.
Keywords: cytokine; DNA synthesis; inflammation; ginger; gingerol; T lymphocyte.

Abbreviations: Ab, antibody; DMSO, dimethylsulfoxide; GM-CSF, granulocyte-macrophage colony-stimulating factor; IFN, interferon;
IL, interleukin; LPS, lipopolysaccharide; NF-κB, nuclear factor κB; OVA, ovalbumin; PBS, phosphate buffered saline; TCR, T cell
3
receptor; [ H]TdR, tritiated thymidine

eosinophilia (Ahui et al., 2008). Ginger is also a rich


INTRODUCTION
source of antioxidants that prevent inflammation caused
by oxidative stress (Dugasani et al., 2010).
Functional foods contain a plethora of bioactive compo- Analysis of fresh ginger rhizome by gas chromatography–
nents, including phytochemicals that have potentially mass spectrometry reveals that gingerols, which are
beneficial effects on human health (Hasler et al., 2004). pungent phenols with different unbranched alkyl side
Ginger (Zingiber officinale Roscoe), which is commonly chain lengths, are the predominant phytochemicals,
used as a spice and condiment, originated in South-East with [6]-gingerol being the most abundant, followed
Asia where the ginger rhizome has a long history of use by [10]-gingerol and [8]-gingerol (Jolad et al., 2004).
in traditional medicine (Butt and Sultan, 2011). Medici- Shoagols, which are the dehydrated form of the corre-
nal uses of ginger include the treatment of chronic sponding gingerols, are present in only small amounts
inflammatory conditions such as arthritis and rheuma- in fresh ginger rhizome. [6]-Gingerol is the most studied
tism, which is in line with increasing evidence that ginger of the gingerols in terms of antiinflammatory activities.
has broad antiinflammatory actions (Grzanna et al., Topical application of [6]-gingerol to mouse skin
2005). In this regard, ginger contains compounds that in- inhibits nuclear factor (NF)-κB-dependent expression of
hibit lipopolysaccharide (LPS)-induced prostaglandin E2 cyclooxygenase-2 induced by phorbol 12-myristate 13-
synthesis (Jolad et al., 2004), as well as the production of acetate (Kim et al., 2004). Macrophage synthesis of LPS-
inflammation-promoting cytokines and chemokines by induced production of pro-inflammatory cytokines and
LPS-stimulated macrophages (Tripathi et al., 2008). In other inflammatory mediators such as prostaglandin E2
addition, ginger extract suppresses ovalbumin (OVA)-in- is also inhibited by [6]-gingerol (Tripathi et al., 2007;
duced allergic airway inflammation in mice by interfering Dugasani et al., 2010). Similar antiinflammatory effects
with both Th1 and Th2 cytokine production, as well have been described for [8]-gingerol and [10]-gingerol
as inhibiting IgE production and eotaxin-induced (Dugasani et al., 2010), including inhibition of NF-κB-
dependent pro-inflammatory cytokine expression by
microglial cells (Ho et al., 2013). In addition, [8]-gingerol
* Correspondence to: David W. Hoskin, Department of Microbiology and
Immunology, Dalhousie University, 5850 College Street, PO Box 15000,
suppresses mitogen-driven proliferation of mouse
Halifax, Nova Scotia, Canada B3H 4R2. splenocytes and the OVA-specific humoral immune
E-mail: d.w.hoskin@dal.ca response in OVA-immunized mice (Lu et al., 2011).

Copyright © 2015 John Wiley & Sons, Ltd.


1708 M. BERNARD ET AL.

However, little is known about the effect that gingerols (Auburn, CA). After additional washes, T lymphocytes
have on the activation and effector function of T lympho- were resuspended in complete RPMI 1640 medium. T
cytes, which are involved in the pathogenesis of a number lymphocyte purity was at least 97%.
of autoimmune diseases and other chronic inflammatory
disorders (Cosmi et al., 2014). In this study, we investi-
gated the effect of [6]-gingerol, [8]-gingerol, and [10]- Dendritic cell generation. Bone marrow was flushed
gingerol on T lymphocyte activation and proliferation, from the tibia and femur of euthanized C57BL/6 mice
as well as cytokine production and signaling via the inter- with ice-cold PBS and forced through an 18 gauge
leukin (IL)-2 receptor. needle to create a single cell suspension. Bone marrow
cells were washed and cultured for 7 days in complete
RPMI 1640 medium containing 20 ng/mL GM-CSF to
promote dendritic cell differentiation. Cells were then
MATERIALS AND METHODS washed and cultured for an additional 24 h in medium
containing 10 ng/mL GM-CSF and 1 μg/mL LPS in
Animals. Female C57BL/6 mice aged 6–8 weeks were order to generate mature dendritic cells.
purchased from Charles River Laboratory (Lasalle,
PQ) and housed at the Carleton Animal Care Facility
of Dalhousie University. Animal protocols were consis- T lymphocyte proliferation. T lymphocytes in complete
tent with the Canadian Council on Animal Care Guide- RPMI 1640 medium were plated in triplicate at 2.5 × 105
lines and were approved by the Dalhousie University cells/well into 96-well round-bottom plates, treated as indi-
Committee on Laboratory Animals. All mice used in cated, and activated with Dynabeads® at a ratio of 1 bead
these experiments were 8–12 weeks old. for every 2 T lymphocytes. In some experiments, syngeneic
dendritic cells (6.4 × 103 cells/well) and anti-TCRβ Ab
(20 μg/mL) were used instead of Dynabeads® to activate
Reagents. [6]-Gingerol (purity > 95%) was purchased T lymphocytes. Cultures were maintained for 48, 72, or
from Dalton Pharma Services (Toronto, ON). [8]-Gingerol 96 h at 37 °C in a humidified 5% CO2 atmosphere, and
and [10]-gingerol (purity > 98%) were from Chengdu pulsed with 0.2 μCi tritiated thymidine ([3H]TdR) from
Biopurify Phytochemicals Ltd. (Chengdu, Sichuan, China). MP Biomedicals (Irvine, CA) 6 h before the end of the
Stock solutions of each gingerol were prepared at 300 mM experiment. Cultures were then harvested onto fiberglass
in dimethylsulfoxide (DMSO) and stored at 20 °C. filters with a Titertek® Cell Harvester (Skatron Instru-
Phosphate buffered saline (PBS), LPS, bovine serum ments, Sterling,VA) and [3H]TdR incorporation into
albumin, and DMSO were purchased from Sigma-Aldrich newly synthesized DNA was measured using a Beckman
Canada (Oakville, ON). Functional grade hamster anti- LS6000IC liquid scintillation counter (Beckman Coulter
mouse T cell receptor (TCR) β, phycoerythrin-conjugated Inc., Mississauga, ON).
anti-CD25, fluorescein isothiocyanate-conjugated anti-
CD69, phycoerythrin-conjugated rat IgG1, and fluorescein
isothiocyanate-conjugated Armenian hamster IgG anti- Cell viability. A Trypan Blue dye exclusion assay was
bodies (Ab) were from eBioscience Inc. (San Diego, used to measure T lymphocyte and dendritic cell viabil-
CA). Recombinant mouse IL-2 was purchased from ity after culture for 24 h in the absence or presence of
PeproTech Inc. (Rocky Hill, NJ). Recombinant mouse [6]-gingerol, [8]-gingerol, or [10]-gingerol. Cells were
granulocyte-macrophage colony-stimulating factor washed and resuspended in medium and Trypan Blue
(GM-CSF) was from R&D Systems, Inc. (Minneapolis, dye at a 1:1 ratio (v/v). A hemocytometer and light mi-
MN). Dynabeads® (anti-CD3 and anti-CD28 Ab- croscope were used to count live and dead cells; % live
coated microbeads) were from Invitrogen Canada Inc. cells = C / T × 100, where C is the number of live
(Burlington, ON) unstained cells and T is the total number of cells.

Cell line. IL-2-dependent mouse CTLL-2 CD8+ T lym- Flow cytometry. Cells were washed with ice-cold PBS,
phocytes (Gillis and Smith, 1977) were obtained from resuspended in ice-cold flow cytometry buffer consisting
American Tissue Culture Collection (Manassas, VA) and of 0.2% sodium azide (w/v) and 1% bovine serum albu-
maintained at 37 °C in a humidified incubator with 5% min in PBS, and labeled with the desired fluorochrome-
CO2. CTLL-2 cells were cultured in RPMI 1640 medium conjugated Ab or isotype-matched Ab (10 μg/mL) for
(Sigma-Aldrich Canada) supplemented with 5% heat- 30 min at 4 °C in the dark. Cells were then washed twice
inactivated fetal calf serum, 1% penicillin–streptomycin, with flow cytometry buffer, fixed with 1% paraformal-
2 mM L-glutamine, and 5 mM HEPES (Invitrogen Canada dehyde (v/v) in PBS, and analyzed with a FACSCalibur
Inc.), hereafter referred to as complete RPMI 1640 me- flow cytometer (BD Biosciences). Data were processed
dium, and 50 U/mL recombinant mouse IL-2. using FCS Express software (version 3.0; De Novo Soft-
ware, Thornhill, ON).

T lymphocyte isolation. Spleens were harvested from eu-


thanized C57BL/6 mice and homogenized in ice-cold PBS Cytokine measurement. T lymphocytes in complete
to create a single cell suspension. Spleen cells were washed RPMI 1640 medium were plated in triplicate at
in PBS, and erythrocytes were removed by hypo-osmotic 2.5 × 105 cells/well into 96-well round-bottom plates
shock. CD3+ T lymphocytes were isolated using the Pan and treated as indicated. After 24 h culture, cell-free
T Cell Isolation MACS® kit from Miltenyi Biotech supernatants were collected and cytokine content was
Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)
T LYMPHOCYTE INHIBITION BY GINGEROLS 1709

determined by enzyme-linked immunosorbent assay. In- to provide costimulatory signals (Fig. 1B). Interestingly,
terferon (IFN)-γ and IL-2 were measured using kits [10]-gingerol showed increased inhibitory activity under
from BD Biosciences (Mississauga, ON). IL-4 was mea- these conditions. There was no significant effect of
sured with a kit from eBioscience Inc. (San Diego, CA). gingerols at the concentrations used in this study on T
Absorbance was read at 450 nm on an Expert 96 Micro- lymphocyte and dendritic cell viability after 24-h culture
plate Reader (Biochrom ASYS, Cambridge, UK) and (data not shown).
the concentration of cytokine in pg/mL was determined
using SOFTmax® Pro software (version 4.3; Molecular
Devices Corp. Sunnyvale, CA).
Effect of [6]-gingerol, [8]-gingerol, and [10]-gingerol on
CD25 and CD69 expression

Statistical analysis. Statistical analysis was performed We next examined the effect of gingerols on the expres-
using a one-way analysis of variance (ANOVA) with a sion of CD25 and CD69, which are markers of T lym-
Tukey–Kramer multiple parameters post-test using phocyte activation (Biselli et al., 1992). Fig. 2 shows
GraphPad Prism analysis software (GraphPad Software that the amount of CD25 expressed by Dynabead®-
Inc., La Jolla, CA). Data were considered statistically stimulated T lymphocytes was decreased in the presence
significant when p < 0.05. of [6]-gingerol, [8]-gingerol, or [10]-gingerol, but was
only statistically significant for 50-μM [8]-gingerol and
[10]-gingerol. The percentage of CD25-expressing T
lymphocytes following Dynabead® stimulation was not
RESULTS substantially affected by any of the gingerols. Similar re-
sults were obtained for CD69 expression, i.e. 50-μM [8]-
gingerol and [10]-gingerol caused a significant reduction
Effect of [6]-gingerol, [8]-gingerol, and [10]-gingerol on
in CD69 expressed by Dynabead®-stimulated T lympho-
T lymphocyte proliferation
cytes while none of the gingerols affected the percentage
of CD69-expressing T lymphocytes.
To determine the effect of gingerols with different chain
lengths on activation-induced T lymphocyte prolifera-
tion in the absence of antigen-presenting cells, mouse
T lymphocytes were stimulated with Dynabeads® in Effect of [6]-gingerol, [8]-gingerol, and [10]-gingerol on
the absence or presence of different concentrations of T lymphocyte cytokine synthesis
[6]-gingerol, [8]-gingerol, or [10]-gingerol. DNA synthe-
sis after 48, 72, and 96 h of culture was measured by [3H] Significant inhibition of IFN-γ production by
TdR incorporation. As shown in Fig. 1A, there was a Dynabead®-stimulated T lymphocytes was observed
time- and dose-dependent decrease in the proliferation with 25 or 50-μM [6]-gingerol, [8]-gingerol, or [10]-
of gingerol-treated T lymphocytes, regardless of the gingerol (Fig. 3A). In contrast, only [8]-gingerol and
gingerol chain length. However, at a concentration of [10]-gingerol at 50 μM showed significant inhibition of
100 μM, both [8]-gingerol and [10]-gingerol were more IL-2 production by Dynabead®-stimulated T lympho-
inhibitory than [6]-gingerol. Similar results were ob- cytes (Fig. 3B). None of the gingerols were able to in-
tained when T lymphocytes were stimulated with anti- hibit IL-4 synthesis by Dynabead®-stimulated T
TCRβ Ab in the presence of syngeneic dendritic cells lymphocytes (Fig. 3C).

Figure 1. Gingerols inhibit DNA synthesis by activated T lymphocytes. (A) T lymphocytes were cultured in the presence of the DMSO vehicle
or the indicated concentrations of [6]-, [8]-, or [10]-gingerol, and stimulated with Dynabeads® for 48, 72, and 96 h. (B) T lymphocytes were
co-cultured with syngeneic dendritic cells in the presence of the DMSO vehicle or the indicated concentrations of [6]-gingerol, [8]-gingerol, or
3
[10]-gingerol, and stimulated with anti-TCRβ Ab for 48, 72, and 96 h. (A, B) Cultures were pulsed with [ H]TdR for the last 6 h of culture and
harvested for liquid scintillation counting. Data shown are the average cpm ± SEM of at least three independent experiments; * p < 0.05
compared to the vehicle at the corresponding time point by one-way ANOVA with a Tukey–Kramer multiple parameters post-test.

Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)
1710 M. BERNARD ET AL.

Figure 2. [8]-Gingerol and [10]-gingerol inhibit T lymphocyte expression of CD25 and CD69. T lymphocytes were cultured in the presence of
the DMSO vehicle or the indicated concentrations of [6]-gingerol, [8]-gingerol, or [10]-gingerol, and stimulated with Dynabeads® for 24 h. T
lymphocytes were then stained with phycoerythrin-conjugated anti-CD25 Ab, fluorescein isothiocyanate-conjugated anti-CD69 Ab, or the
appropriate isotype control Ab, fixed, and analyzed by flow cytometry. Data shown are the median fluorescence intensities (MFI) and the
% of (A) CD25-positive T cells or (B) CD69-positive T cells ± SEM from at least three independent experiments; * p < 0.05 when compared
to the vehicle at the corresponding time point by one-way ANOVA with a Tukey–Kramer multiple parameters post-test.

Effect of exogenous IL-2 on gingerol-mediated (vehicle—27 123 ± 3582 cpm versus [6]-gingerol—24 712
inhibition of T lymphocyte proliferation ± 3448 cpm, p > 0.05). Inhibition of IL-2-dependent
CTLL-2 proliferation by [8]-gingerol and [10]-gingerol
Because [8]-gingerol and [10]-gingerol inhibited IL-2 was not because of a reduction in IL-2 receptor expres-
synthesis, we explored the effect of exogenous IL-2 on sion because constitutive expression of CD25 by CTLL-
the Dynabead®-stimulated proliferation of gingerol- 2 cells was not decreased in the presence of 100-μM [8]-
treated T lymphocytes. As shown in Fig. 4, the addition gingerol or [10]-gingerol (data not shown).
of IL-2, at concentrations that are typically present in
activated T lymphocyte cultures, failed to substantially
affect the inhibition of Dynabead®-stimulated T lym-
phocyte proliferation caused by [8]-gingerol and [10]- DISCUSSION
gingerol; however, IL-2 enhanced the proliferation of
T lymphocytes that were stimulated with Dynabeads® Gingerols have potent antiinflammatory activities that may
in the presence of [6]-gingerol, in spite of the failure of be beneficial in individuals with chronic inflammatory
[6]-gingerol to decrease IL-2 production by these cells. disorders (Tripathi et al., 2007; Dugasani et al., 2010). How-
ever, most studies to date have focused on the effect of
[6]-gingerol on pro-inflammatory functions of macrophages
Effect of [6]-gingerol, [8]-gingerol, and [10]-gingerol on with little attention being paid to the impact of gingerols on
IL-2 receptor signaling T lymphocytes, which contribute to the pathogenesis of
many chronic inflammatory diseases (Cosmi et al., 2014).
To determine whether any of the gingerols affected IL-2 This lack of information prompted us to compare the
receptor signaling, we cultured IL-2-dependent CTLL-2 effects of [6]-gingerol, [8]-gingerol, and [10]-gingerol on
cells in the presence of 50 U/mL exogenous IL-2 with or the activation and effector function of T lymphocytes.
without increasing concentrations of [6]-gingerol, [8]- Non-cytotoxic concentrations of all three gingerols
gingerol, or [10]-gingerol. DNA synthesis was measured inhibited mouse T lymphocyte proliferation induced by
after 48-h culture by [3H]TdR incorporation. There was Dynabeads®, although [8]-gingerol and [10]-gingerol
a statistically significant inhibition of IL-2-dependent were generally more potent than [6]-gingerol. Gingerols
CTLL-2 cell proliferation in the presence of 100-μM with different unbranched alkyl side chain lengths there-
[8]-gingerol (vehicle—26 626 ± 3611 cpm versus [8]- fore have a direct inhibitory effect on T lymphocytes,
gingerol—4772 ± 1802 cpm, p < 0.005) and [10]-gingerol which is consistent with [8]-gingerol-mediated inhibition
(vehicle—27 123 ± 3582 cpm versus [10]-gingerol—5531 of concanavalin A-driven mouse splenocyte prolifera-
± 1396 cpm, p < 0.01). In contrast, 100-μM [6]-gingerol tion (Lu et al., 2011). Interestingly, only [10]-gingerol
did not affect IL-2-induced proliferation of CTLL-2 cells showed a further increase in inhibitory activity when T
Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)
T LYMPHOCYTE INHIBITION BY GINGEROLS 1711

Figure 3. Differential effect of gingerols on T lymphocyte cytokine synthesis. T lymphocytes were cultured in the presence of the DMSO ve-
hicle or the indicated concentrations of [6]-gingerol, [8]-gingerol, or [10]-gingerol, and stimulated with Dynabeads® for 24 h. Cell-free culture
supernatants were then collected for analysis by enzyme-linked immunosorbent assay. Data shown are the mean cytokine production of (A)
IFN-γ, (B) IL-2, and (C) IL-4, normalized to the medium control ± SEM from at least three independent experiments; * p < 0.05 when com-
pared to the vehicle at the corresponding time point by one-way ANOVA with a Tukey–Kramer multiple parameters post-test.

lymphocytes were stimulated with anti-TCRβ Ab in the α subunit of the high affinity interleukin-2 receptor
the presence of syngeneic dendritic cells to provide (Gaffen, 2001), it follows that T lymphocyte utilization
costimulatory signals, suggesting possible inhibition of of IL-2 will be impaired in the presence of [8]-gingerol
dendritic cell costimulatory function in addition to direct or [10]-gingerol. Furthermore, both [8]-gingerol and
inhibition of T lymphocytes. [10]-gingerol inhibited the IL-2 synthesis by T lympho-
We also observed substantial inhibition of activation- cytes. Because IL-2 drives the clonal expansion of T
induced T lymphocyte expression of CD25 and CD69 lymphocytes (Bayer et al., 2013), diminished utilization
by [8]-gingerol and [10]-gingerol, but not [6]-gingerol, of limited amounts of IL-2 likely accounts for much of
which was in line with the stronger anti-proliferative ef- the inhibitory effect of [8]-gingerol and [10]-gingerol
fect of [8]-gingerol and [10]-gingerol. Because CD25 is on T lymphocyte proliferation. However, [8]-gingerol

Figure 4. Differential effect of exogenous IL-2 on gingerol-mediated inhibition of DNA synthesis by T lymphocytes. T lymphocytes were cul-
tured in the presence of the DMSO vehicle or 50-μM [6]-gingerol, [8]-gingerol, or [10]-gingerol without or with the indicated concentrations
3
of IL-2 and stimulated with Dynabeads® for 96 h. Cultures were pulsed with [ H]TdR for the last 6 h of culture and harvested for liquid scin-
tillation counting. Data shown are the average cpm ± SEM of at least three independent experiments; * p < 0.05 compared to the vehicle at
the corresponding time point by one-way ANOVA with a Tukey–Kramer multiple parameters post-test, n.s. denotes not significant.

Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)
1712 M. BERNARD ET AL.

and [10]-gingerol also interfered with IL-2 receptor sig- et al., 2008) could be because of gingerol-mediated inhibi-
naling, which accounts for the failure of exogenous IL-2 tion of IFN-γ production by OVA-reactive T lympho-
to restore the proliferative response of gingerol-treated cytes. However, it is important to note that IFN-γ
T lymphocytes. production by Th1 cells is also important in protection
The profound inhibitory effect of [8]-gingerol and against intracellular pathogens such as viruses
[10]-gingerol on IL-2 synthesis and signaling suggests a (Romagnani, 1994), which our data suggest might be neg-
potential impact on regulatory T lymphocytes, which re- atively impacted by gingerols. In addition, the failure of
quire IL-2 for their development and immunosuppressive [6]-gingerol, [8]-gingerol, and [10]-gingerol to interfere
function (de la Rosa et al., 2004; Burchill et al., 2007). Such with IL-4 synthesis in our hands suggests that another
an effect could at least partially counteract the inhibitory component of ginger extract is responsible for interfering
effect of [8]-gingerol and [10]-gingerol on chronic inflam- with the generation of Th2 cytokines during allergic air-
mation caused by inappropriate or excessive T lym- way inflammation (Ahui et al., 2008). Because the NF-κ
phocyte activation and effector function. In contrast, B family of transcription factors are essential for the acti-
[6]-gingerol did not substantially affect IL-2 synthesis, in- vation of T lymphocytes (Ruland and Mak, 2003), and
ducible CD25 expression, or IL-2 receptor signaling, and [6]-gingerol, [8]-gingerol, and [10]-gingerol inhibit NF-κ
the addition of exogenous IL-2 to T lymphocytes that B in other cell types (Kim et al., 2004; Ho et al., 2013),
were stimulated in the presence of an inhibitory concen- it seems likely that gingerol-mediated inhibition of NF-κ
tration of [6]-gingerol resulted in increased DNA synthe- B is at least in part responsible for the direct suppression
sis. Although our data suggest that [6]-gingerol should not of T lymphocyte activation and effector function by
interfere with regulatory T lymphocytes, tumor-bearing [6]-gingerol, [8]-gingerol, and [10]-gingerol.
mice treated with [6]-gingerol have a reduced number of In conclusion, the present study has established that
tumor-infiltrating regulatory T lymphocytes (Ju et al., gingerols with different unbranched alkyl side chain
2012). In contrast, numbers of tumor-infiltrating CD4+ lengths directly inhibit mouse T lymphocyte prolifera-
and CD8+ T lymphocytes are dramatically increased fol- tion and cytokine production, albeit with some notable
lowing administration of [6]-gingerol; however, it is not differences in potency and effects. Our findings suggest
clear whether this effect was because of [6]-gingerol or a that gingerols should be further investigated for their
[6]-gingerol metabolite (Nakazawa and Ohsawa, 2002). possible use in the treatment of autoimmune diseases
T lymphocytes are an important source of cytokines and other chronic inflammatory disorders caused by
that drive immune responses; IFN-γ and IL-4 are repre- inappropriate or excessive T lymphocyte responses.
sentative of Th1 and Th2 responses, respectively (Zhu
et al., 2010). Interestingly, [6]-gingerol, [8]-gingerol and
[10]-gingerol inhibited T lymphocyte synthesis of IFN-γ Acknowledgements
whereas none of these gingerols affected IL-4 produc-
tion. These gingerols therefore have the potential to pref- This work was supported by a grant to DH from the Natural Sciences
erentially inhibit Th1 responses that contribute to the and Engineering Research Council of Canada.
development of certain autoimmune diseases and other
chronic inflammatory disorders (Cosmi et al., 2014).
Our findings also suggest that reduced IFN-γ levels Conflict of Interest
reported in bronchoalveolar lavage fluids from
OVA-immunized and OVA-challenged mice (Ahui The authors have declared that there is no conflict of interest.

REFERENCES
Ahui MLB, Champy P, Ramadan A, et al. 2008. Ginger prevents lymphocyte culture of normal murine spleen cells. J Exp Med
Th2-mediated immune responses in a mouse model of airway 146: 468–482.
inflammation. Int Immunopharmacol 8: 1626–32. Grzanna R, Lindmark L, Frondoza CG. 2005. Ginger—an herbal
Bayer AL, Pugliese A, Malek TR. 2013. The IL-2/IL-2R system: medicinal product with broad anti-inflammatory actions.
from basic science to therapeutic applications to enhance J Med Food 8: 125–132.
immune regulation. Immunol Res 57: 197–209. Hasler CM, Bloch AS, Thomson CA, et al. 2004. Position of the
Biselli R, Matricardi PM, D’Amelio R, et al. 1992. Multiparametric American Dietetic Association: functional foods. J Am Diet
flow cytometric analysis of the kinetics of surface molecule Assoc 104: 814–826.
expression after polyclonal activation of human peripheral Ho SC, Chang KS, Lin CC. 2013. Anti-neuroinflammatory capacity
blood T lymphocytes. Scand J Immunol 35: 439–447. of fresh ginger is attributed mainly to 10-gingerol. Food Chem
Burchill MA, Yang J, Vogtenhuber C, et al. 2007. IL-2 receptor 141: 3183–3191.
β-dependent STAT5 activation is required for the development Jolad SD, Lantz RC, Solyom AM, et al. 2004. Fresh organically grown
+
of Foxp3 regulatory T cells. J Immunol 178: 280–290. ginger (Zingiber officinale): composition and effects on LPS-
Butt MS, Sultan MT. 2011. Ginger and its health claims: molecular induced PGE2 production. Phytochemistry 65: 1937–1954.
aspects. Crit Rev Food Sci Nutr 51: 383–393. Ju SA, Park SM, Lee YS, et al. 2012. Administration of 6-gingerol
Cosmi L, Maggi L, Santarlasci V, et al. 2014. T helper cells plastic- greatly enhances the number of tumor-infiltrating lympho-
ity in inflammation. Cytom Part A 85: 36–42. cytes in murine tumors. Int J Cancer 130: 2618–2628.
De la Rosa M, Rutz S, Dorninger H, et al. 2004. Interleukin-2 is Kim SO, Chun K-S, Kundu JK, et al. 2004. Inhibitory effects of [6]-
+ +
essential for CD4 CD25 regulatory T cell function. Eur gingerol on PMA-induced COX-2 expression and activation of
J Immunol 34: 2480–2488. NF-κB and p38 MAPK in mouse skin. Biofactors 21: 27–31.
Dugasani S, Pichika MR, Nadarajah VD, et al. 2010. Comparative anti- Lu J, Guan S, Shen X, et al. 2011. Immunosuppressive activity of
oxidant and anti-inflammatory effects of [6]-gingerol, [8]-gingerol, 8-gingerol on immune responses in mice. Molecules 16:
[10]-gingerol and [6]-shogaol. J Ethnopharmacol 127: 515–520. 2636–2645.
Gaffen SL. 2001. Signaling domains of the interleukin 2 receptor. Nakazawa T, Ohsawa K. 2002. Metabolism of [6]-gingerol in rats.
Cytokine 14: 63–77. Life Sci 70: 2165–2175.
Gillis S, Smith KA. 1977. In vitro generation of tumor-specific Romagnani S 1994. Lymphokine production by human T cells in
cytotoxic lymphocytes. Secondary allogeneic mixed tumor disease states. Annu Rev Immunol 12: 227–257.

Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)
T LYMPHOCYTE INHIBITION BY GINGEROLS 1713

Ruland J, Mak TW. 2003. Transducing signals from antigen Tripathi S, Bruch D, Kittur DS. 2008. Ginger extract inhibits LPS
receptors to nuclear factor κB. Immunol Rev 193: 93–100. induced macrophage activation and function. BMC Complement
Tripathi S, Maier KG, Bruch D, et al. 2007. Effect of 6-gingerol on Altern Med 8: 1.
pro-inflammatory cytokine production and costimulatory mol- Zhu J, Yamane H, Paul WE. 2010. Differentiation of effector CD4 T
ecule expression in murine peritoneal macrophages. J Surg cell populations. Annu Rev Immunol 28: 445–489.
Res 138: 209–213.

Copyright © 2015 John Wiley & Sons, Ltd. Phytother. Res. 29: 1707–1713 (2015)

You might also like