ArticlePDF Available

Identification and characterization of [6]-shogaol from ginger as inhibitor of vascular smooth muscle cell proliferation

Authors:

Abstract and Figures

ScopeVascular smooth muscle cell (VSMC) proliferation is involved in the pathogenesis of cardiovascular disease, making the identification of new counteracting agents and their mechanisms of action relevant. Ginger and its constituents have been reported to improve cardiovascular health, but no studies exist addressing a potential interference with VSMC proliferation.Methods and resultsThe dichloromethane extract of ginger inhibited VSMC proliferation when monitored by resazurin metabolic conversion (IC50 = 2.5 μg/mL). The examination of major constituents from ginger yielded [6]-shogaol as the most active compound (IC50 = 2.7 μM). In the tested concentration range [6]-shogaol did not exhibit cytotoxicity toward VSMC and did not interfere with endothelial cell proliferation. [6]-shogaol inhibited DNA synthesis and induced accumulation of the VSMC in the G0/G1 cell-cycle phase accompanied with activation of the nuclear factor-erythroid 2-related factor 2 (Nrf2)/HO-1 pathway. Since [6]-shogaol lost its antiproliferative activity in the presence of the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin IX, HO-1 induction appears to contribute to the antiproliferative effect.Conclusion This study demonstrates for the first time inhibitory potential of ginger constituents on VSMC proliferation. The presented data suggest that [6]-shogaol exerts its antiproliferative effect through accumulation of cells in the G0/G1 cell-cycle phase associated with activation of the Nrf2/HO-1 pathway.
Content may be subject to copyright.
Mol. Nutr. Food Res. 2015, 59, 843–852 843
DOI 10.1002/mnfr.201400791
RESEARCH ARTICL E
Identification and characterization of [6]-shogaol from
ginger as inhibitor of vascular smooth muscle cell
proliferation
Rongxia Liu1,2,ElkeH.Heiss
1, Nadine Sider1, Andreas Schinkovitz3, Barbara Groblacher3,
Dean Guo2, Franz Bucar3, Rudolf Bauer3, Verena M. Dirsch1and Atanas G. Atanasov1
1Department of Pharmacognosy, University of Vienna, Vienna, Austria
2Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory
for TCM Standardization Technology, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological
Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
3Institute of Pharmaceutical Sciences, Department of Pharmacognosy, Karl-Franzens-University Graz, Graz,
Austria
Received: November 3, 2014
Revised: January 16, 2015
Accepted: January 16, 2015
Scope: Vascular smooth muscle cell (VSMC) proliferation is involved in the pathogenesis
of cardiovascular disease, making the identification of new counteracting agents and their
mechanisms of action relevant. Ginger and its constituents have been reported to improve
cardiovascular health, but no studies exist addressing a potential interference with VSMC
proliferation.
Methods and results: The dichloromethane extract of ginger inhibited VSMC proliferation
when monitored by resazurin metabolic conversion (IC50 =2.5 g/mL). The examination of ma-
jor constituents from ginger yielded [6]-shogaol as the most active compound (IC50 =2.7 M).
In the tested concentration range [6]-shogaol did not exhibit cytotoxicity toward VSMC and
did not interfere with endothelial cell proliferation. [6]-shogaol inhibited DNA synthesis and
induced accumulation of the VSMC in the G0/G1cell-cycle phase accompanied with activa-
tion of the nuclear factor-erythroid 2-related factor 2 (Nrf2)/HO-1 pathway. Since [6]-shogaol
lost its antiproliferative activity in the presence of the heme oxygenase-1 (HO-1) inhibitor tin
protoporphyrin IX, HO-1 induction appears to contribute to the antiproliferative effect.
Conclusion: This study demonstrates for the first time inhibitory potential of ginger con-
stituents on VSMC proliferation. The presented data suggest that [6]-shogaol exerts its antipro-
liferative effect through accumulation of cells in the G0/G1cell-cycle phase associated with
activation of the Nrf2/HO-1 pathway.
Keywords:
Ginger / Heme oxygenase-1 / Nrf2 / [6]-Shogaol / Vascular smooth muscle cell
Correspondence: Dr. Atanas G. Atanasov, Department of Phar-
macognosy, Faculty of Life Sciences, University of Vienna, Al-
thanstrasse 14, A-1090 Vienna, Austria
E-mail: atanas.atanasov@univie.ac.at
Abbreviations: BrdU, bromodeoxyuridine; EBM, endothelial cell
basal medium; FCS, fetal calf serum; HO-1, heme oxygenase-1;
HUVEC, human umbilical vein endothelial cells; LDH, lactate de-
hydrogenase; MEF, mouse embryonic fibroblasts; Nrf2, nuclear
factor-erythroid 2-related factor 2; PDGF, platelet-derived growth
factors; PI, propidium iodide; SAR, structure-activity-relationship;
VSMC, vascular smooth muscle cell; WT, wild type
1 Introduction
Ginger, Zingiber officinale Roscoe (Zingiberaceae), is widely
used as a spice in foods and beverages, and has also a long
history of use in traditional medicine for the treatment of
inflammation, rheumatic disorder, indigestion, vomiting,
and fever, among others [1–4]. Many pharmacological activ-
ities have been reported for this plant and its major pungent
principles, including anti-inflammatory, anti-tumorigenic,
anti-apoptotic, anti-hyperglycemic, cancer-chemopreventive,
anti-lipidemic, and anti-emetic effects [4–9].
Additional corresponding author: Dr. Elke H. Heiss
E-mail: elke.heiss@univie.ac.at
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in
any medium, provided the original work is properly cited.
844 R. Liu et al. Mol. Nutr. Food Res. 2015, 59, 843–852
Cardiovascular disease is the number one cause of death
in the world, mainly elicited by atherosclerosis together with
hypertension [10]. Aberrant and accelerated vascular smooth
muscle cell (VSMC) proliferation not only contributes to ini-
tial atherosclerotic plaque formation but also to restenosis
(pathological renarrowing of the vessel lumen) after surgical
interventions like percutaneous transluminal coronary an-
gioplasty or bypass surgery. To overcome restenosis, drug-
eluting stents have been developed, aiming at inhibiting
VSMC growth by the release of antiproliferative substances.
The most prominent drugs used in drug-eluting stents so
far have been paclitaxel (a microtubules stabilizing agent)
and sirolimus (a mTOR inhibitor). These compounds, how-
ever, exhibit a number of unresolved drug-related issues such
as impaired reendothelialization and delayed thrombosis in-
duction [11,12], which makes the discovery of novel effective
compounds and molecular mechanisms suppressing VSMC
proliferation highly relevant.
Plant derived natural products proved to be an excellent
resource for the identification of new lead compounds [13].
While ginger is reported to possess vasoprotective effects
[14, 15], its impact on VSMC proliferation in particular has
not been studied so far. In this study, we therefore examined
the antiproliferative potential of ginger extract and some
of its major active components ([6]-gingerol, [6]-shogaol,
zingerone, [6]-paradol, and rac-[6]-dihydroparadol) in VSMC,
and characterized in more detail the cellular mode of action
of the most active identified compound, [6]-shogaol.
2 Materials and methods
2.1 Chemicals and reagents
Primary rat aortic VSMC were purchased from Lonza
(Braine-L’Alleud, Belgium) and the human umbilical
vein endothelial cells (HUVECtert) were provided by
Dr. Hannes Stockinger (Medical University of Vienna, Aus-
tria) [16]. Wild type (WT) and isogenic nuclear factor-erythroid
2-related factor 2 (Nrf2)–/– mouse embryonic fibroblasts
were kindly provided by Dr. T. Kensler, University of
Pittsburgh, USA. Platelet-derived growth factors (PDGF)-BB
was supplied from Bachem (Weilheim, Germany). The heme
oxygenase-1 (HO-1) inhibitor tin protoporphyrin IX dichlo-
ride was from Enzo Life Sciences (Lausen, Switzerland). [6]-
Parodol and rac-[6]-dihydroparadol were isolated as described
[17]. All other used reagents were from analytical grade and
obtained from Sigma-Aldrich (Vienna, Austria). The anti-HO-
1 antibody was from Stressgene (purchased via Enzo, Lausen,
Switzerland), the anti-actin antibody [mouse anti-actin,
monoclonal (Clone: C4); #69100] was from mpbio (Eschwege,
Germany), and the secondary horseradish-peroxidase-
coupled antibodies came from Cell Signaling (Heidelberg,
Germany).
2.2 Ginger extraction
Root material of Zingiber officinale (Zingiberaceae) was pur-
chased from PLANTASIA (Oberndorf, Austria). A voucher
sample is kept at the University of Graz, Department of
Pharmacogonosy under reference number 730149. Ground
root material (3.17 g) was mixed with 0.97 g diatomaceous
earth (Dionex Corporation, Sunnyvale, CA, USA) before
being extracted with HPLC grade dichloromethane using an
Accelerated Solvent Extractor (ASE 200, Dionex Corporation,
Sunnyvale, CA, USA). Extraction conditions were as follows:
Temperature: 44C, cell preheating time: 1 min, pressure:
68.9 bar, static time: 5 min, flush volume: 150%. Three
extraction cycles were performed yielding 168 mg of dry
extract (5.3%) after solvent evaporation.
2.3 Cell culture
VSMC were cultivated in DMEM–F12 (1:1) supplemented
with 20% fetal calf serum (FCS), 30 g/mL gentamicin, and
15 ng/mL amphotericin B. VSMC with passage number be-
tween 6 and 14 were used in this study. HUVEC were grown
in endothelial cell basal medium (EBM) supplemented with
10% FCS, 100 U/mL penicillin, 100 g/mL streptomycin,
1% amphotericin B and EBMTM SingleQuotsR, containing re-
combinant human epidermal growth factor, hydrocortisone,
gentamicin sulfate, amphotericin B and 0.4% bovine brain
extract. Mouse embryonic fibroblasts (MEF) were grown in
DMEM supplemented with 10% FCS, 100 U/mL penicillin,
and 100 g/mL streptomycin.
2.4 Resazurin conversion assay
VSMC were seeded in 96-well plates at 5 ×103cells/well.
After 24 h, cells were serum-starved for 24 h to render them
quiescent. Quiescent cells were pretreated for 30 min with
ginger extract, compounds, or vehicle (0.1% DMSO) as indi-
cated, and subsequently stimulated for 48 h with PDGF-BB
(20 ng/mL). To measure the number of metabolically active
VSMC by resazurin conversion [18, 19], cells were washed
with PBS and incubated in serum-free medium containing
10 g/mL resazurin for 2 h. Total metabolic activity was
measured by monitoring the increase in fluorescence at a
wavelength of 590 nm using an excitation wavelength of
535 nm in a 96-well plate reader (Tecan GENios Pro).
HUVECtert cells [16, 20] were seeded in 96-well plates at
5×103cells/well. After 24 h, HUVECtert cells were treated
with compounds or vehicle (0.1% DMSO) as indicated and
incubated for 48 h. Then, cells were washed with PBS and in-
cubated in culture medium containing 10 g/mL resazurin
for 2 h. The detection step is performed as described above.
2.5 Crystal violet biomass staining
VSMC were seeded in 96-well plates at 5 ×103cells/well.
Twenty-four hours later, cells were serum starved for 24 h
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2015, 59, 843–852 845
to render them quiescent. Quiescent cells were pretreated
for 30 min with ginger extract, compounds, or vehicle (0.1%
DMSO) as indicated and subsequently stimulated for 48 h
with PDGF-BB (20 ng/mL). To determine the total biomass
by crystal violet staining, cells were then incubated in 100 L
of crystal violet staining solution (0.5% crystal violet, 20%
methanol) for 15 min and then washed with ddH2O. After
drying, 100 L EtOH/Na-citrate solution were added (EtOH:
0.1M Na-citrate =1:1) and the absorbance of the samples was
measured at 595 nm in a 96-well plate reader (Tecan sunrise).
2.6 5-Bromo-2-deoxyuridine (BrdU) incorporation
assay
VSMC were seeded in 96-well plates at 5 ×103cells/well.
Twenty-four hours later, cells were serum starved for 24 h to
render them quiescent. Quiescent cells were pretreated for
30 min with compounds, or vehicle (0.1% DMSO) as
indicated and subsequently stimulated with PDGF-BB
(20 ng/mL). To estimate de novo DNA synthesis in VSMC
[21, 22], BrdU was added 2 h after PDGF stimulation, and
the incorporation amount was determined 22 h afterwards
according to the manufacturer’s instructions (Roche Diag-
nostics).
2.7 Assessment of cytotoxicity
VSMC were seeded in 96-well plates at 5 ×103cells/well.
Twenty-four hours later, cells were serum starved for 24 h
to render them quiescent. Quiescent cells were pretreated
for 30 min with compounds, or vehicle (0.1% DMSO) as
indicated, and subsequently stimulated for 24 h with PDGF-
BB (20 ng/mL). Loss of cell membrane integrity as a sign
for cell death can be quantified by the release of the soluble
cytosolic protein lactate dehydrogenase (LDH) [20, 23]. For
this, the supernatant of the treated cells was assessed for LDH
activity. For estimation of the total LDH, identically treated
samples were incubated for 45 min in the presence of 1%
Triton X-100. The released and total LDH enzyme activity was
measured for 30 min at the dark in the presence of 4.5 mg/mL
lactate, 0.56 mg/mL NAD+, 1.69 U/mL diaphorase, 0.004%
w/v BSA, 0.15% w/v sucrose, and 0.5 mM 2-p-iodophenyl-3-
nitrophenyl tetrazolium chloride. The enzyme reaction was
stopped with 1.78 mg/mL oxymate and the absorbance was
measured at 490 nm. Potential effects on cell viability were
estimated as percentage of extracellular LDH enzyme activity.
The cytotoxic natural product digitonin (100 g/mL) was used
as a positive control.
2.8 Cell-cycle analysis
VSMC were seeded in 12-well plates at 1 ×104cells/well.
Twenty-four hours later, cells were serum starved for 24 h
to render them quiescent. Quiescent cells were preincu-
bated with [6]-shogaol (10 M) or vehicle (1% DMSO) for
30 min. PDGF-BB (20 ng/mL) was added, and 16 h later
cells were trypsinized, washed once with PBS, and resus-
pended in a hypotonic propidium iodide (PI) solution con-
taining 0.1% v/v Triton X-100, 0.1% w/v sodium citrate,
and 50 g/mL PI. After incubation at 4C overnight, PI-
stained nuclei were analyzed by flow cytometry (excitation
488 nm, emission 585 nm; FACScalibur; BD Biosciences,
Germany).
2.9 Immunoblot analysis
Cells (MEF or VSMC) were seeded onto six-well plates
((3–4) ×105cells/well). Twenty-four hours later, MEF were
treated with vehicle (0.1% DMSO) or [6]-shogaol at the indi-
cated concentrations for 20 h; VSMC were serum starved for
another 24 h, then quiescent cells were pretreated for 30 min
with vehicle (0.1% DMSO) or [6]-shogaol as indicated and
subsequently stimulated for 20 h with PDGF-BB (20 ng/mL).
Then, cells were lysed and protein extracts were subjected
to SDS-PAGE electrophoresis and immunoblot analysis as
described [20, 22]. Proteins were visualized using enhanced
chemiluminescence reagent and an LAS-3000 luminescent
image analyzer (Fujifilm) with AIDA software (Raytest) for
densitometric evaluation.
2.10 Cell counting
VSMC and HUVECtert were seeded and treated as described
in chapter 2.4 (“Resazurin conversion assay”). Cell numbers
were determined at different time points (with time point
zero corresponding to the treatment with the solvent vehicle,
0.1% DMSO) upon trypan blue staining with a ViCell counter
(Beckman Coulter, Brea, CA).
2.11 Statistical analysis
Statistical analysis was performed by analysis of variance
/Bonferroni test or by ttest (when comparing just two ex-
perimental groups). The number of experiments is given in
the figure legends, and a probability value <0.05 was con-
sidered significant. All tests were performed using GraphPad
PRISM software, version 4.03.
3 Results
3.1 Ginger extract and its major bioactive
components inhibit VSMC proliferation
For evaluating whether ginger contains compounds able
to inhibit PDGF-induced proliferation of VSMC, a
dichloromethane extract of Z. officinale roots was applied
at different concentrations (0.3–30 g/mL; Fig. 1), and the
total amount of metabolically active cells was measured
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
846 R. Liu et al. Mol. Nutr. Food Res. 2015, 59, 843–852
Figure 1. Metabolic activity of VSMC exposed to different con-
centrations of ginger extract. Quiescent VSMC were pretreated
with the indicated concentrations of ginger dichloromethane ex-
tract or an equal volume of the solvent vehicle (0.1% DMSO)
for 30 min, and then stimulated with 20 ng/mL PDGF-BB for
48 h. The metabolic activity was determined at the end of
the stimulation period by the resazurin conversion method. All
values are normalized to the signal obtained from the PDGF-
stimulated, vehicle (0.1% DMSO)-treated cells. The data groups
represent means ±SD from three independent experiments (n.s.,
not significant; ***p<0.001; *p<0.05; analysis of variance
(ANOVA)/Bonferroni).
after 48 h by the resazurin conversion method [18, 19]. The
extract suppressed VSMC proliferation concentration depen-
dently with an IC50 of 2.5 g/mL. The highest tested con-
centration (30 g/mL) decreased the signal to the basal level
of the untreated growth-arrested cells (Fig. 1). To investi-
gate which compounds from ginger are potentially mediat-
ing this antiproliferative effect, five major bioactive ginger
compounds, [6]-gingerol, [6]-shogaol, zingerone, [6]-paradol,
and rac-[6]-dihydroparadol (Fig. 2) were tested. Among these
five compounds, [6]-shogaol showed best antiproliferative ac-
Figure 2. Structures of the major ginger components studied in
this work.
tivity with an IC50 of 2.7 M in the resazurin conversion
assay (Table 1 and Fig. 3A). [6]-gingerol, [6]-paradol, and
rac-[6]-dihydroparadol, also inhibited VSMC proliferation but
less potently than [6]-shogaol (with IC50s in the range 5.3–
13.2 M; Table 1), while zingerone showed no activity up to
100 M. The resazurin conversion method is based on the
Ta b l e 1 . Comparison of the activity of a ginger extract and several major pure compounds in VSMC and HUVECtert. Antiproliferative effects
were evaluated by the resazurin metabolic conversion assay or crystal violet biomass staining. Cell death was evaluated by a
colorimetric LDH activity assay as described in details in the “Materials and methods” section. The data shown represent means
of three to five independent experiments each in triplicate or quadruplet. IC50 were determined by the GraphPad Prism software
version 4.03 (GraphPad Software Inc., USA). ANOVA/Bonferroni analysis was used for evaluation of the statistical significance
(n.s. indicates p>0.05)
VSMCs HUVECs
Resazurin metabolic Crystal violet Cell death Resazurin metabolic
conversion (IC50,g/mL or M) biomass staining (IC50,M) (LDH release) conversion
DCM extract 2.51 g/mL
[6]-Gingerol 13.2 M 8.77 n.s. (up to 100 M) n.s. (up to 30 M)
[6]-Shogaol 2.72 M 1.13 n.s. (up to 10 M) n.s. (up to 10 M)
Zingerone >100 M—
[6]-Paradol 5.3 M 3.34 n.s. (up to 30 M) n.s. (up to 30 M)
rac-[6]-Dihydroparadol 10 M 4.11 n.s. (up to 30 M) n.s. (up to 30 M)
—: not detected.
n.s.: no significant difference.
DCM, dichloromethane.
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2015, 59, 843–852 847
Figure 3. Effect of [6]-shogaol on VSMC proliferation and cell
death. Quiescent VSMC were pretreated with the indicated con-
centrations of [6]-shogaol or an equal volume of the solvent vehi-
cle (0.1% DMSO) for 30 min, and then stimulated with 20 ng/mL
PDGF-BB. Cell proliferation was quantified by the resazurin con-
version method (A) and crystal violet biomass staining (B). Cell
death was estimated by measuring the cell membrane integrity
through quantification of the percentage of the cytosolic enzyme
lactate dehydrogenase (LDH) detected inside cells and in the ex-
tracellular medium (C). The cytotoxic natural product digitonin
(100 g/mL) was used as a positive control in the cell death assay
(C). The data groups represent means ±SD from three indepen-
dent experiments (n.s., not significant; ***p<0.001; **p<0.01;
*p<0.05; ANOVA/Bonferroni).
Figure 4. HUVECtert viability in the presence of [6]-shogaol. HU-
VECtert were treated with the indicated concentrations of [6]-
shogaol or equal volume of the solvent vehicle (0.1% DMSO) for
48 h. Cell numbers were estimated at the end time point by the
resazurin conversion method. All values are normalized to the
signal obtained from vehicle (0.1% DMSO)-treated cells. The an-
tiproliferative natural product paclitaxel (1 M)wasusedaspos-
itive control. The data groups represent means ±SD from three
independent experiments (n.s., not significant; ***p<0.001;
ANOVA/Bonferroni).
metabolic conversion of resazurin, which generally correlates
well to the cell number but could be potentially sensitive to
redox-active chemicals or treatments modulating the cellular
metabolic capacity. To confirm the antiproliferative effects of
the investigated compounds with a method independent of
cell metabolism and redox reactions, we quantified total cel-
lular biomass by crystal violet staining. The obtained results
were in line with the data from the resazurin conversion assay
(Table 1 and Fig. 3B). To further assure that the decreased
VSMC number upon treatment with test compounds is not
due to cytotoxicity, we also quantified cell death by measur-
ing LDH inside cells and in cell supernatants. No significant
changes in cell viability were detected in the investigated con-
centration range (Table 1 and Fig. 3C).
As vascular health also depends on a functional and intact
endothelium, an optimal vasoprotective compound inhibits
VSMC activation, but does not interfere with endothelial
cell viability. We therefore investigated the impact of the
four most active compounds on endothelial viability using
metabolic activity as readout. In contrast to paclitaxel (1 M),
none of the four ginger compounds that clearly influence
VSMC proliferation showed a negative effect on endothelial
cells when used in the same concentration range, (Table 1,
Fig. 4). Interestingly, paclitaxel had an even stronger anti-
proliferative effect in endothelial cells (IC50 =4 nM) than
in VSMC (IC50 =108 nM; not shown). To assure that the
observed differences in the potency of ginger compounds
and paclitaxel in VSMC and endothelial cells are not due to
different growth rates between the two cell types, we have
determined the doubling times of the two cell types under
the used experimental conditions. Cell counting revealed
comparable proliferation rates for the PDGF-BB stimulated
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
848 R. Liu et al. Mol. Nutr. Food Res. 2015, 59, 843–852
Figure 5. Effect of [6]-shogaol on cell-cycle distribution and
DNA synthesis of VSMC. Quiescent VSMC were stimulated with
20 ng/mL PDGF-BB for 20 h, and cell proliferation was quantified
by the BrdU incorporation in newly synthesized DNA in the pres-
ence of the indicated treatments (A); or the cells were incubated
with [6]-shogaol (10 M) or equal volume of the solvent vehicle
(0.1% DMSO) for 30 min, then stimulated with 20 ng/mL PDGF-BB
for 16 h, followed by PI nuclear staining and flow cytometry
analysis (B). The data groups represent means ±SD from three in-
dependent experiments (n.s., not significant; ***p<0.001; **p<
0.01; *p<0.05; ANOVA/Bonferroni (A), 2-tailed paired t-test (B)).
VSMC (cell doubling time 30.1 h) and the HUVECtert (cell
doubling time 33.1 h).
Based on the observed promising antiproliferative profile
of action on the VSMC, we have chosen the most potent iden-
tified compound, [6]-shogaol, for further mechanistic studies.
3.2 [6]-Shogaol leads to accumulation of VSMC in
G0/G1
In order to examine the mechanism of action of [6]-shogaol
in VSMC, we first investigated whether it blocks DNA
synthesis by quantification of BrdU incorporation in treated
cells. [6]-shogaol potently blunted PDGF-stimulated DNA
synthesis in a concentration-dependent manner exhibiting
an IC50 of 3.0 M, which is in the same range as the IC50
values obtained in the resazurin conversion and the crystal
violet assay (Fig. 5A). In line with the observed inhibition
of DNA synthesis, [6]-shogaol (10 M) counteracted the
PDGF-induced transition of VSMC from the G0/G1cell-cycle
phase to the G2/M phase, as revealed by flow cytometry
analysis of PI stained nuclei (Fig. 5B).
3.3 [6]-Shogaol blocks VSMC proliferation by
Nrf2-dependent HO-1 induction
In a consequent experiment the molecular events underly-
ing the observed cell-cycle arrest in VSMC upon shogaol
exposure were investigated. Among other bioactivities, [6]-
shogaol is reported to be an activator of nuclear factor E2
related factor 2 (Nrf2) [24, 25]. Nrf2 is a transcription fac-
tor that is activated by a vast variety of stressors including
oxidative insults, inadequate nutrient supply or electrophilic
agents. Activated Nrf2 then launches a transcriptional pro-
gram that mainly aims at detoxification and cellular stress
resistance including cell-cycle control and metabolic adapta-
tion (e.g. reviewed in [26, 27]). Activated Nrf2 and elevated
expression and activity of the Nrf2-target gene HO-1 have
been linked with reduced VSMC proliferation and preven-
tion of cardiovascular disease [28–31]. We therefore were
prompted to analyse whether Nrf2 activation and subsequent
HO-1 induction could possibly account for the growth arrest
observed in shogaol-treated VSMC. For this, HO-1 expres-
sion in proliferating VSMC upon [6]-shogaol exposure was
evaluated. Concentrations of 3 and 10 M [6]-shogaol signif-
icantly elevated HO-1 levels compared to vehicle treated cells
(Fig. 6A). Moreover, using WT and isogenic Nrf2-/- fibrob-
lasts, a strictly Nrf2-dependent increase of HO-1 upon [6]-
shogaol treatment (Fig. 6B) was observed, confirming Nrf2
activation by [6]-shogaol and excluding involvement of other
transcription factors in the HO-1 induction in fibroblasts and
most likely also in VSMC. In order to prove causality be-
tween the [6]-shogaol-induced HO-1 induction and prolifer-
ation stop in VSMC we made use of the HO-1 inhibitor tin
protoporphyrin IX. As shown in Fig. 6C and D, [6]-shogaol
loses its antiproliferative influence on VSMC in the presence
of the HO-1 inhibitor. These findings indicate that [6]-shogaol
induces HO-1 that then contributes to a reduced proliferation
rate in VSMC.
4 Discussion
The present study reveals for the first time the inhibition
of VSMC proliferation by a ginger extract and four ma-
jor ginger constituents exhibiting IC50 values from 2.72 to
13.2 M. [6]-shogaol, the most potent identified inhibitor,
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2015, 59, 843–852 849
Figure 6. [6]-Shogaol induces HO-1 in Nrf2-dependent manner and HO-1 inhibition abolishes the antiproliferative effect of [6]-shogaol.
(A) Quiescent VSMC were pretreated with the indicated concentrations of [6]-shogaol or an equal volume of the solvent vehicle (0.1%
DMSO) for 30 min, and then stimulated with 20 ng/mL PDGF-BB for 20 h before their lysates were subjected to immunoblot analysis
for HO-1 and actin as a loading control. Representative blots of three performed independent biological experiments (with one technical
replicate) with consistent results are depicted. The bar graph shows compiled densitometric data of all performed experiments (mean ±
SD, n.s., not significant; **p<0.01; *p<0.05; ANOVA/Bonferroni). (B) WT and Nrf2/mouse embryonic fibroblasts were treated with
[6]-shogaol (10 or 3 M) for 24 h before their lysates were subjected to immunoblot analysis for HO-1 and actin as a loading control.
Representative blots are shown, the bar graph depicts compiled densitometric analyses from three biological replicates with one technical
replicate each (mean ±SD, n.s., not significant; ***p<0.001; ANOVA/Bonferroni). (C and D) Quiescent VSMC were pretreated with or
without the HO-1 inhibitor SnPP (10 M) for 1 h, then treated with [6]-shogaol (3 M) or an equal volume of the solvent vehicle (0.1%
DMSO) for 30 min, and then stimulated with 20 ng/mL PDGF-BB. Cell proliferation was determined by the resazurin conversion method.
The data groups represent mean ±SD from three independent experiments (n.s., not significant; ***p<0.001; ANOVA/Bonferroni).
induces growth stop associated with accumulation of cells in
G0/G1and induction of HO-1 expression.
Ginger has been widely used as a culinary spice as well
as in traditional oriental medicine for centuries. In recent
years ginger attracted increasing attention due to a variety
of newly described bioactivities promoting its use as a safe
and effective medicinal plant [1,2,4, 32]. The nonvolatile pun-
gent components, such as gingerols, shogaols, paradols, and
zingerone, have been reported to be responsible for many of
the reported biological effects of the plant [33–35]. In fresh
ginger, gingerols are the major pungent components, with [6]-
gingerol being the most abundant. Gingerols are not stable,
and their dehydration may yield large amounts of shogaols
during prolonged ginger storage. Zingerone also cooccurs
with shogaols in stored ginger. Shogaols may undergo reduc-
tion to form paradols that are also present in ginger. Shogaols
are minor components in fresh ginger, while predominant
pungent constituents in the dried ginger, with the ratio of
[6]-shogaol to [6]-gingerol being around 1:1 in dried ginger
[32–34,36,37]. In commercially available ground ginger pow-
ders the amount of [6]-shogaol was reported to be in the range
of 353–1459 g/g [36, 38]. After a single oral administration
of ginger oleoresin (300 mg/kg) to rats, the peak plasma
concentration of [6]-shogaol was reported to be 0.11 g/L
(equalling 0.40 M) [39]. It is important to note that
the anti-proliferative effect of [6]-shogaol on VSMC was
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
850 R. Liu et al. Mol. Nutr. Food Res. 2015, 59, 843–852
observed at higher concentrations in our study, with the IC50s
being 2.72 M, 1.13 M, and 3.0 M upon quantification of
metabolic activity (Fig. 3A), total biomass (Fig. 3B), and DNA
synthesis (Fig. 5A), respectively. Therefore, although the here
described anti-proliferative potential of [6]-shogaol might be
of relevance upon local application (e.g. as a coating agent in
drug-eluting stents), the question of whether acute or chronic
oral administration of ginger could result in beneficial anti-
restenotic effects requires further research.
Since all investigated ginger constituents are struc-
turally related (Fig. 2), a comparison of their IC50 values
(Table 1) allows deducting a structure-activity relationship
(SAR). All compounds possess a vanillyl moiety (4-hydroxy-
3-methoxyphenyl) and an alkyl side chain, as shown in
Fig. 2. They differ in the substitution pattern of the side
chain at positions C3 or C5. [6]-shogaol, which was most ac-
tive against PDGF-induced VSMC proliferation, possesses an
,-unsaturated ketone at C3. [6]-paradol, only differing from
[6]-shogaol by a saturation of the double bond, showed lower
activity. [6]-gingerol, differing from [6]-paradol by having an
additional hydroxy substituent at C5, showed even less po-
tency. The activity of rac-[6]-dihydroparadol with a hydroxyl
group at C3 was in a similar range as that of [6]-gingerol.
Zingerone, also having a carbonyl group at C3, but a short-
ened and therefore less lipophilic alkyl side chain, showed
little activity. Having all this in mind, the presence of an
,-unsaturated ketone and the length of the alkyl side-chain
seems to have significant impact on the extent of the observed
antiproliferative activity. Interestingly, the investigated com-
pounds showed a similar SAR when being investigated for
anti-oxidative and anti-inflammatory effects [40], cytotoxicity
and apoptosis induction in human promyelocytic leukemia
(HL-60) cells [41], and protection of neuronal cells from -
amyloid insult [42].
Noteworthy, the Michael system of the ,-unsaturated
carbonyl type is involved in Nrf2 activation by [6]-shogaol and
other small molecules [25, 43]. Moreover, HO-1 induction
in VSMC by [6]-shogaol appears to be dependent on Nrf2
suggesting that the capacity of activating Nrf2/HO-1 is the
basis for the observed SAR between the compounds.
Activation of Nrf2/HO-1 signaling also interferes with
VSMC migration (e.g. [44]) and cholesterol accumulation in
macrophages (e.g. [45]), as well as prevents endothelial dys-
function (e.g. [46]) and inflammation (e.g. [47]). These fea-
tures make the hub an attractive target for the prevention of
atherosclerosis by influencing different steps in the etiology
of the disease. Analyzing whether [6]-shogaol could exert such
pleiotropic vasoprotection by activation of Nrf2/HO-1 may
therefore represent an interesting subject of future research.
At this point it should be noted that although the causal-
ity between HO-1 induction and inhibited proliferation was
demonstrated, it cannot be excluded that also other factors
might be involved in the observed antiproliferative activity
of [6]-shogaol. Likewise, inhibition of STAT3 or activation of
PPARwere reported for [6]-shogaol [48, 49] and could also
contribute to the cell-cycle arrest in VSMC [22, 50].
Despite its potent effect on VSMC, [6]-shogaol did ob-
viously not impair endothelial viability and proliferation
(Fig. 4). Reendothelialization is a key step toward success-
ful vascular healing after therapeutic interventions such as
angioplasty or bypass surgery [51, 52]. Currently, the only
clinical available treatment for restenosis is drug-eluting stent
coated with rapamycin or paclitaxel [53]. Both drugs not
only inhibit VSMC, but also endothelial cells, causing im-
paired reendotheliazation and lengthening the healing of the
wounded vessels [54, 55]. Our results on the apparent prefer-
ential activity of ginger compounds towards VSMC (Table 1)
may therefore inspire the future search for novel stent coating
agents. Interestingly, [6]-shogaol exhibited its potent antipro-
liferation effect on VSMC without affecting the proliferation
of endothelial HUVECtert cells.
In conclusion, the present study examines for the first
time the inhibitory potential of ginger and its constituent
[6]-shogaol towards VSMC proliferation. Furthermore, the
structure-function relationship of several closely related gin-
ger constituents is characterized and it is demonstrated that
[6]-shogaol exerts its antiproliferative effect through accu-
mulation of cells in G0/G1associated with the activation of
Nrf2/HO-1 pathway.
This work was supported by European Union Seventh Frame-
work Programme (EU-FP7) Marie Curie Fellowship, by Vienna
University “Back-to-Research Grant” program (to R. Liu), and
by the Austrian Science Fund (FWF): S10704/S10705 (NFN-
project “Drugs from Nature Targeting Inflammation”), P25971-
B23 and P23317-B11.
The authors have declared no conflict of interest.
5 References
[1] Kubra, I. R., Rao, L. J., An impression on current develop-
ments in the technology, chemistry, and biological activities
of ginger (Zingiber officinale Roscoe). Crit. Rev. Food Sci.
Nutr. 2012, 52, 651–688.
[2] Afzal, M., Al-Hadidi, D., Menon, M., Pesek, J., Dhami, M.
S., Ginger: an ethnomedical, chemical and pharmacological
review. Drug Metab. Drug Inter. 2001, 18, 159–190.
[3] Butt, M. S., Sultan, M. T., Ginger and its health claims: molec-
ular aspects. Crit. Rev. Food Sci. Nutr. 2011, 51, 383–393.
[4] Ali, B. H., Blunden, G., Tanira, M. O., Nemmar, A., Some phy-
tochemical, pharmacological and toxicological properties of
ginger (Zingiber officinale Roscoe): a review of recent re-
search. Food Chem. Toxicol. 2008, 46, 409–420.
[5] Miyoshi, N., Nakamura, Y., Ueda, Y., Abe, M. et al., Dietary
ginger constituents, galanals A and B, are potent apoptosis
inducers in Human T lymphoma Jurkat cells. Cancer Lett.
2003, 199, 113–119.
[6] Lee, S. H., Cekanova, M., Baek, S. J., Multiple mechanisms
are involved in 6-gingerol-induced cell growth arrest and
apoptosis in human colorectal cancer cells. Mol. Carcinogen.
2008, 47, 197–208.
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2015, 59, 843–852 851
[7] Son, M. J., Miura, Y., Yagasaki, K., Mechanisms for antidia-
betic effect of gingerol in cultured cells and obese diabetic
model mice. Cytotechnology 2014, DOI: 10.1007/s10616-014-
9730-3.
[8] Mahluji, S., Ostadrahimi, A., Mobasseri, M., Ebrahimzade
Attari, V., Payahoo, L., Anti-inflammatory effects of Zingiber
officinale in type 2 diabetic patients. Adv. Pharm. Bull. 2013,
3, 273–276.
[9] Marx, W. M., Teleni, L., McCarthy, A. L., Vitetta, L. et al., Gin-
ger (Zingiber officinale) and chemotherapy-induced nausea
and vomiting: a systematic literature review. Nutr. Rev. 2013,
71, 245–254.
[10] Lusis, A. J., Atherosclerosis. Nature 2000, 407, 233–241.
[11] Raja, S. G., Drug-eluting stents and the future of coronary
artery bypass surgery: facts and fiction. Annals Thoracic
Surg. 2006, 81, 1162–1171.
[12] Degertekin, M., Serruys, P. W., Foley, D. P., Tanabe, K.
et al., Persistent inhibition of neointimal hyperplasia af-
ter sirolimus-eluting stent implantation: long-term (up to 2
years) clinical, angiographic, and intravascular ultrasound
follow-up. Circulation 2002, 106, 1610–1613.
[13] Kinghorn, A. D., Pan, L., Fletcher, J. N., Chai, H., The rel-
evance of higher plants in lead compound discovery pro-
grams. J. Nat. Products 2011, 74, 1539–1555.
[14] Verma, S. K., Singh, M., Jain, P., Bordia, A., Protective ef-
fect of ginger, Zingiber officinale Rosc on experimental
atherosclerosis in rabbits. Indian J. Exp. Biol. 2004, 42, 736–
738.
[15] Fuhrman, B., Rosenblat, M., Hayek, T., Coleman, R., Avi-
ram, M., Ginger extract consumption reduces plasma choles-
terol, inhibits LDL oxidation and attenuates development of
atherosclerosis in atherosclerotic, apolipoprotein E-deficient
mice. J. Nutr. 2000, 130, 1124–1131.
[16] Schiller, H. B., Szekeres, A., Binder, B. R., Stockinger, H.,
Leksa, V., Mannose 6-phosphate/insulin-like growth factor
2 receptor limits cell invasion by controlling alphaVbeta3
integrin expression and proteolytic processing of urokinase-
type plasminogen activator receptor. Mol. Biol. Cell 2009, 20,
745–756.
[17] Groblacher, B., Maier, V., Kunert, O., Bucar, F., Putative my-
cobacterial efflux inhibitors from the seeds of Aframomum
melegueta.J. Nat. Products 2012, 75, 1393–1399.
[18] Kurin, E., Atanasov, A. G., Donath, O., Heiss, E. H. et al., Syn-
ergy study of the inhibitory potential of red wine polyphe-
nols on vascular smooth muscle cell proliferation. Planta
Medica 2012, 78, 772–778.
[19] Joa, H., Vogl, S., Atanasov, A. G., Zehl, M. et al., Identification
of ostruthin from Peucedanum ostruthium rhizomes as an
inhibitor of vascular smooth muscle cell proliferation. J. Nat.
Products 2011, 74, 1513–1516.
[20] Fakhrudin, N., Waltenberger, B., Cabaravdic, M., Atanasov,
A. G. et al., Identification of plumericin as a potent new in-
hibitor of the NF-kappaB pathway with anti-inflammatory
activity in vitro and in vivo. Br. J. Pharmacol. 2014, 171,
1676–1686.
[21] Blazevic, T., Schwaiberger, A. V., Schreiner, C. E., Schachner,
D. et al., 12/15-lipoxygenase contributes to platelet-derived
growth factor-induced activation of signal transducer and
activator of transcription 3. J. Biol. Chem. 2013, 288, 35592–
35603.
[22] Schwaiberger, A. V., Heiss, E. H., Cabaravdic, M., Oberan, T.
et al., Indirubin-3’-monoxime blocks vascular smooth mus-
cle cell proliferation by inhibition of signal transducer and
activator of transcription 3 signaling and reduces neointima
formationinvivo.Arteriosclerosis Thrombosis Vasc. Biol.
2010, 30, 2475–2481.
[23] Sepp, A., Binns, R. M., Lechler, R. I., Improved protocol for
colorimetric detection of complement-mediated cytotoxicity
based on the measurement of cytoplasmic lactate dehydro-
genase activity. J. Immunol. Methods 1996, 196, 175–180.
[24] Bak, M. J., Ok, S., Jun, M., Jeong, W. S., 6-shogaol-rich ex-
tract from ginger up-regulates the antioxidant defense sys-
tems in cells and mice. Molecules 2012, 17, 8037–8055.
[25] Luo, Y., Eggler, A. L., Liu, D., Liu, G. et al., Sites of alkylation
of human Keap1 by natural chemoprevention agents. J. Am.
Soc. Mass Spectrom. 2007, 18, 2226–2232.
[26] Baird, L., Dinkova-Kostova, A. T., The cytoprotective role of
the Keap1-Nrf2 pathway. Arch. Toxicol. 2011, 85, 241–272.
[27] Bryan, H. K., Olayanju, A., Goldring, C. E., Park, B. K., The Nrf2
cell defence pathway: Keap1-dependent and -independent
mechanisms of regulation. Biochem. Pharmacol. 2013, 85,
705–717.
[28] Reuland, D. J., Khademi, S., Castle, C. J., Irwin, D. C. et al.,
Upregulation of phase II enzymes through phytochemical
activation of Nrf2 protects cardiomyocytes against oxidant
stress. Free Rad. Biol. Med. 2013, 56, 102–111.
[29] Reuland, D. J., McCord, J. M., Hamilton, K. L., The role of
Nrf2 in the attenuation of cardiovascular disease. Exercise
Sport Sci. Rev. 2013, 41, 162–168.
[30] Barbagallo, I., Galvano, F., Frigiola, A., Cappello, F. et al.,
Potential therapeutic effects of natural heme oxygenase-1
inducers in cardiovascular diseases. Antioxidants Redox Sig-
naling 2013, 18, 507–521.
[31] Duckers, H. J., Boehm, M., True, A. L., Yet, S. F. et al., Heme
oxygenase-1 protects against vascular constriction and pro-
liferation. Nat. Med. 2001, 7, 693–698.
[32] Baliga, M. S., Haniadka, R., Pereira, M. M., D’Souza, J. J.
et al., Update on the chemopreventive effects of ginger and
its phytochemicals. Crit. Rev. Food Sci. Nutr. 2011, 51, 499–
523.
[33] Jolad, S. D., Lantz, R. C., Solyom, A. M., Chen, G. J. et al.,
Fresh organically grown ginger (Zingiber officinale): compo-
sition and effects on LPS-induced PGE2 production. Phyto-
chemis try 2004, 65, 1937–1954.
[34] Jolad, S. D., Lantz, R. C., Chen, G. J., Bates, R. B., Timmer-
mann, B. N., Commercially processed dry ginger (Zingiber
officinale): composition and effects on LPS-stimulated PGE2
production. Phytochemistry 2005, 66, 1614–1635.
[35] Jiang, H., Xie, Z., Koo, H. J., McLaughlin, S. P. et al., Metabolic
profiling and phylogenetic analysis of medicinal Zingiber
species: Tools for authentication of ginger (Zingiber offici-
nale Rosc). Phytochemistry 2006, 67, 1673–1685.
[36] Shao, X., Lv, L., Parks, T., Wu, H. et al., Quantitative anal-
ysis of ginger components in commercial products using
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
852 R. Liu et al. Mol. Nutr. Food Res. 2015, 59, 843–852
liquid chromatography with electrochemical array detection.
J. Agricult. Food Chem. 2010, 58, 12608–12614.
[37] Govindarajan, V. S., Ginger—chemistry, technology, and
quality evaluation: part 1. Crit. Rev. Food Sci. Nutr. 1982,
17, 1–96.
[38] Park, J. S., Jung, M. Y., Development of high-performance
liquid chromatography-time-of-flight mass spectrometry for
the simultaneous characterization and quantitative analy-
sis of gingerol-related compounds in ginger products. J.
Agricult. Food Chem. 2012, 60, 10015–10026.
[39] Wang, W., Li, C. Y., Wen, X. D., Li, P., Qi, L. W., Simultaneous
determination of 6-gingerol, 8-gingerol, 10-gingerol and 6-
shogaol in rat plasma by liquid chromatography-mass spec-
trometry: application to pharmacokinetics. J. Chromatogr. B
Anal. Technol. Biomed. Life Sci. 2009, 877, 671–679.
[40] Dugasani, S., Pichika, M. R., Nadarajah, V. D., Balijepalli,
M. K. et al., Comparative antioxidant and anti-inflammatory
effects of [6]-gingerol, [8]-gingerol, [10]-gingerol and [6]-
shogaol. J. Ethnopharmacol. 2010, 127, 515–520.
[41] Wei, Q. Y., Ma, J. P., Cai, Y. J., Yang, L., Liu, Z. L., Cytotoxic
and apoptotic activities of diarylheptanoids and gingerol-
related compounds from the rhizome of Chinese ginger. J.
Ethnopharmacol. 2005, 102, 177–184.
[42] Kim, D. S., Kim, J. Y., Side-chain length is important for
shogaols in protecting neuronal cells from beta-amyloid in-
sult. Bioorganic Med. Chem. Lett. 2004, 14, 1287–1289.
[43] Jeong, W.S., Jun, M., Kong, A. N., Nrf2: a potential molecular
target for cancer chemoprevention by natural compounds.
Antioxidants Redox Signaling 2006, 8, 99–106.
[44] Blazevic, T., Schaible, A. M., Weinhaupl, K., Schachner, D.
et al., Indirubin-3’-monoxime exerts a dual mode of inhibi-
tion towards leukotriene-mediated vascular smooth muscle
cell migration. Cardiovasc. Res. 2014, 101, 522–532.
[45] Liu, Z., Wang, J., Huang, E., Gao, S. et al., Tanshinone IIA sup-
presses cholesterol accumulation in human macrophages:
role of heme oxygenase-1. J. Lipid Res. 2014, 55, 201–213.
[46] Heiss, E. H., Schachner, D., Werner, E. R., Dirsch, V. M., Active
NF-E2-related factor (Nrf2) contributes to keep endothelial
NO synthase (eNOS) in the coupled state: role of reactive
oxygen species (ROS), eNOS, and heme oxygenase (HO-1)
levels. J. Biol. Chem. 2009, 284, 31579–31586.
[47] Zakkar, M., Vander Heiden, K., Luong le, A., Chaudhury, H.
et al., Activation of Nrf2 in endothelial cells protects arter-
ies from exhibiting a proinflammatory state. Arteriosclerosis
Thrombosis Vasc. Biol. 2009, 29, 1851–1857.
[48] Saha, A., Blando, J., Silver, E., Beltran, L. et al., 6-Shogaol
from dried ginger inhibits growth of prostate cancer cells
both in vitro and in vivo through inhibition of STAT3 and NF-
kappaB signaling. Cancer Prevention Res. 2014, 7, 627–638.
[49] Tan, B. S., Kang, O., Mai, C. W., Tiong, K. H. et al., 6-
Shogaol inhibits breast and colon cancer cell proliferation
through activation of peroxisomal proliferator activated re-
ceptor gamma (PPARgamma). Cancer Lett. 2013, 336, 127–
139.
[50] Bruemmer, D., Blaschke, F., Law, R. E., New targets for
PPARgamma in the vessel wall: implications for restenosis.
Int. J. Obesity 2005, 29(Suppl 1), S26–S30.
[51] Bauters, C., Isner, J. M., The biology of restenosis. Progress
Cardiovasc. Dis. 1997, 40, 107–116.
[52] Kinlay, S., Libby, P., Ganz, P., Endothelial function and coro-
nary artery disease. Curr. Opin. Lipidol. 2001, 12, 383–
389.
[53] Windecker, S., Remondino, A., Eberli, F. R., Juni, P. et al.,
Sirolimus-eluting and paclitaxel-eluting stents for coronary
revascularization. N. Engl. J. Med. 2005, 353, 653–662.
[54] Iakovou, I., Schmidt, T., Bonizzoni, E., Ge, L. et al., Incidence,
predictors, and outcome of thrombosis after successful
implantation of drug-eluting stents. JAMA 2005, 293, 2126–
2130.
[55] Mehilli, J., Byrne, R. A., Tiroch, K., Pinieck, S. et al., Ran-
domized trial of paclitaxel- versus sirolimus-eluting stents
for treatment of coronary restenosis in sirolimus-eluting
stents: the ISAR-DESIRE 2 (Intracoronary Stenting and
Angiographic Results: Drug Eluting Stents for In-Stent
Restenosis 2) study. J. Am. College of Cardiol. 2010, 55,
2710–2716.
C2015 The Authors. Molecular Nutrition & Food Research published by Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
... This effect might, at least in part, counteract the inhibiting effect of 6-shogaol. A study by Liu et al. (2015) shows that 6-shogaol effectively inhibited the proliferation of rat aortic vascular smooth muscle cells with an IC 50 of 2.7 µM without inducing cytotoxicity. Interestingly, 6-shogaol lost its antiproliferative feature when an HO-1 inhibitor was applied. ...
... Moreover, our study demonstrates the successful 6-shogaolinduced impairment of angiogenesis-related cell functions as indicated by the reduction of endothelial cell proliferation, chemotactic migration, and in vitro and ex vivo sprouting. Liu et al. (2015) reported the inhibition of vascular smooth muscle cell proliferation with an IC 50 of 1 µM. In accordance with this, 6shogaol was able to reduce the proliferation of HUVECs with an IC 50 of 8 µM without exerting cytotoxic effects up to 30 µM over an incubation period of 72 h. ...
Article
Full-text available
Rhizomes from Zingiber officinale Roscoe are traditionally used for the treatment of a plethora of pathophysiological conditions such as diarrhea, nausea, or rheumatoid arthritis. While 6-gingerol is the pungent principle in fresh ginger, in dried rhizomes, 6-gingerol is dehydrated to 6-shogaol. 6-Shogaol has been demonstrated to exhibit anticancer, antioxidative, and anti-inflammatory actions more effectively than 6-gingerol due to the presence of an electrophilic Michael acceptor moiety. In vitro, 6-shogaol exhibits anti-inflammatory actions in a variety of cell types, including leukocytes. Our study focused on the effects of 6-shogaol on activated endothelial cells. We found that 6-shogaol significantly reduced the adhesion of leukocytes onto lipopolysaccharide (LPS)-activated human umbilical vein endothelial cells (HUVECs), resulting in a significantly reduced transmigration of THP-1 cells through an endothelial cell monolayer. Analyzing the mediators of endothelial cell–leukocyte interactions, we found that 30 µM of 6-shogaol blocked the LPS-triggered mRNA and protein expression of cell adhesion molecules. In concert with this, our study demonstrates that the LPS-induced nuclear factor κB (NFκB) promoter activity was significantly reduced upon treatment with 6-shogaol. Interestingly, the nuclear translocation of p65 was slightly decreased, and protein levels of the LPS receptor Toll-like receptor 4 remained unimpaired. Analyzing the impact of 6-shogaol on angiogenesis-related cell functions in vitro, we found that 6-shogaol attenuated the proliferation as well as the directed and undirected migration of HUVECs. Of note, 6-shogaol also strongly reduced the chemotactic migration of endothelial cells in the direction of a serum gradient. Moreover, 30 µM of 6-shogaol blocked the formation of vascular endothelial growth factor (VEGF)-induced endothelial sprouts from HUVEC spheroids and from murine aortic rings. Importantly, this study shows for the first time that 6-shogaol exhibits a vascular-disruptive impact on angiogenic sprouts from murine aortae. Our study demonstrates that the main bioactive ingredient in dried ginger, 6-shogaol, exhibits beneficial characteristics as an inhibitor of inflammation- and angiogenesis-related processes in vascular endothelial cells.
... There is therefore the possibility to provide insights into therapeutic goals and strategies for treating breast cancer in detail on the biology and its molecular mechanics behind the progression of the disease.Ginger is a natural dietary rhizome commonly employed as a herb and aromatic agent. Ginger contains several bioactive components, such as gingerols, shogaols, paradols and zingerone, which show the pharmacological role of anti-inflammatory and antitumor mediators (4).6-gingerol and six-shogaol were widely reported in ginger bioactive ingredients to perform anti-tumor activities in a range of cancers through cell proliferation, migration, and invasion or induction of apoptosis (5). One of the major ginger isolated phenolic compounds, gingerol has been reported to have antitumor activity in inhibiting or inducing apoptosis against oval-, colon-, lung-and prostate cancer cells (6), However, gingerol's effects and molecular mechanisms on the growth and progression of breast cancer cells are poorly understandable. ...
... Mechanistic evaluation studies[6]-gingerol's anti-cancer properties against various kinds of cancer offer valuable insight into their various mechanisms of action to achieve cytotoxic or pro-apoptotic effects in cancer cells. A few recent reports on[5]-gingerol's anticancer activity against colon cancer show various mechanisms to deal with various lines of colon cancer.(18).The MTT test suggests that dose-related action is based on the various immortal cell lines (MCF-7), and that induced by gingerol in MCF-7 cell lines, relatively higher apoptotic cell levels. The estrogen receptor (ER) can be present in MCF-7(19).Depending on the origin and type of cells, the ability to bind with the AMN-3 plasma membrane receptor differ. ...
Article
Full-text available
This study was conducted to determine the role and molecular function of phenolic gingerol in regulating the cell proliferation of MCF-7 breast cancer cells, a compound isolated from Zingiberofficinale. Different gingerol concentrations from (1-1000) μg/ml were prepared to assess gingerol's cytotoxic effect on MCF-7 cell lines, cell line exposure periods were measured in (24) hours.Under complete sterile conditions on a microtitration plate. Results showed that, after exposure to cell line for 24 hours, Cells treated with gingerol showed higher cell-death percentagethan untreated cells as quantified bythe MTT assay, with a 100 μg/mL inhibition rate of MCF-7 of 83.2 percent following 24 hour of exposure.The capacity of gingerol to induce apoptosis to an infected cell under a fluorescence microscope has been studied by crystal violet stain to measure the percentage of induction of apoptosis. A significant increase of the expression of caspase-3, caspase-8 and caspase-9 in comparison with untreated cells has revealed the gingerol effect on the MCF-7 line. The role of apoptosis, its high potential as an antitumor agent, was demonstrated in this study.
... Several in vitro and in vivo studies have demonstrated that ginger extract diminished VSMC proliferation. A study by Liu et al. 76 found that [6]-shogaol exerts its anti-VSMC proliferation effect by blocking the cells at the G0/G1 cell-cycle phase. This was shown to be associated with activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. ...
Article
Ginger (Zingiber officinale Roscoe) is a common and widely used spice. It is rich in various chemical constituents, including phenolic compounds, terpenes, polysaccharides, lipids, organic acids, and raw fibers. Herein, we reviewed its effects on the vascular system. Studies utilizing cell cultures or animal models showed that ginger constituents alleviate oxidative stress and inflammation, increase nitric oxide synthesis, suppress vascular smooth muscle cells proliferation, promote cholesterol efflux from macrophages, inhibit angiogenesis, block voltage-dependent Ca2+ channels, and induce autophagy. In clinical trials, ginger was shown to have a favorable effect on serum lipids, inflammatory cytokines, blood pressure, and platelets aggregation. Taken together, these studies point to the potential benefits of ginger and its constituents in treatment of hypertension, coronary artery disease, peripheral arterial diseases, and other vascular diseases
... Epigallocatechin gallate, an extract from Camelia sinenses has shown similar activities, along with ameliorating effects on inflammation and ED [67][68][69][70]. The polyphenols found in Olea europea can also prevent ERK1/2 activation, impeding the transition from the G1 phase to the S phase of the cell cycle [71]; whereas ginger (Zingiber officinale) appears to prevent the change from phase G2 to phase M in VSMC [72]. ...
Article
At present, cardiovascular disease (CVD) remains the leading cause of morbidity and mortality worldwide, and global trends suggest this panorama will persist or worsen in the near future. Thus, optimization of treatment strategies and the introduction of novel therapeutic alternatives for CVD represent key objectives in contemporary biomedical research. In recent years, phytotherapy—defined as the therapeutic use of whole or minimally modified plant components—has ignited large scientific interest, with a resurgence of abundant investigation on a wide array of medicinal herbs (MH) for CVD and other conditions. Numerous MH have been observed to intervene in the pathophysiology of CVD via a myriad of molecular mechanisms, including anti-inflammatory, anti-oxidant, and other beneficial properties, which translate into the amelioration of three essential aspects of the pathogenesis of CVD: Dyslipidemia, atherosclerosis, and hypertension. Although the preclinical data in this scenario is very rich, the true clinical impact of MH and their purported mechanisms of action is less clear, as large-scale robust research in this regard is in relatively early stages and faces important methodological challenges. This review offers a comprehensive look at the most prominent preclinical and clinical evidence currently available concerning the use of MH in the treatment of CVD from a bench-to-bedside approach.
Article
Full-text available
Chronic pain has a high prevalence and a profound impact on patients and society, and its treatment is a real challenge in clinical practice. Ginger is emerging as a promising analgesic—effective against various types of pain and well-tolerated by patients. However, we are just beginning to understand its complex mechanism of action. A good understanding of its mechanism would allow us to fully utilize the therapeutical potential of this herbal medicine as well as to identify a better strategy for treating chronic pain. To provide this information, we searched PubMed, SCOPUS, and Web of Science for in vitro studies or animal experiments investigating the analgesic effect of ginger extract or its components. The analysis of data was carried out in the form of a narrative review. Our research indicates that ginger extract, through its various active ingredients, suppresses the transmission of nociceptive signals while activating the descendent inhibitory pathways of pain.
Article
Ethnopharmacological relevance Atherosclerosis (AS) is a chronic disease that is associated with high morbidity. However, therapeutic approaches are limited. Wu-Zhu-Yu decoction (WZYD) is a well-known traditional Chinese medicine prescription that is traditionally used to treat headaches and vomiting. Modern studies have demonstrated the cardiotonic effects of WZYD. However, whether WZYD can alleviate AS and its underlying mechanisms remain unclear. Aim of the study This study aims to investigate the antiatherosclerotic efficacy of WZYD and illustrate its potential mechanisms using an integrated approach combining in vivo and in vitro assessments, including metabolomics, network pharmacology, cell experiments, and molecular docking analyses. Materials and methods In this work, an atherosclerotic mouse model was established by administering a high-fat diet to apolipoprotein-E deficient (ApoE-/-) mice for twelve weeks. Meanwhile, the mice were intragastrically administered WZYD at different dosages. Efficacy evaluation was performed through biochemical and histopathological assessments. The potential active constituents, metabolites, and targets of WZYD in atherosclerosis were predicted by metabolomics combined with network pharmacology analysis, the constituents and targets were further assessed through cell experiments and molecular docking analysis. Results WZYD decreased the lipid levels in serum, reduced the areas of aortic lesions, and attenuated intimal thickening, which had antiatherosclerotic effects in ApoE-/- mice. Metabolomics and network pharmacology approach revealed that the ten constituents (6-shogaol, evodiamine, isorhamnetin, quercetin, beta-carotene, 8-gingerol, kaempferol, 6-paradol, 10-gingerol, and 6-gingerol) of WZYD affected 24 metabolites by acting on the candidate targets, thus resulting in changes in five metabolic pathways (sphingolipid metabolism; glycine, serine and threonine metabolism; arachidonic acid metabolism; tryptophan metabolism; and fatty acid biosynthesis pathway). Cell experiments indicated that the ten key compounds showed antiproliferative effects on the vascular smooth muscle cell. Moreover, the key compounds exhibited direct interactions with the key targets, as assessed by molecular docking analysis. Conclusion This study revealed that WZYD exerted therapeutic effects on atherosclerosis, and the potential mechanisms were elucidated. Furthermore, it offered a powerful integrated strategy for studying the efficacy of traditional Chinese medicine and exploring its active components and possible mechanisms.
Article
Ginger (Zingiber officinale) is a famous dietary spice rich in bioactive components like gingerols, and it has been used for a long time as food and medicine. Indeed, clinical studies have confirmed the anti-inflammatory and antioxidant properties of ginger. Thus, ginger seems to be an excellent complementary nutritional strategy for non-communicable diseases (NCD) such as obesity, diabetes, cardiovascular disease and chronic kidney disease. This narrative review aims to discuss the possible effects of ginger on the mitigation of common complications such as inflammation, oxidative stress, and gut dysbiosis in NCD.
Article
Objective We sought to evaluate ginger's cardiovascular and metabolic effects (Zingiberofficinale) add-on therapy in type 2 diabetes patients over six weeks. Methods We performed a single-arm clinical trial. In well-to-moderately controlled Type 2 diabetic patients with unchanged treatment for at least three months, the intervention consisted of 6-week add-on oral supplementation of powdered ginger extracts in capsules at a dose of 399 mg three times per day. Transthoracic Doppler echocardiography, ambulatory blood pressure monitoring (ABPM), glycatedhaemoglobin (HbA1c), lipid profile, kidney and liver function analysis were performed at initial and final visits, with a follow-up visit on day 21. Adherence to treatment, palatability and safety were also assessed. Results Overall, 21 participants (16 females) were included in the analysis. We found a non-significant decrease of E’ wave from 0.05[0.04-0.09] to 0.06[0.05-0.7]cm/s, A-wave from 0.8[0.6-0.8] to 0.7[0.6-0.8] cm/s, and E-wave from 0.6[0.5-0.7] to 0.5[0.425-0.6]cm/s. There was a significant reduction of HbA1c from 49.7[47.0-57.4] to 44.3[38.8-53.0] mmol/mol and triglycerides from 1.6[1.4-1.9] to 1.2[0.9-1.8] mmol/l. A 5% decrease or more was observed for diurnal DBP, diurnal MAP and 24-hour DBP. Conclusion Zingiberofficinale used as add-on therapy tend to improve diastolic function, blood pressure and lipid profile of type 2 diabetes patients. Further studies are needed to define the dosage and duration of this supplementary treatment accurately. Trial Registration number:NCT04222738.
Article
Full-text available
The small molecule indirubin-3'monoxime (I3MO) has been shown to inhibit vascular smooth muscle cell (VSMC) proliferation and neointima formation in vivo. The influence of I3MO on VSMC migration and vascular inflammation, two additional key players during the onset of atherosclerosis and restenosis, should be investigated.Methods and ResultsWe examined the influence of I3MO on VSMC migration, with focus on monocyte-derived leukotrienes (LTs) and platelet-derived growth factor (PDGF) as elicitors. Exogenous LTB4 and cysteinyl-leukotrienes (cysLT) as well as LT-enriched conditioned medium of activated primary human monocytes induced VSMC migration which was inhibited by I3MO. I3MO also blunted migration of VSMC stimulated with PDGF, the strongest motogen tested in this study. Induction of heme oxygenase 1 (HO-1) accounted for this anti-migratory activity of I3MO in VSMC. Notably, I3MO not only interfered with the migratory response in VSMC, but also suppressed production of pro-migratory LT in monocytes. Conditioned media from monocytes that were activated in the presence of I3MO failed to induce VSMC migration. In cell-based and cell-free assays I3MO selectively inhibited 5-lipoxygenase (5-LO), the key enzyme in LT biosynthesis, with an IC50 in the low micromolar range. Our study reveals a novel dual inhibitory mode of I3MO on LT-mediated VSMC migration: (i) I3MO interferes with pro-migratory signaling in VSMC, and (ii) I3MO suppresses LT biosynthesis in monocytes by direct inhibition of 5-LO. These inhibitory actions on both migratory stimulus and response complement the previously demonstrated anti-proliferative properties of I3MO and may further promote I3MO as promising vasoprotective compound.
Article
Full-text available
The transcription factor nuclear factor kappa B (NF-κB) orchestrates many pro-inflammatory signals and its inhibition is considered a promising strategy to combat inflammation. Here we report the characterization of the natural product plumericin as a highly potent inhibitor of the NF-κB pathway with a novel chemical scaffold, which was isolated via a bioactivity-guided approach from Himatanthus sucuuba, an Amazonian plant traditionally used to treat inflammation-related disorders. A NF-κB luciferase reporter gene assay was used to identify NF-κB pathway inhibitors from H. sucuuba extracts. Monitoring of TNF-α-induced expression of the adhesion molecules VCAM-1, ICAM-1, and E-selectin by flow cytometry was used to confirm NF-κB inhibition in endothelial cells, and thioglycollate-induced peritonitis in mice to verify in vivo effectiveness. Western blotting and transfection experiments were used to address the mechanism of action of plumericin. Plumericin inhibits NF-κB-mediated transactivation of a luciferase reporter gene with an IC50 value of 1 μM, abolishes TNF-α-induced expression of the adhesion molecules VCAM-1, ICAM-1, and E-selectin in endothelial cells, and suppresses thioglycollate-induced peritonitis in mice. Plumericin exerts its NF-κB pathway-inhibitory effect by suppression of IκB phosphorylation and degradation. Plumericin also inhibits NF-κB activation induced by transfection with the constitutively active catalytic subunit of IKK (IKK-β), implying IKK in the inhibitory action of this natural product. Plumericin is a potent new scaffold inhibitor of the NF-κB pathway with a potential to be further explored as a novel anti-inflammatory lead.
Article
Full-text available
Purpose: Low-grade inflammation, a common feature in type 2 diabetes (DM2), causes some chronic complications in these patients. The present study was aimed to evaluate the effects of ginger (Zingiber officinale) on pro-inflammatory cytokines (IL-6 and TNF-α) and the acute phase protein hs-CRP in DM2 patients as a randomized double-blind placebo controlled trial. Methods: A total of 64 DM2 patients randomly were assigned to ginger or placebo groups and received 2 tablets/day of each for 2 months. The concentrations of IL-6, TNF-α and hs-CRP in blood samples were analyzed before and after the intervention. Results: Ginger supplementation significantly reduced the levels of TNF-α (P = 0.006), IL-6 (P = 0.02) and hs-CRP (P = 0.012) in ginger group in comparison to baseline. Moreover, the analysis of covariance showed that the group received ginger supplementation significantly lowered TNF- α (15.3 ± 4.6 vs. 19.6 ± 5.2; P = 0.005) and hs-CRP (2.42 ± 1.7 vs. 2.56 ± 2.18; P = .016) concentrations in comparison to control group. While there were no significant changes in IL-6 (7.9 ± 2.1 vs. 7.8 ± 2.9; P > .05). Conclusion: In conclusion, ginger supplementation in oral administration reduced inflammation in type 2 diabetic patients. So it may be a good remedy to diminish the risk of some chronic complications of diabetes.
Article
Full-text available
Accumulation of foam cells in the neointima represents a key event in atherosclerosis. We previously demonstrated that Tanshinone IIA (Tan), a lipophilic bioactive compound extracted from Salvia miltiorrhiza Bunge, inhibits experimental atherogenesis, yet the detailed mechanisms are not fully understood. In this study, we sought to explore the potential effects of Tan on lipid accumulation in macrophage foam cells and the underlying molecular mechanisms. Our data indicate that Tan treatment reduced the content of macrophages, cholesterol accumulation and the development of atherosclerotic plaque in ApoE-/- mice. In human macrophages, Tan ameliorated oxidized low-density lipoporotein (oxLDL) elicited foam cell formation by inhibiting oxLDL uptake and promoting cholesterol efflux. Mechanistically, Tan markedly reduced the expression of class A scavenger receptor (SR-A) and increased the expression of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) in lipid-laden macrophages via activation of extracellular signal regulated kinase (ERK)/nuclear factor-erythroid 2-related factor 2 (Nrf2)/haem oxygenase-1 (HO-1) pathway. Tan treatment induced the phosphorylation and nuclear translocation of Nrf2 and subsequently increased the expression of HO-1, and these effects were abolished by the specific ERK inhibitors-PD98059 and U0126. Moreover, HO-1 small interfering RNA or ZnPP (an HO-1 inhibitor) abrogated Tan-mediated suppression of lipid accumulation in macrophages. Our current findings demonstrate that a novel HO-1-dependent mechanism is involved in the regulation of cholesterol balance by Tan.
Article
Full-text available
We showed previously that the small molecule indirubin-3′-monoxime (I3MO) prevents vascular smooth muscle cell (VSMC) proliferation by selectively inhibiting signal transducer and activator of transcription 3 (STAT3). Looking for the underlying upstream molecular mechanism, we here reveal the important role of reactive oxygen species (ROS) for PDGF-induced STAT3 activation in VSMC. We show that neither NADPH-dependent oxidases (Noxes) nor mitochondria, but rather 12/15-lipoxygenase (12/15-LO) are pivotal ROS sources involved in the redox-regulated signal transduction from PDGFR to STAT3. Accordingly, pharmacological and genetic interference with 12/15-LO activity selectively inhibited PDGF-induced Src activation and STAT3 phosphorylation. I3MO is able to blunt PDGF-induced ROS and 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) production, indicating an inhibitory action of I3MO on 12/15-LO and consequently on STAT3. We identify 12/15-LO as a hitherto unrecognized signaling hub in PDGF-triggered STAT3 activation and show for the first time a negative impact of I3MO on 12/15-LO.
Article
Prostate cancer (PCa) is the most common non-cutaneous neoplasm and is the second leading cause of cancer death in American men. Approximately 1 in 6 men will be diagnosed with PCa during their lifetime and about 1 in 36 will die of PCa. It is estimated that approximately 238,590 new cases of PCa will be diagnosed and 29,720 PCa-related deaths will occur in the United States in 2013. 6-Shogaol (6-SHO), a potent bioactive compound in ginger (Zingiber officinale Roscoe), has been shown to possess anti-inflammatory and anticancer activity. In the present study, the effect of 6-SHO on the growth of PCa cell lines was investigated. 6-SHO effectively reduced survival and induced apoptosis of cultured human (LNCaP, DU145 and PC3) and mouse (HMVP2) PCa cell lines. Mechanistic studies revealed that 6-SHO reduced constitutive and IL-6 induced STAT3 activation and inhibited both constitutive and TNF-α induced NF-κB activity in these cell lines. In addition, 6-SHO decreased the level of several STAT3 and NF-κB regulated target genes at the protein level (including cyclin D1, survivin and cMyc) and modulated mRNA levels of chemokine, cytokine, cell cycle and apoptosis regulatory genes (IL-7, CCL5, BAX, BCL2, p21 and p27). 6-SHO was more effective than two other compounds found in ginger, 6-gingerol and 6-paradol, at reducing survival of PCa cell lines and reducing STAT3 and NF-κB signaling. 6-SHO also showed significant tumor growth inhibitory activity in an allograft model using HMVP2 cells. Overall, the current results lead us to suggest that 6-SHO may have potential as a chemopreventive and/or therapeutic agent for PCa and that further study of this compound is warranted Citation Format: Achinto Saha, Jorge Blando, Eric Silver, Linda Beltran, Jonathan Sessler, John DiGiovanni. 6-Shogaol from dried ginger inhibits growth of prostate cancer cells both in vitro and in vivo. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2138. doi:10.1158/1538-7445.AM2014-2138
Article
There have been studies on health beneficial effects of ginger and its components. However, there still remain certain aspects that are not well defined in their anti-hyperglycemic effects. Our aims were to find evidence of possible mechanisms for antidiabetic action of [6]-gingerol, a pungent component of ginger, employing a rat skeletal muscle-derived cell line, a rat-derived pancreatic β-cell line, and type 2 diabetic model animals. The antidiabetic effect of [6]-gingerol was investigated through studies on glucose uptake in L6 myocytes and on pancreatic β-cell protective ability from reactive oxygen species (ROS) in RIN-5F cells. Its in vivo effect was also examined using obese diabetic db/db mice. [6]-Gingerol increased glucose uptake under insulin absent condition and induced 5' adenosine monophosphate-activated protein kinase phosphorylation in L6 myotubes. Promotion by [6]-gingerol of glucose transporter 4 (GLUT4) translocation to plasma membrane was visually demonstrated by immunocytochemistry in L6 myoblasts transfected with glut4 cDNA-coding vector. [6]-Gingerol suppressed advanced glycation end product-induced rise of ROS levels in RIN-5F pancreatic β-cells. [6]-Gingerol feeding suppressed the increases in fasting blood glucose levels and improved glucose intolerance in db/db mice. [6]-Gingerol regulated hepatic gene expression of enzymes related to glucose metabolism toward decreases in gluconeogenesis and glycogenolysis as well as an increase in glycogenesis, thereby contributing to reductions in hepatic glucose production and hence blood glucose concentrations. These in vitro and in vivo results strongly suggest that [6]-gingerol has antidiabetic potential through multiple mechanisms.
Article
Despite much recent progress, prostate cancer (PCa) continues to represent a major cause of cancer-related mortality and morbidity in men. PCa is the most common non-skin neoplasm and second leading cause of death in men. 6-Shogaol (6-SHO), a potent bioactive compound in ginger (Zingiber officinale Roscoe), has been shown to possess anti-inflammatory and anticancer activity. In the present study, the effect of 6-SHO on the growth of PCa cells was investigated. 6-SHO effectively reduced survival and induced apoptosis of cultured human (LNCaP, DU145 and PC3) and mouse (HMVP2) PCa cells. Mechanistic studies revealed that 6-SHO reduced constitutive and IL-6 induced STAT3 activation and inhibited both constitutive and TNF-α induced NF-κB activity in these cells. In addition, 6-SHO decreased the level of several STAT3 and NF-κB regulated target genes at the protein level, including cyclin D1, survivin and cMyc and modulated mRNA levels of chemokine, cytokine, cell cycle and apoptosis regulatory genes (IL-7, CCL5, BAX, BCL2, p21 and p27). 6-SHO was more effective than two other compounds found in ginger, 6-gingerol and 6-paradol at reducing survival of PCa cells and reducing STAT3 and NF-κB signaling. 6-SHO also showed significant tumor growth inhibitory activity in an allograft model using HMVP2 cells. Overall, the current results suggest that 6-SHO may have potential as a chemopreventive and/or therapeutic agent for PCa and that further study of this compound is warranted.