Sei sulla pagina 1di 8

Helicobacter ISSN 1523-5378

Modulation of Activation-Induced Cytidine Deaminase by


Curcumin in Helicobacter pylori-Infected Gastric Epithelial Cells
Syed Faisal Haider Zaidi,*,† Takeshi Yamamoto,† Alaa Refaat,‡ Kanwal Ahmed,§ Hiroaki Sakurai,‡ Ikuo
Saiki,‡ Takashi Kondo,§ Khan Usmanghani,¶ Makoto Kadowaki† and Toshiro Sugiyama*
*
Department of Gastroenterology and Hematology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama,
Japan, †Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan, ‡Division of Pathogenic
Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama, Japan, §Department of Radiological Sciences, Graduate School of
Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan, ¶Department of Basic Clinical Sciences, Faculty of Eastern Medicine,
Hamdard University, Karachi, Pakistan

Keywords Abstract
H. pylori, activation-induced cytidine
Background: Anomalous expression of activation-induced cytidine
deaminase, curcumin, NF-jB.
deaminase (AID) in Helicobacter pylori-infected gastric epithelial cells has
Reprint requests to: Toshiro Sugiyama, Depart- been postulated as one of the key mechanisms in the development of gastric
ment of Gastroenterology and Hematology, cancer. AID is overexpressed in the cells through nuclear factor (NF)-jB
Graduate School of Medicine and Pharmaceu- activation by H. pylori and hence, inhibition of NF-jB pathway can down-
tical Sciences, University of Toyama, 2630- regulate the expression of AID. Curcumin, a spice-derived polyphenol, is
Sugitani, Toyama 930-0194, Japan. known for its anti-inflammatory activity via NF-jB inhibition. Therefore, it
E-mail: tsugi@med.u-toyama.ac.jp
was hypothesized that curcumin might suppress AID overexpression via
NF-jB inhibitory activity in H. pylori-infected gastric epithelial cells.
Materials and Methods: MKN-28 or MKN-45 cells and H. pylori strain 193C
isolated from gastric cancer patient were used for co-culture experiments.
Cells were pretreated with or without nonbactericidal concentrations of
curcumin. Apoptosis was determined by DNA fragmentation assay. Enzyme-
linked immunosorbent assay was performed to evaluate the anti-adhesion
activity of curcumin. Real-time polymerase chain reaction was employed to
evaluate the expression of AID mRNA. Immunoblot assay was performed for
the analysis of AID, NF-jB, inhibitors of NF-jB (IjB), and IjB kinase (IKK)
complex regulation with or without curcumin.
Results: The adhesion of H. pylori to gastric epithelial cells was not inhibited
by curcumin pretreatment at nonbactericidal concentrations (£10 lmol ⁄ L).
Pretreatment with nonbactericidal concentration of curcumin downregulated
the expression of AID induced by H. pylori. Similarly, NF-jB activation inhib-
itor (SN-50) and proteasome inhibitor (MG-132) also downregulated the
mRNA expression of AID. Moreover, curcumin (£10 lmol ⁄ L) has suppressed
H. pylori-induced NF-jB activation via inhibition of IKK activation and IjB
degradation.
Conclusion: Nonbactericidal concentrations of curcumin downregulated
H. pylori-induced AID expression in gastric epithelial cells, probably via the
inhibition of NF-jB pathway. Hence, curcumin can be considered as a
potential chemopreventive candidate against H. pylori-related gastric
carcinogenesis.

Helicobacter pylori is a gram-negative, spiral-shaped, been recognized as the causative agent of chronic
microaerophilic bacterium that colonizes the human gastritis, gastroduodenal ulcers, mucosa-associated
gastric mucosa while neutralizing the hostile gastric lymphoid tissue lymphoma, and gastric adenocarcinoma
environment [1]. It is estimated to have colonized [2]. H. pylori interaction with gastric epithelial cells
about half of the world’s human population and has resulted in overexpression of pro-inflammatory

588 ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595


Haider et al. Curcumin Downregulates AID in H. pylori-Infected Cells

cytokines and the activation of the transcription factor, cells and whether or not this effect is the result of the
nuclear factor (NF)-jB [3]. NF-jB is a crucial regulator inhibition of NF-jB pathway.
of many cellular processes and mediators including
regulation of inflammatory cytokines, chemokines,
Materials and Methods
adhesion molecules, enzymes, and kinases. NF-jB is
localized in the cytoplasm by binding to a family of
Reagents
cytoplasmic inhibitors, the inhibitors of NF-jB (IjB).
Cell stimulation triggers specific intracellular signaling RPMI-1640 culture medium was procured from Wako
pathways consequently leading to the activation of the (Osaka, Japan). Curcumin was purchased from Nacalai
IjB kinase complex (IKK complex). The activated IKK Tesque, Inc. (Kyoto, Japan). NF-jB activation inhibitor
complex phosphorylates the NF-jB-bound IjB proteins SN-50 and proteasome inhibitor MG-132 was obtained
and targets them for poly-ubiquitination and rapid from Calbiochem (EMD Biosciences, Inc., CA, USA).
degradation. The dissociated NF-jB is translocated into Human tumor necrosis factor (TNF)-a was purchased
the nucleus and regulates NF-jB-dependent gene from R&D systems (Minneapolis, MN, USA). Brucella
expression [4,5]. broth (BBLTM) was procured from BD (Sparks, MD,
Recently, Matsumoto et al. has reported the involve- USA).
ment of NF-jB in the abnormal expression of activation-
induced cytidine deaminase (AID) in H. pylori-infected
Bacterial Strains Culture Conditions
gastric epithelial cells [6]. AID is an enzyme that
produces immune diversity by inducing somatic hyper- H. pylori strain 193C derived from gastric cancer patient
mutations (SHM) and class-switch recombinations (CSR) [20], was cultured in Brucella broth medium supple-
in human immunoglobulin (Ig) genes. The impaction of mented with 10% fetal bovine serum (FBS; i.e.,
AID as a genome mutator could aim at the generation of BB-FBS). The strains were subcultured before co-culture
somatic mutations in various host genes of nonlymphoid experiments in 10 mL Brucella broth liquid culture for
tissues like TP53 tumor suppressor gene and contribute 24–48 hours under microaerophilic conditions (5% O2,
to tumorigenesis. Thus, NF-jB and NF-jB-regulated 10% CO2, and 85% N2 at 37C; Sanyo-Multigas Incuba-
gene products, particularly AID, have been closely tor; SANYO Electric Co., Ltd. Tokyo, Japan) on a gyra-
linked with H. pylori-induced gastric carcinogenesis. tory shaker at 160 rpm with 100% humidity. The
Therefore, agents that suppress NF-jB pathway might concentration of bacteria was estimated by using the
have potential efficacy in preventing the occurrence of formula as an absorbance of 0.1 = 108 bacteria per mL
H. pylori-related cancerous lesions [7,8]. [21].
Since the role of H. pylori infection is to promote car-
cinogenesis rather than to act as a direct carcinogen,
Determination of Nonbactericidal Concentration
the eradication in long-standing infected gastric mucosa
of Curcumin
might be insufficient to halt or reverse the pathogenic
changes of gastric inflammation and tumorigenesis The anti-H. pylori concentration of curcumin was evalu-
[9,10]. A recent surge in nonantimicrobial approach ated by the method described earlier [22] with minor
such as anti-oxidants, probiotics, vitamins, plant modifications. Briefly, the BB-FBS medium routinely
extracts, and phytochemicals has opened unique used for growing H. pylori was prepared by adding
dimensions to control H. pylori-related inflammation various concentrations of curcumin as 1, 5, 10, 20, 40,
and precancerous lesions [11,12]. The anti-H. pylori and 80 lmol ⁄ L. Equal amounts of H. pylori liquid
effects of phytomedicines and their phytochemicals culture were added in each curcumin-supplemented
have been documented in the literature [13–15]. media after adjusting the absorbance at A600. Control
Curcumin, a major constituent of the spice named was prepared for each concentration by adding similar
Curcuma longa, is cited earlier for its anti-inflammatory, concentrations of curcumin in liquid media but without
cancer chemopreventive, and anti-H. pylori activity H. pylori inoculation. Cultures were grown in microaer-
[16,17]. Various biological and signaling effects of ophilic conditions as described before for different time
curcumin have been reported such as cell shrinkage, periods and the absorbance was measured at 600 nm.
chromatin condensation, and inhibition of nitric oxide
synthase, protein kinase C, NF-jB, and cyclin D1
Determination of DNA Fragmentation
[18,19]. In this study, we have examined the effect of
non-bactericidal concentrations of curcumin on the Quantitative DNA fragmentation assay was carried out
expression of AID in H. pylori-infected gastric epithelial according to the method of Sellins and Cohen [23].

ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595 589


Curcumin Downregulates AID in H. pylori-Infected Cells Haider et al.

Briefly, cells pretreated with or without curcumin


Quantitative Analysis of mRNA Expression
were lysed in a lysis buffer (10 mmol ⁄ L Tris,
1 mmol ⁄ L EDTA, 0.2% Triton X-100, pH 7.5) and For the evaluation of AID gene expression, cells were
centrifuged at 13,000 ·g for 10 minutes. Then, each co-cultured with H. pylori in the presence or absence of
DNA sample in the supernatant and the resulting curcumin, SN-50, and MG-132 for 24 hours, then
pellet were precipitated in 12.5% trichloroacetic acid washed thrice with PBS, scrapped by rubber policeman
(TCA) at 4C, and quantified using a diphenylamine and the total RNA was extracted by RNeasy Plus Mini
reagent after hydrolysis in 5% TCA at 90C for kit (Qiagen, Germany) according to the manufacturer’s
20 minutes. The percentage of fragmented DNA for instructions. Reverse transcription was performed using
each sample was calculated as the amount of DNA in the Exscript RT reagent kit (Takara Bio Inc., Shiga,
the supernatant divided by the total DNA for that Japan) and random primers, followed by real-time
sample (supernatant plus pellet). polymerase chain reaction (RT-PCR). Amplification of
AID and glyceraldehyde-3-phosphate dehydrogenase
(GAPDH) was performed using SYBR Premix Ex Taq
Co-Culture of Helicobacter pylori with Gastric
(Takara Bio Inc.). The following primer pairs were used:
Epithelial Cells
AID, forward 5¢-AATTAGCCAGGCGTGGTAGCAG-3¢
Human gastric carcinoma-derived MKN-45 epithelial and reverse 5¢-TGCAACGGCACGATCTCAG-3¢; GAPDH,
cells (Riken Cell Bank, Tsukuba, Japan) were grown in forward 5¢-GCACCGTCAAGGCTGAGAAC-3¢ and
RPMI 1640 containing 2 mmol ⁄ L of l-glutamine and reverse 5¢-TGGTGAAGACGCCAGTGGA-3¢. Real-time
10% FBS at 37C in 5% CO2. MKN-45 cells were (RT)-PCR was carried out using the Takara Thermal
routinely passaged every 3 days. Cells were seeded into Cycler Dice Real time system TP800 (Takara Bio Inc.).
6-cm culture dish and grown for 24 hours followed by AID mRNA was normalized to GAPDH mRNA as an
washing with phosphate-buffered saline (PBS) thrice. internal control in each sample. Results were expressed
The medium RPMI 1640, without antibiotics and FBS, as the ratio relative to the average of uninfected cells.
was added finally 3 hours before addition of H. pylori.
H. pylori were cultured overnight in BB-FBS (10%)
Immunoblot Analysis
under the conditions mentioned before and then
washed twice with PBS. Bacteria were then directly MKN-45 cells were maintained in RMPI-1640 medium
added to MKN-45 cells for the indicated times. as mentioned before. Cultures were kept at 37C in a
humidified atmosphere of 5% CO2 ⁄ 95% air. MKN-45
cells were co-cultured with H. pylori for 45 minutes.
Anti-Adhesion Activity Assay by ELISA
Curcumin was added for 60 minutes before the addi-
MKN-28 and MKN-45 cells, derived from human gas- tion of H. pylori. Whole cell lysates from MKN-45 cells
tric carcinomas, were used for the analysis of H. pylori were prepared with lysis buffer (25 mmol ⁄ L of HEPES
adhesion by the method described earlier [24,25]. pH 7.7, 0.3 mol ⁄ L of NaCl, 1.5 mmol ⁄ L of MgCl2,
Briefly, cells were preincubated with or without Curcu- 0.2 mmol ⁄ L of EDTA, 0.1% Triton X-100, 20 mmol ⁄ L
min (2.5–10 lM) in 96-well plates at 37C under 5% of b-glycerophosphate, 0.1 mmol ⁄ L of sodium ortho-
CO2 for 90 minutes and H. pylori was added in the trea- vanadate, 0.5 mmol ⁄ L of phenylmethylsulfonyl
ted and untreated wells. The cells were then fixed at fluoride, 1 mmol ⁄ L of dithiothreitol, 10 lg ⁄ mL of apro-
4C for 60 minutes and after washing, 100 lL of rabbit tinin, and 10 lg ⁄ mL of leupeptin). Cell lysates were
anti-H. pylori polyclonal antibody (DakoCytomation, resolved by SDS-PAGE and transferred to an
Glostrup, Denmark) was added to each well, and the Immobilon-P nylon membrane (Millipore, MA, USA).
plates were incubated for 2 hours at 37C. After wash- The membrane was treated with BlockAce (Dainippon
ing, 100 lL of peroxidase-conjugated goat anti-rabbit Pharmaceutical Co. Ltd., Suita, Japan) overnight at 4C
Igs (Wako) diluted 1 : 1000 in PBS was added to each and probed with antibodies against human AID from
well and incubated for 2 hours at 37C. After final ProSci Inc. (Poway, CA, USA) and NF-jB p65 (C-20-G),
washing, 100 lL of substrate reagent pack (DY999; IjBa (C-21), actin (C-11) from Santa Cruz Biotechnol-
R&D System Inc., Minneapolis, MN, USA) was added ogy (CA, USA), and NF-jB phospho-p65 (Ser536;
for 15 minutes followed by addition of 100 lL of 93H1), IKKa, and phospho-IKKa ⁄ b (ser176 ⁄ 180) from
1 mol ⁄ L H2SO4 to terminate the reaction. The optical Cell Signaling Technology (Boston, MA, USA). The pri-
density (OD) of the reaction was measured at 490 nm mary antibodies were detected using horseradish perox-
with a microplate reader. The OD represents the idase-conjugated anti-rabbit, or anti-goat IgG (Dako)
amount of H. pylori adhering to target cells. and visualized with the enhanced chemiluminescent

590 ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595


Haider et al. Curcumin Downregulates AID in H. pylori-Infected Cells

system (Amersham Biosciences, Buckinghamshire, UK). incubation when compared with untreated cells. With
Ramos cell lysate (ProSci Inc.) was employed as a posi- 24 hours of incubation, curcumin showed high level of
tive control for AID expression. DNA fragmentation at 50 lmol ⁄ L compared with the
control (data not shown). Hence, concentrations of
curcumin less than 20 lmol ⁄ L were deemed suitable
Statistical Analysis
for co-culture experiments.
The results are expressed as the mean ± SD. Statistic
significance (p < .01, p < .05) was evaluated by one-
Determination of Anti-Adhesion Activity of
way ANOVA followed by Bonferroni post-hoc test for
Curcumin against Helicobacter pylori Binding to
real-time PCR analysis of AID expression.
Gastric Epithelial Cells
Previous studies have demonstrated that colonization of
Results
H. pylori is an initial step for H. pylori-induced gastric
inflammation. After adhesion, H. pylori inject various
Nonbactericidal Concentration of Curcumin
factors by type IV secretion system that are responsible
As a first step, we analyzed the growth of H. pylori in for gastric carcinogenesis. Hence, an agent that could
10% BB-FBS medium supplemented with different curc- inhibit the adhesion of H. pylori to gastric epithelial cells
umin concentrations (1–80 lmol ⁄ L), to determine the might hamper H. pylori-related pathogenesis in stomach
nonbactericidal concentration of curcumin. A previous [25,26]. To examine the anti-adhesion activity of
study by Foryst-Ludwig et al. demonstrated no effect on different nonbactericidal concentrations of curcumin
H. pylori viability upto 80 lmol ⁄ L of curcumin when (2.5–10 lmol ⁄ L), enzyme-linked immunosorbent assay
preincubated for 1 and 3 hours [19]. The results of our (ELISA) was employed in two gastric cancer cell lines,
study coincides with these results as curcumin MKN-28 and MKN-45. The results revealed that
(£ 80 lmol ⁄ L) showed no significant effect on H. pylori adhesion of H. pylori to gastric epithelial cells was not
growth up to 5 hours culture. However, H. pylori inhibited by curcumin pretreatment at nonbactericidal
growth was prominently inhibited after 5 hours culture concentrations (Fig. 1). These results indicate that
at a concentration ‡ 20 lmol ⁄ L (data not shown). possible effects of curcumin (£ 10 lmol ⁄ L) in H. pylori-
Concentrations of curcumin at £ 10 lmol ⁄ L showed no infected gastric epithelial cells will not be either a result
bactericidal effect on the growth of H. pylori till 36 hours; of alteration of bacterial viability or by inhibition of
hence, these concentrations were employed for further H. pylori adhesion to epithelial cells.
assays. To test the effects of curcumin on the viability of
the bacteria, cultures containing different concentrations
Curcumin Downregulated AID Expression in
of curcumin (2.5–10 lmol ⁄ L) were plated after serial
Helicobacter pylori-Infected Cells
dilutions onto H. pylori-selective agar plates under micro-
aerophilic conditions, and the numbers of CFU The mRNA expression of AID was analyzed by real-
(CFU ⁄ mL) were determined, as previously described time PCR. As described earlier [6], AID expression is
[22]. The results revealed that the numbers of CFUs
recovered from the liquid cultures containing curcumin
£10 lmol ⁄ L (curcumin 2.5 lmol ⁄ L; 55 · 106 CFU ⁄ mL,
curcumin 5 lmol ⁄ L; 61 · 106 CFU ⁄ mL, and curcumin
10 lmol ⁄ L; 57 · 106 CFU ⁄ mL) were similar to that of the
liquid culture without curcumin (62 · 106 CFU ⁄ mL).

Nontoxic Concentration of Curcumin Evaluated by


DNA Fragmentation
To evaluate whether nonbactericidal concentration of
curcumin possess some toxicity against MKN-45 cells,
Figure 1 Evaluation of anti-adhesion activity of curcumin against
DNA fragmentation, a feature characteristic of apopto-
Helicobacter pylori binding to MKN-28 and MKN-45 cells. Cells were
sis, was measured concentration dependently at 6 and treated with various concentrations of curcumin (2.5–10 lmol ⁄ L) for
24 hours of incubation. The results revealed no signi- 90 minutes. The amount of adherent H. pylori is expressed as the
ficant induction of DNA fragmentation on any concen- percentage of the amount of H. pylori adhering to untreated cells.
tration (5–50 lmol ⁄ L) of curcumin at 6 hours of Each value represents the mean ± SD (n = 3).

ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595 591


Curcumin Downregulates AID in H. pylori-Infected Cells Haider et al.

upregulated via NF-jB activation in H. pylori-infected A


cells; hence, we evaluated the effect of curcumin on
AID expression. Initially, H. pylori was co-cultured with
cells for 6, 12, and 24 hours followed by AID mRNA
expression analysis. AID was not upregulated in 6 and
12 hours co-culture with H. pylori; however, after
24 hours co-culture the AID mRNA was significantly
(p < .01) overexpressed (Fig. 2A). Further, gastric epi-
thelial cells were pretreated with curcumin (5 and
10 lmol ⁄ L) for 1 hour followed by H. pylori co-culture
for 24 hours, and the extracted RNA was employed for
B
real-time PCR analysis. After pretreatment of curcumin
at 5 and 10 lmol ⁄ L, the expression of AID was down-
regulated significantly (p < .05 and p < .01, respec-
tively) in a concentration-dependent manner (Fig. 2B).
Immunoblot analysis showed that AID protein expres-
sion was increased substantially in H. pylori-infected
MKN-45 cells which was considerably inhibited by
curcumin (10 lmol ⁄ L) pretreatment (Fig. 2C). To clarify
whether NF-jB transcription pathway might have
involved in the regulation of AID, NF-jB activation
inhibitor (SN-50) and proteasome inhibitor (MG-132)
C
were examined by real-time PCR at the concentrations
(250 ng ⁄ mL and 0.15 lmol ⁄ L, respectively) mentioned
earlier [6]. The cells were preincubated with SN-50 and
MG-132 for 2 hours followed by addition of H. pylori.
The results revealed almost complete inhibition of AID
expression with both the inhibitors as shown in Fig. 3,
which is suggestive of NF-jB involvement in AID
Figure 2 MKN-45 cells co-cultured with Helicobacter pylori strain
expression in MKN-45 cells.
193C induces AID mRNA and protein expression. (A) Expression of AID
mRNA in H. pylori-infected cells at different time intervals (6, 12, and
Curcumin Suppressed NF-jB Activation by 24 hours). Cells were incubated with or without H. pylori and RNA
was extracted. Data represent the mean values of three independent
Inhibition of IKK Activity and IjB Degradation
experiments. **p < .01 (compared with that of the control). (B) Effect
As previously reported [19] that curcumin inhibits of curcumin on the expression of AID in H. pylori-infected cells.
NF-jB activation by hampering IKK activity and subse- Cells were plated in culture dishes till 80% confluence and incubated
with or without curcumin (5 and 10 lmol ⁄ L) for 60 minutes followed
quent IjB destruction, we further examined this effect
by the addition of H. pylori. Total RNA was isolated after 24 hours of
by Western blot at nonbactericidal concentrations of co-culture and AID mRNA expression was normalized to glyceralde-
curcumin (5 and 10 lmol ⁄ L). As shown in Fig. 4, it can hyde-3-phosphate dehydrogenase mRNA as an internal control in each
be seen that IjBa is phosphorylated and degraded in sample. Results were expressed as the ratio relative to the uninfected
H. pylori-co-cultured and TNF-a-treated cells; in con- cells. Data represents the mean values of three independent experi-
trast, IjBa degradation was inhibited by pretreatment ments; **p < .01 and *p < .05 (compared with H. pylori-infected cells
of curcumin in a concentration-dependent manner. without curcumin). (C) Immunoblot of lysates from MKN-45 cells either
left untreated or treated with curcumin followed by infection with
Similarly, H. pylori- and TNF-a-induced NF-jB p65
H. pylori for 24 hours using human AID antibody. Ramos cell lysate
phosphorylation was also contained by curcumin (RCL) was used as positive control for AID expression.
pretreatment at 5 and 10 lmol ⁄ L. To evaluate whether
this phosphorylation of NF-jB p65 and IjBa is
regulated by IKK activity, we further carried out immu-
Discussion
noblot analysis using phospho-IKKa ⁄ b antibody. The
results showed inhibition of IKK activity by curcumin Previous global cancer studies reported 876,000 new
pretreatment particularly at a concentration of cases and more than 649,000 deaths from gastric cancer
10 lmol ⁄ L while there was no effect on the expression which remains still the second leading cause of cancer
of IKKa. deaths worldwide [27,28]. H. pylori plays a key role in

592 ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595


Haider et al. Curcumin Downregulates AID in H. pylori-Infected Cells

Figure 3 Co-culture of Helicobacter pylori with MKN-45 cells in the


presence or absence of nuclear factor (NF)-jB activation inhibitors
SN-50 (250 ng ⁄ mL) and MG-132 (0.15 lmol ⁄ L). Cells were incubated
with or without inhibitors for 2 hours before H. pylori inoculation. Figure 4 Effects of nonbactericidal concentration of curcumin on
RNA was extracted after 24 hours of co-culture and AID mRNA expres- Helicobacter pylori-induced nuclear factor (NF)-jB activation via
sion was examined. Data represent the mean values of three indepen- modulating degradation of NF-jB inhibitors (IjB), and IjB kinase (IKK)
dent experiments; **p < .01 (compared with H. pylori-infected cells activation. MKN-45 cells were pretreated with or without curcumin at
without inhibitors). the indicated concentrations for 60 minutes followed by infection with
H. pylori for 45 minutes or tumor necrosis factor (TNF)-a stimulation
(10 ng ⁄ mL, 10 minutes). NF-jB phosphorylation, IjBa degradation,
gastric diseases and its presence increases the risk of and IKK activation in the cells were evaluated by immunoblotting using
antibodies against NF-jB phospho-p65 (p-p65), NF-jB p65 (p65),
gastric cancer 20-fold, compared with controls [9].
phospho-IjBa (p-IjBa), IjBa, IKKa, and phospho-IKKa ⁄ b (p-IKKa ⁄ b).
Recent advances in cancer research has opened new
insights in the molecular mechanism of gastric carcino-
genesis especially cancer initiation and promotion. The different Phase I clinical trials performed to determine
involvement of AID enzyme in tumorigenesis is one of safety have reported no toxicity of curcumin at doses as
the milestones in understanding the process of carcino- high as 15 g ⁄ day [37]. A recent study by Mario et al.
genesis providing a novel link between inflammation, comprising of 30 mg ⁄ day curcumin-based 1-week triple
mutagenesis, and cancer development [29]. The causa- therapy for H. pylori eradication mentioned significant
tive factors for upregulation of AID can be a viral infec- improvement of dyspeptic symptoms and reduction of
tion, lipopolysaccharides from bacteria, inflammation gastric inflammatory response even after 2 months of
and, more recently, H. pylori itself [6,7]. AID has been the therapy [11]. These propitious sequels of curcumin
identified as a key enzyme for CSR and SHM leading to prompted us to investigate its effect on AID regulation
chromosomal translocations and genetic mutations. in H. pylori-infected gastric epithelial cells. The results
Therefore, the blockade of AID can be a promising obtained from real-time PCR and immunoblot analysis
chemopreventive approach to control H. pylori-associ- vividly revealed that pretreatment with curcumin even
ated gastric cancers. at low concentrations, downregulated the expression of
Curcumin, a spice-derived polyphenol, is known for AID in H. pylori-treated MKN-45 cells. To elaborate that
its anti-inflammatory and chemopreventive activity. this downregulation of AID might be because of NF-jB
Several lines of evidence, including epidemiological inhibition, we have added NF-jB activation inhibitor
data, preclinical and clinical trials, suggest that curcu- (SN-50) and proteasome inhibitor (MG-132) before
min may be used as a potential candidate for cancer infecting cells with H. pylori. The results showed almost
chemoprevention in diseases like pancreatic cancer, complete downregulation of AID. Although, these
colorectal cancer, familial adenomatous polyposis, and inhibitors are not specific for only NF-jB pathway but
inflammatory bowel disease [30–36]. These studies also the results are suggestive that NF-jB might be responsi-
demonstrated low bioavailability of curcumin after oral ble for AID regulation in H. pylori-infected MKN-45
administration suggesting that the anticancer activity of cells as demonstrated earlier in AGS gastric cancer
oral curcumin may be limited to the gastrointestinal cell line [6]. Next, we examine the regulation of
tract. To evaluate the toxicity of curcumin, three molecules which are involved in NF-jB pathway by

ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595 593


Curcumin Downregulates AID in H. pylori-Infected Cells Haider et al.

low concentrations of curcumin. NF-jB is mainly com- 13 Zaidi SFH, Yamada K, Kadowaki M, Usmanghani K, Sugiyama
posed of a heterodimer of p65 and p50 subunits in most T. Bactericidal activity of medicinal plants, employed for the
treatment of gastrointestinal ailments, against Helicobacter pylori.
cell types and IKK can directly phosphorylate NF-jB
J Ethnopharm 2009;121:286–91.
p65 at Ser-536 residue which is suggested to be a useful 14 Wang YC, Huang TL. Screening of anti-Helicobacter pylori herbs
marker for detecting the constitutive activation of deriving from Taiwanese folk medicinal plants. FEMS Immunol
NF-jB in cancer cells [38,39]. As demonstrated in this Med Microbiol 2005;43:295–300.
study, curcumin suppressed H. pylori-induced NF-jB 15 Zaidi SFH, Yoshida I, Butt F, Yusuf MA, Usmanghani K,
Kadowaki M, Sugiyama T. Potent bactericidal constituents from
p65 and IjB phosphorylation by inhibiting IKK activity
Mallotus philippinensis against clarithromycin and metronidazole
in a concentration-dependent manner. resistant strains of Japanese and Pakistani Helicobacter pylori.
Taken together, low concentrations of curcumin Biol Pharm Bull 2009;32:631–6.
downregulated H. pylori-induced AID expression which 16 Aggarwal BB, Harikumar KB. Potential therapeutic effects
might be because of the inhibition of NF-jB activation of curcumin, the anti-inflammatory agent, against neuro-
degenerative, cardiovascular, pulmonary, metabolic, autoim-
in H. pylori-infected MKN-45 gastric cancer cells. Further
mune and neoplastic diseases. Int J Biochem Cell Biol
studies are required to explicit this potential role of 2009;41:40–59.
curcumin in preventing the development of cancer that 17 Mahady GB, Pendland SL, Yun G, Lu ZZ. Turmeric (Curcuma
can be employed in countries with high gastric cancer longa) and curcumin inhibit the growth of Helicobacter pylori, a
prevalence like East Asian countries including Japan. group 1 carcinogen. Anticancer Res 2002;22:4179–81.
18 Thayyullathil F, Chathoth S, Hago A, Patel M, Galadari S.
Rapid reactive oxygen species (ROS) generation induced by
References curcumin leads to caspase-dependent and -independent apo-
ptosis in L929 cells. Free Radic Biol Med 1999;27:945–50.
1 Goodwin CS, Armstrong JA. Microbiological aspects of Helico- 19 Foryst-Ludwig A, Neumann M, Schneider-Brachert W,
bacter pylori (Campylobacter pylori). Eur J Clin Microbiol Infect Dis Naumann M. Curcumin blocks NF-jB and the motogenic
1990;9:1–13. response in Helicobacter pylori-infected epithelial cells. Biochem
2 Covacci A, Telford JL, Giudice GD, Parsonnet J, Rappuoli R. Biophys Res Commun 2004;316:1065–72.
Helicobacter pylori virulence and genetic geography. Science 20 Marchildon PA, Sugiyama T, Fukuda Y, Peacock JS, Asaka M,
1999;284:1328–33. Shimoyama T, Graham DY. Evaluation of the effects of
3 Keates S, Hitti YS, Upton M, Kelly CP. Helicobacter pylori infec- strain-specific antigen variation on the accuracy of serological
tion activates NF kappa B in gastric epithelial cells. Gastroenter- diagnosis of Helicobacter pylori infection. J Clin Microbiol
ology 1997;113:1099–109. 2003;41:1480–5.
4 Karin M. Nuclear factor-jB in cancer development and pro- 21 Meyer F, Wilson KT, James SP. Modulation of innate cytokine
gression. Nature 2006;441:431–6. responses by products of Helicobacter pylori. Infect Immunol
5 Karin M, Neriah YB. Phosphorylation meets ubiquitination: 2000;68:6265–72.
the control of NF-jB activity. Annu Rev Immunol 2000;18:621– 22 Loh JT, Torres VJ, Cover TL. Regulation of Helicobacter pylori
63. cagA expression in response to salt. Cancer Res 2007;67:4709–
6 Matsumoto Y, Marusawa H, Kinoshita K, Endo Y, Kou T, 15.
Morisawa T, Azuma T, Okazaki IM, Honjo T, Chiba T. 23 Sellins KS, Cohen JJ. Gene induction by gamma-irradiation
Helicobacter pylori infection triggers aberrant expression of leads to DNA fragmentation in lymphocytes. J Immunol
activation-induced cytidine deaminase in gastric epithelium. 1987;139:3199–206.
Nat Med 2007;13:470–6. 24 Hayashi S, Sugiyama T, Yachi A, Yokota K, Hirai Y, Oguma K,
7 Marusawa H. Aberrant AID expression and human cancer Fuji N. A rapid and simple method to quantify Helicobacter pylori
development. Int J Biochem Cell Biol 2008;40:1399–402. adhesion to human gastric MKN-28 cells. J Gastroenterol Hepatol
8 Ralhan R, Pandey MK, Aggarwal BB. Nuclear factor-kappa B 1997;12:373–5.
links carcinogenic and chemopreventive agents. Front Biosci 25 Hayashi S, Sugiyama T, Amano K, et al. Effect of Rebamipide,
2009;1:45–60. a novel antiulcer agent, on Helicobacter pylori adhesion to
9 Malfertheiner P, Sipponen P, Naumann M, Moayyedi P, gastric epithelial cells. Antimicrob Agents Chemother 1998;42:
Megraud F, Xiao SD, Sugano K, Nyren O. Helicobacter pylori 1895–9.
eradication has the potential to prevent gastric cancer: a state- 26 Backert S, Ziska E, Brinkmann V, Zimny-Arndt U, Faucon-
of-the-art critique. Am J Gastroenterol 2005;100:2100–15. nier A, Jungblut PR, Naumann M, Meyer TF. Translocation
10 Han SU, Kim YB, Joo HT, Hahm KB, Lee WH, Cho YK, Kim of the Helicobacter pylori CagA protein in gastric epithelial
DY, Kim MW. Helicobacter pylori infection promotes gastric cells by a type IV secretion apparatus. Cell Microbiol
carcinogenesis in a mice model. J Gastroenterol Hepatol 2000;2:155–64.
2002;17:253–61. 27 Parkin DM, Bray FI, Devesa SS. Cancer burden in the year
11 Mario FD, Cavallaro LG, Nouvenne A, et al. A curcumin-based 2000. The global picture. Eur J Cancer 2001;37:S4–S66.
1-week triple therapy for eradication of Helicobacter pylori infec- 28 Murray CJ, Lopez AD. Alternative projections of mortality and
tion: something to learn from failure?. Helicobacter disability by cause 1990–2020: Global Burden of Disease Study.
2007;12:238–43. Lancet 1997;349:1498–504.
12 Lee SY, Shin YW, Hahm KB. Phytoceuticals: mighty but 29 Marusawa H. Aberrant AID expression and human cancer
ignored weapons against Helicobacter pylori infection. J Dig Dis development. Int J Biochem Cell Biol 2008;40:1399–402.
2008;9:129–39.

594 ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595


Haider et al. Curcumin Downregulates AID in H. pylori-Infected Cells

30 Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin and quercetin of adenomas in familial adenomatous
curcumin: preclinical and clinical studies. Anticancer Res polyposis. Clin Gastroenterol Hepatol 2006;4:1035–8.
2003;23:363–98. 35 Holt PR, Katz S, Kirshoff R. Curcumin therapy in inflammatory
31 Cheng AL, Hsu CH, Lin JK, et al. Phase I clinical trial of bowel disease: a pilot study. Dig Dis Sci 2005;50:2191–3.
curcumin, a chemopreventive agent, in patients with 36 Lopez-Lazaro M. Anticancer and carcinogenic properties of
high-risk or pre-malignant lesions. Anticancer Res 2001;21:2895– curcumin: considerations for its clinical development as a can-
900. cer chemopreventive and chemotherapeutic agent. Mol Nutr
32 Dhillon N, Aggarwal BB, Li L, Chiao P, Sarkar F, Wolf RA, Food Res 2008;52:S103–27.
Kurzrock R. Phase II trial of curcumin, an NF-jB inhibitor, in 37 Aggarwal BB, Sung B. Pharmacological basis for the role of
patients with advanced pancreatic cancer. J Clin Oncol curcumin in chronic diseases: an age-old spice with modern
2006;24:14151. targets. Cell 2008;30:85–94.
33 Sharma RA, McLelland HR, Hill KA, Ireson CR, Euden SA, 38 Sakurai H, Chiba H, Miyoshi H, Sugita T, Toriumi W. IjB kin-
Manson MM, Pirmohamed M, Marnett LJ, Gescher AJ, ases phosphorylates NF-jB p65 subunit on serine 536 in the
Steward WP. Pharmacodynamic and pharmacokinetic study of transactivation domain. J Biol Chem 1997;274:30353–6.
oral curcuma extract in patients with colorectal cancer. 39 Suzuki S, Singhirunnusorn P, Mori A, Yamaoka S, Kitajima I,
Clin Cancer Res 2001;7:1894–900. Saiki I, Sakurai H. Constitutive activation of TAK1 by HTLV-1
34 Cruz-Correa M, Shoskes DA, Sanchez P, Zhao R, Hylind LM, tax-dependent overexpression of TAB2 induces activation of
Wexner SD, Giardiello FM. Combination treatment with JNK-ATF2 but not IKK-NF-jB. J Biol Chem 2007;282:25177–81.

ª 2009 Blackwell Publishing Ltd, Helicobacter 14: 588–595 595

Potrebbero piacerti anche