You are on page 1of 17

Article

Epigenetic imprinting by commensal


probiotics inhibits the IL-23/IL-17 axis in an
in vitro model of the intestinal mucosal
immune system
Darab Ghadimi,*,1 Ulf Helwig,† Juergen Schrezenmeir,‡ Knut J. Heller,* and Michael de Vrese*
*Department of Microbiology and Biotechnology, Max Rubner-Institute, Kiel, Germany; †Partnership for Internal Medicine,
Oldenburg, Germany; and ‡Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institute, Karlsruhe, Germany
RECEIVED JUNE 11, 2011; REVISED MAY 30, 2012; ACCEPTED MAY 31, 2012. DOI: 10.1189/jlb.0611286

ABSTRACT the nuclear translocation of NF-␬B, as demonstrated by


The pathophysiology of IBD is characterized by a com- a decrease in the expression of MyD88, degradation of
plex interaction between genes and the environment. IRAK-1 and I␬B␣ expression of the nuclear NF-␬B p50/
Genetic and environmental differences are attributed to p65 subunits, p-p38 MAPK and p-MEK1, and NF-␬B-de-
the heterogeneity of the disease pathway and to the pendent luciferase reporter gene activity in LPS-stimu-
epigenetic modifications that lead to altered gene ex- lated cells. B. breve and LGG may exert their anti-in-
pression in the diseased tissues. The epigenetic ma- flammatory effects in the gut by down-regulating the
chinery consists of short interfering RNA, histone modi- expression of the IBD-causing factors (IL-23/IL-17/
fications, and DNA methylation. We evaluated the ef- CD40) associated with epigenetic processes involving
fects of Bifidobacterium breve (DSMZ 20213) and LGG the inhibition of histone acetylation and the optimal en-
(ATCC 53103), as representatives of commensal probi- hancement of DNA methylation, reflected in the limited
otics on the expression of IL-17 and IL-23, which play access of NF-␬B to gene promoters and reduced NF-
an important role in IBD, and on the epigenetic machin- ␬B-mediated transcriptional activation. We describe a
ery in a 3D coculture model composed of human intes- new regulatory mechanism in which commensal pro-
tinal HT-29/B6 or T84 cells and PBMCs. The cells were biotics inhibit the NF-␬B-mediated transcriptional acti-
treated with LPS in the presence or absence of bacte- vation of IBD-causing factors (IL-23/IL-17/CD40),
ria for 48 h, and the expression of IL-17, IL-23, and thereby simultaneously reducing histone acetylation
CD40 at the mRNA and protein levels was assessed and enhancing DNA methylation. J. Leukoc. Biol. 92:
using TaqMan qRT-PCR and ELISA, respectively. West- 895–911; 2012.
ern blotting was used to assess the expression of the
MyD88, the degradation of IRAK-1 and I␬B␣, the expres-
sion of the NF-␬B p50/p65 subunits, the p-p38 MAPK Introduction
and p-MEK1, as well as histone modifications. NF-␬B It has been suggested recently that the IL-23/IL-17 axis is in-
activity was assessed by NF-␬B-dependent luciferase volved in IBD [1– 4]. Inhibiting IL-23 expression may have a
reporter gene assays. The accumulation of Ac-H4 and therapeutic potential in IBD [4, 5], as it regulates the Th1/
DNA methylation was quantitatively assessed using col-
Th17 response in UC and CD [2– 4]. An elevation of IL-23 is
orimetric assays. B. breve and LGG diminished the
also associated with pathogen bacteria-induced gastritis [6].
LPS-induced expression of IL-17, IL-23, CD40, and his-
tone acetylation, while slightly enhancing DNA methyl- In addition to IL-17 and IL-23, CD40 costimulatory protein
ation. These effects were paralleled by a decrease in is an important IBD-causing factor [7]. With respect to the
cellular sources of these IBD-causing factors during IBD and
in the in vitro model of intestinal mucosal immune systems, it
can be concluded from previous and recent studies that IL-23
Abbreviations: 2/3D⫽two/three-dimensional, Ac⫽acetylated,
ATCC⫽American Type Culture Collection, CBP⫽CREB-binding protein, can be produced preferentially by IECs [8] and only to a
CD⫽Crohn’s disease, DSMZ⫽German Collection of Microorganisms and lesser extent by immune cells, such as PBMCs, under stimula-
Cell Cultures, GM⫽gentamycin, H3/H4⫽histone 3/4, IBD⫽inflammatory tion with LPS and inflammation conditions [9, 10]. Consider-
bowel disease, IEC⫽intestinal epithelial cell, LAB⫽lactic acid bacteria,
ing the major source of IL-17, Th17 cells and PBMCs are the
LGG⫽Lactobacillus rhamnosus GG, MD2⫽myeloid differentiation
protein 2, MEK1⫽MAPK/ERK kinase 1, MRS⫽de Man, Rogosa, Sharpe pivotal basis of IL-17 in physiologic and pathologic conditions
broth, OD600, OD450, and OD600⫽OD at 600, 450, and 570 nm, re-
spectively, p⫽phosphorylated, qRT-PCR⫽quantitative real-time
RT-PCR, SI⫽stimulation index, TEER⫽transepithelium electrical resis- 1. Correspondence: Department of Microbiology and Biotechnology, Max
tance, TSA⫽trichostatin A, UC⫽ulcerative colitis, WST-1⫽water- Rubner-Institute, Hermann-Weigmann-Strasse 1, 24103 Kiel, Germany.
soluble tetrazolium E-mail: darab.ghadimi@mri.bund.de

0741-5400/12/0092-895 © Society for Leukocyte Biology Volume 92, October 2012 Journal of Leukocyte Biology 895
[11, 12]. More recently, however, intestinal Paneth cells and of intestinal Th17 cells under inflammatory conditions. This
colonic epithelial cells have been reported to express mRNA 3D coculture model may mimic the in vivo situation, in which
levels of IL-17A and IL-17F, respectively [13]. These data, beneficial commensal probiotics interact with the host im-
therefore, suggest that there is an extracellular cross-talk be- mune cells via enterocytes. With respect to cytokine expression
tween IECs and PBMCs in the production of IL-17 and IL-23 and inflammatory responses, previous in vitro and in vivo stud-
under aberrant inflammation conditions and stimulation with ies have demonstrated that compared with conventional 2D
LPS, as IL-23 is reported to be sufficient for differentiation of cell cultures, scaffold-based, 3D-cultured cells (e.g., PBMCs)
human CD4⫹ T cells to Th17 cells and their marked produc- may be more physiologically relevant based on their superior
tion of IL-17 [11], which enhances the inflammatory responses ability to emulate the in vivo environment [25]. The levels of
of the IECs to LPS in vitro—a potential mechanism by which cell proliferation and cytokine secretion in the in vitro scaf-
IL-17 causes mucosal damage by CD [2]. With respect to fold-based, 3D-cultured PBMCs are well correlated with the
source of CD40, it is well known that CD40 can be produced extent of inflammatory responses observed in vivo [26]. Addi-
by IECs and immune cells, such as PBMCs. For example, LPS tionally, the 3D cultures have significantly higher expression
induces CD40 expression in human PBMCs through the activa- levels of key genes that function in cytokine activity, cell
tion of NF-␬B [14, 15]. Furthermore, CD40 expression was growth, and differentiation regulation [27] and interestingly,
demonstrated not limited to immune system cells but was also exhibit different LPS receptor expression, LPS-induced cyto-
expressed in human colon primary tumor samples and human kine responses, and CD14/MD2 expression [28]. Indeed, it
colon cell lines [16]. Although carcinoma intestinal cell lines has been reported that signal transduction in 3D coculture
do not represent the complex function of IEC in IBD, similar differs from that in 2D coculture. Additionally, 3D coculture
to mucosal tissue of IBD patients and freshly isolated human environments promote polarity and differentiation in normal
IECs, they express at least two major hallmarks of IBD, namely epithelial cells and enhance cell biological activities as com-
TLR4 and CD40 [7]. pared with 2D substrates [29]. Furthermore, it has been sug-
Recent studies indicate that the epigenetic machinery-de- gested that the 3D in vitro cell culture model of PBMCs is a
pendent regulation of mucosal cytokines plays an important valuable tool complementary to other models for studying the
role in the modulation of IBD [17]. Interactions with bacteria cells and mediators involved in the immunological events that
and their outer membrane (endotoxin, LPS component) have lead to the pathologically relevant morphological changes ob-
been shown to affect histone acetylation, the phosphorylation served during infection [30]. For these reasons, we investi-
and methylation states of the TLR4 genes, and the secretion of gated the effects of the LAB in this enterocyte-PBMC coculture
IL-8 in host cells [18]. For instance, Schmeck et al. [19] re- model using 2D and 3D PBMC cultures.
ported that intracellular Listeria induces the Ac-H4, the
p-Ac-H3 at the IL-8 promoter, and the recruitment of the his-
tone acetylase and the transcription CREB in endothelial cells. MATERIALS AND METHODS
Clinical impacts have also been shown for Legionella pneumo-
Propagation of bacteria
phila and Moraxella catarrhalis, both of which induce the aber-
B. breve (DSMZ 20213, Braunschweig, Germany) and LGG (ATCC 53103)
rant expression of proinflammatory cytokines [20] by enhanc-
were used in this study. The bacteria were propagated according to previ-
ing MAPK/NF-␬B activity and reducing histone deacetylase activ- ously published procedures [31]. Briefly, using 0.02% inoculums from bac-
ity [21]. Nonpathogenic, Gram-negative bacteria induce the terial stocks stored at ⫺80°C in 30% glycerol, the bacteria were grown an-
recruitment of NF-␬B to the IL-6 gene promoter in IECs via mod- aerobically in a modular atmosphere-controlled system-VA500 workstation
ulation of histone acetylation [22]. Furthermore, Hamon and with an airlock (Don Whitley Scientific, UK) in MRS broth medium (Lacto-
Cossart [23] reported that the activation of the p38 MAPK path- bacilli MRS, Merck, Darmstadt, Germany) at 37°C for 12 h. The Escherichia
coli TG1 strain (Cat. #BU-00035, Maxim Biotech, S. San Francisco, CA,
way by LPS-stimulated TLR4 induces the p-Ac-H3, which is crucial
USA) was grown at 37°C in Luria broth. The CFU/ml of the bacterial cul-
for recruiting NF-␬B to the promoters of certain genes, such as tures was estimated by measuring the OD600 of a 1:6 dilution and using an
IL-12. Additionally, the in vivo association among histone acetyla- experimentally derived conversion factor for each strain. The conversion
tion, the NF-␬B induction of inflammatory gene expression, and factors were determined by plating stationary-phase cells and determining
IBD has been reported recently [24]. the number of CFU/ml/OD600 U for each strain. The liquid cultures were
With respect to the LAB belonging to the natural intestinal centrifuged at 4°C and 5000 g for 10 min, using appropriate volumes to
microflora, a recently published study indicates that Lactobacil- yield the necessary CFU. After washing three times with endotoxin-free
DPBS (BioWhittaker, Lonza, Basel, Switzerland), the bacterial pellets were
lus plantarum inhibits LPS-induced IL-23 secretion by Caco-2
diluted to 1 ⫻ 107 CFU in 50 ␮l of the appropriate cell culture medium.
cells [8]. However, a role for the epigenetic machinery in me- These bacteria samples were added to the HT-29/B6 or T84 cells in the 3D
diating the influence of probiotic bacteria on the reduction of coculture cells to yield a 10:1 bacteria:cell ratio. The bacterial suspensions
the IL-23/IL-17 axis has not been investigated previously. were also plated on agar plates. The colonies were counted to confirm the
Therefore, we were interested in understanding if the epige- accuracy of their concentrations and the viability of the bacteria. For the
netic machinery might be involved in the in vitro IL-23/IL-17- experiments with heat-killed B. breve, the liquid culture was heated to 95°C
for 30 min.
inhibiting potential of commensal probiotics. We evaluated the
We initially included B. breve and LGG strains as representatives of Gram-
effects of B. breve and LGG on the LPS/TLR4/NF-␬B intracel- positive commensal probiotics and E. coli TG1 as a Gram-negative non-LAB
lular signaling cascades and on histone modifications using an strain and internal control to confirm that the observed effects are specific
in vitro 3D coculture model that uses two well-studied IEC for commensal probiotics. In the cases in which only one LAB strain was
lines—HT-29/B6 and T84 —and PBMCs as the original source tested, B. breve was preferentially used, as it releases metabolites that exert

896 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

an anti-inflammation effect capable of crossing the intestinal barrier [32], nal exposure of the intestinal epithelium to bacteria, the intestinal barrier
and the anti-inflammatory capacity of live and dead B. breve and its soluble function by the epithelial cells, and the translocation of the membrane-
factors has been tested in vitro in mucosal DCs and in vivo. In a murine permeable bacterial components and signaling components from the epi-
model of trinitrobenzene sulfonic acid-induced colitis, B. breve alleviated thelial cells to the subepithelial space containing the immunocytes. It fur-
inflammation, as shown by a statistically significant decrease in the proin- thermore enables the interaction between the PBMCs and epithelial cells
flammatory cytokines IL-23 and IL-6 [33, 34]. Furthermore, it has been via soluble, membrane-permeable substances, e.g., cytokines.
shown that another Bifidobacteria, Bifidobacterium infantis, suppresses proin-
flammatory IL-17 production in murine experiments in vivo [12]. Assessing the effect of the LAB on cytokine
expression
Cell culture Dose- and time-response experiments. To evaluate the impact of the dif-
Polarized HT-29/B6 and T84 cells. Model intestinal epithelia were pre- ferent concentrations of B. breve on the secretion of the IL-17 and IL-23
pared using HT-29/B6 or T84 cells, each plated separately at 1 ⫻ 105 proinflammatory cytokines, the upper compartment bathing the filter-
cells/cm2 on Transwell-COL inserts (Corning, Kaiserslautern, Germany), as grown HT-29/B6 and T84 intestinal epithelial monolayers was filled with
described previously [35, 36]. The cells were grown in RPMI-1640 culture medium alone (control) or medium containing increasing concentrations
medium supplemented with 5% v/v FCS from Gibco (Invitrogen, of B. breve (2⫻106, 5⫻106, 1⫻107, 2⫻107, and 4⫻107 CFU/ml, correspond-
Karlsruhe, Germany) and maintained at 37°C in a humidified 5% CO2 in- ing to bacteria:cell ratios of 1, 2.5, 5, 10, and 20, respectively) in the ab-
cubator. The HT-29/B6 and T84 cells used in this study were assessed peri- sence or presence of basolateral-applied LPS (100 ng/ml) and incubated
odically with a Mycoplasma detection kit (Roche Applied Science, Manheim, for 48 h.
Germany) and found to be free of mycoplasma. To determine qualitatively For the time-response analysis, the cells were incubated with 2 ⫻ 107
if the HT-29/B6 and T84 cells had reached confluence and formed tight CFU/ml B. breve in the absence or presence of basolateral-applied LPS
junctions, the TEER across the monolayer was monitored using an Epithe- (100 ng/ml) for 6, 12, 24, 48, and 96 h in a similar subset of experiments.
lial Voltohmmeter electrode and Millicell-Electrical Resistance System (Mil- After incubation, the supernatants from the lower compartments were
lipore, Schwalbach, Germany). At the beginning of the experiments, the collected and centrifuged at 400 g for 10 min at 4°C to remove cells, and
TEER of all epithelial cells was measured, and for all experiments, only the expression of the IL-17 and IL-23 proteins was assessed by specific
intact, polarized cell monolayers exhibiting a net resistance of ⱖ600 ⍀ ⫻ ELISA kits, as indicated below in “Determination of cytokine and CD40
cm2 (HT-29/B6 cells) or ⱖ1300 ⍀ ⫻ cm2 (T84 cells) were used. The T84 protein levels”.
cells were found to be more potent than the HT-29/B6 cells at increasing Assessing the differences in the 2D and 3D PBMCs cultured separately
their tight junctions in monolayer culture, as revealed by the increased or in coculture with T84 cells and ascertaining the site of LPS action. To
TEER. The main reasons for selecting these two cell lines are that first, the investigate which part of the system was responsible for the secretion of
HT-29/B6 grow as highly differentiated, polarized epithelial monolayers IL-17 and IL-23 and whether the expression of IL-17 differs between the
and possess most of the features of native intestinal epithelia necessary for normal and scaffold-based, 3D-cultured PBMCs, 2D PBMCs (1.7⫻105
maintenance of intestinal barrier function [35]. Secondly, the T84 cells are cells/ml in 96-well cell culture microplates) and scaffold-based 3D PBMCs
crypt-like cells that have been widely used for studying secretion, transcellu- (34,000 cells/200 ␮l/scaffold) were cultured separately in the absence of
lar trafficking, inflammation [36], and the LPS-mediated TLR/NF-␬B sig- T84 cells and treated without or with LPS (100 ng/ml) for 48 h in the ab-
naling pathway [37]. sence or presence of B. breve at a bacteria:cell ratio of 10:1. Following this
The 2D and 3D PBMC culture models. Human PBMCs were prepared incubation, the cell-free supernatants were analyzed for IL-17 and IL-23 by
according to previously published procedures [31]. Briefly, venous whole ELISA.
blood from healthy subjects was drawn into heparinized vacutainers and Furthermore, to ascertain the site of LPS action, HT-29/B6 and T84
diluted 1:1 with pyrogen-free physiological serum (0.9% NaCl). The PBMCs cells (2⫻106 cells/ml) grown in the 12-well Transwell-COL inserts were
were then isolated by density gradient centrifugation (1.077 g/ml) (Lym- treated with 100 ng/ml LPS from E. coli 026:B6 (Sigma, Taufkirchen, Ger-
phoprep, Axis Shield PoC AS, Oslo, Norway) and washed twice in endotoxin- many) [38], which was added to the upper or lower compartment for 48 h
free DPBS without Ca2⫹ and Mg2⫹ containing 5% FBS (Gibco, Invitrogen). in the absence or presence of B. breve at a bacteria:cell ratio of 10:1 (which
All of the PBMCs were ⬎95% viable immediately after purification, as as- was added to the upper compartment). Following incubation, the cell-free
sessed by the microscopic examination of trypan blue exclusion. supernatants from the upper and lower compartments were analyzed for
To prepare the 3D cell culture model, freshly isolated PBMCs (34,000 IL-23 by ELISA.
cells/200 ␮l/scaffold) were cultured in antibiotic-free RPMI-1640 culture Based on our preliminary dose- and time-response experiments, the fol-
medium in 12-well 3D insert scaffolds, according to the manufacturer’s sug- lowing parameters were chosen for the experiments: a bacteria:cell ratio of
gested protocol (3D Biotek, Brunswick, NJ, USA). 10:1, a 48-h incubation period, and 100 ng/ml LPS [38]. When the epithe-
The enterocyte-PBMC coculture model. To establish the 3D coculture lial cells reached confluence and formed tight junctions, the upper com-
model, confluent HT-29/B6 or T84 monolayers (2⫻106 cells/ml), grown partment containing the filter-grown epithelial monolayers was filled with
on the filter membranes of 12-well Transwell-COL inserts were cocultured medium alone or medium containing 2 ⫻ 107 CFU/ml (corresponding to
with 2D (1.7⫻105 cells/well) and 3D (34,000 cells/200 ␮l/scaffold) PBMCs a 10:1 bacteria:cell ratio) B. breve, heat-killed B. breve, LGG, or E. coli TG1.
that were placed in the lower compartment of the coculture wells, i.e., the LPS was added to the lower compartment of this transwell coculture sys-
bottom of the culture plates (Fig. 1). This environment simulates the lumi- tem, which contained the 3D insert scaffolds with the 3D culture of the

Figure 1. Schematic illustration of the 3D


coculture models showing the experimental
Transwell insert setup of the transwell system used to assess
LPS-mediated inflammation. The transwell
inserts on which HT-29/B6 or T84 cells were
Upper compartment HT-29/B6 or T84 cells cultured were inserted into multiple wells
• • • • • • containing the PBMC-seeded 3D insert scaf-
Microporous membrane
Lower compartment folds (placed on the bottom of the culture
• • • •• • • 3D Insert scaffold containing 3D PBMCs plates).

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 897


PBMCs placed at the bottom of the wells. After 48 h incubation, the super- dent experiments. Each TaqMan assay included a fixed standard, a no-tem-
natants from the upper and lower compartments were collected and centri- plate control, and a cDNA sample and was run in triplicate for each sam-
fuged at 1200 g for 10 min at 4°C. The cell-free supernatants were then ple. The data for each sample were normalized to the GAPDH mRNA level
sterilized by passage through a 0.2-␮m pore filter (Millipore, Germany), present in each sample and normalized again between the samples to the
and a 100-␮l aliquot of each cell culture was used for cell viability assays. levels of the IL-23 and IL-17 mRNA present in the control, and then the
The remainder of the cell culture supernatants was stored at ⫺80°C for the relative expression levels were determined.
analysis of cytokine and CD40 protein expression. The monolayer HT-
29/B6 and T84 cells and the 3D PBMCs were harvested for assays to deter-
DNA methylation and histone modification assays
mine mRNA expression, histone modifications, proliferation, NF-␬B activ-
ity, and NF-␬B-dependent luciferase reporter gene activity, as indicated be- In the 3D coculture models, the HT-29/B6 and T84 cells were initially in-
low. In some experiments, 10 ng/ml of a TLR4 antagonist (Cat. # tlrl-rslps, cubated with 2 ⫻ 107/ml B. breve for 30, 60, and 120 min in the absence or
InvivoGen, Toulouse, France) was added to the lower compartments as a presence of 100 ng/ml LPS, which was added to the lower compartment
control and to block the TLR4/MD2 complex pathway. Based on the prod- (basolateral surface). After incubation, cell lysates were prepared, and the
uct data sheet, this TLR4/MD2 complex inhibitor is guaranteed to block histone modifications, including the global Ac-H4 and the p-Ac-H3 on Ser-
the LPS-dependent TLR4/MD2 complex pathway by at least two distinct 10/Lys-14, were assessed by Western blot analysis performed according to a
mechanisms. The main mechanism consists of direct competition between standard protocol (Invitrogen). Briefly, protein samples (50 ␮g/lane) were
the underacylated LPS and hexa-acylated LPS for the same binding site on mixed with an equal volume of protein sample buffer (NuPAGE LDS) and
MD2, and the secondary mechanism involves the ability of the underacy- incubated at 85°C for 2 min. After incubation, the samples were subjected
lated LPS:MD2 complexes to inhibit the function of the hexa-acylated en- to SDS-PAGE (Precast, NuPAGE Novex, 4 –12% Bis-Tris) at 110 V for 80
dotoxin:MD2 complexes via TLR4. min and 130 V for 10 min. After electrophoresis, the proteins were trans-
The viability of the bacteria was determined by bacterial plate counts at ferred for 1.5 h at 400 mA in transfer buffer (NuPAGE transfer buffer)
the time of addition and after incubation. During the coincubation, LGG onto a 0.2-␮ PVDF membrane, which was blocked in blocking buffer (TBS)
and E. coli TG1 were able to survive, and they seemed to tolerate aerobic with 5% BSA for 30 min at room temperature, followed by repeated wash-
conditions for up to 48 h rather well. However, B. breve tended to be less ing. The membranes were then incubated overnight at 4°C and incubated
tolerant to aerobic conditions, and a significant (P⬍0.05) decrease in the with anti-Ac-H4 (Lys8; 1:200), -H4, -p-Ac-H3 (Ser10; 1:1000), or -H3 anti-
B. breve population was observed (from 6.88⫾0.33 log10 to 4.82⫾0.24 log10 bodies (Cell Signaling Technology, Beverly, MA, USA) for 1 h at room
CFU/ml; P⬍0.05). For this reason, we also included the counts of the dead temperature. After incubation, the blots were washed four times in washing
bacteria in our experiments. buffer (TBS containing 0.1% Tween 20 and 5% nonfat dry milk) and then
incubated with the secondary HRP-linked antibody (1:2000; Acris, Hidden-
hausen, Germany) in blocking buffer for 1 h at room temperature. Finally,
RNA extraction, semi-qRT-PCR, and TaqMan the membranes were developed with ECL Plus Western blotting detection
qRT-PCR reagents (Amersham), and the images were recorded with a cooled charge-
Semi-qRT-PCR analysis. The expression of IL-17 and IL-23 was screened coupled device camera (Fuji Film, Japan). In addition, a similar subset of
initially using semi-qRT-PCR. Total RNA was isolated from the HT-29/B6 experiments was performed to evaluate the epigenetic imprinting effects of
and T84 cells and PBMCs with an RNeasy mini kit (Qiagen, Hilden, Ger- B. breve and LGG during long-term incubation. The HT-29/B6 and T84
many) and treated with DNA-free (Ambion, HC, UK). The quality and cells were incubated for 48 h with the potent histone deacetylase inhibitor
quantity of the RNA samples were assessed by spectrophotometry, conven- TSA [40] (0.01 ng/ml; Th. Geyer, Hamburg, Germany), the histone
tional gel electrophoresis, and Agilent RNA 600 Nano Assays in a 2100 bio- deacetylase inducer GM (0.2 mM) [41], B. breve, or LGG, all of which were
analyzer (Agilent Technologies, Santa Clara, CA, USA) with the corre- added to the medium in the upper compartments (apical surface) in the
sponding software (version A.01.16). Total RNA (2 ␮g) was reverse-tran- presence of LPS, which was added to the medium in the lower compart-
scribed to cDNA using the Superscript first-strand synthesis system for RT- ments (basolateral surface). After incubation, the p-Ac-H3 and the Ac-H4
PCR, according to the manufacturer’s recommendations (Invitrogen). The were assessed as indicated above. Additionally, for the quantitative eval-
PCR amplification was performed using Platinum PCR Supermix (Invitro- uations, cell lysates were prepared, and the accumulation of acetylated
gen) on a T1 thermal cycler (Biometra, Germany). The primer pairs for histones and methylated DNA was quantitatively assessed using a
␤-actin (internal control) and for IL-23 and IL-17 were used as described EpiQuik Global Histone H3/H4 Acetylation Assay Kit and a Methylamp
previously [8, 39]. The PCR amplification with all primers was performed Global DNA Methylation Quantification Ultra Assay Kit (Epigentek
for 44 cycles of 95°C for 45 s, 56°C for 45 s, and 72°C for 1 min, followed Group, Farmingdale, NY, USA), respectively, according to the manufac-
by a final extension at 72°C for 10 min. After PCR amplification, 15 ␮l of turer’s instructions.
the products was separated by electrophoresis in a 1.5% agarose gel con-
taining 10 ␮g/ml ethidium bromide and visualized with a UV transillumi-
Assessment of the upstream activators of the NF-␬B
nator.
TaqMan qRT-PCR analysis. To further examine the cytokine expression
pathway, transfection, and the NF-␬B-dependent
profile, qRT-PCR was performed using the TaqMan system (Applied Biosys- luciferase reporter gene assay
tems, Darmstadt, Germany). The expression levels of IL-23, IL-17, and Aliquots of the cell lysates obtained after 2- and 48-h incubation periods
GAPDH (internal reference) were measured by PCR using TaqMan gene were also analyzed by immunoblotting for the degradation of IRAK-1 and
expression assays. A total of 1 ␮g RNA was transcribed to cDNA using I␬B␣, the expression of MyD88 and nuclear NF-␬B p50/p65 subunits, and
TaqMan reverse transcription reagents, and TaqMan qRT-PCR was per- the p-p38 MAPK and p-MEK1 using anti-IRAK-1, anti-MyD88, anti-p50, anti-
formed in 50 ␮l vol for IL-23, IL-17, and GAPDH using the TaqMan Uni- p65, anti-p-p38, anti-p38, anti-p-MEK1 (Ser 298), anti-I␬B␣ (Ser32/36; 5A5;
versal PCR Master Mix containing AmpliTaq Gold polymerase. The real- 1:2000), I␬B␣ (112B2; 1:1000), and rabbit anti-␤-actin mAb (Cell Signaling
time reactions had a final volume of 50 ␮l containing 25 ␮l TaqMan Uni- Technology).
versal PCR Master Mix, 22.5 ␮l TaqMan assays-on-demand primer/probe For the transcription activity assays, confluent HT-29/B6 and T84 mono-
mixture (GAPDH: Hs99999903_m1; IL-23: Hs01629265_sl; IL-17: layer cells were transiently transfected for 16 h with a NF-␬B-dependent lu-
Hs99999082_ml), and 2.5 ␮l ddH2O containing cDNA corresponding to ciferase reporter gene (Ad5␬B-LUC; Vector Biolabs, Philadelphia, PA,
10 –20 ng RNA (depending on the target gene) and run on an ABI PRISM USA) in serum-free medium (Gibco, Invitrogen) at different MOIs, as de-
7000 sequence detection system (all reagents were purchased from Applied scribed previously [36]. Ad5␬B-LUC, consisting of three consensus NF-␬B-
Biosystems). The results were analyzed with the SDS software package ver- binding sites, was linked to luciferase. After transfection, the medium was
sion 2.1. The TaqMan qRT-PCR was performed in at least three indepen- removed, and the cells were rinsed with PBS. Fresh complete medium was

898 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

added before the stimulation. The apical surface of the cells was then port, under basal, and more extensively, under inflammatory con-
treated with B. breve or LGG (bacteria:cell ratios 0, 5, 10) for 24 h in the ditions after LPS challenge [32].
presence or absence of 100 ng/ml LPS, which was added to the medium in
As maximum, significant decrease in the basal and LPS-in-
the lower compartments (basolateral surface). The cell extracts from the trans-
fected epithelial cells were prepared with an enhanced luciferase assay kit (BD
duced IL-23 and IL-17 levels was observed at 2 ⫻ 107 CFU/ml
PharMingen, Heidelberg, Germany), according to the manufacturer’s (corresponding to a 10:1 bacteria:cell ratio), this concentra-
instructions. The NF-␬B-dependent luciferase activity was measured on a tion was used for all subsequent experiments. Although delete-
GENios microplate luminometer (Tecan, Crailsheim, Germany), and the rious effects on the cells were not observed with a higher dose
results were normalized to the extract protein concentrations as measured of bacteria (4 ⫻ 107 CFU/ml), this high dose of bacteria did
with a Bio-Rad protein assay kit. The NF-␬B-dependent luciferase assays not inhibit the basal cytokines secretion relative to the 10:1
were performed in triplicate with cell extracts from at least three separate
bacteria:cell ratio. This dose was also used for LGG and E. coli
experiments, and each result is expressed as the mean value of the three
independent experiments ⫾ sem.
TG1, although the optimal doses of these three strains needed
to modulate cytokine production are likely different. Fig. 2C
and D shows that B. breve inhibited the basal and LPS-induced
Determination of cytokine and CD40 protein levels secretion of IL-23 and IL-17 in the lower compartments in a
The levels of the IL-17, IL-23, and CD40 proteins in the cell-free superna- time-dependent manner.
tants were quantified using specific human IL-17, IL-23, and CD40 ELISA
Markedly, in the presence of LPS, IL-17 and IL-23 produc-
development kits (PeproTech, Hamburg, Germany; eBioscience, NatutTec,
tion was inhibited significantly as early as 24 h, with maximal
Frankfurt, Germany; and Abnova, Heidelberg, Germany, respectively). The
detection limit of the assay was 4 pg/ml for IL-17, 16 pg/ml for IL-23, and inhibitory effect after 48 h of culture, and lasted for 96 h. As
⬍1 pg/ml for CD40. The OD values of the samples were OD450 and OD570 the maximal, significant decreases in the cytokine levels were
on an ELISA plate reader (Molecular Devices, Munich, Germany). Each observed at 48 h (P⬍0.05), this incubation time was used for
ELISA was performed in triplicate with cell-fee supernatants from at least all of the subsequent experiments.
three separate experiments, and each result is expressed as the mean value
of three independent experiments ⫾ sem. The differences between the 2D and 3D PBMCs
cultured separately or in coculture with T84 cells and
Cell viability and proliferation assays ascertaining the site of action of LPS
In all experiments, the cell viability and cytotoxicity were determined To determine which part of the system was responsible for the
routinely using the trypan blue exclusion and WST-1 assays, which were secretion of IL-17/IL-23, and if there was a difference in the
conducted according to the manufacturer’s instructions (Roche Applied IL-17 expression between the conventional (2D) - and 3D-cul-
Science). Cell proliferation was evaluated by measuring DNA synthesis
tured PBMCs, we separately cultured 2D and 3D PBMCs with-
using a cell proliferation Biotrak ELISA kit (GE Healthcare, Munich,
Germany) as described previously [42]. Proliferation is expressed as the
out IECs. As shown in Fig. 2E, LPS significantly enhanced the
SI, which was calculated as the ratio of the mean OD of the stimulated: production of IL-17 and IL-23 in the 2D-cultured PBMCs.
unstimulated cultures, and a proliferative response with an SI ⬎2 was However, the basal secretion of IL-17 in the 3D-cultured
regarded as positive. PBMCs was significantly higher than that observed in the con-
ventional 2D PBMCs. These data imply that the PBMCs pro-
Statistical analysis duce IL-17 and IL-23 in response to LPS and that the IECs are
not the only source of IL-23.
The data were analyzed by ANOVA, and the comparisons among the
groups were performed using the Kruskall-Wallis ANOVA and the Moreover, to determine the site of LPS action, the apical
Mann-Whitney U-test. All of the experimental data are expressed as the versus basolateral LPS-stimulating activity was tested in polar-
mean ⫾ sem. The data were considered statistically significant when P ⱕ ized HT-29/B6 and T84 cells grown on Transwell filters. In
0.05. this model, the total amounts of IL-17 and IL-23 secreted into
the medium of the upper and lower compartments were mea-
sured after apical and basolateral LPS stimulation, respectively.
RESULTS We observed that the basal secretion of IL-17 by the HT-29/B6
and T84 cells was negligible, and its level was always lower
B. breve inhibits IL-23/IL-17 secretion in a dose- and than the detection limit indicated by the manufacturer. LPS
time-dependent manner applied apically or basolaterally was also not able to induce
As shown in Fig. 2A and B, B. breve inhibited the secretion of IL-17 in the HT-29/B6 and T84 cells. Similarly, as shown in
basal and LPS-induced IL-17 and IL-23 in the 3D coculture Fig. 2F, LPS added to the medium in the upper compartments
model constructed with the HT-29/B6/T84 IEC line and PBMCs. (apical surface) did not significantly affect the IL-23 levels in
In the case of IL-17 and IL-23 basal levels, a significant decrease the supernatants from the upper and lower compartments. In
was only observed at 2 ⫻ 107 CFU/ml (10:1 bacteria:cell ratio) in contrast, when LPS was added to the medium in the lower
both cell lines. In the presence of LPS, however, significant de- compartments (basolateral surface), a significant induction of
creases in the IL-23 and IL-17 levels were observed at 1 ⫻ 107, IL-23 release was observed in the supernatants from the upper
2 ⫻ 107, and 4 ⫻ 107 CFU/ml (5:1, 10:1, and 20:1 bacteria:cell and lower compartments. However, the basolateral secretion
ratios) with a maximal effect observed at 2 ⫻ 107 CFU bacteria/ was significantly greater than the apical secretion upon baso-
ml. These results are in agreement with a previous study showing lateral LPS stimulation (Fig. 2F). These results describe the
that anti-inflammatory capacity of B breve and Streptococcus thermo- rationale for choosing basolateral LPS stimulation in subse-
philus conditioned media was retained after transepithelial trans- quent experiments and confirm previous studies with animal

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 899


A B
T84 cells, B.breve T84 cells, B.breve
180 250 T84 cells, LPS+ B.breve
T84 cells, LPS+B.breve
HT-29/B6 cells, B.breve HT-29/B6 cells, B.breve
160 HT-29/B6 cells, LPS+B.breve HT-29/B6 cells, LPS+ B.breve

200
140

120

IL-23 (pg/ml)
IL-17(pg/ml)

§
150
100
§
§
80 § §
§ 100 §

60 *
* *
40 50
*
20

0 0
0 2x10*6 5x10*6 1x10*7 2x10*7 4x10*7 0 2x10*6 5x10*6 1x10*7 2x10*7 4x10*7
--------------------------------------------------------------------- ----------------------------------------------------------------------
B.breve concentrations(CFU/ml) B.breve concentrations(CFU/ml)

C D
T84 cells, B.breve T84 cells, B.breve
T84 cells, Control T84 cells, Control
280 400
T84 cells, LPS T84 cells, LPS
T84, LPS+B.breve T84 cells, LPS+ B.breve
HT-29/B6 cells, B.breve HT-29, B.breve
240
HT-29/B6 cells, Control HT-29/B6 cells, Control
320
HT-29/B6 cells, LPS HT-29/B6 cells, LPS
200 HT-29/B6 cells, LPS+B.breve HT-29/B6 cells, LPS+B.brve
IL-23(pg/ml)
IL-17(pg/ml)

240
160
§ §
§
§
120 §
§ 160

80

80
**
40 **
* **

0 0
6 12 24 48 96 6 12 24 48 96

Incubation periods (h) Incubation periods(h)

E F
1500 1200
IL-23 in 2D PBMCs Apical,T84 cells
IL-17 in 2D PBMCs Apical, HT-29/B6 cells
1250 IL-17 in 3D PBMCs 1000 *ϕ Basolateral, T84 cells
* Basolateral, HT-29/B6 cells
Cytikines (pg/ml)

1000 800
*
IL-23 (pg/ml)

*
§ϕ
750 § 600

* §
__#__
500 400 §
§ §
§ §
§
§
ϕ § § §
250 200
§ *

0 0
Control LPS TLR4 antagonist B.breve
______________________ ol
Contr PS(ap
ic) aso) c)
aso
)
pic
)
aso
) _______
apic baso
api
L S(b ist( nist(b nist(a nist(b LPS
LPS LP agon g o a g o g o
n t nta 4 a n t nt a +B.breve(apic)
4a 4a 4a
T L R TLR ) + T L R + T L R
p i c s o )
S(a PS(ba
LP L

900 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

and human IECs showing that administration of LPS to the and PBMCs, although this was not statistically significant.
basolateral surface of rat IEC-6 cells disrupted the barrier func- These differences may reflect the physiological differences in
tion of tight junction, in terms of TEER, whereas LPS adminis- the response of the HT-29/B6 and T84 cell lines to the treat-
tered to the apical surface altered neither the barrier function ments [36] and/or to the PBMC-derived mediators. With re-
nor the localization of 7H6 antigen in rat IEC-6 cells [43]. gard to IL-23 secretion, the profiles of the IL-23 levels were
This is a result of the fact that the expression of TLR4, its co- similar to those observed for IL-17 (Fig. 3B). These data show
receptor MD2, or both, which are necessary for LPS respon- that polarized HT-29/B6 and T84 cells preferentially and con-
siveness in IECs, is polarized to the basolateral membrane in stitutively secreted basal IL-23 into the basolateral side in com-
IECs. Hence, LPS sensing and LPS responsiveness occur along parison with the apical side, and IL-23 concentrations in baso-
on the basolateral membrane of polarized IECs in culture, lateral media were significantly higher than that observed in
such as the human colonic adenocarcinoma T84 cell line [37, the apical side, suggesting no potential flow of medium be-
44]. Moreover, bacterial LPS, added to the apical surface of tween the apical and basolateral compartments. However, ba-
polarized IECs, has previously been shown to be transported solateral-to-apical differences in IL-23 production were dimin-
from the apical to the basolateral pole of the epithelium [45]. ished by LPS, alone or in combination with E. coli TG1. In
Finally, when Caco-2 cells were cocultured in the presence of contrast, B. breve and LGG inhibited the basal and LPS- in-
PBMCs, apical stimulation of polarized Caco-2 cells with LPS duced secretion of IL-23 into upper and lower compartments
had no effect on permeability of Caco-2 cells, as well as on ba- and notably, retained the basolateral-to-apical differences in
solateral secretion of proinflammatory cytokines, even at con- IL-23 production. This may simply reflect the greater barrier
centrations as high as 1 ␮g/ml [14]. integrity of cells, which strongly blocks any potential flow of
medium between the apical and basolateral compartments. To
B. breve and LGG inhibit the LPS-induced expression confirm that the observed effects were specific for B. breve and
of IL-17 and IL-23 mRNA and protein in the 3D LGG, we included a Gram-negative non-LAB strain, E. coli TG1,
coculture model constructed with IECs and PBMCs as an internal control. E. coli TG1, given alone, had no effect
In the 3D coculture system constructed with HT-29/B6 or T84 but resulted in an additive increase in IL-17 secretion in com-
cells and PBMCs, LPS, added only to the lower compartments, bination with LPS, overall, in accordance with a previous study
significantly induced the secretion of IL-17 in the lower com- [46], showing that E. coli TG1 as a proinflammatory control
partments when compared with the control. B. breve and LGG strain stimulated bone marrow-derived DCs to produce large
significantly inhibited the basal and LPS-induced secretion of amounts of proinflammatory cytokines. E. coli TG1 did not in-
IL-17 (Fig. 3A). As it is known that only Th17 cells produce hibit IL-23/IL-17 expression but rather, additively aggravated
IL-17, we measured the IL-17 concentration in the cell-free the LPS-induced IL-23/IL-17 expression in the HT-29/B6 and
supernatants of the lower compartments, where IL-17 may T84 cells. Therefore, the subsequent experiments were con-
be generated from naïve T cells upon LPS stimulation of the ducted without this strain.
PBMCs. Fig. 3A also shows that the IL-17 concentrations in the Like IL-23, B. breve and LGG diminished LPS-stimulated
3D cocultures of HT-29/B6 cells and PBMCs are relatively dif- CD40 protein expression (Fig. 3C). As IECs produce IL-23 in
ferent from those observed in the 3D cocultures of T84 cells response to LPS via the TLR4/MD2 complex [2, 37, 47], we

4
Figure 2. Effect of B. breve on IL-17 and IL-23 secretion in HT-29/B6 or T84 cells and PBMCs cultured either in 3D coculture models or sepa-
rately. (A–D) The dose- and time-dependent effects of B. breve on IL-23/IL-17 production by the 3D coculture models constructed with HT-29/B6
or T84 cells and PBMCs. The HT-29/B6 or T84 cells (2⫻106 cells/ml) were cultured in triplicate in 12-well Transwell-COL inserts, and freshly iso-
lated PBMCs (34,000 cells/200 ␮l/scaffold) were cultured in 12-well 3D insert scaffolds and placed on the bottom of the culture plates (lower
compartment). The upper compartment that bathes the filter-grown HT-29/B6 or T84 epithelial monolayers, was filled with medium alone or me-
dium containing increasing concentrations of B. breve in the absence or presence of basolateral-applied LPS (100 ng/ml) for 48 h. The cell-free
supernatants from the lower compartments were harvested, and the (A) IL-17 or (B) IL-23 concentrations were measured in triplicate by specific
ELISA. In the same set of experiments, the cells were cultured with 2 ⫻ 107 CFU/ml (a bacteria:cell ratio of 10:1) B. breve in the absence or pres-
ence of basolateral-applied LPS (100 ng/ml) for the indicated incubation periods, and the concentrations of IL-17 (C) and IL-23 (D) in the cell-
free supernatants of the lower compartments were measured as indicated above. *Significantly different when compared with control cell cultures
(in the case of HT-29/B6 and T84 cells) without addition of LPS, P ⬍ 0.05; §significantly different when compared with control cell cultures (in
the case of HT-29/B6 and T84 cells) with addition of LPS, P ⬍ 0.05. (E) Separately and without HT-29/B6 or T84 cells, the 2D PBMCs (1.7⫻105
cells/ml in 96-well cell culture microplates) and scaffold-based 3D PBMCs (34,000 cells/200 ␮l/scaffold) were treated without or with 100 ng/ml
LPS for 48 h in the absence or presence of B. breve at a bacteria:cell ratio of 10:1. Following the incubation, the cell-free supernatants were ana-
lyzed for IL-17 and IL-23 expression by ELISA. (F) To determine the site of LPS action, HT-29/B6 and T84 cells (2⫻106 cells/ml) were grown in
12-well Transwell-COL inserts and then stimulated for 48 h with 100 ng/ml LPS on the apical (upper compartment) or basolateral (lower com-
partment) pole in the absence or presence of B. breve at a bacteria:cell ratio of 10:1, which was added to the medium of the upper compartments.
Following the incubation, the cell-free supernatants from the upper and lower compartments were analyzed for IL-23 expression by ELISA. Each
ELISA was run in triplicate with cell-free supernatants from at least three separate experiments, and each result is expressed as the mean value of
three independent experiments ⫾ sem. *P ⬍ 0.05 versus control (in the case of F, it means significant for HT-29/B6 and T84 cells);
§P ⬍ 0.05 versus LPS alone (in the case of F, it means significant for HT-29/B6 and T84 cell). ␸P ⬍ 0.05 versus apical levels (in the case of F, it means
significant for HT-29/B6 and T84 cells); #. P ⬍ 0.05 vs. 2D cultured PBMCs; apic, Apical (upper compartment of the filters); baso, basolateral (lower
compartment of the filters).

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 901


A B
700 1400
HT-29/B6 cells AP, HT-29 cells
T84 cells + BL, HT-29/B6 cells
600 + 1200 AP,T84 cells +
BL, T84 cells +

500 1000 +
+
+
*

IL-23 (pg/ml)
IL-17(pg/ml)

400
* 800
* *
**
300 600
§
§
§ § § §
200 400
φ §
φ § §
§ § §
200 φ φ* φ§ φ§
100 φ φ * §
* § § * * §
* * * * * * * *
0 0
ve G 1 ol
LP .brev
S e G ve GG
1
G1 is
t
TG
l S ntr
LP .bre LG .bre
ve

tro TG
G

LG
1

st +L +T on
G

n Co
LG
re

ni
Co B B
T

B S S g
go
.b

S+

LP LP ta
S+

S+
B

an
ta
LP

LP
LP
S+

an

R 4
LP

L
4

+T
R
TL

PS
S+

L
LP

C D
* 1000
IL-23 in HT-29/B6 cells
600 AP, HT-29/B6 cells
BL,HT-29/B6 cells * * CD40 in HT-29/B6 cells
*
Basolateral concentrations (pg/ml)

AP, T84 cells IL-17 in HT-29/B6 cells


500 * BL,T84 cells 800 * IL-23 in T84 cells
* CD40 inT84 cells
IL-17 in T84 cells
CD40 (pg/ml)

400
600 *
*
300 § * §
§ §
§ § 400 §
§ § §
200 § §
§ §
§ 200
100 § * *
§§

0 0
rol LPS
t
ev
e G ve G ol S
ev
e
ev
e
t nis LG br
e LG nt r LP
Con go .br B. Co B.
br br
B.
t a B +
4a
n + PS d
ed
S L ille ill
LR LP t -k -k
S +T H ea at
LP he
S+
LP

Figure 3. Effect of LAB on IL-17, IL-23, and CD40 production in response to LPS in the coculture models constructed with HT-29/B6 or T84
cells in the upper compartment and 3D PBMCs in the lower compartments. The HT-29/B6 or T84 cells (2⫻106 cells/ml) were cultured in 12-well
Transwell-COL inserts, and freshly isolated PBMCs (34,000 cells/200 ␮l/scaffold) were cultured in 12-well 3D insert scaffolds and placed on the
bottom of the culture plates. LPS (100 ng/ml) was added to the culture medium of the lower compartments for 48 h in the absence or presence
of B. breve, heat-killed B. breve, or LGG (added to the culture medium of the upper compartment) at a bacteria:cell ratio of 10:1. As a control, 10
ng/ml of the TLR4/MD2 complex antagonist was added to the lower compartments. After the incubation, the supernatants from the upper and
lower compartments were harvested and analyzed by ELISA for the secretion of (A) IL-17 in the lower compartments IL-23 (B) and CD40 (C) in
the upper and lower compartments. In the case of heat-killed B.breve, IL-17, IL-23, and CD40 were assessed only in the lower compartments (D).
Each ELISA was performed in triplicate with cell-free supernatants from at least three separate experiments, and each result is expressed as the
mean value of three to five (in the case of A and B) independent experiments ⫾ sem. ␸P ⬍ 0.05 versus apical; *P ⬍ 0.05 versus control; §P ⬍
0.05 versus LPS alone; ⫹P ⬍ 0.05 versus TG1 alone. AP, Apical (upper compartment of the filters); BL, basolateral (lower compartment of the
filters).

902 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

used a TLR4 antagonist to block the TLR4/MD2 pathway and protein expression of IL-17, IL-23, and CD40 into the baso-
measured the protein levels of the inflammation markers IL- lateral compartments of 3D coculture models constructed
17, IL-23, and CD40 in the upper and lower compartments. with HT-29/B6 or T84 cells and PBMCs. In the case of the
Fig. 3A–C shows that the TLR4 antagonist largely abolished heat-killed B.breve, we measured IL-17, IL-23, and CD40 only
the LPS-stimulated production of these inflammation markers. in the lower compartments, as B.breve releases metabolites
Previous studies have revealed that probiotic bacteria and their that can cross the intestinal barrier under normal or inflam-
components inhibit LPS/TLR4/NF-␬B pathway-mediated aber- matory conditions. Furthermore, its soluble factors usually
rant inflammation [32, 48, 49]; however, anaerobic B. breve was retain their anti-inflammatory properties after transepithe-
not able to cope with the aerobic conditions of the cell cul- lial transport across intestinal cell monolayers, mainly in
ture. Therefore, we included heat-killed B. breve to determine inflammatory conditions [32].
whether the effects of the live B. breve to lower aberrant in- Furthermore, our preliminary, semi-qRT-PCR results demon-
flammation could also be mediated by dead B. breve. Fig. 3D strated that B. breve and LGG inhibited the LPS-stimulated ex-
shows that the heat-killed B. breve inhibited LPS-stimulated pression of IL-23 in the HT-29/B6 and T84 monolayers, as

A B
LPS+B. breve

10 HT-29/B6 cells
LPS+ LGG

T84 cells
B. breve

* *
Control

8
LGG

TaqMan real-time RT-PCR


LPS

* *
(Relative to GAPDH)
IL-23 mRNA

IL-23 in HT-29/B6 cells


4 §

§
§
§
2
IL-23 in T84 cells §
§
* * * *
0
Control LPS B.breve LGG B.breve LGG
---------------------
LPS
IL-23 in 3D PBMCs

Figure 4. Semi-qRT-PCR and TaqMan qRT-PCR analysis of IL-23 and


IL-17 in 3D PBMCs IL-17 mRNA expression in response to B. breve and LGG. The coculture
model was constructed with polarized monolayers of filter-grown HT-
29/B6 or T84 cells in the upper compartments and 3D PBMCs placed on
the bottom of the culture plates in the lower compartment of the filters.
β-actin B. breve and LGG (bacteria:cell ratio of 10:1) were added to the medium
of the upper compartments in the absence or presence of 100 ng/ml
LPS, which was added to the medium of the lower compartments. After a
C 48-h incubation, total RNA was extracted and reverse-transcribed to
cDNA, and the mRNA levels of IL-17, IL-23, and ␤-actin were analyzed
IL-23 in 3D PBMCs cocultured with HT-29/B6
IL-17 in 3D PBMCs cocultured with HT-29/B6
initially by semi-qRT-PCR using specific primers for IL-17, IL-23, and ␤-ac-
6 IL-23 in 3D PBMCs cocultured with T84 cells tin. After PCR amplification, 15 ␮l of the products was separated by elec-
IL-17 in 3D PBMCs cocultured with T84 cells trophoresis in a 1.5% agarose gel containing 10 ␮g/ml ethidium bromide
and visualized with a UV transilluminator (A). One representative result
Cytokines mRNA levels
(Relative to GAPDH)

*
from three independent experiments is shown. The IL-23 and IL-17
*
4 * mRNA levels were also analyzed in more detail by TaqMan qRT-PCR (B
*
* TaqMan real-time RT-PCR and C). The data for each sample are normalized to the GAPDH mRNA
* level present in each sample, normalized again between the samples to
the levels of IL-23 and IL-17 mRNA present in the control, and the rela-
§ tive expression levels are shown. Each TaqMan assay was performed in
§ §
2
§ §
§ triplicate for each sample. The data shown are the means ⫾ sem of three
§ §
independent experiments. *P ⬍ 0.05 versus control; §P ⬍ 0.05 versus LPS
alone.

0 * * * * * * *
Control LPS B.breve LGG B.breve LGG
-----------------------
LPS

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 903


A B

T84 cells HT-29/B6 cells


Time [min] - 30 60 120 Time [min] - 30 60 120
LPS +B. breve -p- Ac- H3 LPS+B. breve -p- Ac- H3

LPS -p- Ac- H3 LPS -p- Ac- H3

B. breve -p- Ac- H3 B. breve -p- Ac- H3


- H3
- H3

LPS +B. breve - Ac- H4 LPS+B. breve - Ac- H4

- Ac- H4 - Ac- H4
LPS LPS
B. breve - Ac- H4 B. breve - Ac- H4
- H4 - H4

C D
-p-Ac-H3
-p-Ac-H3
-H3
-H3
-Ac-H4 -Ac-H4

-H4
-H4
- + - - - - GM - + - - - - GM
- - + - - - TSA - - + - - - TSA
- - - + - + LPS - - - + - + LPS
- - - - + + B. breve - - - - + + LGG

E
50 HT-29/B6 cells
F
Histone H3 acetylation[ng/mg protein]

60 HT-29/B6 cells
T84 cells T84 cells * *
*
DNA metylation[ng/mg protein]

40 ** * 50

§ § §
§ § §
40
30 *
* * 30

20 §
§
§ § 20
* *
10
* * * 10

*
0
0 Control TSA GM LPS B.breve L.GG LPS+ LPS+
Control TSA GM LPS B.breve L.GG LPS+ LPS+
B.breve L.GG
B.breve L.GG

904 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

well as IL-17 and IL-23 in the 3D PBMCs (Fig. 4A). The IL-23 cumulation of Ac-H4. Finally, Fig. 5F shows that LPS decreased
and IL-17 mRNA levels were also analyzed using TaqMan qRT- DNA methylation as compared with the control. B. breve and
PCR, which is more reliable and sensitive. As shown in Fig. 4B, LGG slightly enhanced the DNA methylation in the untreated
B. breve and LGG significantly inhibited the LPS-stimulated cells and significantly restored LPS-reduced DNA methylation.
IL-23 mRNA levels in the HT-29/B6 and T84 cells. Further-
more, B. breve and LGG inhibited the IL-23 and IL-17 mRNA
B. breve and LGG inhibit LPS-induced NF-␬B
levels in the LPS-treated 3D PBMCs when cocultured with HT-
transcription activity
29/B6 or T84 cells (Fig. 4C).
As histone acetylation mediates the bacteria-, LPS-, TLR4-, and
B. breve and LGG decrease the global Ac-H4 and the cytokine-mediated induction of the NF-␬B-dependent tran-
p-Ac-H3 at Ser-10/Lys-14 and enhance DNA scription activity, causing the aberrant expression of inflamma-
methylation tory genes [50 –54], the activation of NF-␬B, through its classi-
cal pathways, was evaluated by determining the key signaling
To evaluate the levels of histones and their acetylated forms in
molecules that are generally used to mediate the activation of
the 3D coculture experiments, the T84 and HT-29/B6 mono-
the transcription factors NF-␬B, MAPK, and NF-␬B-dependent
layers were incubated initially with 2 ⫻ 107/ml B. breve, which
was added to the medium in the upper compartments (apical luciferase reporter gene activity. As shown in Fig. 6A and B,
surface) or to the medium in the lower compartments (baso- Western blot analysis and quantitative densitometry of the blot
lateral surface) in the presence of LPS for 30, 60, and 120 indicated that LPS alone induced the expression of MyD88,
min, and the cell lysates were subjected to Western blot analy- the degradation and disappearance of IRAK1 and I␬B␣, the
sis. As shown in Fig. 5A (T84 cells) and B (HT-29/B6 cells), expression of the nuclear NF-␬B p50/p60 subunits, and the
LPS alone induced the global Ac-H4 and the p-Ac-H3 at Ser- p-p38 MAPK and p-MEK1, which were correlated with in-
10/Lys-14 in a time-dependent manner. The maximum in- creased NF-␬B-dependent luciferase gene reporter activity (Fig.
crease in the Ac-H4 and the p-Ac-H3 occurred at 120 min. B. 6D). B. breve inhibited the LPS-induced expression of the nu-
breve inhibited the LPS-induced Ac-H4 and p-Ac-H3. Addition- clear NF-␬B p50/p65 subunits and MyD88, increased the level
ally, to evaluate these effects in more detail and to assess the of the IRAK1 and I␬B␣ proteins by decreasing LPS-induced
epigenetic imprinting effects of B. breve and LGG during long- IRAK1/I␬B␣ degradation, and abolished the LPS-induced
term incubation, we next incubated the HT-29/B6 and T84 p-p38 MAPK and p-MEK1 (Fig. 6A and B). Additionally, as B.
cells for 48 h with the histone deacetylase inducer GM, the breve and LGG were able to inhibit the hyper-Ac-H3/H4 in the
potent histone deacetylase inhibitor TSA, B. breve, or LGG in T84 cells in the 3D cocultures with PBMCs during long-term
the presence of LPS. Fig. 5C and D shows that GM inhibited (48-h) incubation of the cells with LPS, we also analyzed the
the global Ac-H4 and the p-Ac-H3 at Ser-10/Lys-14. TSA in- cell lysates following the 48-h incubation period by immuno-
duced the global Ac-H4 and the p-Ac-H3 at Ser-10/Lys-14. LPS blotting of the above signaling molecules in HT-29/B6 and
alone induced the global Ac-H4 and the p-Ac-H3 at Ser-10/ T84 cells. Fig. 6C shows that LGG inhibited the LPS-induced
Lys-14. B. breve and LGG (Fig. 5C and D, respectively) inhib- expression of MyD88 and the nuclear NF-␬B p50/p65 sub-
ited the LPS-induced global Ac-H4 and p-Ac-H3 at Ser-10/ units, the degradation of the I␬B␣ and IRAK-1 proteins, and
Lys-14 in the T84 cells incubated for 48 h. Further, to confirm the p-p38 MAPK and p-MEK1 in HT-29/B6 and T84 cells.
these results, the accumulation of acetylated histones and Consistent with the inhibition of the LPS-induced NF-␬B activ-
methylated DNA was also quantitatively assessed in the cell ly- ity, Fig. 6D shows that B. breve and LGG dose-dependently re-
sates. As shown in Fig. 5E, quantitative analysis of these cell pressed the LPS-induced NF-␬B-dependent luciferase activity in
lysates revealed that TSA and LPS enhanced the accumulation the HT-29/B6 and T84 cells, reaching a maximum attenuation
of Ac-H4, whereas GM decreased the accumulation of Ac-H4. at a dose of 10:1 bacteria/cell. The TLR4 antagonist abolished
B. breve significantly decreased the accumulation of Ac-H4 in the NF-␬B-dependent luciferase activity in the unstimulated
the untreated and LPS-treated HT-29/B6 and T84 cells, and LPS-stimulated cells. The TLR4 antagonist-mediated inhi-
whereas LGG significantly inhibited only the LPS-induced ac- bition of the basal NF-␬B-dependent luciferase activity may be

4
Figure 5. Effect of B. breve and LGG on histone modifications and DNA methylation in response to LPS in the 3D coculture models constructed
with polarized monolayers of Transwell filter-grown IECs in upper compartments and 3D PBMCs in lower compartments, placed on the bottom of
culture plates. T84 cells (A) or HT-29/B6 cells (B), at a density of 2 ⫻ 106 cells/ml, were treated initially for 30, 60, and 120 min with B. breve
added to the medium of the upper compartments (apical surface; bacteria:cell ratio of 10:1) in the presence of 100 ng/ml LPS, which was added
to the medium of the lower compartments (basolateral surface). After incubation, the cell lysates from the T84 and HT-29/B6 cells were prepared
and subjected to Western blotting to analyze the levels of global H4, Ac-H4, global H3, and p-Ac-H3. To evaluate these effects in more detail and
to assess the epigenetic imprinting effects of B. breve and LGG during long-term incubation, the HT-29/B6 and T84 cells were next incubated for
48 h with 0.2 mM GM (a histone acetylation inhibitor), 0.01 ng/ml TSA (a potent histone acetylation inducer), and B. breve or LGG (bacteria:cell
ratio of 10:1) in the presence of 100 ng/ml LPS added to the medium of the lower compartments. After the incubation, the prepared cell lysates
were subjected to Western blot analysis to assess the effects of B. breve (C) and LGG (D) on the Ac-H3 and the p-Ac-H3 in the LPS-stimulated cells.
The data are representative of three independent experiments. To confirm these results, the accumulation of Ac-H4 (E) and methylated DNA (F)
were also quantitatively assessed in the prepared cell lysates, as described in Materials and Methods. Each value represents the mean ⫾ sem of four
independent experiments performed in duplicate. *P ⬍ 0.05 versus control; §P ⬍ 0.05 versus LPS alone.

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 905


p50
A B

LPS+TLR4 antagonist
p65
5
MyD88

β-Actin normalised densitometric units


TLR4 antagonist
IRAK-1

LPS+ B. breve
IkBa
4 * Phospho-p38 MAPK
Phospho-MEK1
*

B. breve
*
Control
* * §
3

LPS
§
§
§ §
§
Phospho-MEK1 2 §
§§

Phospho-p38 MAPK §§ §
1 * §
* §
IkBα

IRAK1 0
e
ntro
l S nis
t
rev ist ve
LP on re
MyD88 Co tago B.b tag
B .b
an an
R4 S+
TL LR4 LP
T
p65 S+
LP
p50
D 4
HT-29/B6 cells
β-actin T84 cells

*
3
*
Luciferase activity

C
LPS+ LGG

( Relative value)
Control

2
LGG

§
LPS

§ §§
§
§
Phospho-MEK1 1
Phospho- p38 MAPK
IkBα **
0
l e 5 10 0 5 10
ev LGG
0
ro /ml) TLR4
---------------------------------
IRAK1 nt br antagonist B.breve LGG
B.
Co 0ng ---------------
HT-29/B6 (1 __________________
[ 10ng /ml ] Bacteria - to- cell ratios
MyD88 ist 0:
1) LPS
g on (1
ta t io [ 100 ng / ml ]
p65 an ra
R4 ell
TL o -c
t
p50 ia-
er
ct
Ba
β-actin
Figure 6. Effect of B. breve and LGG on the LPS-stimulated transcriptional activity of
Phospho-MEK1 NF-␬B. (A) T84 cells were incubated with medium alone (Control) or LPS in the ab-
Phospho-p38 MAPK sence or presence of B. breve at a bacteria:cell ratio of 10:1. The cell lysates obtained
from the 2-h incubation period were analyzed by immunoblotting for the protein ex-
IkBα pression of the nuclear NF-␬B p50/p65 subunits and MyD88, the degradation of I␬B␣
and IRAK-1, and the p-p38 MAPK and p-MEK1. ␤-Actin was included as a standard for
IRAK1 T84 protein loading. The data are representative of three independent experiments. (B)
Quantitative densitometric analysis of the Western blot is presented, with the ␤-actin-
MyD88 normalized densitometric units for each protein plotted against the treatments. The
data are presented as the means ⫾ sem (indicated with error bars) from three inde-
p65
pendent experiments. *P ⬍ 0.05 versus control; §P ⬍ 0.05 versus LPS alone. (C) HT-
p50 29/B6 cells or T84 cells were treated with 100 ng/ml LPS, which was added to the
medium of the upper compartments in the absence or presence of LGG (bacteria:cell
β-actin
ratio of 10:1). The cell lysates from the 48-h incubation period were analyzed by West-
ern blotting for the protein expression of the nuclear NF-␬B p50/p65 subunits and MyD88, the degradation levels of I␬B␣ and IRAK-1, and the p-p38 MAPK
and p-MEK1. ␤-Actin was included as a standard for protein loading. The data are representative of three independent experiments. (D) The effect of B. breve
or LGG on the NF-␬B-dependent promoter luciferase reporter gene activity in the HT-29/B6 and T84 cells, and HT-29/B6 or T84 cells were transiently trans-
fected with the Ad5␬B-LUC vector. The cells were then incubated with various concentrations of B. breve or LGG (bacteria:cell ratios of 0, 5, and 10), which
were added to the medium of the upper compartments in the presence or absence of 100 ng/ml LPS or 10 ng/ml TLR4 antagonist (added to the medium
of the lower compartments). In the nuclear extracts, the NF-␬B-dependent luciferase activity was measured after 24 h using a Tecan GENios microplate
reader, and the results were normalized to the extract protein concentrations. *P ⬍ 0.05 versus control; §P ⬍ 0.05 versus LPS-stimulated cells. The data
(means⫾sem of triplicate determinations) are representative results of three independent experiments.

906 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

a result of the fact that the culture medium is not absolutely a dose-dependent manner [49]. However, we believe that the
LPS-free. optimum immunomodulating effect by the bacteria at the 10:1
ratio is a result of the average number of PAMP receptors,
Cell viability and proliferation assays such as TLRs and nucleotide binding oligomerization do-
Under the conditions studied, we observed that none of the mains, on the cells [58]. High doses of bacteria may lead to
agents decreased the viability of the HT-29/B6 and T84 mono- the competitive use of the available receptors and false recep-
layers or the 3D PBMCs at the concentrations tested. To ex- tor binding. Therefore, bacteria at high doses failed (in our
clude the possibility that the decreased expression of the IL- hands) to inhibit these cytokines.
23/IL-17 and the decreased activity of the NF-␬B-dependent It has been reported that histone modifications may under-
luciferase reporter gene were a consequence of cell death as a lie the chronic inflammation of UC and CD and that higher
result of exposure to LPS, the TLR4 antagonist, or the tested histone acetylation levels predispose cells to IBD, likely
bacteria, the viability of the cells was assessed routinely, as through the induction of proinflammatory cytokines [18 –20].
stated previously. Microscopic examination of the cells incu- Histone acetylation mediates the bacteria-, LPS-, and cytokine-
bated with bacteria and challenged with agents revealed that induced NF-␬B transcription activity that causes the expression
the viability was ⬎93% in all experiments, demonstrating that of inflammatory genes and is correlated with the NF-␬B tran-
the inhibition of IL-23, IL-17, CD40, and the NF-␬B-dependent scription activity for other genes, including those involved in
luciferase reporter gene by the bacteria and the TLR4 antago- the differentiation, proliferation, and activation of cells [51,
nist is not a result of cell death (Fig. 7A). Our observations are 52, 54]. LPS exposure leads to the phosphorylation/acetyla-
supported by reports from other groups indicating that LPS, at tion of histones at the promoters of proinflammatory genes in
the concentration used in this study, had no cytotoxic effects lung cells [60], macrophages, lymphocytes, and monocytic
on the viability of cell monolayers [55] or PBMCs from cells [61]. Interestingly, in murine macrophages and experi-
healthy and IBD patients [9, 38, 56], even after culturing for mental colitis, LPS strongly stimulates IL-23 expression
48 h. through histone acetylation and NF-␬B activation [52]. LPS is
Notably, LPS alone did not decrease, but rather increased, also known to stimulate the in vivo phosphorylation/acetyla-
the cell viability of the HT-29/B6 and T84 monolayers, and tion of histones and the recruitment of NF-␬B to the CD40
the TLR4 antagonist abolished this effect (Fig. 7A). To deter- promoter, all of which are important events for CD40 gene
mine whether this pattern of LPS-induced cell viability is asso- transcription, indicating that histone acetylation and NF-␬B
ciated with cell proliferation, we also assessed the proliferation activation play critical roles in the transcriptional activation of
of the HT-29/B6 and T84 cells. Fig. 7B shows that the LPS- the CD40 gene by LPS in immune cells [62]. The LPS-stimu-
induced increase in cell viability was related to proliferation lated IL-17 expression in human PBMCs [63] and the in vivo
and that the TLR4 antagonist abolished the LPS-stimulated association among histone acetylation, NF-␬B-induced expres-
proliferation. This corresponds with previous studies showing sion of inflammatory genes, and IBD in humans have been
that LPS induces the proliferation of IEC lines through TLR4- reported recently [24]. In this regard, we first aimed to investi-
mediated signaling, which aids in proliferation and protection gate whether the B. breve- and LGG-mediated inhibition of IL-
against apoptosis and is therefore important for intestinal epi- 23/IL-17 expression is also associated with inhibition of his-
thelial repair from injury [57–59]. tone acetylation in response to LPS. Our experiments revealed
that B. breve and LGG inhibited LPS-induced histone acetyla-
tion in the IECs. LPS-mediated TLR4 signaling occurs via a
cascade of events, including the stimulation of MyD88 and
DISCUSSION
IRAK-1, which in turn simultaneously activate the IKK com-
In this study, we used an in vitro 3D coculture model com- plex. These events then lead to the degradation of I␬B␣, the
posed of IECs and PBMCs to explore the molecular mecha- p-NF-␬B p65/p50 subunits, and NF-␬B and MAPK activity, re-
nism by which B. breve and LGG modulate the IL-23/IL-17 axis, sulting in p-MEK1 (the upstream kinase of ERK1) and p-p38
which is involved in IBD. Our experiments demonstrated that MAPK. This ultimately results in aberrant inflammatory re-
B. breve and LGG inhibited the LPS-stimulated mRNA and pro- sponses by inducing proinflammatory cytokines in vitro and in
tein expression of the IL-23/IL-17 axis and CD40 in the 3D vivo [50, 53]. Therefore, we next evaluated the impact of B.
coculture models constructed with HT-29/B6 or T84 cells and breve and LGG on the LPS/TLR4-mediated intracellular signal-
PBMCs. Together with previous studies [8, 12], the above re- ing cascades, including signaling molecules that are generally
sults suggest that B. breve and LGG may exert their anti-inflam- involved in mediating the activation of the NF-␬B and AP-1
matory effects on the epithelium by reducing the expression transcription factors, as well as the MAPKs, and assessed their
of the IL-23/IL-17 axis as a hallmark of IBD. impact on NF-␬B-dependent luciferase reporter gene activity in
Under the conditions studied, we observed that the effective the IECs stimulated with LPS. Our findings were consistent
inhibition of basal IL-17 and IL-23, as well as maximum inhibi- with the in vitro and in vivo observations that probiotic bacte-
tion of LPS-induced IL-23 and IL-17 by B. breve, only occurred ria inhibit LPS/TLR4/NF-␬B-mediated proinflammatory cyto-
at a bacteria:HT-29/B6 or T84 cell ratio of 10:1. Such a ratio kine expression in colonic epithelial cells and in PBMCs that
was used by Foligne et al. [46] for investigating the cytokine are involved in colitis [32, 36, 48, 49, 64 – 67]. Furthermore, B.
profiles of DCs treated with probiotic bacteria. It is also known breve and LGG inhibited the LPS-induced degradation of
that bifidobacteria can regulate cytokine production in IECs in IRAK1 and I␬B␣, the p-p38 MAPK/p-MEK1, and the expres-

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 907


A B 6
Proliferation HT-29/B6 cells
T84 cells

180 5
*

Proliferation (Stimulation Index)


160 *
HT-29/B6 cells *
T84 cells
PBMCs 4
140 *

120
Cell viability (%)

§
§ 3
100 §
§
§
§
80 §
2
60

40 1
§
§
20
* *
0 0
l S st e e t e e ol t t
ev
e e e e
ev
ro S
nt LP ni ev ev is ntr LP nis ev ev nis ev ev
Co go br br on br . br Co go .br .br go .br B.
br
B. . .
g t a B B t a B
B B +
ta ta +B an led a n S e d
an ed an d R4 kil 4 LP ill
4 ill 4 PS lle TL at- LR t-k
R - k R L -k
i
He +T Hea
TL at TL at LP
S
S+
He P S+ he LP
L
S+
LP

Figure 7. Effect of B. breve on cell viability and proliferation in the coculture models constructed with HT-29/B6 or T84 IECs in the upper com-
partment and 3D PBMCs in the lower compartment, placed on the bottom of culture plates. The apical surface of the HT-29/B6 or T84 cells was
incubated with B. breve and heat-killed B. breve (bacteria:cell ratio of 10:1) in the presence or absence of basolaterally applied LPS (100 ng/ml) or
TLR4 antagonist (10 ng/ml) for 48 h. After incubation, the cell-viability percentages of the HT-29/B6 and T84 cells as well as PBMCs (A) and the
proliferation of the HT-29/B6 and T84 cells (B) were assessed by WST-1 assays and the measurement of DNA synthesis, as described in Materials
and Methods. With respect to proliferation, a significant response was observed only when the T84 cells were stimulated with LPS (P⬍0.05). The
TLR4 antagonist, B. breve, and heat-killed B. breve abolished the LPS-induced proliferation (P⬍0.05). Note the absence of a T84 proliferation re-
sponse when the T84 cells were treated with B. breve or heat-killed B. breve. The results are shown as the mean ⫾ sem of four separate
experiments.*P ⬍ 0.05 versus control; §P ⬍ 0.05 versus LPS alone.

sion of the nuclear NF-␬B p50/p65 subunits and MyD88. Ac- quently leading to the inhibition of LPS-induced NF-␬B activa-
cordingly, the B. breve- and LGG-mediated inhibition of the tion and promoter activity [36, 49]. From these results, we can-
LPS-induced histone acetylation was associated with decreased not rule out the possibility that the activation of the IL-23/
expression of the NF-␬B-dependent luciferase reporter gene, IL-17 axis may also be a result of an increase in viability (or
which is reflected by the decreased, LPS-stimulated production proliferation) of cells upon LPS challenge or that the inhibi-
of IL-17, IL-23, and CD40, as well as proliferation of HT-29/B6 tion of the IL-23/IL-17 axis by B. breve involves the attenuation
and T84 cells. These data provide evidence that B. breve and of the LPS-induced proliferation of cells.
LGG repress conserved inflammatory pathways via the inhibi- As there is a reciprocal interplay between histone acetylation
tion of NF-␬B activity and by specifically inhibiting the access and DNA methylation in the regulation of proinflammatory
of NF-␬B to the promoters of cytokine genes, thereby alleviat- cytokines and as LPS induces the proinflammatory IL-1 and
ing inflammatory responses. In agreement with our results, TNF-␣ cytokines by the molecular inhibition of DNA methyl-
others [48, 49] have shown that under inflammatory condi- ation, while enhancing histone acetylation [5], we further in-
tions, the inflammation-alleviating effects of probiotic bacteria vestigated the effects of B. breve and LGG on DNA methylation
are greater than those observed in basal conditions and are in IECs. These bacteria enhanced DNA methylation slightly in
specific for LPS-induced NF-␬B activation in IECs. untreated cells and abolished the LPS-reduced DNA methyl-
As LPS stimulates the proliferation of human IECs [59, 68, ation. The inhibition of the LPS-mediated reduction of DNA
69] through TLR4, which is important for intestinal epithelial methylation by B. breve and LGG may be explained by B. breve/
repair from injury by aiding in proliferation and protecting LGG-mediated folate production [70], which enhances DNA
against apoptosis [68], we also evaluated cell proliferation. In- methylation [71].
deed, the B. breve-mediated inhibition of LPS-induced prolifer- Based on our findings, B. breve and LGG alleviate the patho-
ation of T84 cells, in the absence of cytotoxic effects, corre- genic factors for IBD by lowering LPS-induced NF-␬B activity
lated well with the B. breve-mediated inhibition of LPS-induced and by decreasing LPS-induced histone modifications. The ca-
histone acetylation and NF-␬B-dependent luciferase reporter pacity of these probiotics to attenuate the expression of IL-23,
gene activity. This effect is presumably a result of the inhibi- IL-17, and CD40, which are causes and risk factors of IBD, at
tion of LPS-mediated histone modifications [60 – 62], subse- the epigenetic level may have two explanations. First, B. breve

908 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

and LGG repressed LPS-induced histone acetylation and NF-␬B Considering the functional similarities between the in vivo
activity. This may occur as a result of the close and positive situation and the in vitro model used, our experiment may be
interplay/correlation between histone acetylation/phosphory- regarded as somewhat artificial. For example, the model does
lation status and NF-␬B transcriptional activity. Indeed, NF-␬B not include the intraepithelial lymphocyte component, the
binding to transcriptionally active chromatin is influenced by sensing by the luminal processes of the DCs, or the trafficking
the status of histone acetylation/phosphorylation, as well as of the bacterial components by the M cells. Therefore, extrap-
the signaling cascades that impact the levels of histone acetyla- olations to in vivo effects must be considered with caution.
tion and thereby control chromatin remodeling and gene ex- However, even more simple in vitro models have been shown
pression [22, 54]. Second, we observed that B. breve and LGG to predict in vivo results [75]. Regardless, these results support
inhibited the LPS-induced degradation of IRAK1 and I␬B␣ the suggestion that therapeutic interventions aimed at sup-
and interestingly, the p-p38 MAPK and p-MEK1, which are key pressing the IL-23/IL-17 axis may be beneficial in IBD [3, 5,
direct and indirect upstream activators of the NF-␬B pathway, 76] and complement the in vitro and in vivo observations of
and subsequent cytokine gene expression in vitro and in vivo the anti-inflammatory effects of the LAB, as B. breve and LGG
[50, 53, 57]. Principally, the p38 MAPK and MEK1 inhibitors attenuated the IL-23/IL-17 axis in the LPS-stimulated IECs.
block the phosphorylation/acetylation of histones and proin- The reduction in histone modifications coinciding with the
flammatory cytokines, whereas LPS stimulates the cytoplasmic- induction of DNA methylation suggests that this anti-inflamma-
to-nuclear translocation of NF-␬B by inducing the degradation tory effect is, to some extent, mediated by epigenetic pro-
of I␬B␣ and p-p38 MAPK and p-MEK1. These activities con- cesses.
tribute to the LPS-induced NF-␬B activation that is correlated In conclusion, using a 3D coculture system composed of
with the p-Ac-H3/H4 and the subsequent production of proin- IECs and PBMCs as an in vitro model of the intestinal mucosal
flammatory cytokines in pathogen bacteria-infected endothelial immune system, we have described a novel regulatory mecha-
cells [19, 22]. Interestingly, our time-course experiment re- nism by which commensal probiotics inhibit the NF-␬B-medi-
vealed that the B. breve-mediated inhibition of IL-17 and IL-23 ated transcriptional activation of inflammatory genes (IL-23,
is not restricted to the 48-h incubation period but was main- IL-17, and CD40), thereby reducing histone acetylation and
tained even after 96 h of incubation. This reflects the B. breve/ simultaneously enhancing DNA methylation in LPS-treated
LGG-mediated inhibition of histone acetylation and slight in- IECs.
duction of DNA methylation, as the nature of epigenetic regu-
lation is essentially a long-lasting modulation of gene
regulation. Based on these data, reasonable molecular mecha- REFERENCES
nisms underlying the inhibitory effects of B.breve and LGG on 1. Caruso, R., Pallone, F., Monteleone, G. (2007) Emerging role of IL-23/
LPS-induced histone acetylation are the inhibition of the nu- IL-17 axis in H. pylori-associated. pathology. World J. Gastroenterol. 13,
5547–5551.
clear translocation of NF-␬B, achieved by decreasing the ex- 2. Hölttä, V., Klemetti, P., Sipponen, T., Westerholm-Ormio, M., Kociubin-
pression of MyD88, the degradation of IRAK-1 and I␬B␣, and ski, G., Salo, H., Räsänen, L., Kolho, K. L., Färkkilä, M., Savilahti, E.,
Vaarala, O. (2008) IL-23/IL-17 immunity as a hallmark of Crohn’s dis-
particularly, the expression of the nuclear NF-␬B p50/p65 sub- ease. Inflamm. Bowel Dis. 14, 1175–1184.
units, as well as a decrease in p-p38 MAPK and p-MEK1, all of 3. Kobayashi, T., Okamoto, S., Hisamatsu, T., Kamada, N., Chinen, H.,
Saito, R., Kitazume, M. T., Nakazawa, A., Sugita, A., Koganei, K., Isobe,
which are important and ultimate mediators for LPS-induced K., Hibi, T. (2008) IL-23 differentially regulates the Th1/Th17 balance
histone acetylation [18 –20, 52, 62, 72]. Indeed, in LPS-stimu- in ulcerative colitis and Crohn’s disease. Gut 57, 1682–1689.
4. Maloy, K. J. (2008) The interleukin-23/interleukin-17 axis in intestinal
lated cells, transcription factors, such as NF-␬B, are activated inflammation. J. Intern. Med. 263, 584 –590.
and translocate to the nucleus, bind DNA, and subsequently 5. Sarra, M., Pallone, F., Macdonald, T. T., Monteleone, G. (2010) IL-23/
recruit coactivator molecules, such as CBP, p300, general con- IL-17 axis in IBD. Inflamm. Bowel. Dis. 16, 1808 –1813.
6. Tan, Z. Y., Bealgey, K. W., Fang, Y., Gong, Y. M., Bao, S. (2009) Inter-
trol nonderepressible 5, and p300/CBP-associated factor. leukin-23: immunological roles and clinical implications. Int. J. Biochem.
These coactivator molecules are part of multiprotein com- Cell Biol. 41, 733–735.
7. Borcherding, F., Nitschke, M., Hundorfeam, G., Rupp, J., von Smolinski,
plexes that are recruited by DNA-bound transcription factors D., Bieber, K., van Kooten, C., Lehnert, H., Fellermann, K., Büning, J.
to the transcription start site and act as molecular switches. (2010) The CD40-CD40L pathway contributes to the proinflammatory
function of intestinal epithelial cells in inflammatory bowel disease. Am.
When transcription factors bind to these coactivator mole- J. Pathol. 176, 1816 –1827.
cules, intrinsic histone acetyltransferase activity is triggered on 8. Paolillo, R., Romano, Carratelli, C., Sorrentino, S., Mazzola, N., Rizzo, A.
(2009) Immunomodulatory effects of Lactobacillus plantarum on human
these proteins, resulting in acetylation on core histone pro- colon cancer cells. Int. Immunopharmacol. 9, 1265–1267.
teins [24]. 9. Dobreva, Z. G., Stanilova, S. A. (2007) The immunomodulatory activity
of C3 binding glycoprotein (C3bgp) is mediated by the complement
Although we did not include pathogenic bacteria in our ex- receptor type III and mitogen-activated protein kinase signal transduc-
periments, it seems plausible that the observed effects of B. tion pathways. Immunopharmacol. Immunotoxicol. 29, 549 –562.
10. Jyonouchi, H., Geng, L., Cushing-Ruby, A., Monteiro, I. M. (2010) Aber-
breve and LGG are specific because pathogenic bacteria (e.g., rant responses to TLR agonists in pediatric IBD patients; the possible
Helicobater pylori) have been shown to aberrantly activate the association with increased production of Th1/Th17 cytokines in re-
sponse to candida, a luminal antigen. Pediatr. Allergy Immunol. 21, e747–
IL-23/IL-17 axis in H. pylori-related gastritis [1] and induce the e755.
p-Ac-H3/H4 and proinflammatory cytokines (e.g., IL-8) in en- 11. Li, J., Li, L., Shang, X., Benson, J., Merle Elloso, M., Schantz, A., Bracht,
M., Orlovsky, Y., Sweet, R. (2008) Negative regulation of IL-17 produc-
dothelial cells [18 –20, 73]. Moreover, the molecular induction tion by OX40/OX40L interaction. Cell. Immunol. 253, 31–37.
of histone acetylation and the inhibition of DNA methylation 12. Tanabe, S., Kinuta, Y., Saito, Y. (2008) Bifidobacterium infantis suppresses
proinflammatory interleukin-17 production in murine splenocytes and
lead to a massive increase in the LPS-stimulated IL-1␤ expres- dextran sodium sulfate-induced intestinal inflammation. Int. J. Mol. Med.
sion and an increase in the basal production of TNF-␣ [74]. 22, 181–185.

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 909


13. Iwakura, Y., Ishigame, H., Saijo, S., Nakae, S. (2011) Functional special- molysin induces active chloride secretion in colon epithelial cells (HT-
ization of interleukin-17 family members. Immunity 34, 149 –162. 29/B6). Infect. Immun. 72, 4848 –4858.
14. De Palma, G., Cinova, J., Stepankova, R., Tuckova, L., Sanz, Y. (2010) 36. Ghadimi, D., de Vrese, M., Heller, K. J., Schrezenmeir, J. (2010) Effect
Pivotal Advance: Bifidobacteria and Gram-negative bacteria differentially of natural commensal-origin DNA on toll-like receptor 9 (TLR9) signal-
influence immune responses in the proinflammatory milieu of celiac ing cascade, chemokine IL-8 expression, and barrier integrity of polar-
disease. J. Leukoc. Biol. 87, 765–778. ized intestinal epithelial cells. Inflamm. Bowel Dis. 16, 410 –427.
15. Wu, W., Alexis, N. E., Bromberg, P. A., Jaspers, I., Peden, D. B. (2009) 37. Vamadevan, A. S., Fukata, M., Arnold, E. T., Thomas, L. S., Hsu, D.,
Mechanisms of LPS-induced CD40 expression in human peripheral Abreu, M. T. (2010) Regulation of Toll-like receptor 4-associated MD-2
blood monocytic cells. Biochem. Biophys. Res. Commun. 379, 573–577. in intestinal epithelial cells: a comprehensive analysis. Innate Immun. 16,
16. Wu, Y., Wang, L., He, X., Xu, H., Zhou, L., Zhao, F., Zhang, Y. (2008) 93–103.
Expression of CD40 and growth-inhibitory activity of CD40 ligand in 38. Von Haefen, C., Mei, W., Menk, M., Klemz, R., Jones, A., Wernecke,
colon cancer ex vivo. Cell. Immunol. 253, 102–109. K. D., Spies, C. D. (2010) Ethanol changes gene expression of transcrip-
17. Gonsky, R., Deem, R. L., Landers, C. J., Derkowski, C. A., Berel, D., tion factors and cytokine production of CD4⫹ T cell subsets in PBMCs
McGovern, D. P., Targan, S. R. (2011) Distinct IFNG methylation in a stimulated with LPS. Alcohol Clin. Exp. Res. 35, 621–631.
subset of ulcerative colitis patients based on reactivity to microbial anti- 39. Lenarczyk, A., Helsloot, J., Farmer, K., Peters, L., Sturgess, A., Kirkham,
gens. Inflamm Bowel Dis. 17, 171–178. B. (2000) Antigen-induced IL-17 response in the peripheral blood
18. Angrisano, T., Pero, R., Peluso, S., Keller, S., Sacchetti, S., Bruni, C. B., mononuclear cells (PBMC) of healthy controls. Clin. Exp. Immunol. 122,
Chiariotti, L., Lembo, F. (2010) LPS-induced IL-8 activation in human 41–48.
intestinal epithelial cells is accompanied by specific histone H3 acetyla- 40. Dje N’Guessan, P., Riediger, F., Vardarova, K., Scharf, S., Eitel, J., Opitz,
tion and methylation changes. BMC Microbiol. 10, 172. B., Slevogt, H., Weichert, W., Hocke, A.C., Schmeck, B., Suttorp, N.,
19. Schmeck, B., Beermann, W., van Laak, V., Zahlten, J., Opitz, B., Witzen- Hippenstiel, S. (2009) Statins control oxidized LDL-mediated histone
rath, M., Hocke, A. C., Chakraborty, T., Kracht, M., Rosseau, S., Suttorp, modifications and gene expression in cultured human endothelial cells.
N., Hippenstiel, S. (2005) Intracellular bacteria differentially regulated Arterioscler. Thromb. Vasc. Biol. 29, 380 –386.
endothelial cytokine release by MAPK-dependent histone modification. 41. Chen, F. Q., Schacht, J., Sha, S. H. (2009) Aminoglycoside-induced his-
J. Immunol. 175, 2843–2850. tone deacetylation and hair cell death in the mouse cochlea. J. Neuro-
20. Schmeck, B., Lorenz, J., N=guessan, P. D., Opitz, B., van Laak, V., Zahl- chem. 108, 1226 –1236.
ten, J., Slevogt, H., Witzenrath, M., Flieger, A., Suttorp, N., Hippenstiel, 42. Michils, A., Dutry, D., de Beyl, V. Z., Remmelink, M., de Maertelaer, V.,
S. (2008) Histone acetylation and flagellin are essential for Legionella Rocmans, P. (2002) Peripheral blood mononuclear cell proliferation to
pneumophila-induced cytokine expression. J. Immunol. 181, 940 –947. heat shock protein-70 derived from autologous lung carcinoma. Am. J.
21. Slevogt, H., Schmeck, B., Jonatat, C., Zahlten, J., Beermann, W., van Respir. Crit. Care Med. 166, 749 –753.
Laak, V., Opitz, B., Dietel, S., N=Guessan, P. D., Hippenstiel, S., Suttorp, 43. Kimura, H., Sawada, N., Tobioka, H., Isomura, H., Kokai, Y., Hirata, K.,
N., Seybold, J. (2006) Moraxella catarrhalis induces inflammatory re- Mori, M. (1997) Bacterial lipopolysaccharide reduced intestinal barrier
sponse of bronchial epithelial cells via MAPK and NF-␬B activation and function and altered localization of 7H6 antigen in IEC-6 rat intestinal
histone deacetylase activity reduction. Am. J. Physiol. Lung Cell. Mol. crypt cells. J. Cell. Physiol. 171, 284 –290.
Physiol. 290, L818 –L826. 44. Abreu, M. T., Thomas, L. S., Arnold, E. T., Lukasek, K., Michelsen,
22. Haller, D., Holt, L., Kim, S. C., Schwabe, R. F., Sartor, R. B., Jobin, C. K. S., Arditi, M. (2003) TLR signaling at the intestinal epithelial inter-
(2003) Transforming growth factor-␤ 1 inhibits non-pathogenic Gram face. J. Endotoxin Res. 9, 322–330.
negative bacteria-induced NF-␬ B recruitment to the interleukin-6 gene 45. Beatty, W. L., Méresse, S., Gounon, P., Davoust, J., Mounier, J., Sanson-
promoter in intestinal epithelial cells through modulation of histone etti, P. J., Gorvel, J. P. (1999) Trafficking of Shigella lipopolysaccharide
acetylation. J. Biol. Chem. 278, 23851–23860. in polarized intestinal epithelial cells. J. Cell Biol. 145, 689 –698.
23. Hamon, M. A., Cossart, P. (2008) Histone modifications and chromatin 46. Foligne, B., Zoumpopoulou, G., Dewulf, J., Ben Younes, A., Chareyre, F.,
remodeling during bacterial infections. Cell Host Microbe 4, 100 –109. Sirard, J. C., Pot, B., Grangette, C. (2007) A key role of dendritic cells
24. Tsaprouni, L. G., Ito, K., Powell, J. J., Adcock, I. M., Punchard, N. in probiotic functionality. PLoS One 2, e313.
(2011) Differential patterns of histone acetylation in inflammatory 47. Cario, E., Rosenberg, I. M., Brandwein, S. L., Beck, P. L., Reinecker,
bowel diseases. J. Inflamm. (Lond) 8, 1. H. C., Podolsky, D. K. (2000) Lipopolysaccharide activates distinct sig-
25. Lai, Y., Asthana, A., Cheng, K., Kisaalita, W. S. (2011) Neural cell 3D naling pathways in intestinal epithelial cell lines expressing Toll-like re-
microtissue formation is marked by cytokines’ up-regulation. PLoS One ceptors. J. Immunol. 164, 966 –972.
6, e26821. 48. Heyman, M., Terpend, K., Menard, S. (2005) Effects of specific lactic
26. Seo, Y. K., Yoon, H. H., Park, Y. S., Song, K. Y., Lee, W. S., Park, J. K. acid bacteria on the intestinal permeability to macromolecules and the
(2009) Correlation between scaffold in vivo biocompatibility and in vitro inflammatory condition. Acta Paediatr. Suppl. 94, 34 –36.
cell compatibility using mesenchymal and mononuclear cell cultures. 49. Riedel, C. U., Foata, F., Philippe, D., Adolfsson, O., Eikmanns, B. J.,
Cell. Biol. Toxicol. 25, 513–522. Blum, S. (2006) Anti-inflammatory effects of bifidobacteria by inhibition
27. Liu, H., Lin, J., Roy, K. (2006) Effect of 3D scaffold and dynamic cul- of LPS-induced NF-␬B activation. World J. Gastroenterol. 12, 3729 –3735.
ture condition on the global gene expression profile of mouse embry- 50. Chandrakesan, P., Ahmed, I., Anwar, T., Wang, Y., Sarkar, S., Singh, P.,
onic stem cells. Biomaterials 27, 5978 –5989. Peleg, S., Umar, S. (2010) Novel changes in NF-␬B activity during pro-
28. Liu, J., Abate, W., Xu, J., Corry, D., Kaul, B., Jackson, S.K. (2011) Three- gression and regression phases of hyperplasia: role of MEK, ERK, and
dimensional spheroid cultures of A549 and HepG2 cells exhibit differ- p38. J. Biol. Chem. 285, 33485–33498.
ent lipopolysaccharide (LPS) receptor expression and LPS-induced cyto- 51. Clarke, D. L., Sutcliffe, A., Deacon, K., Bradbury, D., Corbett, L., Knox,
kine response compared with monolayer cultures. Innate Immun. 17, A. J. (2008) PKC␤II augments NF-␬B-dependent transcription at the
245–255. CCL11 promoter via p300/CBP-associated factor recruitment and his-
29. Kim, J. B., Stein, R., O’Hare, M. J. (2004) Three-dimensional in vitro tone H4 acetylation. J. Immunol. 181, 3503–3514.
tissue culture models of breast cancer. Breast Cancer Res. Treat. 85, 281– 52. Sheikh, S. Z., Matsuoka, K., Kobayashi, T., Li, F., Rubinas, T., Plevy,
291. S. E. (2010) IFN-␥ is a negative regulator of IL-23 in murine macro-
30. Seitzer, U., Gerdes, J. (2003) Generation and characterization of multi- phages and experimental colitis. J. Immunol. 184, 4069 –4073.
cellular heterospheroids formed by human peripheral blood mononu- 53. Watkins, L. R., Hutchinson, M. R., Rice, K. C., Maier, S. F. (2009) The
clear cells. Cells Tissues Organs 174, 110 –116. “toll” of opioid-induced glial activation: improving the clinical efficacy
31. Ghadimi, D., de Vrese, M., Heller, K. J., Schrezenmeir, J. (2010) Lactic of opioids by targeting glia. Trends Pharmacol. Sci. 30, 581–591.
acid bacteria enhance autophagic ability of mononuclear phagocytes by 54. Yu, Z., Kone, B. C. (2006) Targeted histone H4 acetylation via phospho-
increasing Th1 autophagy-promoting cytokine (IFN-␥) and nitric oxide inositide 3-kinase- and p70s6-kinase-dependent pathways inhibits iNOS
(NO) levels and reducing Th2 autophagy-restraining cytokines (IL-4 and induction in mesangial cells. Am. J. Physiol. Renal Physiol. 290, F496 –
IL-13) in response to Mycobacterium tuberculosis antigen. Int. Immunophar- F502.
macol. 10, 694 –706. 55. Vinderola, G., Matar, C., Perdigon, G. (2005) Role of intestinal epithe-
32. Menard, S., Candalh, C., Bambou, J. C., Terpend, K., Cerf-Bensussan, lial cells in immune effects mediated by gram-positive probiotic bacteria:
N., Heyman, M. (2004) Lactic acid bacteria secrete metabolites retain- involvement of Toll-like receptors. Clin. Diagn. Lab. Immunol. 12, 1075–
ing anti-inflammatory properties after intestinal transport. Gut 53, 821– 1084.
828. 56. Martinesi, M., Treves, C., d’Albasio, G., Bagnoli, S., Bonanomi, A. G.,
33. Heuvelin, E., Lebreton, C., Grangette, C., Pot, B., Cerf-Bensussan, N., Stio, M. (2008) Vitamin D derivatives induce apoptosis and downregu-
Heyman, M. (2009) Mechanisms involved in alleviation of intestinal in- late ICAM-1 levels in peripheral blood mononuclear cells of inflamma-
flammation by bifidobacterium breve soluble factors. PLoS One 4, e5184. tory bowel disease patients. Inflamm. Bowel Dis. 14, 597–604.
34. Heuvelin, E., Lebreton, C., Bichara, M., Cerf-Bensussan, N., Heyman, M. 57. Courtois, F., Seidman, E. G., Delvin, E., Asselin, C., Bernotti, S., Ledoux,
(2010) A bifidobacterium probiotic strain and its soluble factors allevi- M., Levy, E. (2003) Membrane peroxidation by lipopolysaccharide and
ate chloride secretion by human intestinal epithelial cells. J. Nutr. 140, iron-ascorbate adversely affects Caco-2 cell function: beneficial role of
7–11. butyric acid. Am. J. Clin. Nur. 77, 744 –750.
35. Epple, H. J., Mankertz, J., Ignatius, R., Liesenfeld, O., Fromm, M., Zeitz, 58. Ghadimi, D., Fölster-Holst, R., de Vrese, M., Winkler, P., Heller, K. J.,
M., Chakraborty, T., Schulzke, J. D. (2004) Aeromonas hydrophila ␤-he- Schrezenmeir, J. (2008) Effects of probiotic bacteria and their genomic

910 Journal of Leukocyte Biology Volume 92, October 2012 www.jleukbio.org


Ghadimi et al. Epigenetic regulation of the IBD-causing IL-23/IL-17 axis by commensal probiotics

DNA on TH1/TH2-cytokine production by peripheral blood mononu- 68. Fukata, M., Chen, A., Klepper, A., Krishnareddy, S., Vamadevan, A. S.,
clear cells (PBMCs) of healthy and allergic subjects. Immunobiology 213, Thomas, L. S., Xu, R., Inoue, H., Arditi, M., Dannenberg, A. J., Abreu,
677–692. M. T. (2006) Cox-2 is regulated by Toll-like receptor-4 (TLR4) signal-
59. Ruemmele, F. M., Beaulieu, J. F., Dionne, S., Levy, E., Seidman, E. G., ing: role in proliferation and apoptosis in the intestine. Gastroenterology
Cerf-Bensussan, N., Lentze, M. J. (2002) Lipopolysaccharide modulation 131, 862–877.
of normal enterocyte turnover by Toll-like receptors is mediated by en- 69. Grimoud, J., Durand, H., de Souza, S., Monsan, P., Ouarné, F., The-
dogenously produced tumour necrosis factor ␣. Gut 51, 842–848. odorou, V., Roques, C. (2010) In vitro screening of probiotics and synbi-
60. Yao, H., Hwang, J. W., Moscat, J., Diaz-Meco, Mt., Leitges, M., Kishore, otics according to anti-inflammatory and anti-proliferative effects. Int. J.
N., Li, X., Rahman, I. (2010) Protein kinase C␨ mediates cigarette Food Microbiol. 144, 42–50.
smoke/aldehyde- and lipopolysaccharide-induced lung inflammation 70. Pompei, A., Cordisco, L., Amaretti, A., Zanoni, S., Matteuzzi, D., Rossi,
and histone modifications. J. Biol. Chem. 285, 5405–5416. M. (2007) Folate production by bifidobacteria as a potential probiotic
61. Tsaprouni, L. G., Ito, K., Adcock, I. M., Punchard, N. (2007) Suppres- property. Appl. Environ. Microbiol. 73, 179 –185.
sion of lipopolysaccharide- and tumour necrosis factor-␣-induced inter- 71. Pompei, A., Cordisco, L., Amaretti, A., Zanoni, S., Raimondi, S., Mat-
leukin (IL)-8 expressions by glucocorticoids involves changes in IL-8 teuzzi, D., Rossi, M. (2007) Administration of folate-producing bifido-
promoter acetylation. Clin. Exp. Immunol. 150, 151–157. bacteria enhances folate status in Wistar rats. J. Nutr. 137, 2742–2746.
62. Qin, H., Wilson, C. A., Lee, S. J., Zhao, X., Benyeniste, E. N. (2005) LPS 72. Xiao, W. (2004) Advances in NF-␬B signaling transduction and tran-
induces CD40 gene expression through the activation of NF-␬B and scription. Cell. Mol. Immunol. 1, 425–435.
STAT-1␣ in macrophages and microglia. Blood 106, 3114 –3122. 73. Ding, S. Z., Fischer, W., Kaparakis-Liaskos, M., Liechti, G., Merrell, D. S.,
63. Moschen, A. R., Geiger, S., Krehan, I., Kaser, A., Tilg, H. (2008) Inter- Grant, P. A., Grant, P. A., Ferrero, R. L., Crowe, S. E., Haas, R., Hat-
feron-␣ controls IL-17 expression in vitro and in vivo. Immunobiology 213, akeyama, M., Goldberg, J. B. (2010) Helicobacter pylori-induced histone
779 –787. modification, associated gene expression in gastric epithelial cells, and
64. Lee, J. H., Lee, B., Lee, H. S., Base, E. A., Lee, H., Ahn, Y. T., Lim, its implication in pathogenesis. PLoS One 5, e9875.
K. S., Huh, C. S., Kim, D. H. (2009) Lactobacillus suntoryeus inhibits pro- 74. Sato, T. A., Mitchell, M. D. (2006) Molecular inhibition of histone
inflammatory cytokine expression and TLR-4-linked NF-␬B activation in deacetylation results in major enhancement of the production of IL-1␤
experimental colitis. Int. J. Colorectal Dis. 24, 231–237. in response to LPS. Am. J. Physiol. Endocrinol. Metab. 290, E490 –E493.
65. Petrof, E. O., Claud, E. C., Sun, J., Abramova, T., Guo, Y., Waypa, T. S., 75. Foligne, B., Nutten, S., Grangette, C., Dennin, V., Goudetcourt, D.,
Nakagawa, Y., Chang, E. B. (2009) Bacteria-free solution derived from Poiret, S., Dewulf, J., Brassart, D., Mercenier, A., Pot, B. (2007) Correla-
Lactobacillus plantarum inhibits multiple NF-␬B pathways and inhibits tion between in vitro and in vivo immunomodulatory properties of lactic
proteasome function. Inflamm. Bowel Dis. 15, 1537–1547. acid bacteria. World J. Gastroentrol. 13, 236 –343.
66. Stetinová, V., Smetanova, L., Kvetina, J., Smetana, J., Svoboda, Z., Zidek, 76. Zaph, C., Du, Y., Saenz, S. A., Nair, M. G., Perrigoue, J. G., Taylor,
Z., Tlaskalova-Hogenova, H. (2010) Caco-2 cells monolayer integrity and B. C., Troy, A. E., Kobuley, D. E., Kastelein, R. A., Cua, D. J., Yu, Y., Ar-
effect of probiotic Escherichia coli Nissle 1917 components. Neuro Endocri- tis, D. (2008) Commensal-dependent expression of IL-25 regulates the
nol. Lett. 31 (Suppl. 2), 51–56. IL-23-IL-17 axis in the intestine. J. Exp. Med. 205, 2191.
67. Ueno, N., Fujiya, M., Segawa, S., Nata, T., Moriichi, K., Tanabe, H.,
Mizukami, Y., Kobayashi, N., Ito, K., Kohgo, Y. (2011) Heat-killed body
of lactobacillus brevis SBC8803 ameliorates intestinal injury in a murine
model of colitis by enhancing the intestinal barrier function. Inflamm KEY WORDS:
Bowel Dis. 17, 2235–2250. bacteria 䡠 histone modification 䡠 inflammation 䡠 cytokines

www.jleukbio.org Volume 92, October 2012 Journal of Leukocyte Biology 911

You might also like