You are on page 1of 11

Multiple TLRs Are Expressed in Human

Cholangiocytes and Mediate Host Epithelial


Defense Responses to Cryptosporidium
parvum via Activation of NF-κB
This information is current as
of June 9, 2017. Xian-Ming Chen, Steven P. O'Hara, Jeremy B. Nelson,
Patrick L. Splinter, Aaron J. Small, Pamela S. Tietz, Andrew
H. Limper and Nicholas F. LaRusso
J Immunol 2005; 175:7447-7456; ;
doi: 10.4049/jimmunol.175.11.7447
http://www.jimmunol.org/content/175/11/7447

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


References This article cites 58 articles, 36 of which you can access for free at:
http://www.jimmunol.org/content/175/11/7447.full#ref-list-1
Subscription Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2005 by The American Association of
Immunologists All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Multiple TLRs Are Expressed in Human Cholangiocytes and


Mediate Host Epithelial Defense Responses to Cryptosporidium
parvum via Activation of NF-␬B1

Xian-Ming Chen,* Steven P. O’Hara,* Jeremy B. Nelson,* Patrick L. Splinter,*


Aaron J. Small,* Pamela S. Tietz,* Andrew H. Limper,† and Nicholas F. LaRusso2*
Infection of epithelial cells by Cryptosporidium parvum triggers a variety of host-cell innate and adaptive immune responses
including release of cytokines/chemokines and up-regulation of antimicrobial peptides. The mechanisms that trigger these host-cell
responses are unclear. Thus, we evaluated the role of TLRs in host-cell responses during C. parvum infection of cultured human
biliary epithelia (i.e., cholangiocytes). We found that normal human cholangiocytes express all known TLRs. C. parvum infection
of cultured cholangiocytes induces the selective recruitment of TLR2 and TLR4 to the infection sites. Activation of several
downstream effectors of TLRs including IL-1R-associated kinase, p-38, and NF-␬B was detected in infected cells. Transfection of

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


cholangiocytes with dominant-negative mutants of TLR2 and TLR4, as well as the adaptor molecule myeloid differentiation
protein 88 (MyD88), inhibited C. parvum-induced activation of IL-1R-associated kinase, p-38, and NF-␬B. Short-interfering RNA
to TLR2, TLR4, and MyD88 also blocked C. parvum-induced NF-␬B activation. Moreover, C. parvum selectively up-regulated
human ␤-defensin-2 in directly infected cells, and inhibition of TLR2 and TLR4 signals or NF-␬B activation were each associated
with a reduction of C. parvum-induced human ␤-defensin-2 expression. A significantly higher number of parasites were detected
in cells transfected with a MyD88 dominant-negative mutant than in the control cells at 48 –96 h after initial exposure to parasites,
suggesting MyD88-deficient cells were more susceptible to infection. These findings demonstrate that cholangiocytes express a
variety of TLRs, and suggest that TLR2 and TLR4 mediate cholangiocyte defense responses to C. parvum via activation of
NF-␬B. The Journal of Immunology, 2005, 175: 7447–7456.

C ryptosporidium parvum, an intracellular parasite within resolution of cryptosporidiosis and resistance to infection (8), the
the protist phylum Apicomplexa, is one of the most com- mechanisms by which host epithelial cells elicit immune responses
monly reported enteric pathogens in both immunocom- are not well understood. Specific attachment to the apical epithelial
petent and immunocompromised individuals worldwide (1). The cell surface by C. parvum sporozoites, as well as parasite mole-
parasite infects gastrointestinal epithelia to produce diarrhea that is cules inserted into epithelia after its attachment (9, 10), appear to
self-limited in immunocompetent subjects but potentially life- activate host-cell secondary signal pathways and thereby alter cell
threatening in immunocompromised individuals, primarily those function (10 –12). The invasion of epithelial cells in vitro by C.
with AIDS (2). Infection by this parasite accounts for ⬃6% of all parvum results in the rapid expression of anti-microbial peptides
diarrheal disease in immunocompetent individuals and occurs in (e.g., ␤-defensins) and the inflammatory chemokines including
⬃24% of AIDS patients with diarrhea worldwide (3). C. parvum is IL-8, TNF-␣, and prostaglandin E2, etc. (12–14). We previously
also the single most common identifiable pathogen in the biliary reported that one intracellular signal pathway activated by C. par-
tract in patients with AIDS-cholangiopathy, an important biliary vum and associated with the cytokine/chemokine release in in-
disorder caused by opportunistic infection of the biliary epithelium fected biliary epithelial cells (i.e., cholangiocytes) is the NF-␬B
and resulting in significant morbidity and mortality in AIDS pa- signal pathway (12). Moreover, we showed that activation of
tients (4). Despite the magnitude and severity of C. parvum-in- NF-␬B accounts for the antiapoptotic activation in C. parvum di-
duced infection, the pathogenesis is poorly understood, and there is rectly infected cholangiocytes and thus benefits parasite propaga-
currently no fully effective therapy (5–7). tion (12). How C. parvum activates such host-cell intracellular
The proportion of exposed individuals developing cryptospori- signaling pathways such as NF-␬B and mediates host-cell inflam-
diosis reflects both parasite infectivity and host immune responses. matory and immune responses is still unclear.
Whereas both innate and adaptive immunity are involved in the TLRs are an evolutionarily conserved family of cell surface
molecules, which are key to innate immunity by detecting invading
*The Center for Basic Research in Digestive Diseases, Division of Gastroenterology
pathogens (15). Most of the known TLRs, upon recognition of
and Hepatology and †Division of Pulmonary and Critical Care Medicine, Mayo Clinic discrete pathogen-associated molecular patterns, activate a com-
College of Medicine, Rochester, MN 55905 mon set of adaptor proteins, such as myeloid differentiation protein
Received for publication April 28, 2005. Accepted for publication September 9, 2005. 88 (MyD88), and protein kinases including IL-1 receptor-associ-
The costs of publication of this article were defrayed in part by the payment of page ated kinase (IRAK).3 IRAK, in turn, activates downstream effec-
charges. This article must therefore be hereby marked advertisement in accordance tors including p-38, ERK1/2, JNK, and I␬B kinases (16). Activa-
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
tion of the kinases leads to the nuclear translocation of
This work was supported by National Institutes of Health Grants DK57993 and
DK24031 (to N.F.L.), HL062150 (to A.H.L.), and by the Mayo Foundation.
2 3
Address correspondence and reprint requests to Dr. Nicholas F. LaRusso, Center for Abbreviations used in this paper: IRAK, IL-1R-associated kinase; HBD, human
Basic Research in Digestive Diseases, Mayo Clinic, 200 First Street, Southwest, ␤-defensin; siRNA, short-interfering RNA; DN, dominant negative; DAPI, 4⬘, 6-dia-
Rochester, Minnesota 55905. E-mail address: larusso.nicholas@mayo.edu midino-2-phenylindole; MBP, myelin basic protein.

Copyright © 2005 by The American Association of Immunologists, Inc. 0022-1767/05/$02.00


7448 TLRs IN C. parvum INFECTION

corresponding nuclear transcription factors, such as AP-1 and NF- liver harvested for transplant and have been extensively characterized (24).
␬B, and thus regulates host-cell responses to pathogens (15, 16). For experiments, H69 cells were used between passage 23 and 30 and
One important host epithelial defense response upon microbial in- maintained for three passages without coculture cells to ensure that the
culture was free of 3T3 fibroblasts.
fection is the production of a variety of small cationic anti-
microbial peptides including ␤-defensins (17). Six human ␤-de- In vitro models and infection assay
fensins (HBDs) have been identified and cloned in human
epithelial cells. HBD-2 and -4 are identical and HBD-5 and -6 are Infection with C. parvum was done in a culture medium consisting of
DMEM-F12, 100 U/ml penicillin, and 100 ␮g/ml streptomycin (Invitrogen
primarily expressed in the epididymis (17, 18). The induction of Life Technologies), and freshly excysted C. parvum sporozoites (1 ⫻ 106
HBD-2 transcription by IL-1 is dependent on a specific NF-␬B sporozoites/per slide well or culture plate). Inactivated organisms (treated
binding site (⫺205 to ⫺186) and its interaction with p-65-p50 at 65°C for 30 min) were used for sham infection controls. An attachment
NF-␬B heterodimer (17, 19, 20). Whereas regulation of HBD ex- model and an attachment/invasion model were used to assay the attachment
pression has been well documented in epithelial cells upon micro- and invasion of C. parvum as described previously (23). Infection assays
(attachment rate or attachment/invasion rate) were conducted after incu-
bial infection (17, 21–22), the role of TLRs in this process has not bation for 2 h with the parasite using an indirect immunofluorescent tech-
been fully explored. Moreover, expression and functions of TLRs nique as described previously (23). For the inhibitory experiments, two
in the liver, particularly in cholangiocytes, which are the target selective inhibitors of NF-␬B, MG-132 and SN50 (25), were added in the
epithelial cells of a group of clinically important diseases, some of medium at the same time as C. parvum. A concentration of 1 ␮M MG-132
or 50 ␮g/ml SN50, which showed no cytotoxic effects on H69 cells or on
which are of infectious nature, have not been characterized. C. parvum sporozoites, was selected for the study. For the experiments to
In the present study, we explored the role of TLR signals in test whether TLRs and TLR-associated signals are involved in cholangio-
cholangiocyte responses to C. parvum infection. We found that cyte defense responses against C. parvum infection, cells in T25 flasks
normal human cholangiocytes express all known TLRs. Whereas were incubated with an equal number of C. parvum sporozoites (5 ⫻ 106)

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


C. parvum attachment to and invasion of cultured cholangiocytes for 2 h, washed with culture medium to remove nonattached and nonin-
ternalized parasites, and were then further cultured for up to 96 h, followed
appear not to be dependent on host-cell TLRs, C. parvum infection by PCR analysis for C. parvum.
of cholangiocytes induces the recruitment of TLR2 and TLR4 to
the infection sites, resulting in activation of IRAK and phosphor- RT-PCR
ylation of p-38. Moreover, activation of NF-␬B by C. parvum in
Total cellular RNA was extracted from the cells using Tri-Reagent (Sigma-
directly infected cells is dependent on both TLR2 and TLR4 sig- Aldrich). Total RNA (5 ␮g) was reverse transcribed to cDNA by using a
nals. In addition, a TLR2- and TLR4-dependent and NF-␬B-asso- Moloney Murine Leukemia Virus Reverse Transcriptase Kit (Invitrogen
ciated induction of HBD-2, but not HBD-1 and HBD-3, occurs in Life Technologies). After reverse transcription, cDNA was amplified using
cells infected with C. parvum, whereas MyD88-deficient cells are PCR with gene-specific primers designed to amplify a portion of the coding
more susceptible to C. parvum infection in vitro. These findings sequences (Table I). The PCR consisted of one cycle of 10-min denatur-
ation at 94°C; 18 –35 cycles of 1 min at 94°C, 1 min at 48 –55°C, and 1 min
demonstrate that cholangiocytes express a variety of TLRs, and at 72°C, and a final extension step at 72°C for 10 min. rRNA (18s) was also
TLR2 and TLR4 signals mediate cholangiocyte responses to C. amplified to confirm that an equal amount of total cDNA was used for each
parvum via activation of NF-␬B, implicating an important role of sample, and all experiments were done in duplicate. Template cDNA pre-
TLRs in cholangiocyte defense responses to microbial infection in pared from TIB-202, a normal human monocyte cell line that expresses all
known TLRs except TLR3 (15, 26), was used as positive control for TLR
the biliary tree. mRNA detection in H69 cells. Sequencing was performed on all positive
PCR products (Mayo Molecular Core Facility, Rochester, MN) to confirm
Materials and Methods the identity of amplified genes. To quantitatively measure C. parvum in-
C. parvum and H69 cells fection in H69 cells, a quantitative RT-PCR approach using a LightCycler
(Roche Diagnostic Systems) was established by modification of a previous
C. parvum oocysts of the Iowa strain were purchased from a commercial report (27). Briefly, total RNA was harvested from the cells after exposure
source (Bunch Grass Farms). Before infecting cells, oocysts were excysted to C. parvum and reverse transcribed to cDNA and amplified using Am-
to release infective sporozoites as described previously (23). H69 cells (a plitag Gold PCR Master Mix (Roche Diagnostic Systems). Primers specific
gift from Dr. D. Jefferson, Tufts University, Boston, MA) are SV40-trans- for C. parvum 18s ribosomal RNA (Table I) were used to amplify the
formed human bile duct epithelial cells originally derived from a normal cDNA specific to the parasite. Primers specific for human plus C. parvum

Table I. Primer sequences

Primers

Target mRNA Forward Reverse

TLR1 5⬘-AAACGGTCTCATCCACGTTC-3⬘ 5⬘-GAGCAATTGGCAGCACACTA-3⬘


TLR2 5⬘-GATGCCTACTGGGTGGAGAA-3⬘ 5⬘-CGCAGCTCTCAGATTTACCC-3⬘
TLR3 5⬘-CTTGACCTGGGCCTTAATGA-3⬘ 5⬘-TTTCCAGAGCCGTGCTAAGT-3⬘
TLR4 5⬘-CAACAAAGGTGGGAATGCTT-3⬘ 5⬘-TGCCATTGAAAGCAACTCTG-3⬘
TLR5 5⬘-TGCATCCAGATGCTTTTCAG-3⬘ 5⬘-CCAGCCATTTCCAGAAACAT-3⬘
TLR6 5⬘-TTCCAGAGCTGCCAGAAGAT-3⬘ 5⬘-GAGATACCAGGGCAGATCCA-3⬘
TLR7 5⬘-CCACAACCAACTGACCACTG-3⬘ 5⬘-GATCACACTTTGGCCCTTGT-3⬘
TLR8 5⬘-TGAAGCACATCCCAAATGAA-3⬘ 5⬘-TCAAAGGGGTTTCCGTGTAG-3⬘
TLR9 5⬘-GGACACTCCCAGCTCTGAAG-3⬘ 5⬘-TTGGCTGTGGATGTTGTTGT-3⬘
TLR10 5⬘-GGCACAGGGTTAGGAAAACA-3⬘ 5⬘-GAGATTGTGGTGGGCAAAGT-3⬘
MD-2 5⬘-AGGGGCACGAGGTAAATCTT-3⬘ 5⬘-AACTTCTTTGCGCTTTGGAA-3⬘
Myd88 5⬘-ACAGCTCCCAGGAACAGCTA-3⬘ 5⬘-CACCTAAGACCATGGCACCT-3⬘
IL-8 5⬘-GCCAAGAGAATATCCGAACT-3⬘ 5⬘-AAAGTGCAACCACATGTCCT-3⬘
HBD-1 5⬘-CATGAGAACTTCCTACCTTCTG-3⬘ 5⬘-GCTCACTTGCAGCACTTGGCC-3⬘
HBD-2 5⬘-GACTCAGCTCCTGGTGAAGC-3⬘ 5⬘-GAGACCACAGGTGCCAATTT-3⬘
HBD-3 5⬘-CCAGGTCATGGAGGAATCAT-3⬘ 5⬘-TCTTTCTTCGGCAGCATTTT-3⬘
C. parvum ⫺18s 5⬘-TAGAGATTGGAGGTTGTTCCT-3⬘ 5⬘-CTCCACCAACTAAGAACGGCC-3⬘
The Journal of Immunology 7449

18s (28) were used to determine total 18s cDNA. Data were expressed as rosine phosphatase inhibitors, sodium orthovanadate and sodium fluoride
copies of C. parvum 18s vs total 18s. at 1 mM. The cell lysates were centrifuged at 13,000 ⫻ g for 10 min and
were assayed for protein concentration using the Bradford reagent (Sigma-
Plasmid constructs and short-interfering RNA (siRNA) Aldrich). The cell lysates (1 mg of protein) were then incubated with 1 ␮g
H69 cells stably transfected with constructs of TLR2-DN (dominant neg- of anti-IRAK (Upstate Biotechnology) for 2 h at 4°C. Fifty microliters of
ative), TLR4-DN, and MyD88-DN were obtained by transfecting cells with 10% protein A-Sepharose (Sigma-Aldrich) was then added and incubated
the constructs followed by antibiotic selection. Cells were plated at 50 – for an additional 2 h at 4°C. The beads were then washed once with lysis
80% confluency and transfected with the plasmid DNA using the Lipo- buffer and twice with kinase buffer (10 mM HEPES (pH 7.6), 20 mM
fectamine Plus reagent kit (Invitrogen Life Technologies) according to the MgCl2, 20 mM ␤-glycerophosphate, 20 mM p-nitrophenyl phosphate, 1
manufacturer’s instructions. TLR2-DN and TLR4-DN are kinase-inactive mM EDTA, and 1 mM benzamidine). The beads were incubated for 30 min
(DN) mutants of TLR2 and TLR4, respectively, and were kindly provided at 30°C in a final volume of 20 ␮l in the presence of 2 ␮g of myelin basic
by Dr. M. F. Smith (University of Virginia, Charlottesville, VA). protein (MBP) (Sigma-Aldrich), 100 ␮M ATP, and 5 ␮Ci of [␥-32P]ATP
MyD88-DN is a DN mutant of MyD88 and was a gift from Prof. J. (DuPont NEN Research Products). SDS sample buffer was added to protein
Tschopp (University of Lausanne, Lausanne, Switzerland). Stable-trans- A beads and boiled for 5 min, and then subjected to SDS-PAGE analysis.
fected clones were selected by adding Zeocin (600 ␮g/ml) (Invitrogen Life The gels were dried, and the intensity of the radioactive signal was quan-
Technologies) to the culture medium. Selected transfected cells were cul- tified using the Molecular Analyst software (Bio-Rad).
tured with Zeocin in the medium and then grown on 4-well chamber slides Western blotting
or T25 flasks for the experiments.
Specific siRNAs to TLR2, TLR4, and MyD88, which target human H69 cells were grown in T25 flasks to 95% confluence and exposed to C.
TLR2, TLR4, and MyD88 mRNA sequences were designed. For siRNA parvum sporozoites. Cells were then lysed with the M-PER mammalian
transfection, H69 cells were grown to 40 – 60% confluency on 10-mm protein extraction reagent (Pierce), and protein concentrations were deter-
dishes and were transfected with the siRNAs using the siPORT lipid trans- mined using Bradford reagent according to the instructions of the supplier
fection agent (Ambion). siRNAs that showed the most inhibition of TLR2, (Sigma-Aldrich). Twenty micrograms of lysate protein per lane were sep-
TLR4, or MyD88 protein expression were selected with approaches we arated by SDS-PAGE under reducing conditions and blotted onto nitrocel-

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


previously reported (28) and used in this study as follows: TLR2, AATAT lulose membranes. Membranes were incubated with the primary Abs to
TCGTGTGCTGGATAAGCCTGTCTC (sense) and AACTTATCCAGC TLR2 and TLR4 (Imgene), MyD88 (Imgene), p-p-38 (Cell Signaling Tech-
ACACGAATACCTGTCTC (antisense); TLR4, AACTGCTGCTTCAAG nology), p-ERK1/2 (Santa Cruz Biotechnology), p-JNK (Santa Cruz Bio-
ATATACCCTGTCTC (sense) and AAGTATATCTTGAAGCAGCAGC technology), I␬B␣ (Santa Cruz Biotechnology), and HBD-2 (Alpha Diag-
CTGTCTC (antisense); and MyD88, AACTGCTGCTTCAAGATATA nostic), and then with 0.2 ␮g/ml HRP-conjugated secondary Ab, and
CCCTGTCTC (sense) and AAGTATATCTTGAAGCAGCAGCCTGTC revealed with ECL light substrate (ECL; Amersham Biosciences).
TC (antisense). These siRNA oligonucleotides were determined to have no
significant overlap with homologous gene sequences. Nonspecific siRNAs ELISA
containing the same nucleotides but in irregular sequence (i.e., scrambled To analyze the production of NF-␬B-associated proinflammatory cytokines
siRNAs) were used as the controls. The siRNAs were further labeled with by H69 cells in response to C. parvum infection, H69 cells were grown to
Cy3 using the Silencer siRNA labeling kit (Ambion) for the identification subconfluence in 4-well chamber slides. After incubation with assay me-
of transfected cells by confocal microscopy. dium containing freshly excysted sporozoites and various specific inhibi-
Immunohistochemistry and immunofluorescent microscopy tors at 37°C for 12 h, supernatants were collected to assay the cytokines,
and cells were harvested for protein determination. IL-8 concentration in
For immunohistochemistry, paraffin-embedded normal human liver tissue the supernatants was determined by ELISA using a commercial kit (R&D
samples (Institutional Review Board no. 725-00) were used for TLRs im- Systems). Cells were lysed with the M-PER mammalian protein extraction
munohistochemistry. Tissue sections of 5 ␮m were deparaffinized in xy- reagent (Pierce), and protein concentrations were assessed with Bradford
lene and rehydrated in ethanol followed by water and PBS. Endogenous reagent (Sigma-Aldrich). The concentrations of IL-8 detected were calcu-
peroxidase was blocked by immersion in 3% hydrogen peroxide. The tissue lated from standard curves prepared with the recombinant proteins and
sections were then incubated with TLR Abs at a concentration of 5 ␮g/ml expressed as picogram of IL-8 per microgram cell protein.
for 1 h at room temperature. Seven TLR Abs currently available and spe-
cific for TLR2–9 were obtained and used in the study (Imgenex). Control
Statistical analysis
sections were incubated with PBS containing normal goat serum without a All values are given as mean ⫹ SE. Means of groups were compared with
primary Ab. Immunostaining was then detected with the Vectastain per- the Student’s t test (unpaired) or ANOVA test when appropriate. p values
oxidase kit (Vector Laboratories) and developed with diaminobenzidine ⬍0.05 were considered statistically significant.
tetrahydrochloride. The sections were counterstained with hematoxylin fol-
lowed by light microscopy. Results
For immunofluorescent microscopy, H69 cells were exposed to C. par- Normal human cholangiocytes express multiple TLRs and
vum sporozoites as described above. After 2 h of incubation, cells were
fixed (0.1 mol/L 1,4-piperazinediethanesulfonic acid (pH 6.95), 1 mmol/L accessory molecules
EGTA, 3 mmol/L magnesium sulfate (Sigma-Aldrich), and 2% parafor- mRNA expression of TLRs and accessory molecules MD-2 and
maldehyde) at 37°C for 20 min and then permeabilized with 0.2% (v/v)
Triton X-100 in PBS. For double-immunofluorescent labeling, fixed cells
MyD88 was assessed by RT-PCR in a SV40-transformed normal
were incubated with primary mAbs to associated proteins mixed with a human cholangiocyte cell line, H69. TIB-202 cells, a human
polyclonal Ab against C. parvum sporozoite membrane proteins followed monocyte cell line that expresses all known TLRs except TLR3
by rhodamine-labeled anti-mouse and fluorescein-labeled anti-rabbit Abs (15, 26), was used as the positive control. TLR1–10 mRNA was
(Molecular Probes). Some cells were incubated with polyclonal Abs to detected in H69 cells (Fig. 1, A and B). Consistent with previous
associated proteins mixed with a mAb against C. parvum (2H2; Immu-
nuCell) followed by rhodamine-labeled anti-rabbit and fluorescein-labeled reports, mRNA of all known TLRs except TLR3 was confirmed in
anti-mouse Abs. In some experiments, 4⬘,6-diamidino-2-phenylindole TIB-202 cells (Fig. 1, A and B). Both MD-2 and MyD88 mRNA
(DAPI; 5 ␮M) was used to stain cell nuclei. Labeled cells were rinsed three were also detected in H69 cells (Fig. 1B). Specificity of the PCR
times with PBS and once with distilled water, then mounted with mounting products was confirmed by sequencing. Expression of selected
medium (H-1000; Vector Laboratories), and assessed by confocal laser
scanning microscopy. Images obtained from the Zeiss 510 confocal mi-
TLRs (i.e., TLR2 and TLR4) and MyD88 protein in H69 cells was
croscope (Zeiss) were manipulated uniformly for contrast and intensity also detected by Western blotting (Fig. 1C).
using the Adobe (Adobe Systems) Photoshop software. To further test expression of TLRs at the protein level in human
Immunoprecipitation and activity of IRAK cholangiocytes, we assessed TLR protein expression in cholangio-
cytes in normal human liver tissues with Abs currently available to
H69 cells were grown in 10-cm dishes to 95% confluence and exposed to TLR2–9 by immunohistochemistry. Although the negative con-
C. parvum sporozoites at 37°C for 2 h. Cells were then rinsed with PBS and
scraped into 1 ml of cold lysis buffer (50 mM Tris-HCl (pH 8.0), 150 mM
trols, stained with normal goat serum and the secondary Ab,
NaCl, 1% Nonidet P-40, 0.5% deoxycholic acid, and 0.1% SDS) supple- showed no specific staining in cholangiocytes (Fig. 2A), strong
mented with 1 mM PMSF, leupeptin and pepstatin at 20 ␮g/ml, and ty- expression of TLR2–9 proteins was detected in cholangiocytes
7450 TLRs IN C. parvum INFECTION

FIGURE 1. Expression of TLRs and accessory molecules in cultured


human cholangiocytes. A SV40-transformed normal human cholangiocyte FIGURE 2. Expression of TLR2–9 proteins in cholangiocytes of nor-
cell line, H69, was screened for the mRNA expression of all known human mal human liver tissues. Expression of TLR2–9 proteins in normal human
TLRs and accessory molecules, MD-2 and MyD88, by RT-PCR. TIB-202 liver tissue sections was assessed by immunohistochemistry followed by
cells, a normal human monocyte cell line that expresses all known TLRs light microscopy. Although the negative controls, stained with normal goat

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


except TLR3 (15, 26), was used as the positive control. Expression of serum and the secondary Ab, showed no specific staining in cholangiocytes
TLR1–10 (A and B), MD-2 and MyD88 (B) was detected in H69 cells. along the biliary duct (A), strong expression of TLR2–9 proteins was de-
Expression of TLR2, TLR4, and MyD88 protein was detected in H69 cells tected in cholangiocytes (B–I). Insets in B–I are high magnification of the
by Western blotting (C). Neg Ctrl, negative control, reverse transcriptase boxed region in B–I. Arrowheads indicate staining at the apical surface.
template from H69 cells. Bar, 10 ␮m.

vum sporozoites (a model in which the organism can attach to but


along the biliary duct (Fig. 2, B–I). Expression was found predom- not invade the fixed cell surface), no significant change in attach-
inately in the apical membrane domain (the luminal domain) (ar- ment rate was found among nontransfected cells and cells trans-
rowheads in Fig. 2, B–I). fected with TLR2-DN, TLR4-DN, or MyD88-DN (Fig. 4A). When
unfixed cholangiocytes were exposed to C. parvum sporozoites (a
C. parvum recruits cholangiocyte TLR2 and TLR4, but not other model in which the organism can both attach to and enter host
TLRs, to the attachment sites in vitro cells), no significant change in attachment/invasion rate was found
Previous studies by us and others (9, 10, 29, 30) demonstrated that between nontransfected and transfected cells (Fig. 4B); these re-
C. parvum recruits a variety of host-cell receptors and exchangers sults suggest that C. parvum sporozoite attachment to and invasion
to the infection sites facilitating host-cell invasion by the parasite. of cholangiocytes is not dependent upon host-cell TLR2 and TLR4
To test whether C. parvum induces membrane translocation of and their signaling pathways.
TLRs in host-cells, accumulation of TLRs at the infection sites was
measured by dual immunofluorescent labeling using Abs to Activation of downstream effectors of TLR2 and TLR4 signal
TLR2–9 and C. parvum. Strong accumulation of TLR2 (in red, pathways
arrowhead in Fig. 3A2) and TLR4 (in red, arrowhead in Fig. 3B2) To determine whether TLR signal pathways are activated during
was observed at the infection site of C. parvum (in green, arrow- C. parvum infection of cholangiocytes, we measured the activation
heads in Fig. 3, A1 and B1). No obvious accumulation of TLR5 of various components of downstream effectors of TLRs. A sig-
(Fig. 3, C1–C3), TLR9 (Fig. 3, D1–D3), and other analyzed TLRs nificant increase of IRAK activity, an immediate downstream ef-
(data not shown) was found at C. parvum infection sites. No mem- fector for both TLR2 and TLR4 (16), was detected in cholangio-
brane accumulation of TLR2–9 was found in the sham-infected cytes upon C. parvum infection as seen by phosphorylation of
cells, and no positive staining was observed in the parasite itself MBP (31). A significant inhibition of IRAK activity was found in
with the procedure used in the experiments (data not shown). To cells transfected with TLR2-DN, TLR4-DN, or MyD88-DN by
test whether TLR2 and TLR4 activity is required for their accu- quantitation of 32P labeling of the substrate MBP after SDS-PAGE
mulation at the infection site, we stably transfected cells with DN (Fig. 5A). To further test which downstream effectors of IRAK are
mutants of TLR2 and TLR4 and then exposed to C. parvum. Func- activated, cholangiocytes were exposed to C. parvum followed by
tional inhibition by transfection of DNs was evidenced by the in- Western blotting using Abs to the activated phosphorylated forms
hibition of activation of downstream effectors as described below. of various IRAK downstream kinases (16). A significant increase
A similar accumulation of TLR2 and TLR4 was detected in cells of phosphorylation of p-38, but not JNK and ERK1/2, was de-
transfected with TLR2-DN and TLR4-DN, respectively. Quanti- tected by immunoblotting using specific Abs against those phos-
tative analysis of TLR accumulation is shown in Fig. 3E. phorylated kinases (Fig. 5, B and C). A significant inhibition of
p-38 phosphorylation was found in cells transfected with TLR2-
C. parvum infection of cholangiocytes in vitro is not TLR2- and DN, TLR4-DN, or MyD88-DN (Fig. 5, B and C).
TLR4-dependent Activated IRAK can further activate the NF-␬B pathway (16).
To determine whether TLR2 and TLR4 and their signal pathways When cholangiocytes were exposed to C. parvum for 2 h, a sig-
are involved in the infection process of C. parvum into host epi- nificant decrease of I␬B␣ protein was detected (Fig. 6A). In con-
thelial cells, C. parvum attachment to and invasion of cells with trast, a partial but significant inhibition of I␬B␣ decrease was
functional inhibition of TLR2, TLR4, and MyD88 was assessed. found in cells transfected with TLR2-DN or TLR4-DN. A com-
When cholangiocytes were prefixed and then exposed to C. par- plete blockage of I␬B␣ decrease upon C. parvum infection was
The Journal of Immunology 7451

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


FIGURE 4. C. parvum attachment to and invasion of human cholangio-
cytes in culture is not dependent upon host-cell TLR2 and TLR4. H69 cells
FIGURE 3. Accumulation of cholangiocyte TLR2 and TLR4, but not or cells stably transfected with functional inhibitory DN mutants of TLR2,
other TLRs, at the attachment sites during C. parvum infection. H69 cells TLR4, or MyD88 were exposed to C. parvum for 2 h followed by immu-
were exposed to C. parvum for 2 h followed by immunofluorescent mi- nofluorescent confocal microscopy. A, Attachment assay in prefixed cells
croscopy. A1–D3, Representative confocal micrographs by dual labeling of shows no significant difference of C. parvum attachment in all conditions.
C. parvum (in green, left panel), TLRs (in red, middle panel), and the B, Attachment/invasion assay in nonfixed cells after exposure to C. parvum
merged images (right panel). Accumulation of TLR2 (arrowhead in A2) sporozoites. No significant change in attachment/invasion rate was found
and TLR4 (arrowhead in B2) was found at the host-cell-parasite interface. between nontransfected and transfected cells. Ctrl, control.
No obvious accumulation of TLR5 (arrowhead in C2), TLR9 (arrowhead
in D2), and other TLRs (data not shown) was observed at the infection
sites. E, Quantitative analysis of accumulation of TLR2 and TLR4 at the
C. parvum sporozoite preparation because NF-␬B activation by C.
infection sites. No significant difference was found in the accumulation of
parvum is limited only to directly infected cells, whereas in the
TLR2 and TLR4 at the infection sites between cells transfected with DN
mutants of TLR2 or TLR4 and cells transfected with empty vector controls. control experiment, LPS activated NF-␬B in virtually all cells.
Bar, 2 ␮m. Taken together, the data suggest that various downstream effectors
of TLR2 and TLR4 and associated intracellular signals such as
NF-␬B are activated in cholangiocytes upon C. parvum infection.
found in MyD88-DN-transfected cells (Fig. 6A). To further test the
nuclear translocation of NF-␬B in cells upon C. parvum infection, C. parvum induces expression of IL-8 and HBD-2 in
we used an Ab to p-65, a component of the NF-␬B complex that cholangiocytes via TLR2- and TLR4-mediated NF-␬B activation
displays nuclear translocation in directly infected cholangiocytes To further confirm the role of TLR2 and TLR4 in C. parvum-
upon C. parvum infection (12), to measure NF-␬B activation in C. induced activation of the NF-␬B pathway, expression and secre-
parvum-infected cells by confocal immunofluorescent microscopy. tion of IL-8, a well-known gene product of NF-␬B-dependent tran-
Consistent with results of our previous studies, nuclear transloca- scription (32), was assessed in cholangiocytes transfected with
tion of p-65 was found in directly infected cells, not in bystander DNs of TLR2, TLR4, and MyD88 in response to C. parvum in-
noninfected cells (Fig. 6B). No nuclear translocation of p-65 was fection. Cells were incubated with C. parvum for 12 h, and ex-
found in both directly infected and bystander noninfected cells in pression of IL-8 mRNA was measured by RT-PCR. An increase of
the TLR2-DN-, TLR4-DN-, and MyD88-DN-transfected cells IL-8 mRNA expression was detected in H69 cells after exposure to
(Fig. 6, C–E). Whereas scrambled siRNAs had no effect on C. C. parvum. C. parvum-induced IL-8 mRNA expression was sig-
parvum-induced NF-␬B nuclear translocation (Fig. 6, F and G), no nificantly reduced in cells transfected with TLR2-DN and
nuclear translocation of p-65 was found in infected cells trans- TLR4-DN (Fig. 7A), revealing only a slight increase over the
fected with siRNAs to TLR4 (Fig. 6, H and I), TLR2, and MyD88 sham-infected control H69 cells, whereas IL-8 mRNA expression
(data not shown). In contrast, a control experiment using 100 in response to C. parvum was completely inhibited in MyD88-
ng/ml Escherichia coli LPS, a well-known ligand for TLR4, DN-transfected cells (Fig. 7A). Inhibition of NF-␬B by two selec-
showed nuclear translocation of p-65 in most treated nontrans- tive functional inhibitors, SN50 and MG-132 (25), also diminished
fected and empty vector-transfected cells, but not in TLR4-DN- C. parvum-induced IL-8 mRNA expression in cholangiocytes.
and MyD88-DN-transfected cells (data not shown), confirming the Consequently, when cells were incubated with C. parvum for 12 h,
existence of the TLR4/MyD88/NF-␬B pathway in H69 cells. a significant increase of IL-8 secretion was found in the superna-
Equally important, these results exclude LPS contamination in our tants from cells incubated with C. parvum as measured by ELISA.
7452 TLRs IN C. parvum INFECTION

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


FIGURE 5. C. parvum activates TLR2- and TLR4-associated signals in
human cholangiocytes in culture. H69 cells or cells stably transfected with
functional inhibitory DN mutants of TLR2, TLR4, or MyD88 were ex-
posed to C. parvum for 2 h. A, Activation of IRAK as measured by im-
munoprecipitation followed by incubation with [␥-32P]ATP using MBP as
a substrate. IRAK was also blotted by Western blot to confirm an equal FIGURE 6. C. parvum activates the NF-␬B pathway in directly infected
amount of total IRAK used for the assay. The intensity of the radioactive cholangiocytes via TLR2 and TLR4 signals. A, Degradation of I␬B␣ as
signals was quantified and expressed as densitometric arbitrary values, rep- assessed by quantitative Western blot. H69 cells were exposed to C. par-
resentative of three distinct experiments. B, Phosphorylation of p-38, JNK, vum for 2 h, and I␬B␣ in the cell lysates was determined by Western blot
and ERK1/2 as assessed by immunoblotting using specific Abs. To confirm using an Ab against I␬B␣. Actin was also blotted to confirm an equal
equal loading, membranes were reprobed with an actin Ab. C, Quantitative loading. The intensity of the signals was quantified using a PhosphorIm-
analysis of phosphorylation of p-38, JNK, and ERK1/2. The intensity of the ager and expressed as densitometric arbitrary values. Data are representa-
signals was quantified using a PhosphorImager and expressed as densito- tive of three distinct experiments. B–F and H, Nuclear translocation of p-65
metric arbitrary values. Data are representative of three distinct experi- of the NF-␬B complex. Cells were exposed to C. parvum for 2 h followed
ments. ⴱ, p ⬍ 0.05, compared with sham-infection control; #, p ⬍ 0.05, by immunofluorescent staining using a mAb against p-65 and a polyclonal
compared C. parvum-infected cells. Ab to C. parvum. Nuclear translocation of p-65 was found in directly
infected cells (p-65 in green, nuclear translocation indicated by asterisks;
C. parvum in red by arrowheads in B), but not in bystander noninfected
cells. No nuclear translocation of p-65 was found in both directly infected
A significant decrease of IL-8 release was detected in the super- (asterisks in C–E) and nondirectly infected cells in the TLR2-DN-, TLR4-
natants from cells transfected with TLR2-DN, TLR4-DN, and DN-, and MyD88-DN-transfected cells. Whereas nuclear translocation of
MyD88-DN or cells treated with NF-␬B inhibitors (Fig. 7B). p-65 was found in infected cells transfected with a scrambled siRNA (F),
To test the role of TLRs in C. parvum-induced HBD production, it was absent in directly infected cells (G) transfected with Cy3-tagged
expression of HBD-1–3 was measured in cells after exposure to C. TLR4-siRNA. G and I show merged images of the DAPI and Cy3 channels
parvum for 12 h. A constitutive expression of ⌯〉D-1 and ⌯〉D-3 of the same field in F and H, respectively. C. parvum was identified with
mRNA was found in the sham-infected, C. parvum-infected non- DAPI staining of its nucleus in blue (arrowheads in G and I). Cells trans-
transfected cells, cells treated with NF-␬B inhibitors, and cells fected with siRNA were confirmed by positive Cy3 signal in red (asterisks
in G and I). ⴱ, p ⬍ 0.05, compared with sham-infection control; #, p ⬍
transfected with TLR2-DN, TLR4-DN, and MyD88-DN (Fig. 8A).
0.05, compared C. parvum-infected cells. Bar, 2 ␮m.
In contrast, a low expression of HBD-2 was found in the sham-
infected cells, and a significant increase of HBD-2 mRNA was
The Journal of Immunology 7453

FIGURE 8. C. parvum induces HBD2 expression in cholangiocytes via

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


TLR2- and TLR4-associated activation of NF-␬B. H69 cells were exposed
to C. parvum for 12 h, and expression of HBDs was determined by RT-
PCR, Western blot, and immunofluorescent microscopy. A, Expression of
mRNA of HBD-1–3 as assessed by RT-PCR. 18s rRNA was amplified to
confirm an equal amount of total mRNA used for each sample. B, Quan-
titative analysis of HBD mRNA signals using a PhosphorImager. C, Ex-
pression of HBD-2 as determined by Western blot using an Ab against
HBD-2. The intensity of the signals was quantified and expressed as
densitometric arbitrary values. D, Expression of HBD-2 in C. parvum
directly infected cells. H69 cells were exposed to C. parvum for 12 h
followed by triple immunofluorescent staining using a polyclonal Ab to
label HBD-2 (in green), a mAb to C. parvum (in red as indicated by
FIGURE 7. C. parvum induces IL-8 release in cholangiocytes via arrowheads), and DAPI to label cell nucleus. ⴱ, p ⬍ 0.05, compared
TLR2- and TLR4-associated activation of NF-␬B. A, Expression of IL-8 with sham-infection control; #, p ⬍ 0.05, compared C. parvum-infected
mRNA. H69 cells were exposed to C. parvum for 2 h followed by RT-PCR. cells. Data are representative of three distinct experiments. Bar, 5 ␮m.
18s rRNA was amplified to confirm an equal amount of total mRNA used
for each sample. The intensity of the signals was quantified using a Phos- and uninfected cells transfected with TLR2-DN, TLR4-DN, and
phorImager and expressed as densitometric arbitrary values. Data are rep- MyD88-DN, or cells treated with NF-␬B inhibitors (data not
resentative of three distinct experiments. B, IL-8 concentration in the su- shown).
pernatants of the cell cultures. H69 cells were exposed to C. parvum for
12 h, and supernatants were collected. IL-8 concentration was determined Expression of MyD88-DN in cultured cholangiocytes hampers
by ELISA. Data are expressed as picograms of IL-8 per milligram of total epithelial defense against C. parvum infection
protein and representative of three distinct experiments. ⴱ, p ⬍ 0.05, com-
pared with sham-infection control; #, p ⬍ 0.05, compared C. parvum- To test whether TLRs and TLR-associated signals are involved in
infected cells. cholangiocyte defense responses against C. parvum infection, we
assessed the number of parasites detected over time in H69 cells
stably transfected with a control empty vector or MyD88-DN. Af-
detected in cells after exposure to C. parvum. C. parvum-induced ter incubation with an equal number of C. parvum sporozoites for
HBD-2 mRNA expression was significantly reduced in cells trans- 2 h, cells were washed with culture medium to remove nonattached
fected with TLR2-DN, TLR4-DN, and MyD88-DN (Fig. 8A). In- and noninternalized parasites and were then further cultured for up
hibition of NF-␬B by SN50 and MG-132 also diminished C. par- to 96 h. We then isolated RNA from the cells to measure C. par-
vum-induced HBD-2 mRNA expression in cholangiocytes (Fig. 8, vum infection in cells by quantitative RT-PCR using C. parvum-
A and B). Consistent with mRNA expression of HBD-2, expres- specific primers. We found that the number of C. parvum detected
sion of HBD-2 peptide was detected in infected cholangiocyte cul- in cells transfected with the control vector gradually but consis-
ture by Western blotting (Fig. 8C). A low expression of HBD-2 tently declined over time (Fig. 9, u), consistent with previous stud-
peptide was detected in the sham-infected cells, and strong expres- ies (23). We also detected a significantly higher number of para-
sion was found in cells exposed to the parasite. A decreased ex- sites in MyD88-deficient cells than in cells transfected with the
pression of HBD-2 was found in cells transfected with TLR2-DN, control vector at 48, 72, and 96 h after initial infection (Fig. 9, f).
TLR4-DN, and MyD88-DN after exposure to C. parvum. To fur- Although other interpretations are possible, these data suggest that
ther clarify the relationship between NF-␬B activation and HBD-2 cholangiocytes exhibit innate epithelial defense responses to in-
expression induced by C. parvum, we measured HBD-2 expression hibit C. parvum infection, and that MyD88 functional inhibition in
in C. parvum-infected cells by immunofluorescent microscopy. cholangiocytes hampers this epithelial defense against C. parvum.
Consistent with nuclear translocation of NF-␬B in directly infected
cells, expression of HBD-2 was found only in cells directly in- Discussion
fected by the parasite not in bystander noninfected cells (Fig. 8D). In the work described in this study, we demonstrated that normal
No obvious HBD-2 expression was found in both directly infected human cholangiocytes in vivo and in culture express all known
7454 TLRs IN C. parvum INFECTION

capacity to quickly recognize and respond to microorganisms in


the bile, initiating the first defense machinery in the biliary tree to
keep the bile sterile and maintain the integrity of the biliary epi-
thelial barrier.
TLRs participate in epithelial immune responses to a variety of
invading pathogens including parasites, such as Trypanosoma
cruzi, malaria parasites, and Leishmania (43– 46). Whether TLRs
also play a role in C. parvum infection of any epithelia is unclear.
C. parvum sporozoites attach to and invade host cells, resulting in
the formation of a parasitophorous vacuole via a host-cell actin-
dependent mechanism (23, 47– 49). We previously showed that a
variety of host-cell membrane-associated kinases, e.g., c-Src and
PI3K, and actin-associated proteins, e.g., cortactin, and Arp2/3
FIGURE 9. Transfection of cholangiocytes with MyD88-DN hampers
epithelial defense against C. parvum infection. H69 cells stably transfected
complex, are recruited to the infection sites and induce actin re-
with a control empty vector or MyD88-DN construct were exposed to an modeling and facilitate parasite invasion of host epithelial cells
equal number of C. parvum for 2 h followed by extensive washing and (10, 23, 29, 30). TLR4 has been reported as a coreceptor for E. coli
continued culture. A significantly greater number of parasites were de- P fimbriae attachment to host epithelial cells (50). In this study, we
tected in MyD88 functionally deficient cells compared with the control found that TLR2 and TLR4, but not other TLRs, are recruited to
cells after exposure to C. parvum for 48 –96 h. Quantitative PCR was the infection sites. However, C. parvum attachment to and invasion
performed using a LightCycler, and data were expressed as copies of of cholangiocytes seems independent of TLRs, because DN mu-

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


C. parvum 18s vs total 18s. tation of TLR2 and TLR4 did not affect parasite attachment and
invasion. Instead, we identified that a set of downstream effectors
of TLR2 and TLR4 are activated in cholangiocytes during C. par-
TLRs. Moreover, C. parvum infection of human cholangiocytes in vum infection, suggesting that parasite host-cell interactions may
culture induces the recruitment of TLR2 and TLR4, but not other trigger cholangiocyte reactivity in response to C. parvum via ac-
TLRs, to the infection sites. However, whereas neither TLR2 nor tivation of TLR2- and TLR4-mediated intracellular signaling
TLR4 appears necessary for C. parvum attachment to or invasion pathways.
of cholangiocytes in vitro, the parasite activates several down- The signaling events following ligation of different TLRs in-
stream effectors of TLR2 and TLR4. More specifically, TLR2- and volve the activation of a common set of adaptor proteins (e.g.,
TLR4-induced signaling is required for the activation of NF-␬B MyD88) and protein kinases (15, 16). Activation of IRAK, an
induced by C. parvum in directly infected cells. More importantly, immediate downstream kinase to MyD88, was detected in cholan-
a TLR2- and TLR4-dependent and NF-␬B-associated induction of giocytes upon C. parvum infection. Downstream effectors of IRAK
HBD-2, but not HBD-1 and HBD-3, was found in C. parvum di- in epithelia include p-38, JNK, ERK1/2, and I␬B kinase (15, 16).
rectly infected cells. MyD88-deficient cells are more susceptible to Phosphorylation of p-38, but not JNK and ERK1/2, and activation
C. parvum infection in vitro. These findings demonstrate that of the NF-␬B signaling pathway were found in cells exposed to C.
cholangiocytes express a variety of TLRs, and TLR2 and TLR4 parvum, suggesting a differential activation of downstream effec-
mediate cholangiocyte defense responses to C. parvum via activa- tors of IRAK in cholangiocytes during C. parvum infection, sim-
tion of NF-␬B. ilar to results of previous studies in respiratory epithelial cells dur-
The family of TLRs plays a key role in controlling innate im- ing Pneumocystis infection (51) or Pseudomonas aeruginosa
mune responses. Recent studies revealed a broad and regulated flagella activation (52). We showed previously that C. parvum ac-
expression of TLRs in human tissues. Most tissues express at least tivates the NF-␬B system via degradation of I␬B␣ in directly in-
one TLR, whereas phagocytes and B cells in particular show abun- fected epithelial cells resulting in host-cell secretion of associated
dant and constitutive expression of all known TLRs (33–36). In- gene products (e.g., IL-8) (12). In this study, we found that C.
testinal epithelial cells normally express very low levels of TLRs parvum-induced degradation of I␬B␣ was blocked in cells trans-
such as TLR2 and TLR4 (37, 38). In contrast, TLR5 is expressed fected with TLR2-DN, TLR4-DN, and MyD88-DN. Nuclear trans-
exclusively on the basolateral surface of the intestinal epithelial location of p-65 was inhibited by transfection of cells with TLR2-
cells (39). The characteristic of TLR expression in intestinal epi- DN, TLR4-DN, and MyD88-DN. Activation of NF-␬B by C.
thelial cells, which are consistently exposed to microbiota includ- parvum was also inhibited by specific siRNAs to TLR2, TLR4,
ing commensal bacteria in the intestine, may explain why the com- and MyD88. Consequently, IL-8 release from cholangiocytes upon
mensal bacteria usually do not induce inflammatory responses (38, C. parvum infection was inhibited. Moreover, our data suggest that
40). Although human bile is sterile under physiological conditions, both TLR2 and TLR4 contribute to C. parvum-induced NF-␬B
duodenal microorganisms are believed to be a major source of activation, because neither TLR2-DN nor TLR4-DN alone could
bacterial infection in several biliary diseases. In particular, enteric completely block induced NF-␬B activation and IL-8 release,
bacteria, demonstrable in bile, may be responsible for chronic pro- whereas transfection with a DN mutant of their common adaptor
liferative cholangitis associated with hepatolithiasis (41). Never- protein MyD88 completely inhibited NF-␬B activation. Taken to-
theless, whereas expression of TLR2–5 was recently shown in gether, these data indicate the following: 1) that C. parvum acti-
normal murine cholangiocytes and in several human cholangiocar- vates TLR2- and TLR4-dependent signal pathways in infected
cinoma cell lines (42), expression of TLRs in the normal human cholangiocytes; and 2) that both TLR2 and TLR4 are involved in
liver, particularly in cholangiocytes, has never been systemically activation of the NF-␬B signaling pathway in directly infected
examined. Extensive expression and cellular distribution of mul- cholangiocytes in which the adaptor protein MyD88 plays an es-
tiple TLRs in human cholangiocytes, as suggested by our results, sential role. Whether host-cell endosomes and intracellular pattern
indicated that TLRs may play a key role in cholangiocyte innate recognition receptors, such as nucleotide-binding oligomerization
immune responses. These TLRs participate in a complex pattern- domain 1 (NOD1) and NOD2 (53, 54), also play a role in host-cell
recognition system (15) and, therefore, provide cholangiocytes the activation by C. parvum is unclear. Whether C. parvum expresses
The Journal of Immunology 7455

TLR ligands such as LPS and peptidoglycans (55) and why direct 3. Guerrant, R. L. 1997. Cryptosporidiosis: an emerging, highly infectious threat.
Emerg. Infect. Dis. 3: 51–57.
parasite-host-cell interactions are required for TLR activation need
4. Wilcox, C. M., and K. E. Monkemuller. 1998. Hepatobiliary diseases in patients
further investigation. with AIDS: focus on AIDS cholangiopathy and gallbladder disease. Dig. Dis. 16:
HBDs are key elements of the innate immune system against 205–213.
invading pathogens in both respiratory and gastrointestinal epithe- 5. Manabe, Y. C., D. P. Clark, R. D. Moore, J. A. Lumadue, H. R. Dahlman,
P. C. Belitsos, R. E. Chaisson, and C. L. Sears. 1998. Cryptosporidiosis in pa-
lial cells (17). Whereas HBD-1 is extensively expressed in these tients with AIDS: correlates of disease and survival. Clin. Infect. Dis. 27:
epithelia under physiological conditions, HBD-2 expression is in- 536 –542.
duced by exposure to microbes or cytokines, such as TNF-␣ (17– 6. Griffiths, J. K. 1998. Treatment for AIDS-associated cryptosporidiosis. J. Infect.
Dis. 178: 915–916.
20). A constitutive expression of HBD-1 and LPS-induced expres- 7. Clark, D. P. 1999. New insights into human cryptosporidiosis. Clin. Microbiol.
sion of HBD-2 has also been reported in human cholangiocytes Rev. 12: 554 –563.
(41). The NF-␬B signaling pathway has been implicated in the 8. McDonald, V., R. Smith, H. Robinson, and G. Bancroft. 2000. Host immune
responses against Cryptosporidium. Contrib. Microbiol. 6: 75–91.
expression of HBD-2 in response to various stimuli (56, 57). 9. Huang, B. Q., X. M. Chen, and N. F. LaRusso. 2004. Cryptosporidium parvum
Whereas TLR signaling-associated HBD-2 expression has been attachment to and internalization by human biliary epithelia in vitro: a morpho-
demonstrated in tracheobronchial and intestinal epithelial cells (21, logic study. J. Parasitol. 90: 212–221.
10. Chen, X. M., B. Q. Huang, P. L. Splinter, H. Cao, G. Zhu, M. A. McNiven, and
22), the role of TLRs in microbial-induced HBD expression in N. F. LaRusso. 2003. Cryptosporidium parvum invasion of biliary epithelia re-
cholangiocytes has not been fully characterized. In this study, we quires host cell tyrosine phosphorylation of cortactin via c-Src. Gastroenterology
provide substantial evidence supporting the notion that C. parvum 125: 216 –228.
11. Deng, M., M. S. Rutherford, and M. S. Abrahamsen. 2004. Host intestinal epi-
induces expression of HBD-2 in cholangiocytes via TLR-mediated thelial response to Cryptosporidium parvum. Adv. Drug Delivery Rev. 56:
NF-␬B activation. First, TLR2 and TLR4 are recruited to the par- 869 – 884.
asite attachment sites. Secondly, TLR2 and TLR4 are associated 12. Chen, X. M., S. A. Levine, P. L. Splinter, P. S. Tietz, A. L. Ganong, C. Jobin,

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


G. J. Gores, C. V. Paya, and N. F. LaRusso. 2001. Cryptosporidium parvum
with activation of the NF-␬B signaling pathway induced by C. activates nuclear factor ␬B in biliary epithelia preventing epithelial cell apoptosis.
parvum, and both nuclear translocation of NF-␬B and expression Gastroenterology 120: 1774 –1783.
of HBD-2 are limited to directly infected cells. More importantly, 13. Laurent, F., L. Eckmann, T. C. Savidge, G. Morgan, C. Theodos, M. Naciri, and
M. F. Kagnoff. 1997. Cryptosporidium parvum infection of human intestinal
DN mutants of TLR2, TLR4, and MyD88, or inhibition of NF-␬B epithelial cells induces the polarized secretion of C-X-C chemokines. Infect. Im-
by specific inhibitors block C. parvum-induced HBD-2 expression. mun. 65: 5067–5073.
Both HBD-1 and HBD-2 have anti-C. parvum activity, as evi- 14. Zaalouk, T. K., M. Bajaj-Elliott, J. T. George, and V. McDonald. 2004. Differ-
ential regulation of ␤-defensin gene expression during Cryptosporidium parvum
denced by a decrease of C. parvum sporozoite viability after in- infection. Infect. Immun. 72: 2772–2779.
cubation with recombinant HBD-1 or HBD-2 (14). Indeed, we 15. Takeda, K., T. Kaisho, and S. Akira. 2003. Toll-like receptors. Annu. Rev. Im-
detected a significantly higher number of parasites in cells trans- munol. 21: 335–376.
16. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nat. Rev. Immunol.
fected with MyD88-DN than in the control cells at 48 –96 h after 4: 499 –511.
initial exposure to an equal number of parasites. Besides HBD-1, 17. Ganz, T. 2003. Defensins: antimicrobial peptides of innate immunity. Nat. Rev.
a constitutive expression of HBD-3 was also detected in both C. Immunol. 3: 710 –720.
18. Yamaguchi, Y., T. Nagase, R. Makita, S. Fukuhara, T. Tomita, T. Tominaga,
parvum-infected and sham-infected cultured cholangiocytes, sug- H. Kurihara, and Y. Ouchi. 2002. Identification of multiple novel epididymis-
gesting that both HBD-1 and HBD-3 may play a general antimi- specific ␤-defensin isoforms in humans and mice. J. Immunol. 169: 2516 –2523.
crobial role in the defense of the biliary system, similar to that 19. O’Neil, D. A., E. M. Porter, D. Elewaut, G. M. Anderson, L. Eckmann, T. Ganz,
and M. F. Kagnoff. 1999. Expression and regulation of the human ␤-defensins
documented in other epithelia (58). In contrast, TLR-induced ex- hBD-1 and hBD-2 in intestinal epithelium. J. Immunol. 163: 6718 – 6724.
pression of HBD-2, as demonstrated by our results, may provide 20. Tsutsumi-Ishii, Y., and I. Nagaoka. 2003. Modulation of human ␤-defensin-2
epithelial cells (e.g., cholangiocytes) a critical local defense re- transcription in pulmonary epithelial cells by lipopolysaccharide-stimulated
mononuclear phagocytes via proinflammatory cytokine production. J. Immunol.
sponse against microbial infection. 170: 4226 – 4236.
In conclusion, our study demonstrated that human cholangio- 21. Hertz, C. J., Q. Wu, E. M. Porter, Y. J. Zhang, K. H. Weismuller, P. J. Godowski,
cytes normally express TLR1–10. Using an in vitro model of bil- T. Ganz, S. H. Randell, and R. L. Modlin. 2003. Activation of Toll-like receptor
2 on human tracheobronchial epithelial cells induces the antimicrobial peptide
iary cryptosporidiosis, we found that C. parvum recruits TLR2 and human ␤ defensin-2. J. Immunol. 171: 6820 – 6826.
TLR4, but not other analyzed TLRs, to the host-cell-parasite in- 22. Vora, P., A. Youdim, L. S. Thomas, M. Fukata, S. Y. Tesfay, K. Lukasek, K,
terface, an event that results in activation of NF-␬B thus triggering S. Michelsen, A. Wada, T. Hirayama, M. Arditi, and M. T. Abreu. 2004. ␤-De-
fensin-2 expression is regulated by TLR signaling in intestinal epithelial cells.
host-cell responses such as cytokine/chemokine release and HBD J. Immunol. 173: 5398 –5405.
expression. Future studies should define the molecular mecha- 23. Chen, X. M., and N. F. LaRusso. 2000. Mechanisms of attachment and internal-
nisms by which C. parvum activates host-cell TLRs and investi- ization of Cryptosporidium parvum to biliary and intestinal epithelial cells. Gas-
troenterology 118: 368 –379.
gate the potential synergistic effects of C. parvum and other patho- 24. Grubman, S. A., R. D. Perrone, D. W. Lee, S. L. Murray, L. C. Rogers,
gen (e.g., HIV-1) infection in the pathogenesis of biliary disease L. I. Wolkoff, A. E. Mulberg, V. Cherington, and D. M. Jefferson. 1994. Regu-
via the TLR pathways. lation of intracellular pH by immortalized human intrahepatic biliary epithelial
cell lines. Am. J. Physiol. 266: G1060 –G1070.
25. Lin, Y. Z., S. Y. Yao, R. A. Veach, T. R. Torgerson, and J. Hawiger. 1995.
Acknowledgments Inhibition of nuclear translocation of transcription factor NF-␬B by a synthetic
We are grateful to Dr. M. Smith for providing the TLR2-DN and peptide containing a cell membrane-permeable motif and nuclear localization
TLR4-DN constructs, Prof. J. Tschopp for the MyD88-DN construct, and sequence. J. Biol. Chem. 270: 14255–14258.
26. Becker, M. N., G. Diamond, M. W. Verghese, and S. H. Randell. 2000. CD14-
Drs. G. Zhu and J. S. Keithly for the antiserum to C. parvum. We thank
dependent lipopolysaccharide-induced ␤-defensin-2 expression in human tra-
Drs. D. K. Podolsky, H. D. Dong, G. J. Gores, and A. Badley for helpful cheobronchial epithelium. J. Biol. Chem. 275: 29731–29736.
and stimulating discussions and D. Hintz for secretarial assistance. 27. Rochelle, P. A., M. M. Marshall, J. R. Mead, A. M. Johnson, D. G. Korich,
J. S. Rosen, and R. De Leon. 2002. Comparison of in vitro cell culture and a
Disclosures mouse assay for measuring infectivity of Cryptosporidium parvum. Appl. Envi-
ron. Microbiol. 68: 3809 –3817.
The authors have no financial conflict of interest.
28. Splinter, P. L., A. I. Masyuk, and N. F. LaRusso. 2003. Specific inhibition of
AQP1 water channels in isolated rat intrahepatic bile duct units by small inter-
References fering RNAs. J. Biol. Chem. 278: 6268 – 6274.
1. O’Donoghue, P. J. 1995. Cryptosporidium and cryptosporidiosis in man and an- 29. Chen, X. M., B. Q. Huang, P. L. Splinter, J. D. Orth, D. D. Billadeau,
imals. Int. J. Parasitol. 25: 139 –195. M. A. McNiven, and N. F. LaRusso. 2004. Cdc42 and the actin-related protein/
2. Chen, X. M., J. S. Keithly, C. V. Paya, and N. F. LaRusso. 2002. Cryptospori- neural Wiskott-Aldrich syndrome protein network mediate cellular invasion by
diosis. N. Engl. J. Med. 346: 1723–1731. Cryptosporidium parvum. Infect. Immun. 72: 3011–3021.
7456 TLRs IN C. parvum INFECTION

30. Chen, X. M., P. L. Splinter, P. S. Tietz, B. Q. Huang, D. D. Billadeau, and efficient control of infection with the protozoan parasite Leishmania major. In-
N. F. LaRusso. 2004. Phosphatidylinositol 3-kinase and frabin mediate Crypto- fect. Immun. 72: 1920 –1928.
sporidium parvum cellular invasion via activation of Cdc42. J. Biol. Chem. 279: 45. De Trez, C., M. Brait, O. Leo, T. Aebischer, F. A. Torrentera, Y. Carlier, and
31671–31678. E. Muraille. 2004. Myd88-dependent in vivo maturation of splenic dendritic cells
31. Guillot, L., S. Medjane, K. Le-Barillec, V. Balloy, C. Danel, M. Chignard, and induced by Leishmania donovani and other Leishmania species. Infect. Immun.
M. Si-Tahar. 2004. Response of human pulmonary epithelial cells to lipopoly- 72: 824 – 832.
saccharide involves Toll-like receptor 4 (TLR4)-dependent signaling pathways: 46. Campos, M. A., M. Closel, E. P. Valente, J. E. Cardoso, S. Akira,
evidence for an intracellular compartmentalization of TLR4. J. Biol. Chem. 279: J. I. Alvarez-Leite, C. Ropert, and R. T. Gazzinelli. 2004. Impaired production of
2712–2718. proinflammatory cytokines and host resistance to acute infection with Trypano-
32. Roebuck, K. A. 1999. Regulation of interleukin-8 gene expression. J. Interferon soma cruzi in mice lacking functional myeloid differentiation factor 88. J. Im-
Cytokine Res. 19: 429 – 438. munol. 172: 1711–1718.
33. Zarember, K. A., and P. J. Godowski. 2002. Tissue expression of human Toll-like 47. Forney, J. R., D. B. DeWald, S. Yang, C. A. Speer, and M. C. Healey. 1999. A
receptors and differential regulation of Toll-like receptor mRNAs in leukocytes in role for host phosphoinositide 3-kinase and cytoskeletal remodeling during Cryp-
response to microbes, their products, and cytokines. J. Immunol. 168: 554 –561. tosporidium parvum infection. Infect. Immun. 67: 844 – 852.
34. Iwasaki, A., and R. Medzhitov. 2004. Toll-like receptor control of the adaptive 48. Elliott, D. A., and D. P. Clark. 2000. Cryptosporidium parvum induces host cell
immune responses. Nat. Immunol. 5: 987–995. actin accumulation at the host-parasite interface. Infect. Immun. 68: 2315–2322.
35. Bourke, E., D. Bosisio, J. Golay, N. Polentarutti, and A. Mantovani. 2003. The 49. Elliott, D. A., D. J. Coleman, M. A. Lane, R. C. May, L. M. Machesky, and
Toll-like receptor repertoire of human B lymphocytes: inducible and selective D. P. Clark. 2001. Cryptosporidium parvum infection requires host cell actin
expression of TLR9 and TLR10 in normal and transformed cells. Blood 102: polymerization. Infect. Immun. 69: 5940 –5942.
956 –963. 50. Frendeus, B., C. Wachtler, M. Hedlund, H. Fischer, P. Samuelsson, M. Svensson,
36. Hornung, V., S. Rothenfusser, S. Britsch, A. Krug, B. Jahrsdorfer, T. Giese, and C. Svanborg. 2001. Escherichia coli P fimbriae utilize the Toll-like receptor
S. Endres, and G. Hartmann. 2002. Quantitative expression of Toll-like receptor 4 pathway for cell activation. Mol. Microbiol. 40: 37–51.
1–10 mRNA in cellular subsets of human peripheral blood mononuclear cells and 51. Lebron, F., R. Vassallo, V. Puri, and A. H. Limper. 2003. Pneumocystis carinii
sensitivity to CpG oligodeoxynucleotides. J. Immunol. 168: 4531– 4537. cell wall ␤-glucans initiate macrophage inflammatory responses through NF-␬B
activation. J. Biol. Chem. 278: 25001–25008.
37. Cario, E., and D. K. Podolsky. 2000. Differential alteration in intestinal epithelial
52. Adamo, R., S. Sokol, G. Soong, M. I. Gomez, and A. Prince. 2004. Pseudomonas
cell expression of Toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel
aeruginosa flagella activate airway epithelial cells through asialoGM1 and Toll-
disease. Infect. Immun. 68: 7010 –7017.

Downloaded from http://www.jimmunol.org/ by guest on June 9, 2017


like receptor 2 as well as Toll-like receptor 5. Am. J. Respir. Cell Mol. Biol. 30:
38. Melmed, G., L. S. Thomas, N. Lee, S. Y. Tesfay, K. Lukasek, K. S. Michelsen, 627– 634.
Y. Zhou, B. Hu, M. Arditi, and M. T. Abreu. 2003. Human intestinal epithelial 53. Kim, J. G., S. J. Lee, and M. F. Kagnoff. 2004. Nod1 is an essential signal
cells are broadly unresponsive to Toll-like receptor 2-dependent bacterial ligands: transducer in intestinal epithelial cells infected with bacteria that avoid recogni-
implications for host-microbial interactions in the gut. J. Immunol. 170: tion by Toll-like receptors. Infect. Immun. 72: 1487–1495.
1406 –1415. 54. Barnich, N., J. E. Aguirre, H. C. Reinecker, R. Xavier, and D. K. Podolsky. 2005.
39. Gewirtz, A. T., T. A. Navas, S. Lyons, P. J. Godowski, and J. L. Madara. 2001. Membrane recruitment of NOD2 in intestinal epithelial cells is essential for nu-
Cutting edge: bacterial flagellin activates basolaterally expressed TLR5 to induce clear factor-␬B activation in muramyl dipeptide recognition. J. Cell Biol. 170:
epithelial proinflammatory gene expression. J. Immunol. 167: 1882–1885. 21–26.
40. Noverr, M. C., and G. B. Huffnagle. 2004. Does the microbiota regulate immune 55. Schwandner, R., R. Dziarski, H. Wesche, M. Rothe, and C. J. Kirschning. 1999.
responses outside the gut? Trends Microbiol. 12: 562–568. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by Toll-
41. Harada, K., K. Ohba, S. Ozaki, K. Isse, T. Hirayama, A. Wada, and like receptor 2. J. Biol. Chem. 274: 17406 –17409.
Y. Nakanuma. 2004. Peptide antibiotic human ␤-defensin-1 and -2 contribute to 56. Wehkamp, J., J. Harder, K. Wehkamp, B. Wehkamp-von Meissner, M. Schlee,
antimicrobial defense of the intrahepatic biliary tree. Hepatology 40: 925–932. C. Enders, U. Sonnenborn, S. Nuding, S. Bengmark, K. Fellermann, et al. 2004.
42. Harada, K., S. Ohira, K. Isse, S. Ozaki, Y. Zen, Y. Sato, and Y. Nakanuma. 2003. NF-␬B- and AP-1-mediated induction of human ␤ defensin-2 in intestinal epi-
Lipopolysaccharide activates nuclear factor-␬B through Toll-like receptors and thelial cells by Escherichia coli Nissle 1917: a novel effect of a probiotic bac-
related molecules in cultured biliary epithelial cells. Lab. Invest. 83: 1657–1667. terium. Infect. Immun. 72: 5750 –5758.
43. Pichyangkul, S., K. Yongvanitchit, U. Kum-arb, H. Hemmi, S. Akira, 57. Kao, C. Y., Y. Chen, P. Thai, S. Wachi, F. Huang, C. Kim, R. W. Harper, and
A. M. Krieg, D. G. Heppner, V. A. Stewart, H. Hasegawa, S. Looareesuwan, et R. Wu. 2004. IL-17 markedly up-regulates ␤-defensin-2 expression in human
al. 2004. Malaria blood stage parasites activate human plasmacytoid dendritic airway epithelium via JAK and NF-␬B signaling pathways. J. Immunol. 173:
cells and murine dendritic cells through a Toll-like receptor 9-dependent path- 3482–3491.
way. J. Immunol. 172: 4926 – 4933. 58. Dunsche, A., Y. Acil, H. Dommisch, R. Siebert, J. M. Schroder, and S. Jepsen.
44. Kropf, P., M. A. Freudenberg, M. Modolell, H. P. Price, S. Herath, S. Antoniazi, 2002. The novel human ␤-defensin-3 is widely expressed in oral tissues. Eur.
C. Galanos, D. F. Smith, and I. Muller. 2004. Toll-like receptor 4 contributes to J. Oral Sci. 110: 121–124.

You might also like