Skip to main content
Advertisement
  • Loading metrics

Characterization of the Giardia intestinalis secretome during interaction with human intestinal epithelial cells: The impact on host cells

  • Showgy Y. Ma’ayeh,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Project administration, Validation, Visualization, Writing – original draft

    Affiliation Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden

  • Jingyi Liu,

    Roles Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden

  • Dimitra Peirasmaki,

    Roles Data curation, Formal analysis, Methodology, Validation

    Affiliation Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden

  • Katarina Hörnaeus,

    Roles Data curation, Formal analysis, Methodology, Validation, Writing – original draft

    Affiliation Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden

  • Sara Bergström Lind,

    Roles Data curation, Funding acquisition, Investigation, Methodology

    Affiliation Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden

  • Manfred Grabherr,

    Roles Data curation, Formal analysis, Investigation, Methodology, Validation, Writing – original draft

    Affiliation Department of Medical Biochemsitry and Microbiology, BMC, Uppsala, Sweden

  • Jonas Bergquist,

    Roles Data curation, Formal analysis, Investigation, Methodology

    Affiliation Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden

  • Staffan G. Svärd

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Writing – original draft

    staffan.svard@icm.uu.se

    Affiliation Department of Cell and Molecular Biology, Uppsala University, BMC, Uppsala, Sweden

Abstract

Background

Giardia intestinalis is a non-invasive protozoan parasite that causes giardiasis in humans, the most common form of parasite-induced diarrhea. Disease mechanisms are not completely defined and very few virulence factors are known.

Methodology

To identify putative virulence factors and elucidate mechanistic pathways leading to disease, we have used proteomics to identify the major excretory-secretory products (ESPs) when Giardia trophozoites of WB and GS isolates (assemblages A and B, respectively) interact with intestinal epithelial cells (IECs) in vitro.

Findings

The main parts of the IEC and parasite secretomes are constitutively released proteins, the majority of which are associated with metabolism but several proteins are released in response to their interaction (87 and 41 WB and GS proteins, respectively, 76 and 45 human proteins in response to the respective isolates). In parasitized IECs, the secretome profile indicated effects on the cell actin cytoskeleton and the induction of immune responses whereas that of Giardia showed anti-oxidation, proteolysis (protease-associated) and induction of encystation responses. The Giardia secretome also contained immunodominant and glycosylated proteins as well as new candidate virulence factors and assemblage-specific differences were identified. A minor part of Giardia ESPs had signal peptides (29% for both isolates) and extracellular vesicles were detected in the ESPs fractions, suggesting alternative secretory pathways. Microscopic analyses showed ESPs binding to IECs and partial internalization. Parasite ESPs reduced ERK1/2 and P38 phosphorylation and NF-κB nuclear translocation. Giardia ESPs altered gene expression in IECs, with a transcriptional profile indicating recruitment of immune cells via chemokines, disturbances in glucose homeostasis, cholesterol and lipid metabolism, cell cycle and induction of apoptosis.

Conclusions

This is the first study identifying Giardia ESPs and evaluating their effects on IECs. It highlights the importance of host and parasite ESPs during interactions and reveals the intricate cellular responses that can explain disease mechanisms and attenuated inflammatory responses during giardiasis.

Author summary

Giardia intestinalis is a major contributor to the enormous burden of diarrheal diseases with 280 million symptomatic human infections (giardiasis) per year, mainly in children. Nonetheless, there is poor insight into how Giardia causes disease; it is not invasive and very few virulence factors are known. Here we use proteomics, large-scale protein identification, to identify proteins released by the parasite and host cells when they interact in vitro. Our analyses show that both cell types respond to interaction by the release of specific proteins. These proteins affect the metabolism of host cells and parasites, and many are involved in the modification of host’s immune responses. The secreted parasite proteins bind to host cells and some are internalized, resulting in the modulation of immune-signaling proteins, cell cycle arrest and the induction of cell death. This study provides new insights into host-parasite interactions during giardiasis and this will be the basis for further studies of specific proteins that might explain disease induction and symptoms during giardiasis.

Introduction

Giardia intestinalis is an intestinal protozoan parasite causing 280 million human diarrheal cases (giardiasis) annually [1]. Giardia has two life-cycle stages: infectious cysts and disease-causing trophozoites [2]. Trophozoites move by flagella and adhere to intestinal epithelial cells (IECs) strongly using a suction cup on the ventral surface [2]. The direct contact of Giardia with IECs induces cellular damage on the apical surface, compromising brush border enzymes function, and affect cellular junctions, leading to an increased intestinal barrier permeability [3]. These changes cause maldigestion, malabsorption and electrolyte imbalance, collectively culminating in causing diarrhea [4]. Symptoms are usually acute but chronicity is not uncommon and some patients develop intestinal and extra-intestinal complications post-infection [1,5]. Many Giardia infections, however, remain asymptomatic and the reason for variable disease outcomes remains obscure.

Early studies of Giardia pathogenicity suggested that the parasite releases proteins that can contribute to disease induction. Excretory-secretory products (ESPs) of Giardia affect intestinal absorption and secretion [6], cause histopathological alterations [7] and induce specific antibodies against glycoproteins [8]. A secreted 58 kDa glycoprotein has been shown to react with antibodies from giardiasis patients and induce fluid accumulation in ligated rabbit ileal loop experiments [9,10]. Nevertheless, the identity of this protein remained unknown as tryptic digests did not align with any proteins in the Giardia Database. Cysteine proteases (CPs) are also yet unidentified Giardia ESPs. Their involvement in virulence, however, is well studied, demonstrated by their association with parasite attachment [11], and the degradation of extracellular matrix (ECM) proteins [12], antibodies [13] and proinflammatory cytokines [14,15]. Arginine metabolising enzymes (AMEs) (e.g. arginine deiminase (ADI) and ornithine carbamoyltransferase (OCT)) are the best-studied examples of secreted virulence factors in Giardia. AMEs outcompete host cells for arginine uptake, inhibiting cellular proliferation [16], abrogating nitric oxide production, a compound cytotoxic to Giardia, [17], and inhibiting cytokine production by dendritic cells [18]. Other reports documented the secretion of elongation factor-1 alpha (EF-1α) [19] and the glycolytic enzyme enolase [17] into Giardia growth medium and the medium of interaction with IECs in vitro, respectively. Their functions in parasite virulence, if any, is still unknown. Finally, variable surface proteins (VSPs) are cysteine-rich proteins that cover trophozoite surface and they are released in large quantities into the growth medium (≈70% within 24h) [20]. VSPs are immunodominant during infection [21] and their constant switching (i.e. antigenic variation) help the parasite evade the host immune system [22].

In view of the simple endomembrane system of Giardia, which lacks a true Golgi apparatus, peroxisomes and endosomal/lysosomal system [2], the secretion of proteins, their sorting and trafficking remain a perplexing aspect of the parasite’s biology. Most of our knowledge about secretion and secreted proteins of Giardia derive from experiments targeting the sorting and trafficking of VSPs to the plasma membrane and cyst-wall proteins in encystation-specific vesicles (ESVs) to the cyst wall during encystation [23]. AMEs, EF-1α and enolase all lack signal peptides (SPPs) but they are still secreted by the parasite. This suggests the presence of non-conventional secretory mechanisms in Giardia such as extracellular vesicles (EVs) carrying protein cargo with no SSP. EVs include the exosomes (30–100 nm) and microvesicles (MVs) (100–1000 nm), which have been shown to play roles in parasites communication, host cell colonization and immune modulation [24,25]. A recent study has identified MVs released from Giardia trophozoites and encysting cells [26]. The released MVs affected trophozoite attachment and dendritic cell activation [26].

In this study, we used proteomics to identify the secretome of the Giardia reference isolates WB and GS during growth and interaction with IECs in vitro in serum-free medium. We also show the release of EVs by Giardia as a part of its secretome. We demonstrate that Giardia ESPs alter gene expression in IECs, cell signalling and the production of proinflammatory cytokines. This study highlights the importance of Giardia ESPs in inducing pathological effects in IECs and suggests how this might have immunomodulatory effects on host immune responses.

Materials and methods

Ethics statement and chemicals

The cell lines used in this study were all obtained from American Type Culture Collection (ATCC): Giardia intestinalis isolates WB clone C6 (ATCC30957; assemblage A) and GS, clone H7 (ATCC50581; assemblage B) were used in this study in combination with the human colonic adenocarcinoma cell line, Caco-2 clone TC7 (ATCC HTB-37). All the experiments were approved by the Programme Review Board at Department of Cell and Molecular Biology, Uppsala University, Sweden. All chemicals were purchased from Sigma-Aldrich (MO, USA), unless otherwise stated.

Parasites and intestinal epithelial cells (IECs)

Giardia trophozoites were cultured in 10 ml and 50 ml tubes filled with TYDK medium (37°C) supplemented with 10% heat inactivated bovine serum (Sigma-Aldrich) as described in [27]. The Caco-2 clone TC7 (passage numbers 7–12) cell line was grown and differentiated as previously described [16].

Medium for collection of excretory/secretory products (ESPs) and trophozoite viability

To collect Giardia ESPs from axenic cultures we used a modified RPMI-1640 medium containing 55.5 mM glucose, 22.8 mM L-arginine, 11.4 mM L-cysteine hydrochloride monohydrate, 11.4 mM ascorbic acid, 2mM Glutamax (Gibco), 1 mM sodium pyruvate (Gibco) and 1x MEM essential amino acids. The medium was filter-sterilized and placed in a water bath (final pH is 6.85 at 37°C) until used.

Confluent Giardia culture tubes, prepared as described above, were used for trophozoite viability assessment. Briefly, the TYDK medium was decanted from each tube followed by three washes in Hank’s Balanced Salt Solution with glucose (HBSS-G) supplemented with 11.4 mM L-cysteine (pH 6.8, 37°C). Culture tubes were replenished with the modified medium, closed tightly and incubated for 2h or 6h (37°C). At the end of each incubation, tubes were placed on ice to detach trophozoites (10 min), followed by counting using the expulsion of Trypan Blue and flagellar motility as viability criteria.

Collection and processing of ESPs from axenic cultures and host parasite interactions

Tubes with Giardia trophozoites in modified RPMI-1640 medium (50 ml, two tubes of each isolate) were used to collect Giardia ESPs in axenic cultures. The tubes and media, following counting and viability assessment, were centrifuged (930 xg, 10 min, 4°C) to pellet trophozoites. Collected media were filter-sterilized (0.22 μM) and treated with protease inhibitor cocktail tablets (cOmplete, Mini EDTA-free, Roche, Sigma-Aldrich). Media were concentrated down to 200–300 μl using the Amicon Ultra 15 mL centrifugal filters with 3kDa cutoff (Merck-Millipore, Darmstadt, Germany). To further eliminate any traces of residual serum, concentrates were processed through the AlbuminOUT kit (G-Biosciences, MO, USA) according to the manufacturer’s instructions. Three biological replicates (2h and 6h) were prepared and stored at -80°C until used for MS analysis. The whole experimental setup is described in Fig 1A.

thumbnail
Fig 1. Experimental setup for proteomic analyses and characterization of Giardia ESPs.

(A) The collection of Giardia intestinalis, isolates WB and GS, excretory-secretory products (ESPs) in a serum-free medium from axenic culture and co-culture with differentiated intestinal epithelial cells (IECs), Caco-2, for 2h and 6h. Samples were processed as described in Materials and Methods followed by liquid chromatography/mass spectrometry to identify secreted proteins. (B) Studies of IECs exposed to G. intestinalis ESPs using differential gene expression analyses by RNA sequencing, quantitative real time PCR, Western blots, ELISA and confocal microscopy. The panel illustrates the experimental setup (left to right) showing IECs exposed to ESPs of either Giardia isolates through a Transwell insert (direct exposure) or different amounts of ESPs collected from axenic culture (i.e. the absence of IECs, 1 μg, 5 μg or 10 μg per ml of medium). Abbreviation: Dulbecco’s Modified Eagles Medium (DMEM).

https://doi.org/10.1371/journal.pntd.0006120.g001

Differentiated Caco-2 cells (75 cm2 flasks) were washed with warm PBS (3x) and incubated with 6 x 107 trophozoites resuspended in 25 ml of DMEM (no FBS) for 2h or 6h. Trophozoites were washed (3x, HBSS-G plus 11.4 mM L-cysteine) prior addition to cells. Caco-2 cells treated as above and incubated alone in DMEM (2h and 6h) served as controls (Fig 1A). All incubations were performed in 10% CO2 humidified tissue culture incubator at 37°C. At the end of each interaction, flasks were placed on ice (10 min) to detach trophozoites for counting and viability assessment. Media were then collected, processed as described earlier and stored at -80°C until used for MS analysis. Three biological replicates were prepared (2h or 6h including controls).

Isolation of Giardia extracellular vesicles (EVs) and electron microscopy

The ESP enrichment experiment for axenic cultures described earlier was repeated to collect Giardia EVs. Samples were processed with the ExoQuick TC Kit (SBI System Biosciences, CA, USA) according to the manufacturer’s instructions. Giardia EVs were pelleted by centrifugation (1500 xg, 30 min) and resuspended in 100 μl PBS buffer and processed immediately for electron microscopy (EM). Samples (5μl) were adsorbed onto a Formvar-coated 200 mesh grid (15 seconds), followed by treatment with 2% molybdenum (10 seconds) and contrasting using 2% uranyl acetate (10 seconds). Grids were dried and visualized using a transmission electron microscope at the BioVis Core Facility, Uppsala University, Uppsala, Sweden. A negative control (media alone) was included and processed similarly.

Liquid chromatography and mass spectrometry (LC/MS) analysis

Before digestion, the buffer in the collected samples was exchanged to a urea-containing lysis buffer (20 mM HEPES, 9 M urea, Complete Mini EDTA-free protease inhibitor cocktail (Roche) using 3 kDa molecular weight cut off filters (Thermo Fisher Scientific, MA, USA). Proteins were then subjected to dithiothreitol (DTT, 12.5 mM) reduction and iodoacetamide (IAA, 6.9 mM) alkylation before digested in-solution with trypsin (1 μg added per sample). The samples were desalted using Pierce C18 Spin Columns (Thermo Fisher Scientific). Eluates were dried and resolved in 15 μL 0.1% formic acid of which 5 μL were injected. The nanoLC-MS/MS experiments were performed using a Q Exactive Orbitrap mass spectrometer (ThermoFisher Scientific, Bremen, Germany) equipped with a nano-electrospray ion source. The peptides were separated by C18 reversed phase liquid chromatography using an EASY-nLC 1000 system (Thermo Fisher Scientific). A set-up of pre-column and analytical column was used. The pre-column was a 2 cm EASYcolumn (ID 100 μm, 5 μm particles) (Thermo Fisher Scientific) while the analytical column was a 10 cm EASY-column (ID 75 μm, 3 μm particles, Thermo Fisher Scientific). Peptides were eluted with a 150-min linear gradient from 4% to 100% acetonitrile at 250 nL min-1. The mass spectrometer was operated in positive ion mode acquiring a survey mass spectrum with resolving power 70,000 (full width half maximum), m/z 400–1750 using an automatic gain control (AGC) target of 3×106. The 10 most intense ions were selected for higher-energy collisional dissociation (HCD) fragmentation (25% normalized collision energy) and MS/MS spectra were generated with an AGC target of 5×105 at a resolution of 17,500. Mass spectrometer worked in data-dependent mode.

Database searches were performed using the Sequest algorithm, embedded in Proteome Discoverer 1.4 (Thermo Fisher Scientific). Proteins in axenic cultures were identified by performing separate searches against the fasta files of the Giardia proteome for each isolate (GS clone containing 4470 entries and WB clone 9667 entries) (available at www.giardiadb.org; giardiadb.org/common/downloads/release5.0/). The fasta file for human proteins was downloaded from Uniprot 2015–05 and contained 20192 entries. Proteins in co-cultures were searched against a combined database of Giardia and human proteins in a MudPIT search combining all three replicates. For both searches, the following parameters were used for data processing: Maximum 10 ppm and 0.02 Da error tolerances for the survey scan and MS/MS analysis, respectively, trypsin as digesting enzyme, carbamidomethylation of cysteins as fixed modifications, oxidation of methionine and deamidation of asparagine and glutamine as variable modifications, maximum of two miss cleavages sites. The target decoy PSM validator was used to calculate false discovery rate. The search criteria for protein identification were set to at least two matching peptides of 95% confidence level per protein.

Identified proteins were subject to gene ontology (GO) enrichment analysis to identify their biological as well as proteins class. The analysis was performed using the PANTHER algorithms provided within the Gene Ontology Consortium website (http://geneontology.org/page/go-enrichment-analysis). The results were processed through the GO slim option for biological GO terms, with Bonferroni correction applied for multiple testing (P < 0.05). In addition, the STRING Database (http://string-db.org/) was also used to search for InterPro domains, applying Bonferroni correction at P < 0.05 as above. Identified proteins were also analyzed for the presence of secretion signal peptides (SSPs) using the SignalP program embedded within the GiardiaDB (http://giardiadb.org/giardiadb/queries_tools.jsp). The search criteria were set to include any of the following; a SignalP-NN conclusion score of 3, SignalP-NN D-score of 0.5 and SignalP-HMM signal probability of 0.5.

Differential gene transcription of differentiated Caco-2 cells in response to Giardia ESPs

We used RNA sequencing (RNA Seq) or quantitative PCR (qPCR) to identify differential gene expression on the RNA level in IECs (i.e. differentiated Caco-2 cells) exposed to Giardia ESPs in two different experimental setups (Fig 1B). First, IECs (6-well plate) were exposed to parasite ESPs via a Transwell insert (0.4 μm pore polyester membrane, Corning, Sigma Aldrich). Here, IECs were physically separated from parasite but directly exposed to their ESPs. For this experiment, we used RNA Seq to identify global changes in gene expression. Second, IECs were exposed to 1, 5 or 10 μg/ml of ESPs from axenic cultures. This aimed at assessing changes in pro-inflammatory gene expression in IECs exposed to ESPs released in axenic culture (i.e. no IECs) and assess how IECs respond to different amounts of Giardia ESPs. Changes of gene expression were studied using qPCR. An illustration of the experiments above is depicted in Fig 1B.

The experiments below were performed using complete DMEM with 10% HI-FBS and all incubations were performed in a 10% CO2 humidified incubator at 37°C. Before the beginning of all experiments, tissue culture flasks or well-plates were washed twice with warm PBS and incubated with fresh media for 2h. Trophozoite cultures (50 ml) were washed once with warm PBS then cold PBS, incubated on ice (10 min), counted and pelleted by centrifugation (930 xg, 10 min, 4°C). Media collected in the insert experiment were centrifuged (930 xg, 10 min, 4°C), filter-sterilized (0.45 μM) and stored at -20°C to be used later for cytokines measurement. Three biological replicates were analyzed with RNA Seq for the insert experiment, whereas four biological replicates were used for qPCR in the other experiment. Each biological replicate had a control (IECs alone in media) and IECs exposed to ESPs of either WB or GS isolate. For RNA collection, IECs were lyzed directly in the tissue culture flasks or well-plates using Trizol reagent (Ambion, Thermo Fisher Scientific) and stored at -80°C until processed later.

In the insert experiment, IECs were washed and replenished with 4 ml of fresh DMEM added directly into the well. Trophozoites were processed as stated above and resuspended in DMEM, from which 2 ml (1 x 107 trophozoites) were added into the Transwell inserts and incubated for 2h or 6h. Media were collected at the end of each time point. In the second experiment, IECs (25 cm2 flasks) were washed and replenished with 10 ml of fresh DMEM. The amounts of protein in ESPs from either the WB and GS isolates were quantified using the Qubit protein kit (Thermo Fisher Scientific) and added to flasks at a concentration of 1, 5 or 10 μg/ml followed by incubation for 2 h or 6h.

Enzyme linked immunosorbent assay (ELISA) for cytokines in collected media

Media collected from Caco-2-trophozoite interactions were analyzed by ELISA to measure the concentration of cytokines released by IECs in response to parasite ESPs. The Human CXCL8/IL-8 Quantikine ELISA Kit, Human CCL20/MIP-3 alpha Quantikine ELISA Kit and Human CXCL1/GRO alpha Quantikine ELISA Kit were purchased from R&D Signalling and the samples were processed for cytokine measurements following the manufacturer’s recommendations. The results were plotted and analyzed using a four Parameter Logistic ELISA curve fitting provided at http://www.elisaanalysis.com.

RNA extraction, library preparation and RNA sequencing

Samples collected as described earlier were processed using the PureLink RNA Mini Kit (Ambion, Thermo Fisher scientific) according to the manufacturer’s instruction. During RNA extraction, a DNaseI treatment step (PureLink DNase Set, Ambion, Thermo Fisher Scientific) was performed on the spin columns to remove genomic DNA. The quality of extracted RNA was assessed by checking the 260/280 and 260/230 ratios using a NanoDrop 1000 Spectrometer (Thermo Fisher Scientific) and by running samples (500 ng) on a 1.5% Tris-Borate-EDTA (TBE) agarose gel prepared with 20 mM of guanidium isothiocyanate (GITC). For cDNA library preparation and RNA Sequencing, RNA (50 ng each) from each sample was reverse transcribed according to the instructions provided in the Ion AmpliSeq Transcriptome Human Gene Expression kit (Revision A.0, Thermo Fisher Scientific) and the generated cDNA was amplified using the Ion AmpliSeq Transcriptome Human Gene Expression core panel (Thermo Fisher Scientific). Primer sequences were partially digested followed by adaptors ligation (Ion P1 Adapter and Ion Xpress Barcode Adapter, Thermo Fisher Scientific) and purification using the Agencourt AMPure XP reagent (Beckman Coulter Inc., CA, USA). Adaptor-ligated amplicons were eluted into the amplification mix (Platinum PCR SuperMix High Fidelity and Library Amplification Primer Mix, Thermo Fisher Scientific) and amplified. Amplicons were subject to size-selection and purification using Agencourt AMPure XP reagent (Beckman Coulter) and were quantified using the Fragment Analyzer instrument (Advanced Analytical Technologies Inc., Ankeny, IA, USA) with DNF-474 High Sensitivity NGS Fragment Analysis Kit (Advanced Analytical Technologies Inc.). Samples were then pooled, followed by emulsion PCR on the Ion OneTouch 2 system with Ion PI Template OT2 200 Kit v3 (Thermo Fisher Scientific) and enrichment using the Ion OneTouch ES (Thermo Fisher Scientific). Samples were loaded on an Ion PI chip Kit v3 and sequenced in the Ion Proton System using the Ion PI Sequencing 200 Kit v3 (Thermo Fisher Scientific).

Bioinformatic analysis of RNA sequencing data

Acquired reads from RNA Seq were analyzed using the hg19 AmpliSeq RNA plugin in the Torrent Suite Server with default settings. This analysis tool produces tables with raw read counts as well as normalized expression values for all genes in the Ion AmpliSeq Human Transcriptome panel. For each experiment, the RPKM values were normalized across samples using the moose2 software (https://github.com/grabherr/moose2). Differential expression was estimated based on Laplace/Normal distributions calculated from replicate samples, computing individual significance values as with ri and rj being the RPKM values, for up-and down-regulation respectively for all given pairwise comparisons i and j, applying a correction term c based on a normal distribution of read counts. The expectations and for comparing all replicates from condition k with l testing for up- or down-regulation in time point k were estimated as for all and n being the number of values, and Bonferroni-corrected to account for multiple testing. For reporting significance, a threshold of E < 0.00001 was applied.

Genes with significant change in the level of transcription were also subject to GO Terms analysis (http://geneontology.org/page/go-enrichment-analysis) to identify biological or molecular functions as well as protein class. We used the GO slim option with Bonferroni correction applied for multiple testing (P < 0.05).

Quantitative polymerase chain reaction (qPCR)

For qPCR experiments, RNA was extracted as above and cDNA was synthesized from 1μg RNA using the RevertAid H Minus First Strand cDNA Synthesis Kit (Thermo Fisher Scientific) following the manufacturer’s instructions. qPCR was performed on the genes, il-8, cxcl1-3, ccl2 and ccl20 with gapdh used as an endogenous control. The Maxima SYBR Green/ROX qPCR Master Mix (Thermo Fisher Scientific) was used in the qPCR reactions with 250 nM of each primer and 10 ng template. Cycling conditions included an initial polymerase heat activation step (10 min, 95°C), and 40 cycles of 15 sec denaturation (95°C), 30 sec annealing (60°C) and 30 sec extension (72°C). The change in gene transcription was calculated using the ΔΔCt method.

Activator protein 1 (AP-1) reporter plasmid luciferase assay

AP-1 is a transcription factor involved in inducing the transcription of inflammatory genes [28,29]. It is activated by MAPKs and could work in conjunction with NFκB to activate gene transcription [28,29]. To detect AP-1 activation in response to Giardia ESPs, Caco-2 cells were transfected with a plasmid pGL4.44[luc2P/AP1 RE/Hygro] expressing the luciferase reporter gene luc2P (Photinus pyralis) under the control of a promoter that has the transcription response element of AP-1. The pGL4.75 Vector (encoding Renilla luciferase) was used as a normalisation control and was co-transfected into Caco-2 cells with the pGL4.44 plasmid. Both plasmids were purchased from Promega (WI, USA). Transfections were performed in a 24-wells plate format according to the supplier’s protocol, using 1.1μg of plasmid DNA/105 cells and the FuGENE HD reagent (Promega). In the beginning of experiment, both parasite and Caco-2 cells were washed and processed as described earlier and the wells were replenished with 500 μl DMEM (no phenol red as it interferes with luminescence reading). Trophozoites were re-suspended in DMEM (no phenol red) and 106 trophozoites were added into the Transwell insert in a total volume of 100 μl and incubated for 2h or 6h (37°C, 10% CO2). Three wells were used for each time point and the controls. At the end of each incubation, the inserts were removed and the cells were washed (3x) with 200 μl PBS at RT, and lyzed with an equal amount of the Dual-Glo Luciferase Assay reagent. A 100 μl was dispensed into the well of 96-well plate in triplicates and luminescence was read from both plasmids following the instructions of Dual-Glo Luciferase Assay reagent and using an Infinite M200 Pro Tecan machine (Tecan Group Ltd., Männedorf, Switzerland). Cells with no plasmid (background fluorescence), cells with plasmid alone (negative control), and cells incubated with Phorbol 12-myristate 13-acetate (PMA) (positive control) served as controls. Three biological replicates were performed in this experiment.

Western blot analyses

To correlate RNA Seq results with those at protein level, we studied the effects of Giardia ESPs on differentiated Caco-2 cells using Western blotting. We tested for the activation/inhibition of mitogen activated protein kinases (MAPKs) and the nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), both of which are involved in inflammatory gene transcription [30,31]. Briefly, the insert experiment was repeated as described earlier with the inclusion of three washes with warm PBS (37°C) at the end of each incubation. In the last wash, cells were scraped off and pelleted by centrifugation (930 xg, 10 min, 4°C) and the pellet was lyzed using the NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Fisher Scientific) according to supplier’s instructions. Both Halt Protease and Phosphatase Inhibitor Cocktails (Thermo Fisher Scientific) were added to lysis buffers during extractions. Cytoplasmic and nuclear fractions were aliquoted and stored at -80°C until later use. To confirm that Giardia ESPs can modulate/inhibit immune signaling in IECs, we repeated the experiment above but IECs were previously treated with lipopolysaccharide (LPS, Sigma-Aldrich) (10 μg/ml) or tumour necrosis factor-alpha (TNF-α, Sigma-Aldrich) (100 ng/ml) for 2h prior adding trophozoites into the inserts. Trophozoites in inserts and IECs, in the presence of inflammatory signal, were left to interact for 2h and 6h, upon which IECs were processed as above for protein collection. IECs treated with LPS and TNF-α were tested for MAPKs activation/inhibition and the nuclear translocation of NF-κB.

Protein samples collected as described above were used for SDS-PAGE and Western blots. Briefly, 30 μg of total proteins were electrophoresed on AnykD-stain-free gels (Bio-Rad) (100 v, 4°C) and transferred onto a PVDF membrane (100 v, 90 min, 4°C). Membranes were washed with Tris-Buffered Saline with 0.1% Tween-20 (TBST) and blocked with 5% non-fat dry milk in TBST for 1h at RT, followed by incubation with primary antibodies overnight (4°C). Blots were then washed with TBST (3x) and incubated for 1h at RT with a secondary anti-mouse HRP conjugated or anti-rabbit HRP conjugated antibodies (1:5000 dilution, Cell Signaling Technology, MA, USA). Blots were developed using the Clarity ECL Western Blotting Substrate (Bio-Rad). For re-probing, blots were stripped using a mild (200 mM glycine, 0.1% w/v SDS, 1% Tween 20, pH 2.2) or harsh stripping buffer (2% SDS, 62.5 mM Tris, pH 6.8, and 114.4 mM beta-mercaptoethanol) and probed for the loading control, which is either an anti-human GAPDH (cat no. MAB5718, R&D Systems, Inc., MN, USA), anti α-tubulin antibody (Cell Signaling Technology) or TATA box binding protein (TBP) for nuclear samples (MA5-14739, Thermo Fisher Scientific). To detect the activation of MAPKs (ERK1/2, P38 and JNK), the following antibodies were used against the phosphorylated and non-phosphorylated forms of kinases: phospho-p44/42 MAPK (Erk1/2) (cat no. 8544) and p44/42 MAPK (Erk1/2) (cat no. 4348), Phospho-p38 MAPK (cat no. 4511) and p38 MAPK (cat no. 8690), Phospho-SAPK/JNK (cat no. 4671) and SAPK/JNK (cat no. 9258), and NF-κB1 p105/p50 (cat no. 13586). Antibodies were purchased from Cell Signaling Technology and used at the dilution recommended by the supplier.

Immunofluorescence of labelled ESPs and confocal microscopy

For ESPs labelling with Alexa Fluor 488, proteins collected from axenic culture were washed (3x) with cold PBS during sample concentration to remove free amino acids and antioxidants that might interfere with protein labelling (recommended in the kit). ESPs (45μg) were incubated with 11.3 nmol/μl of Alexa Fluor-488 and excess dye was removed by gel filtration following the instructions in the Alexa Fluor 488 Microscale Protein Labelling Kit (Thermo Fisher Scientific).

To visualise ESPs interaction with host cells, chamber slides were washed and replenished with fresh DMEM as above, followed by adding labelled ESPs to the wells at 1 or 5 μg/ml and incubation for 2 h or 6 h (37°C, 10% CO2). The amounts of labelled proteins were quantified using a NanoDrop 1000 Spectrometer (Thermo Fisher Scientific). Caco-2 cells incubated in DMEM only (no Labelled ESPs added) served as a control. Slides were washed, fixed (4% paraformaldehyde in PBS) and mounted (Vectashield anti-fade reagent with DAPI, Vector Laboratories, CA, USA), excluding the blocking step. The fluorescent label on IECs was visualised using a Zeiss LSM700 confocal microscope (Zeiss, Oberkochen, Germany) at the BioVis Core Facility, SciLifeLab, Uppsala, Sweden.

Statistical analyses

The results from the cytokine measurements and differential gene expression for qPCR results were analysed using a one-way analysis of variance (ANOVA) to assess overall significance among the tested groups at α < 0.05. When significant, the values from the replicates were processed through the Bonferroni-Holm’s and significance was marked at P < 0.05.

Results

The secretome of Giardia trophozoites in the absence and presence of host cells

In order to identify proteins in the Giardia secretome, we collected ESPs from Giardia trophozoites grown axenically or in the presence of IECs (Fig 1A). The secretomes of axenic WB (assemblage A) and GS (assemblage B) Giardia trophozoites were analyzed in a modified serum-free medium (see Materials and methods) that could support the full viability of trophozoites for 2h and estimated viabilities of 98.74% ± 1.03 (WB, n = 5) and 98.32% ± 0.81 (GS, n = 5) under 6h of incubation. In total, 196 proteins were identified in the secretome of the WB isolate with 149 proteins detected in 2h culture and 180 in 6h culture; 133 proteins overlapped in both time points (Fig 2A). For the GS isolate, 155 proteins were identified including 141 proteins from the 2h culture and 83 proteins from 6h culture; 69 proteins overlapped in both time points (Fig 2A). The reproducibility of the secretome profiles was conserved in the three biological replicates. Full lists of identified proteins, including tryptic peptides, score, coverage and number of peptide spectrum matches (PSM) are presented in S1 Table (WB) and S2 Table (GS). The secretomes of both isolates were also compared leading to the identification of 122 common secreted proteins (S3 Table).

thumbnail
Fig 2. Secretomes of Giardia trophozoites and differentiated Caco-2 cells during interactions.

(A) Number of secreted WB and GS proteins in axenic culture. (B) Number of secreted WB and GS proteins during host cell interactions. (C) Number of secreted Caco-2 cell proteins during interaction with Giardia WB and GS trophozoites. (D) and (E) GO term analyses of WB and GS secretomes during interaction with host cells. (F) and (G) GO term analyses of secreted Caco-2 cell proteins during interactions with Giardia WB and GS trophozoites.

https://doi.org/10.1371/journal.pntd.0006120.g002

We also collected and analyzed the medium from Giardia trophozoites incubated with differentiated Caco-2 cells (Fig 1A). In total, 248 WB proteins were identified in the co-culture medium, 156 at 2h, 240 at 6h and 148 in both time points (Fig 2B, S4 Table). For the GS isolate, 152 proteins in total were identified, 122 at 2h, 128 at 6h and 98 at both time points (Fig 2B, S5 Table). The top 50 proteins, based on their peptide score, are listed in Table 1. These proteins overlap with those in axenic culture (S1 and S2 Tables). A comparison was performed between the secretomes of WB trophozoites incubated with (S4 Table) and without IECs (S1 Table) through which 161 common proteins and 87 interaction-specific proteins were identified (Table 2). Similarly, an analysis of the secreted proteins from GS isolate with (S5 Table) and without IECs (S2 Table) identified 111 common proteins and 41 proteins specific to the interaction (Table 2). Amongst the proteins specific to interactions, 13 proteins overlap between the two isolates (denoted in Table 2) whereas 74 WB trophozoite proteins and 28 GS trophozoite proteins are exclusive to the interaction process and represent an isolate-specific response to IECs. This overall shows that most of the secreted Giardia proteins during interactions with IECs are conserved between the WB and GS isolates but there are assemblage-specific differences.

thumbnail
Table 1. Top 50 identified proteins of Giardia intestinalis isolates, WB and GS, secreted into the medium of interaction with intestinal epithelial cells in vitro.

The proteins are ranked based on their peptide score. Note that all listed proteins were also detected in axenic cultures.

https://doi.org/10.1371/journal.pntd.0006120.t001

thumbnail
Table 2. Proteins specifically released by Giardia intestinalis isolates, WB and GS, during interaction with differentiated Caco-2 cells.

https://doi.org/10.1371/journal.pntd.0006120.t002

Secreted Giardia proteins in co-culture were analyzed for the enrichment of certain biological functions and for protein class. For the GS isolate, since the current online algorithms do not support GO analysis, orthologous genes in WB were used to perform the quest. The results are presented in Fig 2D and 2E. The largest number of detected proteins have functions associated with metabolism (85 proteins in WB and 64 in GS) with different subgroups in the child linages associated with protein metabolism, amino acid catabolic and metabolic processes, glycolysis, monosaccharide metabolic process and the generation of precursor metabolites and energy (Fig 2D). These metabolic groups were also seen in the secretome of axenic cultures (S1 Fig). Other metabolic functions such as the hexose monophosphate shunt and arginine metabolism could also be identified when secreted proteins were manually delineated into different metabolic pathways (S1 Fig). Furthermore, a nuclease with endonuclease I activity (Table 1), possibly involved in nucleic acids salvaging, and proteins involved in pyrimidine salvage (cytidine deiminase and uridine phosphorylase (UPL-1)) as well as in lipid metabolism were identified in secretome of co-culture (S1 Fig). The lipid metabolic proteins included enzymes involved in phospholipids (PLs) remodelling and salvaging fatty acids (phospholipase B, PLB), producing monoacylglycerol (lysophospholipid phosphatase) and generating inositol-3 phosphate (I3P, I3P synthase), the precursor for all inositol containing compounds including PLs [32]. These result show that the major part of Giardia secretome is involved in different metabolic functions.

The majority of proteins in the two isolates metabolo-secretome fell under four major categories: the hydrolase, dehydrogenase, oxidoreductase and protease groups. The oxidoreductase group contained 25 proteins in WB and 14 proteins in GS (Fig 2E). Compared with axenic culture (14 proteins) (S2A Fig), the WB isolate released 11 additional proteins with oxidoreductase activity including peroxidases when incubated with IECs (e.g. PFOR, NADH oxidase lateral transfer candidate, peroxiredoxin 1, FixW protein alcohol dehydrogenases (ALDs), glutamate dehydrogenase and glutamate synthase and a HP GL50803_9355, Table 2). This suggests a response to counteract IECs oxidative defences such as reactive oxygen species. It has been suggested that oxidative stress provides the trigger to initiate encystation [33] and proteins involved in cyst wall synthesis (e.g. glucosamine 6-phosphate deaminase and N-acetylglucosamine 6-phosphate mutase) were released by WB isolate interacting with IECs, corroborating this notion. For GS, on the other hand, peroxidases and antioxidant proteins were already secreted by trophozoites without exposure to IECs (S2 Table) and thus the results above show that the two isolates exhibit different anti-oxidative responses during interaction with IECs.

The protease group contained 13 secreted proteins in both isolates (Fig 2E) and harboured the cysteine- (cathepsins B and L), serine- (Alanyl dipeptidyl peptidases) and metallo-type (Aminoacyl-histidine dipeptidases and Xaa-Pro dipeptidases) of proteases. For WB isolate, two secreted metalloproteases (GL50803_8407 and GL50803_3822) were released in response to IECs whereas the rest of proteases were the same as those in axenic culture. For GS isolate, the same proteases were detected in both co-culture and axenic culture with two serine proteases (GL50581_3181 and GL50581_2704) being non-orthologous to any WB isolate proteins.

Many of the proteins detected in our analysis have been previously reported as immunoreactive proteins during human giardiasis (S6 Table) [21]. Amongst these proteins, the variable surface proteins (VSPs) are immunodominant during infection [2] and together with the high cysteine membrane proteins (HCMPs) they occupied a large portion of the WB isolate secretome (23 and 12, respectively, S1 Table). In the GS isolate secretome, lower numbers of VSPs and HCMPs were detected (4 and 5, respectively, S2 Table), more likely due to the fragmented genome used in protein identification. VSPs and HCMPs are cysteine-rich proteins and those detected in our analysis have an epidermal growth factor (EGF)-like domain in common (InterPro:IPR000742, 39 in WB and 10 in GS) which is also found in other secreted cysteine-rich proteins (e.g. tenascins, high cysteine proteins (HCPs) and neurogenic locus Notch protein precursor, S1 and S2 Tables). EGF-like domains are often present in secreted protein [34] and thus, this promoted us to look for proteins with secretion signal peptides (SSPs). A genome-wide screen in the Giardia database showed that 1361 proteins of the WB isolate have SSPs, amongst which 82 proteins were detected in our co-culture secretome analysis (~29%, S7 Table) with 41 of those being N-glycosylated [35] (S7 Table). For the GS isolate, 422 proteins in Giardia DB contain SSPs, 44 were detected in our analysis (~29%, S7 Table). The majority of proteins with SSPs (S7 Table) contained either an EGF-like domain (38 proteins for WB and 16 for GS) or a peptidase C1A domain (8 proteins for WB and 8 proteins for GS). The detection of a relatively small number of proteins with SSPs in Giardia secretomes was not surprising as this has been previously reported in other protozoan parasites like Trypanosoma. brucei (~20%) and Leishmania donovani (~9%) [36,37], suggesting the presence of alternative secretory pathways. Many protozoan parasites produce extracellular vesicles (MVs and exosomes), which can release proteins without SPPs [38]. MVs have recently been shown to be released from Giardia trophozoites [26] and we could detect two types of vesicles (100–250 nm and 100 nm, S2B Fig) in the supernatants of axenic trophozoite culture. These vesicles might explain the release of proteins without SSPs but further experiments are needed to show how these vesicles are formed and what proteins they contain.

The secretome of differentiated Caco-2 cells during parasite interactions

The secretome of differentiated Caco-2 cells incubated with WB trophozoites was analyzed for proteins released in co-culture. Overall, 384 Caco-2 cell proteins were identified in the medium of interaction with 21 proteins secreted at 2h, 136 at 6h and 227 at both time points (Fig 2C). The identified proteins, together with the number of peptides, score, coverage and PSMs are presented in S4 Table. A comparison between the secretomes of Caco-2 cells incubated alone (i.e. control, S8 Table) and with WB trophozoites (S4 Table) identified 308 overlapping proteins (S9 Table) and 76 proteins released in response to WB trophozoites (Table 3). The secretome of Caco-2 cells incubated with GS trophozoites was also analyzed for the proteins released by host cells in response to interaction. In total, 355 proteins were secreted in co-culture, 60 proteins were secreted at 2h, 78 at 6h and 217 at both time points (Fig 2C). The proteins identified, together with the number of peptides, score, coverage and PSMs are presented in S5 Table. The top 50 proteins, based on their peptide scores, are presented in Table 4. A comparison between the secretomes of co-culture (S5 Table) and control (S8 Table) identified 310 common proteins (S10 Table) and 45 proteins that were specifically released in response to GS trophozoites (Table 3). Amongst the proteins specific to interaction (45 proteins) 31 proteins overlapped in the response to both isolates.

thumbnail
Table 3. Proteins secreted by differentiated Caco-2 cells specific to interaction with Giardia intestinalis isolates, WB and GS.

https://doi.org/10.1371/journal.pntd.0006120.t003

thumbnail
Table 4. Top 50 identified proteins of secreted by differentiated Caco-2 cells co-incubated with Giardia intestinalis isolates, WB and GS in vitro.

The proteins are ranked based on peptide score. Note that all listed proteins were also detected in Caco-2 cells culture without Giardia.

https://doi.org/10.1371/journal.pntd.0006120.t004

GO enrichment analyses of parasitized Caco-2 cell secretomes (both isolates, Fig 2F and 2G) revealed that the majority of proteins have metabolic functions (152 protein in response to WB and 135 in response to GS) such as protein metabolism (including proteolysis), carbohydrate metabolism (including monosaccharides and polysaccharide metabolism as well as glycolysis), nucleobase-containing compounds metabolic process and lipid metabolism (only WB). The same metabolic groups were also seen in the control Caco-2 cell secretome (S2C Fig). One of the big groups that emerged in our analysis was the cytoskeletal proteins (41 proteins in response to WB isolate and 32 proteins in response to GS). This group included the intermediate filament and the actin family cytoskeletal proteins (including non-motor actin binding protein in the child lineage). Except for the actin family cytoskeletal protein group which was not enriched in control Caco-2 cell secretome, the rest of cytoskeletal protein groups contained more proteins compared to the control secretome. This clearly indicates the effect of Giardia colonization on IECs actin cytoskeleton. An interesting finding is that seven microvilli proteins were detected in the secretome of Caco-2 cells incubated with WB trophozoites (GO:0005903, intestinal epithelial brush border) (e.g. villin-1, intelectin-1, myosin heavy chain-9 and -14, beta 1,4- galactosyltransferase polypeptide 1, plectin and LIM domain and actin binding 1). These proteins might have been cleaved off/dissociated from damaged microvilli as a result of trophozoite attachment.

Two of the proteins that we identified in the parasitized Caco-2 cell secretome are associated with immunological functions. The first protein is a cytokine called macrophage migration inhibitory factor (MIF) (Table 3). MIF is a pleiotropic cytokine that regulates the expression of inflammatory genes (e.g. TNF-α, cyclooxygenase 2 and inducible NO synthase (iNOS)) [39], indicating its involvement in innate immune responses. The second protein is complement c3 identified in the secretome of Caco-2 cell incubated with GS only (Table 3), suggesting differences in Giardia isolates abilities to activate the complement system.

In conclusion, we have identified the major secreted proteins of host and parasite during interaction. Our analysis showed that both, the host and parasite, release proteins with similar functions (sugar, protein, lipid and nucleic acid metabolism). Both assemblages/isolates (A and B) induced similar responses but there are assemblage-specific differences that might explain observed differences in infectivity and symptoms.

Transcriptional response of IECs to Giardia ESPs

Interaction between Giardia trophozoites and IECs in vitro induces the transcription of a large number of genes including the chemokines ccl2, ccl20, and cxcl1-3 that are up-regulated up to 100-fold at the RNA level [40]. To test whether Giardia ESPs are capable of inducing similar effects, differentiated Caco-2 cells were exposed to Giardia ESPs through Transwell inserts (Fig 1B) for 2h and 6h. Differentially expressed genes (DEGs) were identified by RNA sequencing (RNA Seq) (S11 Table). Overall, 120 and 87 genes were differentially expressed (DE) in IECs exposed to ESPs of WB and GS trophozoites, respectively, with a global gene expression profile showing differences between the two time points and between the two isolates (S11 Table, Fig 3A).

thumbnail
Fig 3. Gene expression changes in IECs after ESP exposure.

(A) Heat-map of differentially expressed genes in the differentiated human colon carcinoma cell line Caco-2 exposed to Giardia intestinalis excretory secretory products. (B) Induction of AP-1 activity using a luciferase reporter plasmid transfected into Caco-2 cells. The cells were exposed to ESPs of Giardia intestinalis isolate WB or GS for 2h or 6h through a Transwell insert placed in the tissue culture plate (Mean ± SD, n = 4). (C) Relative RNA levels (Mean ± SD, n = 4) of inflammatory genes in differentiated Caco-2 cells incubated with different amounts of ESPs (5μg/ml and 10μg/ml) collected from Giardia intestinalis isolate WB or GS. Significant results in the panels are indicated by (*) for P < 0.05 or (**) for P < 0.01. (D) ELISA measurement (Mean ± SD, n = 3) of interleukin-8 (IL-8), chemokine (C-X-C motif) ligand 1 (CXCL1) and chemokine (C-C motif) ligand 20 (CCL20) in the medium of differentiated human colon carcinoma cell line, Caco-2, exposed to ESPs of G. intestinalis WB or GS isolate through a Transwell insert. No statistical significance was found between controls and treatments (P > 0.05).

https://doi.org/10.1371/journal.pntd.0006120.g003

At 2h, seven genes were DE in IECs exposed to ESPs of the WB isolate, six of which were up-regulated and one gene (il8) was down-regulated (Table 5). The up-regulated genes encode immediate early genes [41], including the protein constituents of AP-1 transcription factor (FBJ murine osteosarcoma viral oncogene homolog, fos and jun B proto-oncogene, junb). To verify AP-1 activation, Caco-2 cells we transfected with a plasmid expressing a luciferase gene under the control of AP-1 transcription response element and luminescence was measured upon cell exposure to Giardia ESPs. At 2h, no significant change was observed in the AP-1 promoter activity with ESPs of either isolates (1.07 for ESPs of WB isolate and 1.12 for ESPs of GS isolate, P > 0.05) (Fig 3B). At 6h, on the other hand, despite the slight increase in AP-1 promoter activity (1.17 for ESPs of WB isolate and 1.15 for ESPs of GS isolate), the average fold change was significant (n = 4, P < 0.05) (Fig 3B). These findings are in line with the RNA Seq results and indicate that AP-1 is activated in response to Giardia ESPs. The gene encoding DUSP1 was the highest up-regulated gene at 2h and the only up-regulated gene in IECs exposed to ESPs of GS trophozoites (Table 5). Its transcriptional level increased at 6h together with dual specificity phosphatases 4 and 5 (dusp4 and dusp5), and tribbles pseudokinase 1 (trib1) (Table 5). The products of these genes play important roles in the regulation of MAPKs’ activity [4244].

thumbnail
Table 5. Selected groups of differentially expressed genes in differentiated human colon carcinoma Caco-2 cells exposed to excretory-secretory products of Giardia intestinalis isolates WB and GS via Transwell insert.

Bold fonts indicate a significantly up-regulated gene whereas italic fonts indicate a significantly down-regulated gene.

https://doi.org/10.1371/journal.pntd.0006120.t005

The majority of DEGs (120 in WB and 87 in GS) were up-regulated at 6h (S11 Table) with a higher fold change for DEGs compared to 2h (Table 5, S11 Table). One exception was that the increase in RNA levels il8 in of IECs exposed to GS trophozoite (1.95 fold) was not significant. Insulin induced gene 1 (insig1) was the highest up-regulated gene in IECs in response to ESPs of both isolates at 6h (Table 5) and this gene encodes a protein that regulates cholesterol metabolism, lipogenesis, and glucose homeostasis [4547]. More genes associated with glucose uptake and response to glucose starvation (Table 5) were DE at 6h, indicating that Giardia releases ESPs that interfere with glucose homeostasis. Nuclear receptor subfamily 4 group A member 1 (nr4a1) was the second top up-regulated gene (ESPs of both isolates, Table 5) and this gene has functions associated with cell cycle, inflammation, and induction of apoptosis [4850]. Another two cell cycle genes associated with response to DNA damage, were amongst the top ten up-regulated genes and these include BTG family member 2 (btg2) [51] and growth arrest and DNA damage inducible alpha (gadd45a) [52] (ESPs of both isolates, Table 5). This suggests that Giardia ESPs induce damaging effects on host DNA and arrests IECs in the cell cycle. The tight junction proteins, claudin 3 and 4 (cldn 3 and cldn 4), were also up-regulated (Table 5) and these proteins play essential roles in the maintenance of the intestinal epithelial barrier (IEB) [53].

To obtain a general overview on the function of DEGs, they were processed through the GO Database for biological and molecular functions. For IECs exposed to ESPs of the WB isolate, cell proliferation (GO:0008283) and MAPK cascade (GO:0000165) constituted the biggest biological groups encompassing 11 genes each. Both chemokines (e.g. CXCL1-3 and IL-8) and growth factors (e.g. amphiregulin, proepiregulin, Proheparin-binding EGF-like growth factor) were contained within the cell proliferation group. All chemokine genes, including ccl2 and ccl20, were listed under chemokine activity (GO:0008009) and cytokine receptor binding (GO:0005126) by molecular function. In addition to these cytokines, the gene clcf1 encoding cardiotrophin-like_cytokine_factor_1 was DE in IECs exposed to ESPs of WB trophozoites. CLCF1 is an IL-6 family cytokine capable of activating NFκB and exerts stimulatory functions on B cells [54]. For MAPK cascade, it was interesting to see genes encoding RNA binding proteins (zfp36 and zfp36l2) within this group, both of which are associated with anti-inflammatory functions [55]. Another DE gene is Regnase 1 (ZC3H12A), an RNase that controls inflammatory responses by inducing mRNA decay of specific cytokine transcripts [56]. The rest of functional groups in this analysis included response to external stimulus (GO:0009605), locomotion (GO:0040011), localization (GO:0051179), behaviour (GO:0007610) and cell death (GO:0008219). For IECs exposed to ESPs of the GS isolate, the groups cell proliferation, MAPK cascade, cell death and endoderm development (GO:0007492) emerged by biological functions and no specific groups emerged by molecular function analysis. Only cxcl2, cxcl3 and clcf1 were significantly induced in IECs exposed to ESPs of the GS trophozoites. Overall, based on the transcriptomic profile we could identify a general response in IECs exposed to Giardia ESPs, which involves inducing and regulating inflammatory responses, glucose homeostasis, apoptosis, cell cycle, and maintenance of IEB.

Next, we examined which amount of ESPs could induce the transcription of pro-inflammatory genes in IECs. ESPs collected from axenic culture (1, 5 and 10 μg/ml) were incubated with IECs for 2h or 6h (Fig 3C) and RNA levels were assessed with qPCR. At 2h, 5 μg/ml of WB isolate ESPs induced a 2 to 2.9-fold increase in RNA levels of il8, ccl20, and cxcl1-3 but not ccl2, scoring the highest for cxcl3 (Fig 3C). The fold change, however, was statistically insignificant for all the genes tested (P > 0.05). The fold change in RNA levels remained below 2 for 1 μg/ml (not shown) and 10 μg/ml of ESPs (Fig 3C). At 6h, all genes were significantly up-regulated (2.84–8.46 fold) in IECs incubated with 5 μg/ml WB isolate ESPs, scoring the highest for ccl20 (P < 0.05) and the genes ccl2, ccl20, cxcl1 and cxcl3 remained significantly up-regulated (2.84–4.76 fold) when IECs were incubated with 10 μg/ml of ESPs (Fig 3C). For ESPs of the GS isolate, the fold change at RNA level for all the genes tested at the three concentrations of ESPs was below 2 (P > 0.05) (Fig 3D). Nevertheless, at 6h, il8, ccl20, cxcl1 and cxcl3 were significantly up-regulated (4.23–4.71 fold) with il8 being significantly up-regulated at all ESPs concentrations tested and ccl20 and cxcl1 at 10 μg/ml (Fig 3C). The results show that ESPs of both isolates induce variable transcriptional responses of inflammatory genes in IECs and these responses differ with the amounts of ESPs IECs are exposed to. However, direct host-cell interactions have a much stronger effect on the induction of chemokine gene transcription [40].

To assess whether the up-regulated chemokine genes were translated into their protein products, we selected three chemokines (IL-8, CXCL1 and CCL20) and measured their levels in the collected media from the insert experiment (Fig 3D). The amounts of IL8, CXCL1 and CCL20 were very low and their concentration was similar or lower than their controls at 6h (50.2, 42.1 and 57.9 pg/ml, respectively). A slight increase in the amounts of CXCL1 and CCL20 amounts could be seen at 2h upon IECs exposure to ESPs of the WB trophozoites (12.9 pg/ml in control versus 18.2 pg/ml) and GS trophozoites (24.7 pg/ml in control versus 27.1 pg/ml), respectively (P > 0.05) (Fig 3D). Overall, the levels of measured chemokines were either close to control or slightly lower and no significant differences could be seen between IECs exposed or unexposed to ESPs (P > 0.05) (Fig 3D). The low levels of measured chemokines indicate that their mRNA levels are post-transcriptionally regulated or the chemokines being degraded upon release from IECs.

The effect of ESPs on cell signaling

Giardia ESPs from axenic cultures were labelled with Alexa Fluor488 and added to the medium to visualize their interaction with IECs. Confocal images taken upon 2h and 6h of incubation with IECs showed that the labelled ESPs of WB and GS trophozoites bind to the IECs surface as well as the intercellular space (i.e. intercellular junctions) and some are internalized into differentiated Caco-2 cells (Fig 4). This indicates the presence of ligands or receptors on IECs surface and a potential of the internalized ESPs to modulate cellular processes and signaling.

thumbnail
Fig 4. The binding and internalization of Giardia intestinalis excreted-secreted products (ESPs) to intestinal epithelial cells (IECs).

Confocal microscopy images of differentiated human colon carcinoma cell line, Caco-2, incubated with Alexa Fluor488-labelled ESPs of G. intestinalis isolate WB or GS. Note that the colour of nucleus, stained with DAPI, was changed to red for better contrast.

https://doi.org/10.1371/journal.pntd.0006120.g004

We consequently examined MAPK signaling in IECs exposed to ESPs via inserts because this pathway emerged in the functional analysis of RNA Seq data. Western blot analyses showed a slight decrease in the phosphorylation levels of ERK1/2 and P38 at 2h but an increased level of nuclear translocation of NFκB compared to control (Fig 5A). Despite the slight decrease in ERK1/2 and P38 phosphorylation, their roles in inducing the nuclear recruitment of NFκB cannot be excluded and it indicates a possibility of other factors contributing to NFκB activation (e.g. growth factors, stress, hypoxia or nutrient depletion). At 6h, we could see a marked reduction in the phosphorylation levels of ERK1/2 and P38 compared to control and this coincided with a decrease in the levels of NFκB nuclear translocation (Fig 5A). The nuclear translocation of NFκB, however, was not completely abolished. The phosphorylated form of JNK could not be detected (S3A Fig). This shows that Giardia ESPs actively modulate MAPK signaling to avert the induction of strong inflammatory responses and this effect is built up upon prolonged exposure of IECs to ESPs. To test whether Giardia ESPs can also inhibit inflammatory signaling, we incubated IECs with LPS or TNF-α for 2h, then added Giardia trophozoites into Transwell inserts in the persistence of inflammatory stimuli for 2h or 6h. While the controls exhibited increased phosphorylation of ERK1/2 and P38 as well as nuclear recruitment of NFκB, these levels were reduced in inflamed tissue upon exposure to Giardia ESPs at both time points (Fig 5B). These results show that Giardia ESPs can attenuate inflammatory signaling mediated by MAPK phosphorylation and NFκB recruitment. It is possible that the parasite senses IECs inflammatory signals and release specific protein factors to subvert such effects.

thumbnail
Fig 5. The effects of ESPs on IEC cell signaling.

(A) Western blot analyses of differentiated Caco-2 cells exposed to G. intestinalis ESPs through a Transwell insert for 2h or 6h in the absence of inflammatory stimuli (e.g. lipopolysaccharides or tumour necrosis factor-alpha). Blots were probed for total proteins (phosphorylated/non-phosphorylated forms) of mitogen activated proteins kinases (MAPKs, ERK1/2 and P38) and nuclear factor kappa beta (NFκB). (B) Western blot analyses after the induction of inflammation with LPS and TNF-alpha.

https://doi.org/10.1371/journal.pntd.0006120.g005

Discussion

Several recent studies have shed the light on the secretomes of different protozoan parasites and their role in parasite physiology, adaptation to host environment, pathogenicity and immune modulation [36,37,5759]. To date, very few Giardia ESPs have been identified, and thus we devised experiments to characterise Giardia secretome, identify secreted proteins that may be involved in virulence and better understand disease mechanism (Fig 1). We also assessed the effect of Giardia colonization on host cells and identified IECs secretory responses.

Our analysis highlighted metabolic functions as a dominant secretory response when Giardia trophozoites interact with IECs (Fig 6). These functions involve carbohydrate (including glycolysis), protein (including arginine metabolism), lipid (phospholipid remodelling) and nucleic acid (purines and pyrimidines salvage) metabolism. One striking observation is that the same types of metabolic proteins are released by Giardia and host cells during interactions, indicating competition for acquiring nutrients (Fig 6).

thumbnail
Fig 6. Proteins secreted during Giardia-host cell interactions.

During Giardia-host cell interactions in vitro specific proteins are released into the growth medium. The same types of proteins, mainly enzymes, are released from both the host and parasite (black) but there are also parasite-specific proteins being released (blue).

https://doi.org/10.1371/journal.pntd.0006120.g006

The release of glycolytic enzymes has been previously reported in Giardia [17] and in many parasites [60] including Trichomonas vaginalis [61], T. brucei [36] and L. donovani [37]. Secreted glycolytic enzymes of parasites have functions associated with virulence, but those of Giardia require further investigation.

The protein metabolic group contained proteins similar to those secreted by L. donovani and T. brucei [36,37]. In this group, a large number of CPs were identified whose activity is associated with disease induction and immune modulation [12,15,62]. Herein, we report the identity of secreted CPs (Fig 6) as well as other proteases (e.g. serine and metalloproteases). We also showed a variation in protease numbers between the isolates; an aspect underlying virulence [63]. Little is known about the serine and metalloproteases of Giardia but studies in Entamoeba invadens [64], Acanthamoeba castellini [65], Acanthamoeba lugdunensis [66] and T. brucei [67] have identified some roles in differentiation [64,68], pathogenesis (e.g. degradation of ECM and immunoglobulins) [6467] and invasion [69]. It will be interesting to characterize the function of these secreted proteases during Giardia infections.

Giardia trophozoites cannot synthesize purines and pyrimidines de novo and rely on exogenous sources for their acquisition [70]. UPL-1 is secreted by Giardia and its gene is up-regulated during interaction with IECs [33,71]. This enzyme is involved in pyrimidine salvage and it is highly active in the small intestines [72] corroborating the notion of host-parasite competition for nutrients (Fig 6). UPL-1 is also an immunogenic protein recognized by sera from giardiasis patients, indicating its potential as a diagnostic and vaccine target [21]. Interacting Giardia and IECs also secrete nucleases but which nucleases’ activity can surpass the other, it is unknown. Nucleases harness nucleic acids for salvaging purines and pyrimidines as reported for E. histolytica [73], L. donovani [37] and T. brucie [36]. Nucleases of T. brucei, have also been proposed to attenuate nucleic acids-induced inflammatory responses [36].

Giardia trophozoites cannot synthesize lipids de novo and acquire those from the host [32]. PLB is an enzyme secreted by the parasite and IECs into the interaction medium (Fig 6). PLB is involved in PLs remodeling in Giardia and it localizes to brush border membranes of mature enterocytes in humans [74]. In virulent Candida albicans strains, PLB is suggested to promote host cell damage and lysis, and the deletion of plb attenuated Candida virulence in murine models [75]. I3PS is another ESP secreted by Giardia and Caco-2 cells. I3PS synthesises I3P, the precursor for all inositol containing compounds including PLs [76]. The deletion of i3ps in Leishmania renders the parasite avirulent in vivo [77] but whether it contributes to Giardia’s virulence this requires investigation. Since the above proteins are required for parasite nutrition and thus survival, it will be interesting to test their potential targets for drugs and/or vaccines.

Giardia releases proteins with antioxidant functions during growth and interaction with IECs (Fig 6). This highlights the microaerophilic nature of Giardia [78] and the mechanism by which the parasite maintains a low redox potential extracellularly leading to enhanced intracellular homeostasis. When readily released by the parasite or in response to interaction with IECs, antioxidant proteins can promptly attenuate host ROS during infection as suggested for E. histolytica [7981]. We previously showed that the transcriptional levels of antioxidant genes vary between the WB and GS isolates during oxidative stress [82] and herein we show differences in the array of secreted antioxidant proteins, further pointing out the differences in oxidative stress responses between assemblage A and B Giardia parasites.

Giardia escapes host and environmental stresses by cysts formation and thus encystation is regarded as a mean of survival for the parasite. In response to IECs, both Giardia isolates released proteins involved in cyst wall formation (Fig 6) and these proteins were not detected in axenic culture indicating that host cells provide the signal to initiate encystation. Interestingly, in a recent study that investigated transcriptional changes in bioluminescent WB isolate trophozoites from high density foci in the mice intestines, the authors reported the up-regulation of encystation, oxidative stress and metabolic genes (carbohydrate, protein, lipid and nucleoside base metabolism) [83]. Herein, we show that the protein products for many of up-regulated genes in that study are being released into the interaction medium. Nevertheless, we did not see any cyst formation in our in vitro model, indicating that stress alone might not be a sufficient stimuli to complete encystation successfully and possibly requires other factors like bile [84].

Giardia VSPs, HCMPs and tenascin were abundant proteins detected in the secretome of both isolates (Fig 6). VSPs are constantly released from trophozoite surface and they are involved in immune evasion during infection [2,20]. HCMPs are similar to VSPs but lack the c-terminal tail. So far, there is no reports on HCMPs function in Giardia except for being transcriptionally up-regulated during interaction with IECs [33,40,71]. Tenascins are also up-regulated during interaction with IECs [85]. Despite the lack of true similarity with human tenascins, the presence of an EGF-like domains in these glycoproteins suggests their involvement in protein-protein interaction, cell signaling or adhesion [86,87]. Tenascins are also able to bind lectin [88,89] and thus a role for Giardia tenascins in host innate immunity have been suggested previously [85]. The fact that 8 out 10 WB isolate “tenascins” are secreted, glycosylated and up-regulated during interaction with IECs [85] might suggest interesting roles during Giardia infection.

Only a small percentage of proteins in the parasite’s secretome were identified with SPP. Recently, several arguments have supported the hypothesis that many parasitic proteins are released in EVs, containing cytosolic and plasma membrane proteins but not proteins from intracellular organelles [60]. In this study, we provided EM evidence on the presence of EVs from Giardia trophozoites (S2 Fig). The sorting of EVs cargo, however, is not well understood but the suggestion is that posttranslational modifications (e.g. glycosylation, ubiquitination, phosphorylation or acylation) could target proteins for vesicular secretion [36,60,90]. Whether this is the case in Giardia, this requires further investigation.

Functional analysis of parasitized IECs secretome indicated that Giardia trophozoites induce cytoskeletal changes in IECs. It is known that Giardia adheres to IECs very strongly, leaving marks on cell surface and affecting the organisation of the actin cytoskeleton [9193]. One interesting finding is that Giardia effect on microvilli structure was pronounced by the detection of seven microvilli proteins in the co-culture medium, one of which is villin. Villin plays an essential role in reorganizing microvilli actin filaments as well as actin bundles assembly, capping and severing [94,95]. Villin binds phosphatidylinositol 4,5-bisphosphate (PIP2), enhancing actin bundling and lysophosphatidic acid preventing all actin modifying activities [96,97]. This protein, however, is cleaved during Giardia infection, uncoupling its protective role from the actin cytoskeleton [62].

The secretome of parasitized IECs contained host proteins with immunological function. Examples are the MIF and complement factor c3. MIF is a pleiotropic cytokine that induces inflammatory responses mediated by TNF-α, interferon-γ (IFN- γ), IL-1β, IL-12, IL-6 and, IL-8, among others [39]. MIF plays a role in resistance to infection with Trypanosoma cruzi [98], T. gondii [99] and Leishmania major [100] but many studies have also reported its contribution in disease pathology [101103]. It will be interesting to identify its role during Giardia infections. The complement factor c3 was specifically released by IECs in response to GS isolate, which was previously shown to activate the complement system [104]. In fact, the complement system contribute to trophozoite killing both in vitro and in vivo [105,106] and mice with non-functional component system (deficient in mannose-binding lectin 2 or complement factor 3a receptor [107]) exhibit delayed parasite clearance [104]. Therefore, while these results show the induction of innate immune responses by parasitized IECs, it also shows that these responses might vary in infections with different isolates/assemblages.

To our knowledge, this is the first study that has investigated transcriptional changes in IECs exposed to Giardia ESPs. Interestingly, transcriptome data brought about and highlighted important pathological processes previously shown to occur during Giardia infection. Specifically, the disturbance in glucose homeostasis [108,109] and IEB integrity [91,92], cell cycle arrest [27] and apoptosis [93,110,111]. It could also indicate another pathomechanism that is Giardia ESPs might interfere with cholesterol and lipid metabolism based on the differential expression of insig1 gene. Of note is that transcriptional changes in IECs were not immediate and were only pronounced at 6h, possibly until enough ESPs had accumulated in the media to induce an effect. By comparison, a similar transcriptional profile was seen in parasitized IECs, but transcriptional changes occurred as early as 1.5h upon trophozoite attachment [40]. Therefore, while Giardia ESPs could also play a role in disease induction, these effects might be slower and less apparent compared to those induced upon trophozoite attachment to IECs.

Giardia ESPs induced the transcription of chemokines genes previously shown to be induced upon trophozoite attachment (e.g. cxcl1-3, ccl2 and ccl20) [40]. When produced locally, chemokines attract immune cells (e.g. B cells, T cells, dendritic cells and macrophages) to the infection site [112]. Giardia ESPs also induced the transcription of il8, an important neutrophils attractant produced during the early phase of infection. NFκB and AP-1 are TFs involved in the induction of inflammatory genes transcription [28,29] and their activity was evident in our results and previously reported by others in response to Giardia ESPs [113]. Despite the activation of both TFs, the upstream signaling pathway leading to their activation (i.e. MAPK phosphorylation) appeared to be attenuated but not abolished, even in the presence of inflammatory stimuli. Our transcriptomic data suggests that this attenuation could be mediated by DEGs encoding, dusp1, dusp4, dusp5 and trib1, all of which are known to regulate/inhibit MAPKs’ activity [4244,114116]. Although the results of ERK1/2 and P38 phosphorylation were inconsistent with a previous report [113], these discrepancies might be due to the different experimental setup used to study ESPs effects on MAPK signaling. In our model, IECs were exposed to parasite ESPs released directly into the medium whereas in the other study ESPs were added directly to IECs (HT29). In fact, we got the same result when ESPs collected in the absence of IECs were added directly to Caco-2 cells (S3B Fig). This could suggest that when Giardia senses inflammatory signals it actively releases ESPs to attenuate the pathways leading to cytokines production whereas ESPs produced in the absence of IECs exert an opposite effect.

The differential up-regulation of inflammatory cytokines/chemokines genes did not correlate with the amounts of cytokines measured in medium of insert experiment. Indeed, in some instances the levels of IL8, CXCL1 and CCL20 were lower than their controls, indicating degradation of their mRNAs post-transcriptionally or upon synthesis and release into the medium. Tristetraprolin (i.e. zfp36 or TTP) among DEGs, encodes a protein that binds the AU rich element (ARE) in the 3’UTR of cytokine transcripts inducing their decay and its role in controlling inflammation is well established in the literature [117,118]. Which parasite factors induce TTP transcription are to be identified. Second, in a previous report, Giardia ESPs, specifically CPs, were able to degrade IL8 and this has been shown to attenuate neutrophils infiltration into mice intestines even in the presence of inflammatory stimuli [14,15]. Based on our findings it is plausible to propose that ESPs mediate the attenuation of inflammatory responses by promoting the decay of cytokine transcripts and/or cytokines degradation.

To conclude, an interesting picture of host-parasite interactions during giardiasis emerge from this and other studies. Parasites produce ESPs that consist of secreted proteins [11,17,19,85], released surface proteins [20] and EVs [26]. These ESPs affect gene expression, secretion, signaling, metabolism and immune responses in IECs [11,16,40,85,113]. Nevertheless, parasite attachment to IECs induces stronger and more complex responses compared to when IECs are exposed to ESPs only [40]. Concurrently, there is a secretory response by parasitized IECs where similar factors are released affecting parasite attachment, metabolism and gene expression [16,85]. So far, most studies of Giardia-host cell interactions have been performed with simplified models using a small selection of human cell lines but it will be important to follow up these results using more complex in vitro host systems like enteroids [119]. This can be complemented by in vivo experiments of secreted proteins in mice where trophozoites are mixed with early encysting cells [83]. Thus, Giardia interaction with IECs is an intricate process and further understanding of this cross-talk could be the key for understanding giardiasis.

Supporting information

S1 Fig. Delineated metabolic pathways associated with the secretome of Giardia intestinalis isolates WB and GS identified in axenic culture and the medium of interaction with the intestinal epithelial cells Caco-2 in vitro.

https://doi.org/10.1371/journal.pntd.0006120.s001

(TIFF)

S2 Fig.

(A) GO term analyses of WB and GS trophozoites in axenic culture. (B) Electron micrographs of Giardia extracellular vesicles. (C) GO term analyses of differentiated Caco-2 cell secretome in the absence of parasite.

https://doi.org/10.1371/journal.pntd.0006120.s002

(TIF)

S3 Fig.

(A) Western blot analyses of differentiated Caco-2 cells exposed to G. intestinalis ESPs through a Transwell insert for 2h or 6h in the absence of inflammatory stimuli. Blots were probed for the phosphorylated/non-phosphorylated forms of JNK. (B) Western blot analyses of differentiated Caco-2 cells exposed to G. intestinalis ESPs collected in axenic culture (i.e. in the absence of intestinal epithelial cells). Blots were probed for total proteins (phosphorylated/non-phosphorylated forms) of mitogen activated proteins kinases (MAPKs, ERK1/2 and P38) and nuclear factor kappa beta (NFκB).

https://doi.org/10.1371/journal.pntd.0006120.s003

(TIF)

S1 Table. All proteins identified in the secretome of Giardia intestinalis WB isolate in the serum-free medium, RPMI-1640.

Proteins are compared between the two time points, 2h and 6h, which are indicated by colours (orange, 2h) and (blue, 6h). Identification criterion for proteins is based on two peptides per protein. Click on (+) sign in each row to see the tryptic peptides identified.

https://doi.org/10.1371/journal.pntd.0006120.s004

(XLSX)

S2 Table. All proteins identified in the secretome of Giardia intestinalis GS isolate in the serum-free medium, 1640.

Proteins are compared between the two time points, 2h and 6h, which are indicated by colours (orange, 2h) and (blue, 6h). Identification criterion for proteins is based on two matching peptides per protein. Click on (+) sign in each row to see the tryptic peptides identified.

https://doi.org/10.1371/journal.pntd.0006120.s005

(XLSX)

S3 Table. Common and isolate-specific proteins identified in the secretome of Giardia intestinalis WB and GS isolates.

Open reading frames together with the protein names are presented in columns based on their annotation in the Giardia Database.

https://doi.org/10.1371/journal.pntd.0006120.s006

(XLSX)

S4 Table. All secreted proteins identified in in the serum-free medium of the differentiated human colon carcinoma cell line, Caco-2, incubated with Giardia intestinalis WB isolate.

Proteins are compared between the two time points, 2h and 6h, which are indicated by colours (orange, 2h) and (blue, 6h). Identification criterion is two peptides per protein. Click on the (+) sign in each row to see the tryptic peptides identified.

https://doi.org/10.1371/journal.pntd.0006120.s007

(XLSX)

S5 Table. All secreted proteins identified in in the serum-free medium of the differentiated human colon carcinoma cell line, Caco-2, incubated with Giardia intestinalis GS isolate.

Proteins are compared between the two time points, 2h and 6h, which are indicated by colours (orange, 2h) and (blue, 6h). Identification criterion is two peptides per protein. Click on the (+) sign in each row to see the tryptic peptides identified.

https://doi.org/10.1371/journal.pntd.0006120.s008

(XLSX)

S6 Table. Immunoreactive proteins of Giardia trophozoites during human infection.

These proteins are detected as parasite secreted products in the medium of interaction with intestinal epithelial cells in vitro.

https://doi.org/10.1371/journal.pntd.0006120.s009

(XLSX)

S7 Table. Proteins identified in Giardia intestinalis WB and GS isolates secretome with secretion signal peptide (SSP).

https://doi.org/10.1371/journal.pntd.0006120.s010

(XLSX)

S8 Table. All proteins identified in the secretome of differentiated colon carcinoma human cell line (Caco2) in the serum-free medium, DMEM.

Proteins are compared between the two time points, 2h and 6h, which are indicated by colours (orange, 2h) and (blue, 6h). Identification criteria for proteins are based on two peptides per protein. Click on the (+) sign in each row to see the tryptic peptides identified.

https://doi.org/10.1371/journal.pntd.0006120.s011

(XLSX)

S9 Table. Secreted proteins of the differentiated human colonic epithelial cell line, Caco2, incubated alone or with Giardia intestinalis WB isolate in a serum-free medium.

Proteins are listed in columns with headings indicating the time point they were detected.

https://doi.org/10.1371/journal.pntd.0006120.s012

(XLSX)

S10 Table. Secreted proteins of the differentiated human colonic epithelial cell line, Caco2, incubated alone or with Giardia intestinalis GS isolate in a serum-free medium.

Proteins are listed in columns with headings indicating the time point they were detected.

https://doi.org/10.1371/journal.pntd.0006120.s013

(XLSX)

S11 Table. Transcriptional profile of differentiated human colon carcinoma cell line Caco-2 exposed to excretory-secretory products (ESPs) of Giardia intestinalis isolates WB and GS through a Transwell insert placed into tissue culture plates.

The table shows transcriptional changes including fold change of genes transcriptions in Caco-2 cells exposed to ESPs of WB or GS isolate for 2h or 6h. The table also contains differentially expressed genes, which are in blue fonts (up-regulated) or red fonts (down-regulated) to indicate their transcriptional activity.

https://doi.org/10.1371/journal.pntd.0006120.s014

(XLSX)

Acknowledgments

Livia Knörr, Karin Sköld, Åsa Eriksson and Alexander Falk are acknowledged for technical assistance.

References

  1. 1. Einarsson E, Ma’ayeh S, Svärd SG. An up-date on Giardia and giardiasis. Curr Opin Microbiol. 2016;34: 47–52. pmid:27501461
  2. 2. Ankarklev J, Jerlström-Hultqvist J, Ringqvist E, Troell K, Svärd SG. Behind the smile: cell biology and disease mechanisms of Giardia species. Nat Rev Microbiol. 2010;8: 413–22. pmid:20400969
  3. 3. Cotton JA, Beatty JK, Buret AG. Host parasite interactions and pathophysiology in Giardia infections. Int J Parasitol. 2011;41: 925–933. pmid:21683702
  4. 4. Rogawski ET, Bartelt LA, Platts-Mills JA, Seidman JC, Samie A, Havt A, et al. Determinants and Impact of Giardia Infection in the First 2 Years of Life in the MAL-ED Birth Cohort. J Pediatric Infect Dis Soc. 2017;6: 153–160. pmid:28204556
  5. 5. Halliez MCM, Buret AG. Extra-intestinal and long term consequences of Giardia duodenalis infections [Internet]. World Journal of Gastroenterology 2013 pp. 8974–8985. pmid:24379622
  6. 6. Samra HK, Ganguly NK, Garg UC, Goyal J, Mahajan RC. Effect of excretory-secretory products of Giardia lamblia on glucose and phenylalanine transport in the small intestine of Swiss albino mice. Biochem Int. 1988;17: 801–12. Available: http://www.ncbi.nlm.nih.gov/pubmed/3254161 pmid:3254161
  7. 7. Jiménez JC, Fontaine J, Grzych J-M, Dei-Cas E, Capron M. Systemic and mucosal responses to oral administration of excretory and secretory antigens from Giardia intestinalis. Clin Diagn Lab Immunol. 2004;11: 152–60. Available: http://www.ncbi.nlm.nih.gov/pubmed/14715563 pmid:14715563
  8. 8. Jiménez JC, Morelle W, Michalsky J-C, Dei-Cas E. Excreted/secreted glycoproteins of G. intestinalis play an essential role in the antibody response. Parasitol Res. Springer-Verlag; 2007;100: 715–720. pmid:17171569
  9. 9. Shant J, Bhattacharyya S, Ghosh S, Ganguly NK, Majumdar S. A potentially important excretory-secretory product of Giardia lamblia. Exp Parasitol. 2002;102: 178–186. pmid:12856314
  10. 10. Kaur H, Ghosh S, Samra H, Vinayak VK, Ganguly NK. Identification and characterization of an excretory-secretory product from Giardia lamblia. Parasitology. 2001;123: 347–56. pmid:11676366
  11. 11. Rodríguez-Fuentes GB, Cedillo-Rivera R, Fonseca-Liñán R, Argüello-García R, Muñoz O, Ortega-Pierres G, et al. Giardia duodenalis: Analysis of secreted proteases upon trophozoite-epithelial cell interaction in vitro. Mem Inst Oswaldo Cruz. 2006;101: 693–696. pmid:17072486
  12. 12. Piña-Vázquez C, Reyes-López M, Ortíz-Estrada G, de la Garza M, Serrano-Luna J. Host-Parasite Interaction: Parasite-Derived and -Induced Proteases That Degrade Human Extracellular Matrix. J Parasitol Res. 2012;2012: 1–24. pmid:22792442
  13. 13. Parenti DM. Characterization of a thiol proteinase in Giardia lamblia. J Infect Dis. 1989;160: 1076–80. Available: http://www.ncbi.nlm.nih.gov/pubmed/2584755 pmid:2584755
  14. 14. Cotton JA, Motta J-PP, Schenck LP, Hirota SA, Beck PL, Buret AG. Giardia duodenalis infection reduces granulocyte infiltration in an in vivo model of bacterial toxin-induced colitis and attenuates inflammation in human intestinal tissue. PLoS One. 2014;9: e109087. pmid:25289678
  15. 15. Cotton JA, Bhargava A, Ferraz JG, Yates RM, Beck PL, Buret AG. Giardia duodenalis cathepsin B proteases degrade intestinal epithelial interleukin-8 and attenuate interleukin-8-induced neutrophil chemotaxis. Infect Immun. 2014;82: 2772–87. pmid:24733096
  16. 16. Stadelmann B, Hanevik K, Andersson MK, Bruserud O, Svärd SG. The role of arginine and arginine-metabolizing enzymes during Giardia—host cell interactions in vitro. BMC Microbiol. 2013;13: 256. pmid:24228819
  17. 17. Ringqvist E, Palm JEED, Skarin H, Hehl AB, Weiland M, Davids BJ, et al. Release of metabolic enzymes by Giardia in response to interaction with intestinal epithelial cells. Mol Biochem Parasitol. 2008;159: 85–91. pmid:18359106
  18. 18. Banik S, Viveros PR, Seeber F, Klotz C, Ignatius R, Aebischer T. Giardia duodenalis arginine deiminase modulates the phenotype and cytokine secretion of human dendritic cells by depletion of arginine and formation of ammonia. Infect Immun. 2013;81: 2309–2317. pmid:23589577
  19. 19. Skarin H, Ringqvist E, Hellman U, Svärd SG. Elongation factor 1-alpha is released into the culture medium during growth of Giardia intestinalis trophozoites. Exp Parasitol. 2011;127: 804–810. pmid:21276445
  20. 20. Papanastasiou P, Hiltpold A, Bommeli C, Köhler P. The release of the variant surface protein of Giardia to its soluble isoform is mediated by the selective cleavage of the conserved carboxy-terminal domain. Biochemistry. 1996;35: 10143–8. pmid:8756478
  21. 21. Palm JED, Weiland ME-L, Griffiths WJ, Ljungström I, Svärd SG. Identification of immunoreactive proteins during acute human giardiasis. J Infect Dis. 2003;187: 1849–59. pmid:12792861
  22. 22. Prucca CG, Slavin I, Quiroga R, Elías E V, Rivero FD, Saura A, et al. Antigenic variation in Giardia lamblia is regulated by RNA interference. Nature. 2008;456: 750–4. pmid:19079052
  23. 23. Faso C, Hehl AB. Membrane trafficking and organelle biogenesis in Giardia lamblia: use it or lose it. Int J Parasitol. 2011;41: 471–80. pmid:21296082
  24. 24. Evans-Osses I, Reichembach LH, Ramirez MI, Ingrid Evans-Osses1,2 & Luis H. Reichembach2 & Marcel I. Ramirez1 2. Exosomes or microvesicles? Two kinds of extracellular vesicles with different routes to modify protozoan-host cell interaction. Parasitol Res. Springer Berlin Heidelberg; 2015;114: 3567–3575.
  25. 25. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015;16: 24–43. pmid:25488940
  26. 26. Evans-Osses I, Mojoli A, Monguió-Tortajada M, Marcilla A, Aran V, Amorim M, et al. Microvesicles released from Giardia intestinalis disturb host-pathogen response in vitro. Eur J Cell Biol. 2017;96: 131–142. pmid:28236495
  27. 27. Stadelmann B, Merino MC, Persson L, Svärd SG. Arginine Consumption by the Intestinal Parasite Giardia intestinalis Reduces Proliferation of Intestinal Epithelial Cells. PLoS One. 2012;7: e45325. pmid:23028934
  28. 28. Fujioka S, Niu J, Schmidt C, Sclabas GM, Peng B, Uwagawa T, et al. NF-kappaB and AP-1 connection: mechanism of NF-kappaB-dependent regulation of AP-1 activity. Mol Cell Biol. American Society for Microbiology (ASM); 2004;24: 7806–19.
  29. 29. Karin M. The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem. American Society for Biochemistry and Molecular Biology; 1995;270: 16483–6.
  30. 30. Whitmarsh AJ. Regulation of gene transcription by mitogen-activated protein kinase signaling pathways. Biochim Biophys Acta—Mol Cell Res. 2007;1773: 1285–1298. pmid:17196680
  31. 31. Vallabhapurapu S, Karin M. Regulation and Function of NF-κB Transcription Factors in the Immune System. Annu Rev Immunol. Annual Reviews; 2009;27: 693–733. pmid:19302050
  32. 32. Yichoy M, Duarte TT, De Chatterjee A, Mendez TL, Aguilera KY, Roy D, et al. Lipid metabolism in Giardia: a post-genomic perspective. Parasitology. NIH Public Access; 2011;138: 267–78. pmid:20880419
  33. 33. Ma’ayeh SY, Brook-Carter PT. Representational difference analysis identifies specific genes in the interaction of Giardia duodenalis with the murine intestinal epithelial cell line, IEC-6. Int J Parasitol. 2012;42: 501–509. pmid:22561399
  34. 34. Davis CG. The many faces of epidermal growth factor repeats. New Biol. 1990;2: 410–9. Available: http://europepmc.org/abstract/MED/2288911 pmid:2288911
  35. 35. Ratner DM, Cui J, Steffen M, Moore LL, Robbins PW, Samuelson J. Changes in the N-glycome, glycoproteins with Asn-linked glycans, of Giardia lamblia with differentiation from trophozoites to cysts. Eukaryot Cell. American Society for Microbiology; 2008;7: 1930–40. pmid:18820077
  36. 36. Geiger A, Hirtz C, Bécue T, Bellard E, Centeno D, Gargani D, et al. Exocytosis and protein secretion in Trypanosoma.
  37. 37. Silverman JM, Chan SK, Robinson DP, Dwyer DM, Nandan D, Foster LJ, et al. Proteomic analysis of the secretome of Leishmania donovani. Genome Biol. 2008;9: R35. pmid:18282296
  38. 38. Marcilla A, Martin-Jaular L, Trelis M, De Menezes-Neto A, Osuna A, Bernal D, et al. Extracellular vesicles in parasitic diseases. Int Heal Res. 2014; pmid:25536932
  39. 39. Rosado J de D, Rodriguez-Sosa M. Macrophage migration inhibitory factor (MIF): a key player in protozoan infections. Int J Biol Sci. 2011;7: 1239–56. Available: http://www.ncbi.nlm.nih.gov/pubmed/22110378 pmid:22110378
  40. 40. Roxström-Lindquist K, Ringqvist E, Palm D, Svärd S. Giardia lamblia-induced changes in gene expression in differentiated caco-2 human intestinal epithelial cells. Infect Immun. 2005;73: 8204–8208. pmid:16299316
  41. 41. Bahrami S, Drabløs F. Gene regulation in the immediate-early response process. Adv Biol Regul. 2016;62: 37–49. pmid:27220739
  42. 42. Wancket LM, Frazier WJ, Liu Y. Mitogen-activated protein kinase phosphatase (MKP)-1 in immunology, physiology, and disease. Life Sci. 2012;90: 237–48. pmid:22197448
  43. 43. Korhonen R, Turpeinen T, Taimi V, Nieminen R, Goulas A, Moilanen E. Attenuation of the acute inflammatory response by dual specificity phosphatase 1 by inhibition of p38 MAP kinase. Mol Immunol. 2011;48: 2059–68. pmid:21764456
  44. 44. Kiss-Toth E, Bagstaff SM, Sung HY, Jozsa V, Dempsey C, Caunt JC, et al. Human tribbles, a protein family controlling mitogen-activated protein kinase cascades. J Biol Chem. American Society for Biochemistry and Molecular Biology; 2004;279: 42703–8. pmid:15299019
  45. 45. Dong X-Y, Tang S-Q. Insulin-induced gene: A new regulator in lipid metabolism. Peptides. 2010;31: 2145–2150. pmid:20817058
  46. 46. Krapivner S, Chernogubova E, Ericsson M, Ahlbeck-Glader C, Hamsten A, van ‘t Hooft FM. Human evidence for the involvement of insulin-induced gene 1 in the regulation of plasma glucose concentration. Diabetologia. 2007;50: 94–102. pmid:17106696
  47. 47. Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, et al. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell. Elsevier; 2002;110: 489–500.
  48. 48. Hamers AAJ, van Dam L, Teixeira Duarte JM, Vos M, Marinković G, van Tiel CM, et al. Deficiency of Nuclear Receptor Nur77 Aggravates Mouse Experimental Colitis by Increased NFκB Activity in Macrophages. Moschetta A, editor. PLoS One. Public Library of Science; 2015;10: e0133598. pmid:26241646
  49. 49. Pei L, Castrillo A, Tontonoz P. Regulation of Macrophage Inflammatory Gene Expression by the Orphan Nuclear Receptor Nur77. Mol Endocrinol. 2006;20: 786–794. pmid:16339277
  50. 50. Li H, Kolluri SK, Gu J, Dawson MI, Cao X, Hobbs PD, et al. Cytochrome c Release and Apoptosis Induced by Mitochondrial Targeting of Nuclear Orphan Receptor TR3. Science (80-). 2000;289. Available: http://science.sciencemag.org/content/289/5482/1159.long
  51. 51. Rouault J-P, Falette N, Guéhenneux F, Guillot C, Rimokh R, Wang Q, et al. Identification of BTG2, an antiproliferative p53–dependent component of the DNA damage cellular response pathway. Nat Genet. Nature Publishing Group; 1996;14: 482–486. pmid:8944033
  52. 52. Smith M, Chen I, Zhan Q, Bae I, Chen C, Gilmer T, et al. Interaction of the p53-regulated protein Gadd45 with proliferating cell nuclear antigen. Science (80-). 1994;266. Available: http://science.sciencemag.org/content/266/5189/1376.long
  53. 53. Lu Z, Ding L, Lu Q, Chen Y-H. Claudins in intestines: Distribution and functional significance in health and diseases. Tissue barriers. Taylor & Francis; 2013;1: e24978. pmid:24478939
  54. 54. Senaldi G, Varnum BC, Sarmiento U, Starnes C, Lile J, Scully S, et al. Novel neurotrophin-1/B cell-stimulating factor-3: a cytokine of the IL-6 family. Proc Natl Acad Sci U S A. National Academy of Sciences; 1999;96: 11458–63.
  55. 55. Brooks SA, Blackshear PJ. Tristetraprolin (TTP): Interactions with mRNA and proteins, and current thoughts on mechanisms of action. Biochim Biophys Acta—Gene Regul Mech. 2013;1829: 666–679. pmid:23428348
  56. 56. Matsushita K, Takeuchi O, Standley DM, Kumagai Y, Kawagoe T, Miyake T, et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature. 2009;458: 1185–1190. pmid:19322177
  57. 57. Wong WK, Tan ZN, Othman N, Lim BH, Mohamed Z, Olivos Garcia A, et al. Analysis of Entamoeba histolytica excretory-secretory antigen and identification of a new potential diagnostic marker. Clin Vaccine Immunol. 2011;18: 1913–7. pmid:21918120
  58. 58. Song M-J, Lee J-J, Nam YH, Kim T-G, Chung YW, Kim M, et al. Modulation of dendritic cell function by Trichomonas vaginalis-derived secretory products. BMB Rep. 2015;48: 103–8. Available: http://www.ncbi.nlm.nih.gov/pubmed/24965578 pmid:24965578
  59. 59. Grébaut P, Chuchana P, Brizard J-P, Demettre E, Seveno M, Bossard G, et al. Identification of total and differentially expressed excreted-secreted proteins from Trypanosoma congolense strains exhibiting different virulence and pathogenicity. Int J Parasitol. 2009;39: 1137–50. pmid:19285981
  60. 60. Gómez-Arreaza A, Acosta H, Quiñones W, Concepción JL, Michels PAM, Avilán L. Extracellular functions of glycolytic enzymes of parasites: unpredicted use of ancient proteins. Mol Biochem Parasitol. 2014;193: 75–81. pmid:24602601
  61. 61. Kucknoor AS, Mundodi V, Alderete JF. The proteins secreted by Trichomonas vaginalis and vaginal epithelial cell response to secreted and episomally expressed AP65. Cell Microbiol. 2007;9: 2586–97. pmid:17590165
  62. 62. Bhargava A, Cotton JA, Dixon BR, Gedamu L, Yates RM, Buret AG. Giardia duodenalis Surface Cysteine Proteases Induce Cleavage of the Intestinal Epithelial Cytoskeletal Protein Villin via Myosin Light Chain Kinase. PLoS One. Public Library of Science; 2015;10: e0136102. pmid:26334299
  63. 63. de Carvalho TB, David EB, Coradi ST, Guimarães S. Protease activity in extracellular products secreted in vitro by trophozoites of Giardia duodenalis. Parasitol Res. 2008;104: 185–90. pmid:18797927
  64. 64. Makioka A, Kumagai M, Kobayashi S, Takeuchi T. Involvement of serine proteases in the excystation and metacystic development of Entamoeba invadens. Parasitol Res. 2009;105: 977–987. pmid:19479279
  65. 65. Na B-K, Kim J-C, Song C-Y. Characterization and pathogenetic role of proteinase from Acanthamoeba castellanii. Microb Pathog. 2001;30: 39–48. pmid:11162184
  66. 66. Kim H-K, Ha Y-R, Yu H-S, Kong H-H, Chung D-I. Purification and characterization of a 33 kDa serine protease from Acanthamoeba lugdunensis KA/E2 isolated from a Korean keratitis patient. Korean J Parasitol. 2003;41: 189. pmid:14699259
  67. 67. Huet G, Richet C, Demeyer D, Bisiau H, Soudan B, Tetaert D, et al. Characterization of different proteolytic activities in Trypanosoma brucei brucei. Biochim Biophys Acta. 1992;1138: 213–21. Available: http://www.ncbi.nlm.nih.gov/pubmed/1547283 pmid:1547283
  68. 68. Bangs JD, Ransom DA, Nimick M, Christie G, Hooper NM. In vitro cytocidal effects on Trypanosoma brucei and inhibition of Leishmania major GP63 by peptidomimetic metalloprotease inhibitors. Mol Biochem Parasitol. 2001;114: 111–117. pmid:11356520
  69. 69. Hainard A, Tiberti N, Robin X, Ngoyi DM, Matovu E, Enyaru JCK, et al. Matrix metalloproteinase-9 and intercellular adhesion molecule 1 are powerful staging markers for human African trypanosomiasis. Trop Med Int Health. 2011;16: 119–26. pmid:20958893
  70. 70. Adam RD. Biology of Giardia lamblia. Clin Microbiol Rev. 2001;14: 447–75. pmid:11432808
  71. 71. Ferella M, Davids BJ, Cipriano MJ, Birkeland SR, Palm D, Gillin FD, et al. Gene expression changes during Giardia-host cell interactions in serum-freeh medium. Mol Biochem Parasitol. 2014;197: 21–3. pmid:25286381
  72. 72. el Kouni MH, el Kouni MM, Naguib FN. Differences in activities and substrate specificity of human and murine pyrimidine nucleoside phosphorylases: implications for chemotherapy with 5-fluoropyrimidines. Cancer Res. 1993;53: 3687–93. Available: http://www.ncbi.nlm.nih.gov/pubmed/8339277 pmid:8339277
  73. 73. McGugan GC, Joshi MB, Dwyer DM. Identification and biochemical characterization of unique secretory nucleases of the human enteric pathogen, Entamoeba histolytica. J Biol Chem. American Society for Biochemistry and Molecular Biology; 2007;282: 31789–802. pmid:17766245
  74. 74. Tojo H, Ichida T, Okamoto M. Purification and characterization of a catalytic domain of rat intestinal phospholipase B/lipase associated with brush border membranes. J Biol Chem. 1998;273: 2214–21. Available: http://www.ncbi.nlm.nih.gov/pubmed/9442064 pmid:9442064
  75. 75. Ghannoum MA. Potential role of phospholipases in virulence and fungal pathogenesis. Clin Microbiol Rev. 2000;13: 122–43, table of contents. Available: http://www.ncbi.nlm.nih.gov/pubmed/10627494 pmid:10627494
  76. 76. Majumder AL, Johnson MD, Henry SA. 1l-myo-Inositol-1-phosphate synthase. Biochim Biophys Acta—Lipids Lipid Metab. 1997;1348: 245–256.
  77. 77. Ilg T. Generation of myo-inositol-auxotrophic Leishmania mexicana mutants by targeted replacement of the myo-inositol-1-phosphate synthase gene. Mol Biochem Parasitol. 2002;120: 151–6. Available: http://www.ncbi.nlm.nih.gov/pubmed/11849714 pmid:11849714
  78. 78. Brown DM, Upcroft JA, Edwards MR, Upcroft P. Anaerobic bacterial metabolism in the ancient eukaryote Giardia duodenalis. Int J Parasitol. 1998;28: 149–64. Available: http://www.ncbi.nlm.nih.gov/pubmed/9504342 pmid:9504342
  79. 79. Sen A, Chatterjee NS, Akbar MA, Nandi N, Das P. The 29-kilodalton thiol-dependent peroxidase of Entamoeba histolytica is a factor involved in pathogenesis and survival of the parasite during oxidative stress. Eukaryot Cell. American Society for Microbiology (ASM); 2007;6: 664–73. pmid:17307964
  80. 80. Cheng X-J, Yoshihara E, Takeuchi T, Tachibana H. Molecular characterization of peroxiredoxin from Entamoeba moshkovskii and a comparison with Entamoeba histolytica. Mol Biochem Parasitol. 2004;138: 195–203. pmid:15555731
  81. 81. Choi M-H, Sajed D, Poole L, Hirata K, Herdman S, Torian BE, et al. An unusual surface peroxiredoxin protects invasive Entamoeba histolytica from oxidant attack. Mol Biochem Parasitol. 2005;143: 80–9. pmid:15996766
  82. 82. Ma’ayeh SY, Knörr L, Svärd SG. Transcriptional profiling of Giardia intestinalis in response to oxidative stress. Int J Parasitol. 2015; pmid:26341007
  83. 83. Pham JK, Nosala C, Scott EY, Nguyen KF, Hagen KD, Starcevich HN, et al. Transcriptomic Profiling of High-Density Giardia Foci Encysting in the Murine Proximal Intestine. Front Cell Infect Microbiol. Frontiers Media SA; 2017;7: 227. pmid:28620589
  84. 84. Gillin FD, Reiner DS, Boucher SE. Small-intestinal factors promote encystation of Giardia lamblia in vitro. Infect Immun. 1988;56: 705–707. pmid:3343054
  85. 85. Emery SJ, Mirzaei M, Vuong D, Pascovici D, Chick JM, Lacey E, et al. Induction of virulence factors in Giardia duodenalis independent of host attachment. Sci Rep. Nature Publishing Group; 2016;6: 20765. pmid:26867958
  86. 86. Balzar M, Briaire-de Bruijn IH, Rees-Bakker HA, Prins FA, Helfrich W, de Leij L, et al. Epidermal growth factor-like repeats mediate lateral and reciprocal interactions of Ep-CAM molecules in homophilic adhesions. Mol Cell Biol. 2001;21: 2570–80. pmid:11259604
  87. 87. Baron M. An overview of the Notch signalling pathway. Semin Cell Dev Biol. 2003;14: 113–9. Available: http://www.ncbi.nlm.nih.gov/pubmed/12651094 pmid:12651094
  88. 88. Aspberg A, Miura R, Bourdoulous S, Shimonaka M, Heinegârd D, Schachner M, et al. The C-type lectin domains of lecticans, a family of aggregating chondroitin sulfate proteoglycans, bind tenascin-R by protein-protein interactions independent of carbohydrate moiety. Proc Natl Acad Sci U S A. 1997;94: 10116–21. pmid:9294172
  89. 89. Aspberg A. The versican C-type lectin domain recognizes the adhesion protein tenascin-R. Proc Natl Acad Sci U S A. 1995;92: 10590–10594. pmid:7479846
  90. 90. Moreno-Gonzalo O, Villarroya-Beltri C, Sánchez-Madrid F. Post-translational modifications of exosomal proteins. Front Immunol. Frontiers Media SA; 2014;5: 383. pmid:25157254
  91. 91. Humen MA, Pérez PF, Liévin-Le Moal V. Lipid raft-dependent adhesion of Giardia intestinalis trophozoites to a cultured human enterocyte-like Caco-2/TC7 cell monolayer leads to cytoskeleton-dependent functional injuries. Cell Microbiol. 2011;13: 1683–1702. pmid:21790940
  92. 92. Maia-Brigagão C, Morgado-Díaz JA, De Souza W. Giardia disrupts the arrangement of tight, adherens and desmosomal junction proteins of intestinal cells. Parasitol Int. 2012;61: 280–287. pmid:22146155
  93. 93. Chin AC, Teoh DA, Scott KG-E, Meddings JB, Macnaughton WK, Buret AG. Strain-dependent induction of enterocyte apoptosis by Giardia lamblia disrupts epithelial barrier function in a caspase-3-dependent manner. Infect Immun. 2002;70: 3673–80. Available: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=128105&tool=pmcentrez&rendertype=abstract pmid:12065509
  94. 94. Khurana S, George SP. Regulation of cell structure and function by actin-binding proteins: Villin’s perspective. FEBS Letters. 2008. pp. 2128–2139. pmid:18307996
  95. 95. Costa de Beauregard M a, Pringault E, Robine S, Louvard D. Suppression of villin expression by antisense RNA impairs brush border assembly in polarized epithelial intestinal cells. EMBO J. 1995;14: 409–21. Available: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=398099&tool=pmcentrez&rendertype=abstract pmid:7859732
  96. 96. Kumar N, Zhao P, Tomar A, Galea CA, Khurana S. Association of Villin with Phosphatidylinositol 4,5-Bisphosphate Regulates the Actin Cytoskeleton. J Biol Chem. 2004;279: 3096–3110. pmid:14594952
  97. 97. Tomar A, George SP, Mathew S, Khurana S. Differential effects of lysophosphatidic acid and phosphatidylinositol 4,5-bisphosphate on actin dynamics by direct association with the actin-binding protein villin. J Biol Chem. 2009;284: 35278–35282. pmid:19808673
  98. 98. Reyes JL, Terrazas LI, Espinoza B, Cruz-Robles D, Soto V, Rivera-Montoya I, et al. Macrophage migration inhibitory factor contributes to host defense against acute Trypanosoma cruzi infection. Infect Immun. American Society for Microbiology (ASM); 2006;74: 3170–9. pmid:16714544
  99. 99. Flores M, Saavedra R, Bautista R, Viedma R, Tenorio EP, Leng L, et al. Macrophage migration inhibitory factor (MIF) is critical for the host resistance against Toxoplasma gondii. FASEB J. The Federation of American Societies for Experimental Biology; 2008;22: 3661–71. pmid:18606868
  100. 100. Satoskar AR, Bozza M, Rodriguez Sosa M, Lin G, David JR. Migration-Inhibitory Factor Gene-Deficient Mice Are Susceptible to Cutaneous Leishmania major Infection. Infect Immun. 2001;69: 906–911. pmid:11159984
  101. 101. Stijlemans B, Leng L, Brys L, Sparkes A, Vansintjan L, Caljon G, et al. MIF contributes to Trypanosoma brucei associated immunopathogenicity development. Mitre E, editor. PLoS Pathog. 2014;10: e1004414. pmid:25255103
  102. 102. Cavalcanti MG, Mesquita JS, Madi K, Feijó DF, Assunção-Miranda I, Souza HSP, et al. MIF participates in Toxoplasma gondii-induced pathology following oral infection. Fritz JH, editor. PLoS One. 2011;6: e25259. pmid:21977228
  103. 103. Santos-Oliveira JR, Regis EG, Leal CRB, Cunha R V., Bozza PT, Da-Cruz AM. Evidence That Lipopolisaccharide May Contribute to the Cytokine Storm and Cellular Activation in Patients with Visceral Leishmaniasis. Carvalho EM, editor. PLoS Negl Trop Dis. 2011;5: e1198. pmid:21765960
  104. 104. Li E, Tako EA, Singer SM. Complement Activation by Giardia duodenalis Parasites through the Lectin Pathway Contributes to Mast Cell Responses and Parasite Control. Appleton JA, editor. Infect Immun. American Society for Microbiology (ASM); 2016;84: 1092–9. pmid:26831470
  105. 105. Deguchi M, Gillin FD, Gigli I. Mechanism of killing of Giardia lamblia trophozoites by complement. J Clin Invest. 1987;79: 1296–302. pmid:3646264
  106. 106. Hill DR, Burge JJ, Pearson RD. Susceptibility of Giardia lamblia trophozoites to the lethal effect of human serum. J Immunol. 1984;132: 2046–52. Available: http://www.ncbi.nlm.nih.gov/pubmed/6699407 pmid:6699407
  107. 107. Dunkelberger JR, Song W-C. Complement and its role in innate and adaptive immune responses. Cell Res. Nature Publishing Group; 2010;20: 34–50. pmid:20010915
  108. 108. Yu LCH, Huang C, Kuo W, Sayer H, Turner JR, Buret AG. SGLT-1-mediated glucose uptake protects human intestinal epithelial cells against Giardia duodenalis-induced apoptosis. Int J Parasitol. 2008;38: 923–934. pmid:18281046
  109. 109. Mahmood S, Sodhi CP, Ganguly NK. Expression of sodium-glucose co-transporter and brush border disaccharidases in Giardia lamblia infected rat intestine. Indian J Biochem Biophys. 2002;39: 185–90. Available: http://www.ncbi.nlm.nih.gov/pubmed/22905389 pmid:22905389
  110. 110. Koh WH, Geurden T, Paget T, O’Handley R, Steuart RF, Thompson RCA, et al. Giardia duodenalis assemblage-specific induction of apoptosis and tight junction disruption in human intestinal epithelial cells: effects of mixed infections. J Parasitol. 2013;99: 353–8. pmid:22924932
  111. 111. Panaro MA, Cianciulli A, Mitolo V, Mitolo CI, Acquafredda A, Brandonisio O, et al. Caspase-dependent apoptosis of the HCT-8 epithelial cell line induced by the parasite Giardia intestinalis. FEMS Immunol Med Microbiol. 2007;51: 302–309. pmid:17714487
  112. 112. Ono SJ, Nakamura T, Miyazaki D, Ohbayashi M, Dawson M, Toda M, et al. Chemokines: roles in leukocyte development, trafficking, and effector function. J Allergy Clin Immunol. Garland, New York; 2003;111: 1185–99; quiz 1200.
  113. 113. Lee H-YH-Y, Hyung S, Lee NY, Yong T-ST-S, Han S-HS-H, PARK S-JS-J. Excretory-secretory products of Giardia lamblia induce interleukin-8 production in human colonic cells via activation of p38, ERK1/2, NF-κB and AP-1. Parasite Immunol. 2012;34: 183–98. pmid:22224945
  114. 114. Patterson KI, Brummer T, O’brien PM, Daly RJ. Dual-specificity phosphatases: critical regulators with diverse cellular targets. Biochem J. 2009;418. Available: http://www.biochemj.org/content/418/3/475.long
  115. 115. Guan KL, Butch E. Isolation and characterization of a novel dual specific phosphatase, HVH2, which selectively dephosphorylates the mitogen-activated protein kinase. J Biol Chem. American Society for Biochemistry and Molecular Biology; 1995;270: 7197–203.
  116. 116. Ishibashi T, Bottaro DP, Michieli P, Kelley CA, Aaronson SA. A novel dual specificity phosphatase induced by serum stimulation and heat shock. J Biol Chem. 1994;269: 29897–902. Available: http://www.ncbi.nlm.nih.gov/pubmed/7961985 pmid:7961985
  117. 117. Clark AR, Dean JLE. The control of inflammation via the phosphorylation and dephosphorylation of tristetraprolin: a tale of two phosphatases. Biochem Soc Trans. 2016;44: 1321–1337. pmid:27911715
  118. 118. Tiedje C, Diaz-Muñoz MD, Trulley P, Ahlfors H, Laaß K, Blackshear PJ, et al. The RNA-binding protein TTP is a global post-transcriptional regulator of feedback control in inflammation. Nucleic Acids Res. 2016;44: 7418–40. pmid:27220464
  119. 119. In JG, Foulke-Abel J, Estes MK, Zachos NC, Kovbasnjuk O, Donowitz M. Human mini-guts: new insights into intestinal physiology and host—pathogen interactions. Nat Rev Gastroenterol Hepatol. Nature Research; 2016;13: 633–642. pmid:27677718