arginine deiminase has multiple regulatory roles in the biology of...

9
2930 Research Article Introduction Giardia lamblia is a ubiquitous unicellular parasite of humans and other vertebrates that commonly causes diarrhea and gastrointestinal upset (Adam, 2001). G. lamblia trophozoites undergo fundamental biological changes in response to adverse environmental conditions. To survive outside the host’s intestine, Giardia trophozoites differentiate into infective cysts, which are then released with the feces and are responsible for the transmission of the disease among susceptible hosts. Another mechanism of adaptation is antigenic variation (a process by which the parasite continuously switches its major surface molecules), allowing the parasite to evade the host’s immune response while residing in the small intestine (Nash, 2002). Only one variant-specific surface protein (VSP), from a pool of approximately 250 genes present in the genome of the parasite, is expressed on the surface of G. lamblia trophozoites at any point in time (Morrison et al., 2007). These antigens are membrane proteins with common characteristics, such as their variable N-terminal extracellular region and a well-conserved C-terminus – which consists of a hydrophobic intramembranous domain that anchors the VSPs to the parasite surface as well as a perfectly conserved five-amino-acid (CRGKA) C-terminal tail located in the cytoplasm (Mowatt et al., 1991; Mowatt et al., 1994). However, the biological purpose of the conserved elements in VSPs and their role in antigenic variation are still unknown. The function of the CRGKA tail of VSPs is unclear, but recent evidence indicates that it is important in VSP biology. It has been previously shown that many VSPs are palmitoylated (Hiltpold et al., 2000; Papanastasiou et al., 1997b), and we recently established that palmitic acid binds specifically to the Cys of the CRGKA tail of the VSPs, thus controlling the segregation of these proteins to detergent-resistant domains at the plasma membrane (Touz et al., 2005). Consequently, VSP mutants unable to bind palmitic acid show abnormal membrane segregation and avoid VSP-specific- antibody cytotoxicity, performing an essential function in the control of VSP-mediated signaling and processing (Touz et al., 2005). These results, taken together, prompted us to investigate whether the CRGKA cytoplasmic tail contains other post- translational modifications or interacting proteins involved in the control of signaling. In this work, we found that G. lamblia arginine deiminase (ADI, encoded by ArcA) bound specifically to the CRGKA sequence. Prior studies in G. lamblia determined that ADI catalyzes the irreversible catabolism of arginine to citrulline in the arginine dehydrolase (ADH) pathway and serves as an important source of energy (Schofield et al., 1990). This pathway has been regarded as being restricted to prokaryotic organisms. By contrast, higher eukaryotes use nitric-oxide synthase (NOS) to convert free L-arginine into citrulline and nitric oxide (NO), and use peptidylarginine deiminases (PADs) to deiminate protein-bound arginine and convert it to citrulline (citrullination) by a Ca 2+ -dependent mechanism. This post- translational modification has a major impact on the structure and function of the target protein (Vossenaar et al., 2003). We discovered that the function of ADI goes beyond energy production because it also functions as a PAD. We present evidence indicating that ADI plays an essential role in the control of antigenic variation via VSP citrullination and influences the process of encystation. Our findings on the participation of ADI in unusual post-translational modification mechanisms during G. lamblia survival are discussed. Results ADI interacts with VSPs Almost all VSP genes encode the type-I integral membrane proteins that have a five-amino-acid cytoplasmic tail, CRGKA. Because this cytoplasmic sequence is highly conserved, we examined the The protozoan parasite Giardia lamblia uses arginine deiminase (ADI) to produce energy from free L-arginine under anaerobic conditions. In this work, we demonstrate that, in addition to its known role as a metabolic enzyme, it also functions as a peptidylarginine deiminase, converting protein-bound arginine into citrulline. G. lamblia ADI specifically binds to and citrullinates the arginine in the conserved CRGKA tail of variant-specific surface proteins (VSPs), affecting both antigenic switching and antibody-mediated cell death. During encystation, ADI translocates from the cytoplasm to the nuclei and appears to play a regulatory role in the expression of encystation-specific genes. ADI is also sumoylated, which might modulate its activity. Our findings reveal a dual role played by ADI and define novel regulatory pathways used by Giardia for survival. Key words: Arginine deiminase, Citrullination, Sumoylation, Antigenic variation, Encystation, Gene regulation Summary Arginine deiminase has multiple regulatory roles in the biology of Giardia lamblia Maria Carolina Touz 1, * ,‡ , Andrea Silvana Rópolo 1, *, Maria Romina Rivero 1 , Cecilia Veronica Vranych 1 , John Thomas Conrad 2 , Staffan Gunnar Svard 3 and Theodore Elliott Nash 2 1 Instituto de Investigación Medica Mercedes y Martín Ferreyra, INIMEC – CONICET, Friuli 2434, Cordoba, Argentina 2 Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA 3 Department of Cell and Molecular Biology, Uppsala University, SE-75 124 Uppsala, Sweden. *These authors contributed equally to this work Author for correspondence (e-mail: [email protected]) Accepted 16 June 2008 Journal of Cell Science 121, 2930-2938 Published by The Company of Biologists 2008 doi:10.1242/jcs.026963 Journal of Cell Science

Upload: dinhkiet

Post on 03-May-2019

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2930 Research Article

IntroductionGiardia lamblia is a ubiquitous unicellular parasite of humans andother vertebrates that commonly causes diarrhea and gastrointestinalupset (Adam, 2001). G. lamblia trophozoites undergo fundamentalbiological changes in response to adverse environmental conditions.To survive outside the host’s intestine, Giardia trophozoitesdifferentiate into infective cysts, which are then released with thefeces and are responsible for the transmission of the disease amongsusceptible hosts. Another mechanism of adaptation is antigenicvariation (a process by which the parasite continuously switchesits major surface molecules), allowing the parasite to evade the host’simmune response while residing in the small intestine (Nash, 2002).Only one variant-specific surface protein (VSP), from a pool ofapproximately 250 genes present in the genome of the parasite, isexpressed on the surface of G. lamblia trophozoites at any point intime (Morrison et al., 2007). These antigens are membrane proteinswith common characteristics, such as their variable N-terminalextracellular region and a well-conserved C-terminus – whichconsists of a hydrophobic intramembranous domain that anchorsthe VSPs to the parasite surface as well as a perfectly conservedfive-amino-acid (CRGKA) C-terminal tail located in the cytoplasm(Mowatt et al., 1991; Mowatt et al., 1994). However, the biologicalpurpose of the conserved elements in VSPs and their role inantigenic variation are still unknown.

The function of the CRGKA tail of VSPs is unclear, but recentevidence indicates that it is important in VSP biology. It has beenpreviously shown that many VSPs are palmitoylated (Hiltpold etal., 2000; Papanastasiou et al., 1997b), and we recently establishedthat palmitic acid binds specifically to the Cys of the CRGKA tailof the VSPs, thus controlling the segregation of these proteins todetergent-resistant domains at the plasma membrane (Touz et al.,2005). Consequently, VSP mutants unable to bind palmitic acid

show abnormal membrane segregation and avoid VSP-specific-antibody cytotoxicity, performing an essential function in thecontrol of VSP-mediated signaling and processing (Touz et al.,2005). These results, taken together, prompted us to investigatewhether the CRGKA cytoplasmic tail contains other post-translational modifications or interacting proteins involved in thecontrol of signaling.

In this work, we found that G. lamblia arginine deiminase (ADI,encoded by ArcA) bound specifically to the CRGKA sequence. Priorstudies in G. lamblia determined that ADI catalyzes the irreversiblecatabolism of arginine to citrulline in the arginine dehydrolase(ADH) pathway and serves as an important source of energy(Schofield et al., 1990). This pathway has been regarded as beingrestricted to prokaryotic organisms. By contrast, higher eukaryotesuse nitric-oxide synthase (NOS) to convert free L-arginine intocitrulline and nitric oxide (NO), and use peptidylarginine deiminases(PADs) to deiminate protein-bound arginine and convert it tocitrulline (citrullination) by a Ca2+-dependent mechanism. This post-translational modification has a major impact on the structure andfunction of the target protein (Vossenaar et al., 2003). We discoveredthat the function of ADI goes beyond energy production becauseit also functions as a PAD. We present evidence indicating that ADIplays an essential role in the control of antigenic variation via VSPcitrullination and influences the process of encystation. Our findingson the participation of ADI in unusual post-translationalmodification mechanisms during G. lamblia survival are discussed.

ResultsADI interacts with VSPsAlmost all VSP genes encode the type-I integral membrane proteinsthat have a five-amino-acid cytoplasmic tail, CRGKA. Because thiscytoplasmic sequence is highly conserved, we examined the

The protozoan parasite Giardia lamblia uses arginine deiminase(ADI) to produce energy from free L-arginine under anaerobicconditions. In this work, we demonstrate that, in addition to itsknown role as a metabolic enzyme, it also functions as apeptidylarginine deiminase, converting protein-bound arginineinto citrulline. G. lamblia ADI specifically binds to andcitrullinates the arginine in the conserved CRGKA tail ofvariant-specific surface proteins (VSPs), affecting both antigenicswitching and antibody-mediated cell death. During encystation,

ADI translocates from the cytoplasm to the nuclei and appearsto play a regulatory role in the expression of encystation-specificgenes. ADI is also sumoylated, which might modulate itsactivity. Our findings reveal a dual role played by ADI anddefine novel regulatory pathways used by Giardia for survival.

Key words: Arginine deiminase, Citrullination, Sumoylation,Antigenic variation, Encystation, Gene regulation

Summary

Arginine deiminase has multiple regulatory roles inthe biology of Giardia lambliaMaria Carolina Touz1,*,‡, Andrea Silvana Rópolo1,*, Maria Romina Rivero1, Cecilia Veronica Vranych1,John Thomas Conrad2, Staffan Gunnar Svard3 and Theodore Elliott Nash2

1Instituto de Investigación Medica Mercedes y Martín Ferreyra, INIMEC – CONICET, Friuli 2434, Cordoba, Argentina2Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD 20892, USA3Department of Cell and Molecular Biology, Uppsala University, SE-75 124 Uppsala, Sweden. *These authors contributed equally to this work‡Author for correspondence (e-mail: [email protected])

Accepted 16 June 2008Journal of Cell Science 121, 2930-2938 Published by The Company of Biologists 2008doi:10.1242/jcs.026963

Jour

nal o

f Cel

l Sci

ence

Page 2: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2931Multiple roles of ADI in Giardia

possibility of CRGKA interaction with regulatory proteins. Pull-down assays using the synthesized peptide NH2-HHHHHHCRGKA-COO– (His6-CRGKA) and further liquidchromatography with tandem mass spectrometry (LC-MS/MS)analysis showed an associated protein corresponding to the enzymearginine deiminase (ADI; GGD 112103) in both isolate GS cloneH7 (GS/H7) and isolate WB clone 1267 (WB/1267) (Fig. 1A). The

85 kDa band corresponded to the modified whole protein (seebelow), and the ~13 kDa band to the protein C-terminus (Fig. 1A).In addition, using the yeast two-hybrid system, we determined thatthe entire VSPH7 [H7-BD (binding domain)] and VSP1267 (1267-BD) proteins, but not VSP1267 lacking the cytoplasmic tailCRGKA (1267-tail-BD), interacted with ADI [ADI-AD (activationdomain)], thus validating the VSP-ADI interaction found by thepeptide pull-down assays (Fig. 1B).

To confirm the interaction of ADI with the tail of VSPs, wecreated WB-strain transgenic trophozoites that stably expresseddifferent VSPH7 versions [H7-HA is VSPH7 with a hemagglutin(HA) epitope sequence at the C-terminus; H7-tail is VSPH7 witha HA epitope right after its transmembrane domain; and ΔR-H7 isVSPH7 containing a point mutation of the R residue of itscytoplasmic tail] (Fig. 1C) and performed immunoprecipitation(IPP) assays. VSPH7 is not genetically codified nor expressed inthe G. lamblia WB/1267, thereby allowing detection of theextracellular portion of VSPH7 using VSPH7-specific monoclonalantibody (mAb) G10/4 and allowing detection of the C-terminusby using anti-HA mAb. Our results showed that H7-HA and ΔR-H7, but not H7-tail, co-immunoprecipitate with ADI polyclonalantibody (pAb) (Fig. 1D). This suggests that the whole tail of theVSPs rather than one amino acid is necessary for ADI-VSPinteraction, as expected from an enzyme-substrate association.

ADI acts as a PADUsing a C-terminal HA-tagged ADI (ADI-HA) andimmunofluorescence assay (IFA), we found that the subcellularlocalization of ADI-HA is cytoplasmic (Fig. 2A). We next examinedwhether ADI localizes close to the tail of VSPs. Confocal imagingshowed that ADI partially colocalizes with VSP9B10 close to theplasma membrane (Fig. 2, merge, top panel). Similar results werefound when anti-ADI pAb, raised against recombinant ADI protein(Ringqvist et al., 2008) was used, thereby confirming thecytoplasmic localization of ADI as well as its partial colocalizationwith VSP9B10 (Fig. 2A, bottom panels).

Two observations suggested that ADI possesses PAD activity.First, by sequence analysis and 3D-structure prediction, we foundthat G. lamblia ADI contains a ‘Cys-His-Glu’ active site similar tothe PADs (data not shown). Second, we discovered that ADI bindsto the cytoplasmic tail of VSPs, a finding not expected of an enzymeinvolved exclusively in the energy pathway. To determine whetherVSPs are citrullinated, western blotting using extracts from clonedtrophozoites expressing VSP1267, VSP9B10 (both from isolateWB), or VSPH7 (from isolate GS) was performed. Fig. 2B showsthat only the full-length VSP1267, VSP9B10 and VSPH7 proteinsare citrullinated. It was previously shown that VSPs are processedby deletion of the conserved C-terminus (Papanastasiou et al.,1997a; Touz et al., 2005). This event explains why the processedN-terminus (Fig. 2B, lower band of VSP1267 and VSPH7), whichis also recognized by their specific mAbs, is not citrullinated.

To demonstrate that ADI possesses PAD activity, we mixed activeADI-HA, purified from transgenic G. lamblia trophozoites, andHis6-CRGKA. After His6-CRGKA purification and westernblotting, we were able to demonstrate that ADI, but not a non-relatedenzyme, citrullinates the His6-CRGKA peptide (Fig. 2C). Therequirements for PAD activity of ADI to citrullinate the VSP tailwere determined by analysis of transfected VSPH7 mutants for thepresence of citrulline. Citrullination was found in H7-HA, but notin H7-tail or ΔR-H7, indicating that citrullination could not occurwithout the presence of the crucial arginine in the CRGKA tail (Fig.

Fig. 1. G. lamblia ADI is associated with VSPs through their cytoplasmic tail.(A) Left: representation of the pull-down assays. Right: SDS-PAGE andCoomassie staining show an ~85 kDa and an ~13 kDa protein from bothGS/H7 (GS) and WB/1267 (WB) isolates. Identification of these bands asbeing ADI was performed by LC/MS-MS. Controls without peptide fail to pulldown any protein. (B) Protein-protein interactions were detected by the abilityof yeast cells (AH109) to grow on selective plates. In the upper-left panel, theexpression of the entire VSPH7 protein (H7-BD), the entire VSP1267 protein(1267-BD) and VSP1267 lacking its cytoplasmic tail (1267-tail-BD) with ADI(ADI-AD) is revealed by the presence of white colonies in minimal mediumlacking leucine and tryptophan (–L/–T medium). Interaction of ADI-AD withboth VSPH7-BD and VSP1267-BD is shown in the bottom-left panel by thegrowth of yeast colonies in plates lacking tryptophan, leucine and histidine[TDO (triple-dropout medium) plates]. No interaction between ADI and 1267-tail-BD is observed. Controls of the methodology include ESCP-BD–MuA-AD interaction (+) and emptyBD–ADI-AD vector (–). (C) Schematicrepresentation of wild-type VSPH7 and transgenic VSPH7 proteins. TheVSPH7 ORF contains a signal peptide, an extracellular domain, atransmembrane domain and a cytoplasmic tail. H7-HA has a HA epitopesequence at the C-terminus. H7-tail possesses the HA epitope right after thetransmembrane domain of H7. ΔR-H7 is VSPH7 containing a point mutationof the R residue of its cytoplasmic tail. (D) H7-HA and ΔR-H7, but not H7-tail, co-immunoprecipitate with ADI. Antibody against ADI was used toimmunoprecipitate comparable amounts of protein from WB transgenic cells.Western blotting of original lysate was stained with anti-HA mAb labeled withalkaline phosphatase (Lysate, bottom panel). UT, untransfected cells; STD,molecular weight standard.

Jour

nal o

f Cel

l Sci

ence

Page 3: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2932

2D). As shown in the bottom panel of Fig. 2D, equal amounts oftransgenic VSPs were detected after IPP.

ADI participates in the control of antigenic variationAntibodies directed against VSPs either induce antigenic variationin, inhibit the growth of or kill recognized trophozoites, allowingthe repopulation of trophozoites expressing other VSPs. In vitro,results depend either on the concentration of the antibodies addedto G. lamblia cultures or the exposure time. It has been observedthat a high concentration of the antibodies cause immediateimmobilization as well as detachment and aggregation of thetrophozoites. By contrast, it was reported that, when a lowconcentration of anti-VSP antibody is added to the culture (or duringa short time), no cytotoxicity is observed, but a rapid accumulationof parasites expressing VSPs that are different to the one that was

Journal of Cell Science 121 (17)

originally expressed arise (Aggarwal et al., 1989; Stager et al., 1997;Touz et al., 2005).

To determine whether citrullination of VSPs abrogates antibody-mediated cytotoxicity, we used specific VSPH7 Abs and WB/1267transfected trophozoites, which expressed ~100% VSPH7 mutants

Fig. 2. VSP citrullination is probably mediated by the PAD activity of ADI.(A) Top panels: confocal direct IFA was performed on permeabilized cells,showing a cytoplasmic distribution of ADI-HA (green) by using FITC-labeledanti-HA mAb and its partial colocalization (yellow in merge) with VSP9B10(red) underneath the plasma membrane of the transgenic trophozoite. Nucleiare stained with DAPI (blue). Bottom panels: the same result was obtainedusing Alexa-Fluor-488–anti-ADI (green) in wild-type cells. Texas-Red–9B10mAb was used to visualize VSP9B10 (red). Scale bars: 10 μm. (B) Westernblotting of G. lamblia homogenates expressing different VSPs that reactedwith anti-citrulline pAb (left panel). The same filter membranes were stripped,cut and reacted with 5C1, 9B10 and G10/4 mAbs against VSP1267, VSP9B10and VSPH7, respectively (right panel), indicating that these VSPs arecitrullinated. STD, molecular weight standard. (C) Dot-blotting to detectcitrullination of the H6-CRGKA peptide after incubation with the purifiedrecombinant ADI-HA (ADI-HA). A non-related purified enzyme ESCP-HAwas used as a negative control. Dot-blotting to detect H6-CRGKA wasperformed using anti-H6 mAb. (D) Top panel: specific citrullination of theCRGKA tail is shown. Western blotting using anti-citrulline pAb performedafter immunoprecipitation with anti-HA mAb of H7-transgenic trophozoites.Bottom panel: the presence of H7-HA, H7-tail and ΔR-H7 afterimmunoprecipitation was analyzed using anti-HA mAb labeled with alkalinephosphatase. UT, untransfected cells.

Fig. 3. ADI participates in the control of cell death, probably by altering VSPswitching. (A) Cytotoxicity assays of G. lamblia trophozoites WB/1267,GS/H7, WB/1267 transfected with H7-HA or WB/1267 transfected with ΔR-H7 (from left to right on x axis) analyzed after 24 hours post-addition of theanti-VSPH7-specific antibodies G10-4 and anti-GS-VSPs pAb by estimatingthe number of adherent viable parasites. Controls include cells withouttreatment (w/o mAb) and the use of a non-related mAb (8G8 mAb). Datarepresent the means ± s.d. for n=2 of three independent experiments.(B) Progenies were analyzed by addition of goat anti-mouse FITC-conjugatedantibody in IFA. Positive cells correspond to trophozoites expressing VSPH7.DIC, differential interference contrast. Scale bars: 10 μm. (C) VSP expressionis established in WB/9B10 wild-type and WB/9B10-ADI+ transgenictrophozoites after a short time exposure to specific VSP9B10 mAb. Controlshad no exposure to the mAb (–). Data represent the means ± s.d. for n=3 oftwo independent experiments. (D) VSP9B10 citrullination is analyzed bywestern blotting in WB/9B10 and WB/9B10-ADI+ trophozoites after a shorttime exposure to the anti-VSP9B10 mAb. The membrane labeled with anti-citrulline pAb (right) was stripped and re-blotted with anti-VSP9B10 mAb(left). A similar amount of loaded VSP9B10 is observed. Molecular mass ofVSP9B10 is indicated on the right.

Jour

nal o

f Cel

l Sci

ence

Page 4: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2933Multiple roles of ADI in Giardia

(Fig. 1C). Controls for cytotoxicity effects included a GS/H7 cloneexpressing ~100% VSPH7 (+ control), and wild-type WB/1267expressing VSP1267 and a non-specific antibody (8G8 mAb) (–controls). An equal surface expression of H7-HA and ΔR-H7 wascontrolled by selecting trophozoites with the same level of surfacefluorescence using a fluorescence-activated cell sorter (FACS) andanti-VSPH7 antibodies (data not shown). Fig. 3A shows that, afteraddition of anti-VSPH7 mAb (G10/4) or pAb (anti-GS/VSP),immobilization, aggregation, detachment and complement-independent cytotoxicity was observed for the native GS/H7 cloneand for H7-HA-transgenic trophozoites. These demonstrated cell-survival rates lower than 5% after 24 hours. By contrast, recoveryof the non-citrullinated ΔR-H7 mutant expressing trophozoitesexposed to the anti-VSPH7 Abs showed no cytotoxicity after 24hours compared to controls. As expected, WB/1267 trophozoitesexposed to anti-VSPH7 Abs and the samples exposed to 8G8 mAbshowed no cytotoxicity (Fig. 3A).

Analysis of the progeny revealed that the surviving GS/H7 (notshown) and H7-HA trophozoites were nearly all VSPH7 negative,whereas the surviving ΔR-H7 trophozoites were almost 100%VSPH7 positive (Fig. 3B). These findings strongly suggest that,similar to palmitoylation, VSP citrullination plays an essential rolein G. lamblia survival (Touz et al., 2005) (this study).

Because an inability to citrullinate VSPs results in decreasedantigenic switching under Ab pressure, increased citrullinationmight result in enhanced antigenic switching in the same situation.To test this hypothesis, wild-type isolate WB clone 9B10trophozoites (WB/9B10) and WB/9B10 transgenic trophozoitesoverexpressing active ADI (WB/9B10-ADI+) were exposed to thespecific anti-VSP9B10 mAb (not used for controls) for only 10minutes, with the cytotoxicity as well the VSP switching beinganalyzed at 24 hours. As expected, no cytotoxicity was observed.However, a significant increase of VSP switching was seen in thetransfected trophozoites, with this being accompanied by enhancedcitrullination (Fig. 3C,D). Similar results were also obtained whenwild type (WB/1267wt) and WB/1267-ADI+ were tested (notshown). The VSP switching was relatively frequent and even smallpopulations of recently cloned organisms contained parasitesexpressing different VSPs. Related to this, we observed thattrophozoites that highly expressed ADI switched faster than wild-type cells under antibody pressure. These results could not beexplained by differences in growth rates because transfected andnon-transfected trophozoites multiply at the same speed.

ADI is post-translationally modifiedADI breakdown products were reported previously (Palm et al.,2003). In this work, analysis of protein expression of either ADI-HA or the native ADI showed that this protein is highly unstableand/or post-translationally modified, because several bandswere observed, including a major 85 kDa band instead of thepredicted 64 kDa band (Fig. 4A). Analysis of ADI failed tofind probable glycosylation, myristolation, prenylation orglycoinositolphospholipid-binding sites. However, phosphorylationwas predicted in several positions and we also found that ADIpossessed two motifs with a high probability of modification bythe 11 kDa SUMO1 protein (small ubiquitin-related modifier, alsocalled Sentrin) at positions K101 (LKYE) and K387 (IKAD). Westernblot assays using anti-SUMO1 mAb detected an 85 kDa proteincorresponding to the ADI higher band (Fig. 4B). In addition, anti-SUMO1 mAb was able to immunoprecipitate the 85 kDa band ofADI, thus confirming the ADI-SUMO1 interaction (Fig. 4C). These

results suggest that G. lamblia SUMO1 binds to ADI, probably tothe K101 and K387 residues, thereby explaining why the ADImolecular mass increased from the predicted 64 kDa to 85 kDa.Nevertheless, the role of sumoylation on the modification of ADIand the biological function of this modification during G. lambliagrowth and differentiation are the subject of on-going experiments.

ADI might play an additional role during G. lamblia encystationPost-translational modifications of proteins occur and are importantin many cellular processes, including cell-cycle progression,apoptosis, cellular proliferation and development, and have morerecently been found in cell differentiation (Dohmen, 2004; Leightet al., 2005; Poulin et al., 2005; Shalizi et al., 2006). These findingsprompted us to investigate whether ADI is involved in the processof encystation, the developmental process that a trophozoite goesthrough to turn it into a cyst. This process is initiated whentrophozoites are deprived of cholesterol or challenged with high bileconcentrations, resulting in the appearance of large secretory granulescalled encystation-specific secretory vesicles (ESVs). These vesiclestransport cyst-wall materials (e.g. CWP1 and CWP2) for thesubsequent release and extracellular assembly of the rigid cyst wallthat protects the parasite outside its host (Fig. 5A). First, we

Fig. 4. Post-translational modifications of ADI. (A) Western blotting usinganti-HA mAb shows ADI-HA in ADI-transgenic trophozoites. Multiple bandsare also obtained using specific anti-ADI pAb in wild-type cells. The band of64-66 kDa corresponding to the predicted protein sequences is observed inboth cases (arrowhead), together with degradation products (gray arrows)including the low-weight band (gray arrow with asterisk) found in the peptidepull-down (see Fig. 1). Also, for both HA-tagged and native ADI, an increasein molecular mass from a 64-66 kDa to a 85 kDa band is shown (black arrow),suggesting that ADI undergoes post-translational modification. STD,molecular weight standard. (B) Western blot assays using anti-SUMO1 mAbrecognize an ~85 kDa band (arrow) in both WB/1267 (WB) and GS/H7 (GS)G. lamblia clones. The same filter membrane was stripped and re-blotted withanti-ADI pAb, showing a perfect match with the higher band that isrecognized by the anti-SUMO1 mAb. To confirm the lack of residual primaryantibodies after stripping, only the secondary antibody was added to thestripped blots, showing no signal (Control). (C) Western blotting using biotin-conjugated anti-ADI pAb was performed to detect ADI bands afterimmunoprecipitation with anti-SUMO1 mAb. (a) ADI in lysate before IPP; (b)ADI after immunoprecipitation by using 0.1 μg of anti-SUMO1 mAb (arrow);(c) ADI after immunoprecipitation by using 1 μg of anti-SUMO1 mAb (arrow);(d) control using a non-related anti-HA mAb; and (e) supernatant of sample c.

Jour

nal o

f Cel

l Sci

ence

Page 5: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2934

characterized ADI by analyzing the level of adi mRNA and proteinexpression in both wild-type (WB/1267wt) and ADI-transgenic(WB/1267-ADI+) trophozoites at 0, 6, 24 and 48 hours ofencystation. Slot-blot assays showed that there was a minor increasein adi mRNA during encystation, compared with cwp2 and the house-keeping gdh, in WB/1267wt (Fig. 5B, left panel). By contrast, ahigher expression of adi mRNA and a lower expression of cwp2mRNA were observed, whereas gdh mRNA levels remained equalduring encystation of WB/1267-ADI+ cells (Fig. 5B, right panel).

Semi-quantitative reverse transcriptase (RT)-PCR assayscorroborated the data obtained by slot-blotting. gdh mRNAexpression showed no variation during encystation for eitherWB/1267wt or WB/1267-ADI+ trophozoites. Furthermore, novariation in mRNA levels was detected for adi during encystationin either case. However, its expression was found to be 2.5-timeshigher in transgenic cells. cwp2 mRNA dramatically increased at

Journal of Cell Science 121 (17)

24 hours in wild-type cells, but not in ADI-transgenic trophozoites,suggesting that ADI inhibited cwp2 expression and therefore mightparticipate in the regulation of the cwp2 gene (Fig. 5C).

Western blotting using specific antibodies showed that, inWB/1267wt and WB/1267-ADI+ trophozoites, ADI levels increaseafter 24 hours of encystation. The expression of VSP1267 wasequivalent in both cell types (VSP1267) but the amount of citrullinedetected in WB/1267-ADI+ trophozoites was higher than inWB/1267wt trophozoites (also demonstrated in Fig. 3D).Surprisingly, although ADI expression increased, VSP1267citrullination was not modified during encystation (Fig. 5D, Cit).As expected, CWP2 rose in WB/1267wt trophozoites duringencystation (Fig. 5D, left panel). Conversely, there was a decreasein CWP expression to almost undetectable levels in WB/1267-ADI+trophozoites, validating a role of ADI in the regulation of CWP2(Fig. 5D, right panel). Generally, WB/1267 trophozoites differentiateinto cysts in about 4-8% of total cells after 48 hours in encystingmedium. Our results showed that overexpression of ADI reducedthe number of cysts produced (approximately 0.1%), compared withwild type (not shown). Even though the amount of formed cyst isdrastically reduced in transgenic trophozoites that highly expressADI, in cells expressing ADI at low levels, the kinetics ofdifferentiation is unaltered and the formed cysts are resistant to harshenvironmental conditions (C.V.V. and A.S.R., unpublished results).

Because Lys residues are also the target for ubiquitylation, it ispossible that ADI sumoylation functions as an inhibitor of ADIproteolysis during differentiation. Another possibility is that theaddition of SUMO1 proteins to ADI allows cytoplasm-nucleitranslocation (Ishov et al., 1999). To test this last hypothesis, thepresence of sumoylated ADI was analyzed in nuclear andcytoplasmic fractions both before and during encystation, bywestern blotting. In wild-type trophozoites, the ADI 85 kDa bandincreased in the nuclear fraction during encystation (Fig. 6A). Totest for cytoplasmic contamination of nuclear fractions, the presenceof VSP1267, a protein not found in the nucleus, was investigatedby western blotting and was undetected in these fractions (Fig. 6A).

Confirmation of the nuclear translocation of ADI was obtainedby IFA in WB/1267wt (Fig. 6A,B) and ADI-HA-transfectedtrophozoites (Fig. 6C), both demonstrating a shift of ADI to thenuclei during encystation. Wild-type organisms only showed thepresence of nuclear ADI late during encystation (when the cellscontained a large number of ESVs) (Fig. 6A,B). Interestingly, thosetransgenic organisms showing a clear translocation of ADI-HA tothe nuclei had few, if any, ESVs (Fig. 6C). The failure of ESVdevelopment together with the decrease in cwp2 mRNA intransfected encysting cells suggest that ADI has a role in the controlof the encystation process.

DiscussionPrevious studies determined that ADI plays a role in the energymetabolism in G. lamblia. In the present work, we show anadditional ADI action that affects or controls essential biologicalactivities of the organism. There are four major findings. First, weestablish that ADI acts as a PAD in addition to its known functionas a metabolic enzyme. Second, ADI influences important processesin G. lamblia. Specifically, by way of its ability to citrullinate VSPs,ADI alters the VSP biology, the response to cytotoxic antibodiesand the antigenic variation. Moreover, ADI appears to play animportant, if not controlling, role in encystation. Lastly, becauseADI is sumoylated, it is likely that this modification alters its activity,degradation and/or localization.

Fig. 5. ADI expression increases during G. lamblia differentiation. (A) IFA andconfocal microscopy show the G. lamblia encystation process. CWP2 (red) issynthesized and transported in ESVs in encysting trophozoites (ET, arrowhead).At the end of the encystation process, CWP2 is found in mature cyst walls(Cyst). Nuclei are stained with DAPI (blue). Scale bars: 10 μm. (B) Slot-blotting qualitatively shows gdh, adi and cwp2 gene expression at 0, 6, 24 and48 hours of encystation in wild-type cells (WB/1267wt) and transgenictrophozoites (WB/1267-ADI+). The assay was performed in triplicate.(C) Analysis of gdh, adi and cwp2 gene expression by RT-PCR and opticaldensity quantified by densiometry comparing both wild-type (WB/1267wt) andADI-transgenic (WB/1267-ADI+) trophozoites. Data represent the means ± s.d.for n=4 of two independent experiments. (D) Western blotting using specificantibodies shows ADI, modified citrulline (Cit), VSP1267 and CWP2 proteinexpression in both wild-type (WB/1267wt) and ADI-transgenic (WB/1267-ADI+) trophozoites. Non-encysting trophozoites (NT, 10 μg) and 24-hour-encysting trophozoites (ET, 10 μg) were used for each sample.

Jour

nal o

f Cel

l Sci

ence

Page 6: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2935Multiple roles of ADI in Giardia

The evidence supporting that VSPs are citrullinated by ADI iscompelling. ADI binds specifically to the CRGKA tail and localizesclose to the VSPs found on the plasma membrane of the trophozoite.Using specific antibodies to modified citrulline, VSPs were shownto be citrullinated specifically, at the arginine residue (R) of theCRGKA cytoplasmic tail. Also, purified ADI citrullinates the argininein the conserved tail in vitro, verifying the PAD activity of ADI. Thismodification has profound effects on the biology of the parasite.

The most commonly cited biological role of antigenic variationin pathogenic microorganisms is immunological escape, in whichthe host antibodies produced against a dominant antigen destroythose organisms bearing it, resulting in the organisms being replacedby ones that possess a variant form of the antigen. However, thefunction of antigenic variation among organisms differs and, in somecases, its analysis is complex. In G. lamblia, antibodies directedagainst VSPs – including VSP-specific surface-reacting mAbs (Nashand Aggarwal, 1986), immune lactogenic IgA (Stager et al., 1998)and serum from infected humans (Nash et al., 1990a; Nash et al.,1990b) or animals – either inhibit the growth of or kill recognizedtrophozoites (Hemphill et al., 1996; Stager and Muller, 1997), thusallowing the repopulation of trophozoites expressing other VSPs.The in vitro effects range from little or no inhibition of growth tocytotoxicity, and the effects appear to depend on the concentration(direct correlation), affinity, and even perhaps the nature of theantibody and target epitopes (Hemphill et al., 1996; Nash andAggarwal, 1986; Stager and Muller, 1997). By contrast, monovalentF(ab�) of the same antibodies exhibited none of these cytologicaleffects (Hemphill et al., 1996; Nash and Aggarwal, 1986), althoughthe VSP switching was not analyzed in these instances

Our results support the findings that exposure of trophozoitesto a high level of specific VSP Abs results in cell death and theemergence of trophozoites expressing an antigenically differentVSP. Most significantly, this event is strongly linked todeimination (citrullination) of the cytoplasmic tail of VSPs,because mutation of the amino acid arginine led to survival oftargeted trophozoites and failure to switch. Conversely, we havenow shown that an increase in VSP citrullination byoverexpression of ADI in the presence of the specific Ab causesderegulation of VSP switching, probably due to an amplificationof a signal-transduction event (Touz et al., 2005) (this study).Therefore, it is now clear that post-translational modifications areimportant for the control of the antigenic variation in G. lamblia.These studies are consistent with the hypothesis that bothimmunological and biological factors act in concert to select whichVSPs are expressed in a particular host.

The functional significance of ADI in cell survival appears notto be restricted to its role in energy production and antigenicvariation. We found that the subcellular localization of ADI in G.lamblia is cytoplasmic and is significantly close to the plasmamembrane but that it is upregulated and translocated to the nucleiwhen the trophozoites are induced to encyst. At least twopossibilities arise from this event: (1) as an arginine deiminase, ADImight be sequestered from the cytoplasm to enter in a ‘stand-by’process during encystation, because the requirement of energyduring this process is lower than that needed during active growth;(2) as a PAD, ADI might be directed into the nuclei for histonemodification and transcription regulation (Wang et al., 2004).However, the results presented in this work suggest that the secondpossibility is more likely. Post-translational histone modifications,such as phosphorylation, acetylation, methylation and citrullination,regulate a broad range of DNA- and chromatin-templated nuclearevents, including transcription (Jenuwein and Allis, 2001; Wang etal., 2004). Because acetylation was shown to be a G. lamblia histonepost-translational modification (Kulakova et al., 2006), we proposethat ADI modifies histones by citrullination, with this post-translational modification being involved in the downregulation ofthe encystation process. This hypothesis is based on the fact thattranslocation of the functionally overexpressed ADI to the nucleiavoids cyst formation. We also suggest that ADI causes genes in

Fig. 6. During encystation, ADI is translocated to the nuclei and inhibits CWPexpression. (A) Results of western blotting that was performed aftercytoplasm- and nuclear-fractionation assays using anti-ADI pAb at 24 and48 hours post-encystation induction in wild-type (ADI) cells. L, cell lysateprevious fractionation; C, cytoplasmic fraction; N, nuclear fraction (upperpanel). Middle panel: anti-VSP1267 mAb is used to detect cytoplasm- ornuclear-fraction contamination. Lower panel: a representative time course ofIFA shows ADI (red) distribution during encystation (ESVs in green).(B) Confocal microscopy and IFA using wild-type cells show that ADI (red)is translocated to the nuclei when the encysting cell is filled with ESVs(arrowheads). CWP1 is stained in green. (C) Top panels: representativedifferential interference contrast (DIC) and DAPI-staining images show astable ADI-transgenic culture (Merge). Middle panels: direct IFA shows thattrophozoites highly overexpressing ADI-HA (green) in the nuclei do notexpress CWP2 (red) during encystation (arrowheads in Merge). Bottompanels: closer analysis of three ADI-transgenic cells demonstrated that thoseexpressing ADI-HA (green) in the nuclei (arrowheads in Merge) do not revealCWP2 (red) expression after 24 hours of encystation, whereas those with lowADI-HA expression do (arrow in Merge). Scale bars: 10 μm.

Jour

nal o

f Cel

l Sci

ence

Page 7: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2936

the CWP family to turn off as an essential requirement tosuccessfully complete the encystation. This effect takes place earlyon in ADI-transgenic cells, in which the high overexpression ofADI together with its nuclear translocation avoids CWP expressionand cyst formation. Nevertheless, we cannot exclude the possibilitythat ADI is also involved in the control of its major surface antigenwhen its life cycle is completed, as was demonstrated by Svard etal. (Svard et al., 1998). Differentiation-associated switching ofsurface antigens could be one reason for the common occurrenceof repeated infections (Gilman et al., 1988) due to thedownregulation of the major VSP expressed during encystation andthe appearance of new VSPs after excystation.

ADI possesses two predicted nuclear localization signals(PQRRREQ and RRGIVMGQFQAPQRRRE) (SignalIP program)(Le Panse et al., 1997), suggesting that these motifs are involvedin the translocation of this protein to nuclei during encystation. Thereis, however, no evidence of the participation of these motifs in thenuclear protein translocation of any protein in this parasite. Thus,further analyses are needed to show how this parasite uses thenuclear signaling motifs that are highly conserved during evolution.Another possibility is that ADI is translocated to the nuclei by meansof SUMO1-protein binding. Recently, it was shown that conjugationof SUMO1 could lead to protein stabilization and protection fromdegradation, but an increasing body of evidence implicatessumoylation in the targeting of certain proteins to the cell nucleusand to the subnuclear structures (Dohmen, 2004). The underlyingmechanisms, however, are still largely unknown. In G. lamblia, themolecular biology of enzymes of the SUMO system has never beenaddressed. In fact, only three putative proteins involved insumoylation have been posted in the Giardia database: SUMO1(or Sentrin, GGD 7760), the dimer of the SUMO-activating enzymeE1 (Uba2, GGD 6288) and the SUMO ligase (GGD 17386).Our results from ‘in silico’ data, western blotting andimmunoprecipitation using anti-SUMO1 mAb indicate that ADI isa sumoylated protein. This protein modification might be essentialin the nuclear translocation of ADI in order to accomplish itsfunction during encystation, because the 85 kDa ADI was mainlylocated in the nuclear subfractions. Further research into thepossibility that ADI function could be regulated by sumoylation

Journal of Cell Science 121 (17)

might provide insight into the biology of this important intestinalparasite and also contribute to the understanding of the evolutionof protein modification in eukaryotic cells. Taking earlier studiesand this work together, we have been able to unravel the multiplefunctions of ADI in the survival of G. lamblia (Fig. 7). One of theseconcerns is the ability to obtain energy by using free arginine asthe preferential fuel under anaerobic conditions. During theproliferative stages of growth, ADI converts arginine into citrulline,with ATP production occurring at the final enzymatic step of theADH pathway (Schofield et al., 1992) (Fig. 7A). In addition to thisfunction, it was reported that G. lamblia uses ADI as a competitorto NOS from the host cell for the free arginine, thereby reducingthe production of NO and the host defense mechanism againstmicrobial infection (Eckmann et al., 2000). Supporting this finding,it was confirmed that ADI is released from G. lamblia to theextracellular space as a 66 kDa protein when the trophozoites arein contact with human colon epithelial cells (Ringqvist et al., 2008)(Fig. 7B). Also, during growth, ADI acts as a PAD on thecytoplasmic tail of VSPs, probably by functioning as a regulatorof antigenic variation (Fig. 7C). During encystation, ADI is locatedin the nuclei and cyst formation is reduced (Fig. 7D). Thistranslocation event is most probably associated with ADIsumoylation, but the possibility of the participation of ADI nuclear-localization signals cannot been discarded.

Changes in the environment (anaerobiosis, the presence of Abs,the presence of the host cell or depletion of cholesterol) define whichfunctions of ADI need to be performed at one point in time. Theunderlying mechanisms, however, are still largely unknown. Webelieve that the analysis of the protein modifications associated withdifferentiation, along with the mediators of these activities reportedhere, represent an important contribution to our understanding ofthe control of gene expression in parasitic protozoa.

Materials and MethodsG. lamblia cultivation and transfectionTrophozoites of the isolate WB, clone 1267 (WB/1267), 9B10 (WB/9B10) and fromthe isolate GS, clone H7 (GS/H7) were cultured in TYI-S-33 supplemented with 10%fetal bovine serum and bovine bile (Keister, 1983). Trophozoites of the WB/1267clone were transfected by electroporation and selected with puromycin (Singer et al.,1998; Yee and Nash, 1995). The transfection of stable WB/1267 cells was 100%, as

determined by IFA and flow cytometry. A two-step encystationprocedure was used by increasing the medium pH and by additionof porcine bile as reported by Boucher and Gillin (Boucher andGillin, 1990).

Pull-down assaysG. lamblia clones GS/H7 and WB/1267 were grown, harvestedand suspended in 1 ml of lysis buffer (50 mM NaH2PO4, 300 mMNaCl, 10 mM imidazol, pH 8.0, 1% Triton X-100, and proteaseinhibitors) for 3 hours at 4°C. After mild sonication using aBranson sonifier 250 (Branson, CT) with an output control of 3

Fig. 7. Schematic representation of the functions of ADIduring growth and encystation. (A) During growth, thetrophozoites acquire free arginine from the extracellularmedium. Inside the cell, cytoplasmic ADI converts arginineinto citrulline, with ATP production occurring at the finalenzymatic step of the ADH pathway. (B) ADI is released tothe extracellular space when the trophozoites are in contactwith human colon epithelial cells and compete with the hostNOS for the free arginine, thereby reducing the productionof NO. (C) Under low exposure to specific antibodies, ADIacts as a PAD on the cytoplasmic tail of VSP, inducing VSPswitching. (D) During the last step of encystation, ADI istranslocated from the cytoplasm to the nuclei, turningencystation-specific genes off and ending the process.

Jour

nal o

f Cel

l Sci

ence

Page 8: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

2937Multiple roles of ADI in Giardia

and a 50% duty cycle (sonication complex), the lysate was centrifuged at 7500 g,for 30 minutes at 4°C. The supernatant was then mixed with 1 mg of His6-CRGKApeptide dissolved in 10 μl of DMSO overnight at 4°C. Controls of specific bindinghad no His6-CRGKA peptide. The lysate and peptide were mixed with 200 μl of Ni-agarose beads (QIAGEN, Valencia, CA) and were incubated for 4 hours at 4°C. Beadswere spun down at 700 g and washed four times with wash buffer (50 mM NaH2PO4,300 mM NaCl, pH 8.0, 0.1% Triton X-100, and protease inhibitors). Bound proteinswere eluted four times with 100 μl elution buffer (50 mM NaH2PO4, 300 mM NaCl,250 mM imidazol, pH 8.0, 0.1% Triton X-100, and protease inhibitors). Peptide-bound proteins were analyzed by SDS-PAGE, stained with Coomassie G-250 (Fig.1A). The detected bands were cut out and submitted to the Research TechnologiesBranch for Protein Identification (NIAID, NIH) for LC-MS/MS analysis. After twoindependent experiments, one protein that was associated with the His6-CRGKApeptide was identified.

Yeast two-hybrid assayThe MATCHMAKER two-hybrid system was used following the protocol suggestedby the company (Clontech, Palo Alto, CA). The two-hybrid pGBKT7(TRP1) vector(GAL4 DNA-binding domain, BD) containing the genes for vsph7, vsp1267 orvsp1267 without the 3� (TGCAGAGGCAAGGCG) gene sequence were used as baitwhile the adi gene was ligated to the pGADT7-Rec(LEU2) vector (GAL4 transcriptionactivation domain, AD), yielding H7-BD, 1267-BD, 1267-tail-BD and ADI-ADvectors, respectively. AH109 transformants were cultured at 30°C for 4-5 days onplates with minimal medium lacking leucine and tryptophan (–L/–T) to test for positivetransformation, or in the absence of leucine, tryptophan and histidine (TDO – tripledropout medium) to study specific protein interactions as previously described (Touzet al., 2004). Controls of the methodology include ESCP-BD–MuA-AD interaction(Touz et al., 2004) and the emptyBD–ADI-AD vector.

Expression of VSPH7 variant and ADI-HA in WB/1267trophozoitesThe construction of VSPH7 and H7-tail was detailed previously (Touz et al., 2005).ΔR-H7 was prepared using a site-directed mutagenesis kit (QuikChange, Stratagene)and the presence of the mutation was confirmed by using dye terminator cyclesequencing (Beckman Coulter). For ADI-HA construction, the vsph7 gene wasreplaced by the adi gene using the forward 5�-GCCTCCATGGCTG ACT TCT -CCAAGGATAAAGAG-3� and 5�-GATGGTTAACCTTGATATCGACGCAGAT -GTC AGC-3� oligonucleotides yielding the ADI-HA vector. ADI-HA, H7-HA, H7-tail and ΔR-H7 vectors were used to transfect the WB/1267 clone in order to generatestable puromycin-selected cells.

AntibodiesAnti-HA mAb (Sigma), anti-HA mAb labeled with alkaline phosphatase (Sigma),anti-ADI pAb (Ringqvist et al., 2008), anti-H6 mAb (Invitrogen), anti-CWP1 FITC-labeled mAb (Warerborne, New Orleans, LA) and CWP2–Texas-Red (Touz et al.,2003) were used. Citrullinated proteins were detected by using a polyclonal antibodyagainst a chemically modified version of citrulline (anti-MC) (Senshu et al., 1992).To detect sumoylated proteins, mouse anti-GMP1 (anti-SUMO1) mAb (ZYMEDLaboratories, Invitrogen) was used. The mAbs 5C1, G10/4 and 9B10 were used todetect VSP1267, VSPH7 and VSP9B10, respectively (Mowatt et al., 1991; Nash etal., 2001; Nash and Mowatt, 1992). Anti-ADI pAb was biotinylated by using the EZ-Link NHS-SS-Biotin (Pierce, Rockford, IL) and detected in western blotting by usingalkaline-phosphatase-labeled streptavidin. For direct IFA, anti-HA FITC-labeled mAb(Sigma) VSP9B10–Texas-Red and anti-CWP2–Texas-Red (Touz et al., 2003) wereused.

Western blotting and IFATotal protein (10 μg) from parasite lysates was incubated with sample buffer, boiledfor 10 minutes and separated on 10% Bistris gels. Samples were transferred tonitrocellulose membranes, blocked with 5% skimmed milk and 0.1% Tween 20 inTBS, and incubated with primary antibody diluted in the same buffer. After incubationwith conjugated secondary antibody, proteins recognized by antibodies were visualizedwith SuperSignal West Pico chemiluminescent substrate (Pierce) and autoradiography,or by using BCIP/NBT substrate (Sigma). Controls included omission of the primaryantibody, use of an unrelated antibody or assays using non-transfected cells. In Fig.3D, the AutoDeblur v9.3 program was used (AutoDeblur v9.3, AutoQuant Imaging,NY).

For IFA of fixed cells, trophozoites cultured in growth medium were harvestedand processed as described (Touz et al., 2005). Briefly, parasites were attached toslides for 30 minutes and then fixed with fresh 4% formaldehyde for 40 minutes.The cells were incubated sequentially with blocking solution (10% goat serum and0.1% Triton X-100 in PBS) at 37°C for 30 minutes and antibodies conjugated withfluorescent dyes for 1 hour. The cells were then washed three times with PBS beforethe addition of mounting medium (VECTASHIELD Mounting Medium). Images werecollected on a Leica TCS-NT/SP confocal microscope (Leica Microsystems, Exton,PA) using 40� or 63� oil-immersion objectives (NA 1.32, zoom X). Fluorochromeswere excited using an argon laser at 488 nm for FITC and a krypton laser at 568 nmfor Texas Red 568. DAPI was excited using a 364 nm argon laser. Detector slits were

configured to minimize any crosstalk between the channels. Differential interferencecontrast (DIC) images were collected simultaneously with the fluorescence imagesby use of a transmitted-light detector. Images were processed using Leica TCS-NT/SP(version 1.6.587), Imaris 3.1.1 (Bitplane AG, Zurich, Switzerland) and AdobePhotoshop 5.5 (Adobe Systems) software.

Immunoprecipitation assay (IPP)Cultured G. lamblia trophozoites were harvested and resuspended in 1 ml of lysisbuffer (50 mM NaH2PO4, 300 mM NaCl, 1% Triton X-100, and protease inhibitors)for 1 hour at 4°C. The lysate was centrifuged at 10,000 g for 10 minutes at 4°C, andthe supernatant mixed with anti-ADI, anti-HA (1 μg) or anti-SUMO1 (0.2 or 1 μg)and incubated overnight at 4°C. Protein A-agarose beads (50 μl; Qiagen, Valencia,CA) was added to each sample before incubation for 4 hours at 4°C. Beads werepelleted at 700 g and washed four times with wash buffer (50 mM NaH2PO4, 300mM NaCl, pH 8.0, 0.1% Triton X-100, and protease inhibitors). Beads weresuspended in sample buffer and boiled for 10 minutes before western blot analysis.

Citrullination assayNon-encysting and trophozoites encysted at different time points were collected,washed in PBS, resuspended in 20 μl of lysis buffer without protease inhibitors (50 mMTris, pH 8.0, 120 nM NaCl, 1% Triton X-100), and incubated with 5 mM dithiothreitolfor 30 minutes at 37°C prior to being subjected to SDS-PAGE and nitrocelluloseblotting using the anti-citrulline (modified) detection kit (Upstate) as was previouslydescribed (Senshu et al., 1995). Controls included omission of the anti-citrullineantibody or absence of the chemically modifying citrulline step.

ADI-activity assayADI activity was assayed by measuring the modification of His6-CRGKA peptide toHis6-CcitGKA. First, trophozoites expressing ADI-HA or ESCP-HA (Touz et al.,2004) were cultured, collected and sonicated in PBS buffer at 4°C until there wereno intact cells before being centrifuged for 10 minutes at 700 g. ADI-HA and ESCP-HA were purified using anti-HA mAb and protein-A/G–agarose as previouslydescribed (Touz et al., 2004). 5 mM of soluble His6-CRGKA peptide (10 mg/ml ofADI activity buffer: 50 mM Tris-HCl pH 7.6, 2 mM DTT and 5 mM CaCl2) wasmixed with either purified ADI-HA or ESCP-HA for 16 hours at 50°C. After sampleswere boiled for 5 minutes, the peptide was purified by using Ni-agarose beads, andwas subjected to dot-blot and citrullination assays.

Cytotoxicity and switching assaysCytotoxicity was assayed as we previously reported (Touz et al., 2005). Briefly, G.lamblia trophozoites expressing native VSPH7 or transgenic VSPH7 at the sameintensity as surface VSPH7 were selected and harvested using CELLQuest softwareand a FACScalibur flow cytometer (Becton Dickinson, San Jose, CA). These cloneswere grown until confluence before a second IFA using G10-4 mAb was performedto test for 100% expression of VSPH7. A WB/1267 clone expressing almost 100%VSP1267 was used as a negative control. After addition of G10-4 mAb (specific forVSPH7), mouse polyclonal antiserum (anti-GS/VSP pAb, raised against GS/H7Giardia clone) or anti-CWP2-8G8 mAb (generated against ESVs) (Lujan et al., 1995)in 1:20 dilution, we analyzed immobilization, aggregation, detachment andcomplement-independent cytotoxicity. Cytotoxicity was determined in triplicate after24 hours by estimating the number of adherent viable parasites. The progeny wereanalyzed by growing the assayed trophozoites for 24 hours and by labeling theremaining H7-positive viable trophozoites by using goat anti-mouse FITC-conjugatedantibody (Cappel). For the VSP switching assay, Giardia ADI-transgenic and non-transgenic parasites expressing approximately 100% VSP9B10 or VSP1267 weremaintained in vitro before the addition of anti-VSP9B10 or anti-VSP1267 (5C1) mAbs,respectively, for only 10 minutes in a 1:20 dilution. VSP expression was analyzedby FACS as previously described (Nash et al., 2001). Controls consisted of identicalcultures not exposed to mAb.

Slot-blot assayTotal RNA (2 μg) extracted from growing and encysting trophozoites was immobilizedonto a Nytran SuperCharge nylon membrane (Schleicher & Schuell, Keene, NH) byusing the MINIFOLD II slot-blot system, according to the manufacturer’s instructions.Membranes were first hybridized with antisense probes specific for the adi gene, andthen stripped and reprobed by using an antisense specific for gdh or cwp2 as thecontrols. Oligonucleotides used: GDH (forward 5�-ATGCCTGCCCAGACGATCGA-3�, reverse 5�-GAGCCAGAAAGAAGGACGTT-3�), ADI (forward 5�-CACTTG -TGGAAATTACGTCT-3�, reverse 5�-CTTGATATCGACGCAGATGT-3�), ADI-HA(forward 5�-CACTTGTGGAAATTACGTCT-3�, reverse 5�-CTATGCATAGTCT -GGTACAT-3�) and CWP2 (forward 5�-ATGATCGCAGCCCTTGTTCT-3�, reverse5�-ATCCATCTCTCTCGAGAGTT-3�).

Semi-quantitative RT-PCRThe total RNA from wild-type and ADI-transgenic trophozoites was isolated usingRNA STAT- 60 reagent (Tel-Test, Friendswood, TX) followed by digestion with 50U DNAse (Roche Diagnostics, Indianapolis, IN) for 15 minutes at 25°C, and a finalpurification was performed by using SV Total RNA Isolation System (Promega,

Jour

nal o

f Cel

l Sci

ence

Page 9: Arginine deiminase has multiple regulatory roles in the biology of ...jcs.biologists.org/content/joces/121/17/2930.full.pdf · 2930 Research Article Introduction Giardia lamblia is

Madison, WI). For reverse transcription and PCR amplification, a one-step RT-PCRkit (Qiagen, Valencia, CA) was used with dilutions of the total RNA from 0.2 ng to20 ng per reaction in a final reaction volume of 50 μl. The reverse-transcription reactionwas done at 50°C for 30 minutes followed by inactivation of the reverse transcriptaseat 95°C for 15 minutes. For PCR, 30 cycles (30 seconds at 94°C, 30 seconds at 50°Cand 1 minute at 72°C) were used followed by 10 minutes at 72°C for final extension.DNA-contamination control was carried out by adding ADI oligonucleotides at thePCR step of the RT-PCR reaction. Aliquots (5 μl) of the RT-PCR reaction were size-separated on 1.2% agarose gel in TAE (E-Gel, Invitrogen Corporation, Carlsbad, CA)pre-stained with ethidium bromide. Amplification products were directly quantifiedby densitometric scanning of the fluorescence intensity under UV light, usingEagleSight software for image acquisition, documentation and analysis (Stratagene,La Jolla, CA). The oligonucleotides used were the same as the ones described forslot-blotting.

Subcellular fractionationNon-encysting and encysting Giardia trophozoites were suspended in buffer A (20mM Tris-HCl, pH 7.6, 5 mM MgCl2, 1.5 mM KCl, 1 mM phenylmethanesulfonylfluoride, 2 mM dithiothreitol and 0.1% Nonidet P-40) and disrupted with sonication.The homogenate was centrifuged at 760 g at 4°C for 10 minutes to separate it intosupernatant (cytoplasmic) and pellet (nuclear) fractions. The supernatant and pelletfractions were subjected to western blotting.

Adrian Hehl is acknowledged for his collaboration in the productionof anti-ADI pAb, and Alfredo Caceres and laboratory members forproviding numerous reagents. This work was partially supported bythe Agencia Nacional para la Promoción de la Ciencia y TecnologíaFONCyT Jóvenes PICT2004. This research was also supported in partby the Intramural Research Program of the National Institutes of Allergyand Infectious Diseases, National Institutes of Health.

ReferencesAdam, R. D. (2001). Biology of Giardia lamblia. Clin. Microbiol. Rev. 14, 447-475.Aggarwal, A., Merritt, J. W., Jr and Nash, T. E. (1989). Cysteine-rich variant surface

proteins of Giardia lamblia. Mol. Biochem. Parasitol. 32, 39-47.Boucher, S. E. and Gillin, F. D. (1990). Excystation of in vitro-derived Giardia lamblia

cysts. Infect. Immun. 58, 3516-3522.Dohmen, R. J. (2004). SUMO protein modification. Biochim Biophys Acta 1695, 113-

131.Eckmann, L., Laurent, F., Langford, T. D., Hetsko, M. L., Smith, J. R., Kagnoff, M.

F. and Gillin, F. D. (2000). Nitric oxide production by human intestinal epithelial cellsand competition for arginine as potential determinants of host defense against the lumen-dwelling pathogen Giardia lamblia. J. Immunol. 164, 1478-1487.

Gilman, R. H., Marquis, G. S., Miranda, E., Vestegui, M. and Martinez, H. (1988).Rapid reinfection by Giardia lamblia after treatment in a hyperendemic Third Worldcommunity. Lancet 1, 343-345.

Hemphill, A., Stager, S., Gottstein, B. and Muller, N. (1996). Electron microscopicalinvestigation of surface alterations on Giardia lamblia trophozoites after exposure to acytotoxic monoclonal antibody. Parasitol. Res. 82, 206-210.

Hiltpold, A., Frey, M., Hulsmeier, A. and Kohler, P. (2000). Glycosylation andpalmitoylation are common modifications of giardia variant surface proteins. Mol.Biochem. Parasitol. 109, 61-65.

Ishov, A. M., Sotnikov, A. G., Negorev, D., Vladimirova, O. V., Neff, N., Kamitani, T.,Yeh, E. T., Strauss, J. F., 3rd. and Maul, G. G. (1999). PML is critical for ND10formation and recruits the PML-interacting protein daxx to this nuclear structure whenmodified by SUMO-1. J. Cell Biol. 147, 221-234.

Jenuwein, T. and Allis, C. D. (2001). Translating the histone code. Science 293, 1074-1080.

Keister, D. B. (1983). Axenic culture of Giardia lamblia in TYI-S-33 medium supplementedwith bile. Trans. R. Soc. Trop. Med. Hyg. 77, 487-488.

Kulakova, L., Singer, S. M., Conrad, J. and Nash, T. E. (2006). Epigenetic mechanismsare involved in the control of Giardia lamblia antigenic variation. Mol. Microbiol. 61,1533-1542.

Le Panse, S., Ayani, E., Nielsen, S., Ronco, P., Verroust, P. and Christensen, E. I. (1997).Internalization and recycling of glycoprotein 280 in epithelial cells of yolk sac. Eur. J.Cell Biol. 72, 257-267.

Leight, E. R., Glossip, D. and Kornfeld, K. (2005). Sumoylation of LIN-1 promotestranscriptional repression and inhibition of vulval cell fates. Development 132, 1047-1056.

Lujan, H. D., Mowatt, M. R., Conrad, J. T., Bowers, B. and Nash, T. E. (1995).Identification of a novel Giardia lamblia cyst wall protein with leucine-rich repeats.Implications for secretory granule formation and protein assembly into the cyst wall. J.Biol. Chem. 270, 29307-29313.

Morrison, H. G., McArthur, A. G., Gillin, F. D., Aley, S. B., Adam, R. D., Olsen, G.J., Best, A. A., Cande, W. Z., Chen, F., Cipriano, M. J. et al. (2007). Genomicminimalism in the early diverging intestinal parasite Giardia lamblia. Science 317, 1921-1926.

Mowatt, M. R., Aggarwal, A. and Nash, T. E. (1991). Carboxy-terminal sequenceconservation among variant-specific surface proteins of Giardia lamblia. Mol. Biochem.Parasitol. 49, 215-227.

Mowatt, M. R., Nguyen, B. Y., Conrad, J. T., Adam, R. D. and Nash, T. E. (1994).Size heterogeneity among antigenically related Giardia lamblia variant-specific surfaceproteins is due to differences in tandem repeat copy number. Infect. Immun. 62, 1213-1218.

Nash, T. E. (2002). Surface antigenic variation in Giardia lamblia. Mol. Microbiol. 45,585-590.

Nash, T. E. and Aggarwal, A. (1986). Cytotoxicity of monoclonal antibodies to a subsetof Giardia isolates. J. Immunol. 136, 2628-2632.

Nash, T. E. and Mowatt, M. R. (1992). Characterization of a Giardia lamblia variant-specific surface protein (VSP) gene from isolate GS/M and estimation of the VSP generepertoire size. Mol. Biochem. Parasitol. 51, 219-227.

Nash, T. E., Conrad, J. T. and Merritt, J. W., Jr (1990a). Variant specific epitopes ofGiardia lamblia. Mol. Biochem. Parasitol. 42, 125-132.

Nash, T. E., Herrington, D. A., Levine, M. M., Conrad, J. T. and Merritt, J. W., Jr(1990b). Antigenic variation of Giardia lamblia in experimental human infections. J.Immunol. 144, 4362-4369.

Nash, T. E., Lujan, H. T., Mowatt, M. R. and Conrad, J. T. (2001). Variant-specificsurface protein switching in Giardia lamblia. Infect. Immun. 69, 1922-1923.

Palm, J. E., Weiland, M. E., Griffiths, W. J., Ljungstrom, I. and Svard, S. G. (2003).Identification of immunoreactive proteins during acute human giardiasis. J. Infect. Dis.187, 1849-1859.

Papanastasiou, P., Bruderer, T., Li, Y., Bommeli, C. and Kohler, P. (1997a). Primarystructure and biochemical properties of a variant-specific surface protein of Giardia.Mol. Biochem. Parasitol. 86, 13-27.

Papanastasiou, P., McConville, M. J., Ralton, J. and Kohler, P. (1997b). The variant-specific surface protein of Giardia, VSP4A1, is a glycosylated and palmitoylated protein.Biochem. J. 322 (Pt 1), 49-56.

Poulin, G., Dong, Y., Fraser, A. G., Hopper, N. A. and Ahringer, J. (2005). Chromatinregulation and sumoylation in the inhibition of Ras-induced vulval development inCaenorhabditis elegans. EMBO J. 24, 2613-2623.

Ringqvist, E., Palm, J. E., Skarin, H., Hehl, A. B., Weiland, M., Davids, B. J., Reiner,D. S., Griffiths, W. J., Eckmann, L., Gillin, F. D. et al. (2008). Release of metabolicenzymes by Giardia in response to interaction with intestinal epithelial cells. Mol.Biochem. Parasitol. 159, 85-91.

Schofield, P. J., Costello, M., Edwards, M. R. and O’Sullivan, W. J. (1990). Thearginine dihydrolase pathway is present in Giardia intestinalis. Int. J. Parasitol. 20,697-699.

Schofield, P. J., Edwards, M. R., Matthews, J. and Wilson, J. R. (1992). The pathwayof arginine catabolism in Giardia intestinalis. Mol. Biochem. Parasitol. 51, 29-36.

Senshu, T., Sato, T., Inoue, T., Akiyama, K. and Asaga, H. (1992). Detection of citrullineresidues in deiminated proteins on polyvinylidene difluoride membrane. Anal. Biochem.203, 94-100.

Senshu, T., Akiyama, K., Kan, S., Asaga, H., Ishigami, A. and Manabe, M. (1995).Detection of deiminated proteins in rat skin: probing with a monospecific antibody aftermodification of citrulline residues. J. Invest. Dermatol. 105, 163-169.

Shalizi, A., Gaudilliere, B., Yuan, Z., Stegmuller, J., Shirogane, T., Ge, Q., Tan, Y.,Schulman, B., Harper, J. W. and Bonni, A. (2006). A calcium-regulated MEF2sumoylation switch controls postsynaptic differentiation. Science 311, 1012-1017.

Singer, S. M., Yee, J. and Nash, T. E. (1998). Episomal and integrated maintenance offoreign DNA in Giardia lamblia. Mol. Biochem. Parasitol. 92, 59-69.

Stager, S. and Muller, N. (1997). Giardia lamblia infections in B-cell-deficient transgenicmice. Infect. Immun. 65, 3944-3946.

Stager, S., Felleisen, R., Gottstein, B. and Muller, N. (1997). Giardia lamblia variantsurface protein H7 stimulates a heterogeneous repertoire of antibodies displayingdifferential cytological effects on the parasite. Mol. Biochem. Parasitol. 85, 113-124.

Stager, S., Gottstein, B., Sager, H., Jungi, T. W. and Muller, N. (1998). Influence ofantibodies in mother’s milk on antigenic variation of Giardia lamblia in the murinemother-offspring model of infection. Infect. Immun. 66, 1287-1292.

Svard, S. G., Meng, T. C., Hetsko, M. L., McCaffery, J. M. and Gillin, F. D. (1998).Differentiation-associated surface antigen variation in the ancient eukaryote Giardialamblia. Mol. Microbiol. 30, 979-989.

Touz, M. C., Lujan, H. D., Hayes, S. F. and Nash, T. E. (2003). Sorting of encystation-specific cysteine protease to lysosome-like peripheral vacuoles in Giardia lamblia requiresa conserved tyrosine-based motif. J. Biol. Chem. 278, 6420-6426.

Touz, M. C., Kulakova, L. and Nash, T. E. (2004). Adaptor protein complex 1 mediatesthe transport of lysosomal proteins from a Golgi-like organelle to peripheral vacuolesin the primitive eukaryote Giardia lamblia. Mol. Biol. Cell 15, 3053-3060.

Touz, M. C., Conrad, J. T. and Nash, T. E. (2005). A novel palmitoyl acyl transferasecontrols surface protein palmitoylation and cytotoxicity in Giardia lamblia. Mol.Microbiol. 58, 999-1011.

Vossenaar, E. R., Zendman, A. J., van Venrooij, W. J. and Pruijn, G. J. (2003). PAD,a growing family of citrullinating enzymes: genes, features and involvement in disease.BioEssays 25, 1106-1118.

Wang, Y., Wysocka, J., Sayegh, J., Lee, Y. H., Perlin, J. R., Leonelli, L., Sonbuchner,L. S., McDonald, C. H., Cook, R. G., Dou, Y. et al. (2004). Human PAD4 regulateshistone arginine methylation levels via demethylimination. Science 306, 279-283.

Yee, J. and Nash, T. E. (1995). Transient transfection and expression of firefly luciferasein Giardia lamblia. Proc. Natl. Acad. Sci. USA 92, 5615-5619.

Journal of Cell Science 121 (17)2938

Jour

nal o

f Cel

l Sci

ence