ArticlePDF Available

Ectopic Expression of a Neospora caninum Kazal Type Inhibitor Triggers Developmental Defects in Toxoplasma and Plasmodium

Authors:

Abstract and Figures

Regulated proteolysis is known to control a variety of vital processes in apicomplexan parasites including invasion and egress of host cells. Serine proteases have been proposed as targets for drug development based upon inhibitor studies that show parasite attenuation and transmission blockage. Genetic studies suggest that serine proteases, such as subtilisin and rhomboid proteases, are essential but functional studies have proved challenging as active proteases are difficult to express. Proteinaceous Protease Inhibitors (PPIs) provide an alternative way to address the role of serine proteases in apicomplexan biology. To validate such an approach, a Neospora caninum Kazal inhibitor (NcPI-S) was expressed ectopically in two apicomplexan species, Toxoplasma gondii tachyzoites and Plasmodium berghei ookinetes, with the aim to disrupt proteolytic processes taking place within the secretory pathway. NcPI-S negatively affected proliferation of Toxoplasma tachyzoites, while it had no effect on invasion and egress. Expression of the inhibitor in P. berghei zygotes blocked their development into mature and invasive ookinetes. Moreover, ultra-structural studies indicated that expression of NcPI-S interfered with normal formation of micronemes, which was also confirmed by the lack of expression of the micronemal protein SOAP in these parasites. Our results suggest that NcPI-S could be a useful tool to investigate the function of proteases in processes fundamental for parasite survival, contributing to the effort to identify targets for parasite attenuation and transmission blockage.
NcPI-S expression in Toxoplasma tachyzoites. (A) Gra1/ssmycNcPIS WT or mutant construct is shown indicating the signal sequence of ROP1, the myc-tag and the NcPIS variants. (B) (left) Western blot analysis of lysates derived from extracellular parasites of transgenic NcPIs_6 and RHhxgprt–strains. An arrow marks the predominant monomeric form of myc-tagged NcPI-S inhibitor (bottom), while asterisk indicates the additional NcPIS dimeric form. SAG1 protein (top) was used as loading control. (Right) Localization of NcPIS (red) and the DG protein GRA3 (green) in intracellular parasites, 24 h p.i. Nuclei stained with DAPI (blue). Scale bars, 1μm. (C) (left) Western blot analysis of lysates derived from extracellular parasites of transgenic NcPIsmut_16 (arrow) and RHhxgprt- strains. GRA1 protein (top) was used as loading control. (right) Localization of mutant NcPI-S (green) and the chaperonin Bip protein (red) in intracellular parasites, 24 h p.i. Nuclei stained with DAPI (blue). Scale bars, 1μm. (D) Inhibitory activity of tachyzoite lysates against Bacillus lechiniformis (Bl) subtilisin. Tachyzoite lysates prepared from RH and NcPIS_6 were incubated with Bl subtilisin in the presence of 0.15 mM CBZ-Gly-Gly-Leu-pNA substrate. Residual hydrolytic activity (R.H.A) was measured. No activity was detected when lysates were incubated with the substrate alone (NcPIs_6/NP) or in reactions in which lysates were incubated with the Bl subtilisin in the presence of 2mM PMSF (NcPIS_6/+PMSF). Error bars represent the means ±SEM. (n = 2 experiments per strain, representative of triplicate samples). (E) Distribution of NcPI-S in soluble (S), membrane salt-eluted (SE) and membrane (M) fractions derived from either extracellular (left) or intracellular (right) parasites. The monomeric and dimeric forms of NcPIS (bottom) are marked with an arrow and asterisk, respectively. MIC4 micronemal protein (upper) and GPI anchored SAG1 (middle) were used as indicators of soluble and membrane associated proteins respectively.
… 
Developmentally arrested NcPIS_3 zygotes/ookinetes show gross abnormalities. (A) Ookinete conversion in NcPIS_3 and NcPISmut parasites versus GFPp (WT). The percentage of ookinetes formed in NcPIS_3 was significantly lower compared to WT and NcPISmut parasites. Ookinetes, macrogametes and zygotes were counted after labeling with an antibody recognizing the Pbs21 surface antigen. Error bars represent ± SEM, n = 3. (B) Whole protein extracts from ~1x106 ookinetes derived from in vitro cultured NcPIS_3, NcPISmut or GFPp (WT) parasites were examined for the presence of NcPI-S or NcPI-Smut respectively using anti-V5 mAb. In NcPIS_3 extracts a predominant ~17 kDa band was detected while a band of ~14 kDa was detected in extracts of NcPISmut. P28 was used as a loading control. (C-D) Subcellular distribution of NcPI-S and NcPI-Smut in ookinetes of transgenic P. berghei parasites. Confocal microscopy using anti-V5 MAb (red) shows the different localization of NcPIS_3 and NcPISmut in parasites. In NcPISmut there is dispersed and perinuclear staining pattern compared to the NcPIS_3 where expression is in close proximity to the nucleus. Nucleus is stained with TO-PRO-3 (blue). Scale bar 5 μM. (E) Transmission electron micrographs (TEM) of arrested NcPIS_3, zygotes/ookinetes show gross abnormalities. The endomembrane system is dilated. Left image shows an arrested parasite with discontinuities in the inner membrane (red arrowheads) while release of content from the nucleus (N) to the cytoplasm is manifested (white arrowhead).
… 
NcPIS_3 Plasmodium ookinetes fail to produce normal oocysts. (A) Oocyst formation of NcPI-S expressing clones (NcPIS_3 and NcPIS_1) compared to that of the parental WT strains (GFPp and ANKA respectively) and of NcPISmut compared to NcPIS_3 7–10 days post infection. Four sets of paired infection experiments were included in the case of NcPIS_3/GFPp, two in the case of NcPISmut/NcPIS_3 and three in the case of NcPIS_1/ANKA 2.34 paired infections. Error bars; SEM; P<0.001, Mann-Whitney test, in all three cases. (B) Confocal sections of representative mosquito midgut epithelial sheets infected with GFPp, NcPIS_3, ANKA or NcPIS_1 parasites, and stained with antibodies against the mosquito serpin6 (SRPN6) (red) and the ookinete surface protein P28 (green). Nuclei are stained with TO-PRO 3 (blue). Arrows indicate the position of ookinetes. NcPIS_1 and 3 ookinetes fail to induce an epithelial response, which in contrast is revealed in the case of ANKA and GFPp infected midguts by SRPN6 specific antibody. Scalebar 10μM. (C) Confocal microscopy of mature oocysts. The two types of NcPIS_3 oocysts (upper panel) are shown alongside the single oocyst that showed a normal morphology 14 days post infection. Main oocyst morphology includes: Normal sized oocysts with diffused nuclear material and a collapsed capsule (CO) and oocysts arrested in an early developmental stage (minute oocysts: MO). Green corresponds to GFP expression; nuclei are stained with TO-PRO 3. (Lower panel) NcPIS_1 abnormal oocysts labeled with the antibody recognizing the oocyst capsule protein PbCap380. Scale bar 20μM.
… 
Content may be subject to copyright.
RESEARCH ARTICLE
Ectopic Expression of a Neospora caninum
Kazal Type Inhibitor Triggers Developmental
Defects in Toxoplasma and Plasmodium
Zoi Tampaki
1,2
, Ramadhan S. Mwakubambanya
1,2¤
, Evi Goulielmaki
1,2
, Sofia Kaforou
1
,
Kami Kim
3
, Andrew P. Waters
4
, Vern B. Carruthers
5
, Inga Siden-Kiamos
1
, Thanasis
G. Loukeris
1
, Konstantinos Koussis
1
*
1Institute of Molecular Biology and Biotechnology, Foundation for Research and TechnologyHellas,
Heraklion, Greece, 2Department of Biology, University of Crete, Heraklion, Greece, 3Department of
Medicine, Albert Einstein College of Medicine, Bronx, United States of America, 4Faculty of Biomedical &
Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, Scotland, United
Kingdom, 5Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor,
United States of America
Deceased.
These authors contributed equally to this work.
¤Current address: Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536-
20115, Egerton, Kenya
*kousis@imbb.forth.gr
Abstract
Regulated proteolysis is known to control a variety of vital processes in apicomplexan para-
sites including invasion and egress of host cells. Serine proteases have been proposed as
targets for drug development based upon inhibitor studies that show parasite attenuation
and transmission blockage. Genetic studies suggest that serine proteases, such as subtili-
sin and rhomboid proteases, are essential but functional studies have proved challenging
as active proteases are difficult to express. Proteinaceous Protease Inhibitors (PPIs) pro-
vide an alternative way to address the role of serine proteases in apicomplexan biology. To
validate such an approach, a Neospora caninum Kazal inhibitor (NcPI-S) was expressed
ectopically in two apicomplexan species, Toxoplasma gondii tachyzoites and Plasmodium
berghei ookinetes, with the aim to disrupt proteolytic processes taking place within the se-
cretory pathway. NcPI-S negatively affected proliferation of Toxoplasma tachyzoites, while
it had no effect on invasion and egress. Expression of the inhibitor in P.berghei zygotes
blocked their development into mature and invasive ookinetes. Moreover, ultra-structural
studies indicated that expression of NcPI-S interfered with normal formation of micronemes,
which was also confirmed by the lack of expression of the micronemal protein SOAP in
these parasites. Our results suggest that NcPI-S could be a useful tool to investigate the
function of proteases in processes fundamental for parasite survival, contributing to the ef-
fort to identify targets for parasite attenuation and transmission blockage.
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 1/23
OPEN ACCESS
Citation: Tampaki Z, Mwakubambanya RS,
Goulielmaki E, Kaforou S, Kim K, Waters AP, et al.
(2015) Ectopic Expression of a Neospora caninum
Kazal Type Inhibitor Triggers Developmental Defects
in Toxoplasma and Plasmodium. PLoS ONE 10(3):
e0121379. doi:10.1371/journal.pone.0121379
Academic Editor: Gordon Langsley, Institut National
de la Santé et de la Recherche MédicaleInstitut
Cochin, FRANCE
Received: September 25, 2014
Accepted: January 31, 2015
Published: March 24, 2015
Copyright: © 2015 Tampaki et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any
medium, provided the original author and source are
credited.
Data Availability Statement: All relevant data are
within the paper and its Supporting Information files.
Funding: This work was performed in the framework
of the BIOSYS research project, Action KRIPIS,
project No. MIS-448301 (2013SE01380036) that was
funded by the General Secretariat for Research and
Technology, Ministry of Education, Greece and the
European Regional Development Fund (Sectoral
Operational Programme: Competitiveness and
Entrepreneurship, NSRF 20072013)/European
Commission. The funders had no role in study
Introduction
The phylum Apicomplexa comprises a number of intracellular parasites causing disease in hu-
mans and animals. Two prominent members are Plasmodium parasites causing malaria and
Toxoplasma that is the causative agent of toxoplasmosis in immunocompromised individuals.
These parasites are characterized by having both invasive and replicative forms. In T.gondii
during the asexual life cycle tachyzoites invade cells and replicate inside a parasitophorous vac-
uole in the host cytoplasm. Newly formed parasites egress from the host cell, and immediately
invade new target cells. During these events the secretory organelles of tachyzoites, micro-
nemes, rhoptries and dense granules (DG) have been found to have important roles. Plasmodi-
um parasites also have a complex life cycle. The parasite goes through a replicative cycle in the
blood of the human host, causing the pathology of the disease. Plasmodium is transmitted by
mosquitoes. After the uptake of sexual forms in a blood meal, the parasite develops into a zy-
gote in the mosquito midgut. The zygote in turn matures into the motile ookinete, which tra-
verses the midgut epithelium and forms the sporogonic oocyst. Sporozoites having developed
inside the cyst are transmitted to a new host during a blood meal. Zygote to ookinete transition
is a crucial point in the life cycle of the parasite as a failure to successfully complete this step
blocks transmission. This transition is accompanied by a radical reorganization of the cell, in-
cluding formation of the micronemes, which have an important role in motility and invasion,
and an extension of the cell through the elongation of the cytoskeleton.
Proteases have been recognized as basic components in the life cycle of apicomplexan para-
sites regulating a plethora of physiological processes such as replication, host invasion, egress
and metabolism [14]. Apart from increasing our understanding of the basic biology of api-
complexans, proteases comprise potential targets for drug development [5] or for interventions
aiming at parasite attenuation or transmission blockage.
Serine proteases have been identified in both Plasmodium and Toxoplasma and primarily
subtilisins and rhomboids have been studied in more detail. In T.gondii 12 genes encoding
subtilisin-like proteases have been identified, [69], while in Plasmodium, three subtilisin-like
proteases have been found [1013]. In the case of rhomboids, 6 are encoded in T.gondii and 8
in Plasmodium spp [14,15]. Genetic studies have shown that many of these proteases are essen-
tial [6,8,1620]. However, with the exception of Plasmodium SUB1, in vitro biochemical assays
to further elucidate their function, identify potential substrates and develop potential inhibitors
are either not available or technically challenging with contradictory data in some cases be-
tween in vitro and in vivo studies [21,22].
Small molecule protease inhibitors (SM-PIs) with a broad range of activity have been em-
ployed in different experimental models to uncover the significance of regulated proteolysis in
developmental and physiological processes. SM-PIs have been also tested against apicomplexan
parasites revealing the overall importance of proteolysis in host cell invasion, egress and intra-
cellular parasite replication [2326]. Among the disadvantages of SM-PIs are that they affect in
parallel proteolytic processes taking place in different cellular compartments (including prote-
olysis in the host cells) and/or involving different types of proteases.
On the other hand Proteinaceous Protease Inhibitors (PPIs) are considered superior inhibi-
tors to SM-PIs. These molecules have evolved as one of the various self-protecting strategies
against assaulting or uncontrolled proteolysis. Firstly, PPIs are more specific inhibitors than
SM-PIs since co-evolution with their target proteases has shaped their specificity, and secondly
because PPIs are proteins their expression/activity can be restricted in a specific stage and/or in
a specific subcellular compartment.
Serine PPIs can be distinguished based on their structure and their mechanism of action to
serpins, canonical or non-canonical inhibitors [27]. A well-studied family of PPIs inhibiting
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 2/23
design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing Interests: The authors have declared
that no competing interests exist.
serine proteases is that of Kazal type inhibitors. The basic domain of Kazal inhibitors has a
characteristic structure dictated by six conserved cysteines forming intra-domain disulfide
bonds [28]. Non-canonical inhibitors such as hirudin are much less abundant; they occur only
in blood sucking organisms and inhibit proteases involved in clot formation [29,30].
The distribution of PPIs in parasites varies. While Neospora and Toxoplasma have canonical
Kazal type inhibitors as well as serpins, other apicomplexan parasites such as Plasmodium and
Babesia do not possess any serine protease inhibitors. A protective role against digestive en-
zymes has been hypothesized for the four-Kazal domain inhibitors, TgPI-1 and TgPI-2, identi-
fied in Toxoplasma [31,32]. Besides putative homologues of TgPI-1 and TgPI-2, N.caninum
expresses the single Kazal domain NcPI-S that strongly inhibits bacterial subtilisins in vitro
[33,34] and it has been speculated that NcPI-S might regulate endogenous subtilisin activity in
N.caninum [34]. Apart from their difference in the number of Kazal-domains, there is no simi-
larity in the amino acid composition of the active sites of these inhibitors [34] suggesting that
they possess distinct substrate specificities.
We investigated the potential of NcPI-S to interfere with proteolytic processes within the se-
cretory apparatus, by ectopically expressing NcPI-S in Toxoplasma and Plasmodium; neither of
which express an NcPI-S homologue. Toxoplasma tachyzoites provided our study with a cellu-
lar and biochemical environment equivalent to that of the native NcPI-S, as the two species T.
gondii and N.caninum are phylogenetically very close. This strategy provided a system for a
fast evaluation of the chosen inhibitor. After confirming that active NcPI-S affected the growth
of T.gondii tachyzoites, we designed a stage specific ectopic expression of NcPI-S in the rodent
model parasite P.berghei that resulted in an arrest of zygote to ookinete transition. Our results
indicate that PPIs can be used as tools to study parasitic proteases and additionally could lead
to the discovery of novel antiparasitic agents.
Materials and Methods
Ethics Statement
All animal work has passed an ethical review process and was approved by the FORTH Ethics
Committee (FEC). All work was carried out in full conformity with Greek regulations: Presi-
dential Decree (160/91) and law (2015/92) which implement the directive 86/609/EEC from
the European Union and the European Convention for the protection of vertebrate animals
used for experimental and other scientific purposes and the new legislation Presidential Decree
56/2013. The experiments were carried out in a certified animal facility license number
EL91-BIOexp-02.
Parasites, cells, mosquitoes
Human foreskin fibroblasts (HFFsATCC SCRC-1041) were maintained in Dulbecco's Modi-
fied Eagles Medium (DMEM) containing 10% heat inactivated Fetal Bovine Serum (FBS, PAA
Laboratories), 2 mM Glutamine, 1% Penicillin/Streptomycin (GIBCO) (D10 Complete). The
T.gondii RH strain and the RHΔhxgprt [35] were maintained by serial passage in HFFs as de-
scribed [36]. Tachyzoites were harvested after lysis of the monolayer, unless otherwise stated,
by scraping, passage through a 26-gauge needle and a 3-μm Nucleopore membrane filter
(Whatman), and collected by centrifugation (400xg, 15 min at 4°C). P.berghei strains were
ANKA HP (also called 15cy1A), and the GFP-expressing 507cl1 clone [37]. Anopheles gambiae
G3 strain was used for mosquito infections.
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 3/23
Plasmid constructions
All primers are listed in S1 Table in supplemental material.
pGra1/ssmycNcPIS. ROP1 signal sequence (ss) was amplified from pRop1myc plasmid
[38], using primers ssRop1_5and ssRop1_3.The myc tag was amplified using mycRI5and
myctoxo3primers. NcPI-S was amplified from pQE/NcPIS [34], using NcPIS_5and
NcPIS_3primers. The fragments were fused in frame and inserted in pGra1/GFP/Gra2-SK
[39], which contains the dhfr_5/HXGPRT/dhfr_3 selection marker, using the NsiI and PacI re-
striction sites (S1A Fig.).
pGra1/ssmycNcPISmut. NcPISmut was generated from NcPI-S by PCR-based mutagene-
sis with primers NcPIsFor and NcPISRev. The construct encoded NcPI-S with changed amino
acids P2, P1 and P1-P4of the Kazal inhibitory domain from SMEYDP to FASGKR. The con-
struct was verified by sequencing. For cloning in the transfection vector NcPIsmut was ampli-
fied with the primers Ncmut_5and Ncmut_3and subcloned into FseI-PacI digested pGra1/
ssmycNcPIS resulting in the exchange of the wild type NcPI-S with the mutant version.
p[CTRP-Sp-V5-NcPIS]DEFSSUToxo. The signal sequence of the PbSUB2 was amplified
from P.berghei gDNA with primers SPsub-2F and SPsub-2R. The V5 epitope was amplified
from the pIZ/V5-His vector (Invitrogen) using V5for and V5Rev primers. NcPI-S (excluding
the signal sequence) was amplified from the pQE/NcPIS plasmid with primers NcPIS_5and
NcPIS-R. The fragments were joined in an intermediate vector and subsequently inserted be-
tween the CTRP promoter and P28 3UTR in the vector pCp*S[40]. The derived [CTRP-Sp-
V5-NcPIS] cassette was finally inserted into the pDEF-TgDHFR/D-SSU (RV) transformation
vector (kindly provided by Dr. Franke Fayard).
NcPISmut. NcPI-Smut was inserted in the p[CTRP-Sp-V5-NcPIS]DEFSSUToxo using
FseI and PacI/NotI restriction enzymes. The sequences encoding the PbSUB2 signal sequence
and V5 remained the same.
Transfection, selection and cloning of stable transformants
To generate stable transformants freshly released RHΔhxgprt parasites were electroporated as
described [41] with 100 μg of Gra1/ssmycNcPIS and Gra1/ssmycNcPISmut constructs. Tachy-
zoites were transfected in presence of NotI restriction enzyme [42] and parasites were selected
with mycophenolic acid and xanthine as described [43]. Resistant parasites were cloned by lim-
iting dilution. Clones NcPI-S_6 and NcPI-S_8 from two independent transfection experiments,
and clone NcPI-Smut_16 were selected.
P.berghei schizonts were transfected using standard procedures [44]. Pulse Field Inversion
Gel Electrophoresis (PFIGE) and genomic PCR genotyping was used to characterize the trans-
genic clones (S2B, S2C Fig.). For PFIGE-Southern, total gDNA was separated on 1% agarose
gel under Pulsed-Field Inversion Gel Electrophoresis (PFIGE) conditions, transferred and hy-
bridized with P.berghei radio-labeled dhfr-ts 3UTR probe as described [44](S2B Fig.). Geno-
mic PCR genotyping was performed using integration specific primer sets [45]: L665 and L740.
Clone purity and determination of the integration site (cor dssu locus), were verified by using
forward primers cssu specific L270 and dssu specific L260 in combination with the reverse
L740 primer common to both loci [46](S2C Fig.).
Two independent clones expressing NcPI-S were studied; NcPIS_3, in the GFP507cl1
(GFPp) recipient strain [37] and NcPIS_1, the latter derived from a transfection using the
ANKA 15cy1A reference strain as a recipient. The NcPISmut line expressed the mutated form
of NcPI-S and was derived from the GFP507cl1 (GFPp) recipient strain. The strain ANKA 2.34
was used as a WT control in oocyst experiments.
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 4/23
T.gondii assays
Equal numbers of freshly released filtered tachyzoites were used to infect HFF cells during rep-
lication assays. After sedimentation and incubation for 1 h, cultures were washed and incubat-
ed for another 24 h. Coverslips were labeled with anti-SAG1 antibody and 4'-6-Diamidino-
2-phenylindole (DAPI). Vacuoles containing different numbers of parasites (1, 2, 4, 8 or 16)
were counted (total 150 vacuoles/sample). Three independent experiments in duplicates
were performed.
Invasion competence of the parasites, was determined according to previously described
protocols [47]. Freshly lysed parasites from all strains were allowed to invade HFF cell mono-
layers grown on coverslips for 10 min and the number of extracellular vs. intracellular parasites
was determined. Extracellular tachyzoites were revealed after labeling with anti-SAG1 without
permeabilization and intracellular tachyzoites with subsequent staining with anti-GRA3
after permeabilization.
The percentage of egressed vacuoles was determined after inducing egress with calcium ion-
ophore A23187. Intracellular replicating tachyzoites (3640 h p.i.) were treated with 1 μM
A23187 or DMSO (solvent control) for 5 min [48] and egressed vacuoles were enumerated in
each sample.
P.berghei assays
P.berghei ookinete cultures and purification was performed as previously described [49]. Con-
version rates were calculated as previously described, using a mouse monoclonal antibody
(13.1), which recognizes the P28 protein on the surface of female gametes, zygotes and ooki-
netes [50]. Positive cells were counted on a Zeiss Axioscope2plus microscope (Carl Zeiss, Unit-
ed States) equipped with an AxioCam color Zeiss CCD camera. For oocyst counting,
mosquitoes were fed on anaesthesized infected mice for gametocyte feeding. Alternatively,
membrane feeders were used for feeding in vitro cultured ookinetes. Oocysts were counted on
day 10 post-feeding.
Transmission Electron Microscopy
For TEM observations, ookinetes were pelleted at 720xg and fixed in 2% glutataraldehyde/2%
paraformaldehyde in 0.1 M sodium cacodylate buffer pH 7.4 for 45 min at 4°C, post fixed in
1% osmium tetroxide, dehydrated in graded ethanol, stained with uranyl acetate 1% and finally
embedded in graded Durcupan:propylene oxide series (1:3, 1:1, 3:1, 4:0). Ultrathin-sections
(50100nm) were taken on a Leica LKB2088 ultramicrotome and examined under JEM 100C/
JEOL/Japan Transmission Electron Microscope. Microphotographs were obtained with an
ES500W Erlangshen camera and analysed by the DigitalMicrograph software (Gatan,
Germany).
Antibodies
For immunoblotting of T.gondii the antibodies were: 9B10 anti-myc (1:5000 dilution) (Cell
Signaling Technology), anti-MIC2 (6D10) [51], anti-SAG1 (gift of Jean Francois Dubremetz),
anti-ROM4 (gift of Dominique Soldati-Favre) and anti-GRA1 (gift of Louis Weiss), all 1:1000
dilution. For Western blot experiments of P.berghei anti-V5 (Invitrogen, diluted 1:2000), rab-
bit anti-SOAP (developed in the Loukeris laboratory, diluted 1:500), the monoclonal antibody
13.1, which detects the P28 surface antigen of P.berghei ookinetes [52] (1:20000), anti-Bip
(1:2000-gift of Ellen Knuepfer, NIMR, UK) were used. For IFA of T.gondii mouse a-myc 9B10
(1:2000, Cell Signaling Technology), rabbit anti-GRA3 (1:300, gift of Jean Francois
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 5/23
Dubremetz), rabbit anti-PfBip (MR4 ATTC, MRA-20 1:1000) were used. Antibodies used for
IFA of P.berghei were anti-serpin6 (1:1000, provided by G. Christophides) and PbCAP380 that
recognizes the oocyst capsule [53]. Secondary antibodies were conjugated to Alexa 555 or
Alexa 488 (Molecular Probes).
Fractionation and Immuno blot analysis
For analysis of T.gondii one infected HFF culture was left to lyse spontaneously, and extracellu-
lar tachyzoites were purified from the culture medium. For a second parallel culture, intracellu-
lar tachyzoites were force released (2022 h p.i.) by trypsinolysis, passage through a 26-gauge
needle, and filtering through a 3 μm Nucleopore membrane filter. Tachyzoite pellets were re-
suspended in Tris-Cl pH 8.5, lysed by freeze-thaw and sonication, centrifuged for 30 min at
320xg, and the supernatant was removed (Soluble fraction, S). The pellet was resuspended in
0.1 M Na
2
CO
3
pH 11.5, incubated on ice for 1 h, and centrifuged at 320xg for 30 min in order
to remove peripheral membrane proteins. The supernatant (Salt-eluted fraction, SE) was sepa-
rated from the remaining pellet that was resuspended in PBS (Membrane fraction, M). β-mer-
captoethanol was added to the lysates and samples were separated on 15% SDS-PAGE gels and
transferred to nitrocellulose membranes.
For detection of NcPI-S in P.berghei zygotes/ookinetes, parasites were resuspended in 10
volumes of RIPA buffer (50 mM Tris-Cl pH 8.0, 150 mM NaCl, 2 mM EDTA, 1% NP-40, 1%
Sodium Deoxycholate, 0.1% SDS) supplemented with Protease Inhibitor Cocktail (Sigma).
After incubation on ice for 30 min and centrifugation 8000xg for 30 min, the cleared lysates
were separated on SDS-PAGE gels and processed for Western blot analysis. For P.berghei frac-
tionation, parasite pellets were initially resuspended in 10 volumes of 100 mM Tris-Cl pH 8.0
and incubated on ice for 1 h. After centrifugation at 8000xg for 30 min, supernatants were col-
lected, while the pellet was resuspended in 10 volumes of RIPA buffer, loaded on SDS-PAGE
gels and processed for Western blot analysis. To detect phosphorylation of PbeIF2a in the dif-
ferent strains, parasite pellets were resuspended in 10 volumes modified RIPA buffer (50 mM
Tris-Cl pH 8.0, 150 mM NaCl, 2 mM EDTA, 1% NP-40, 0.1% SDS) supplemented with Prote-
ase Inhibitor Cocktail (Sigma), 50 mM NaF and 1mM phenylmethylsulfonyl fluoride (PMSF).
Subsequent treatment was as described above.
Indirect immunofluorescence microscopy
All manipulations were carried out at room temperature. T.gondii tachyzoite-infected HFF
cells on 24-well chamber slides were fixed with 100% cold methanol for 10 min, followed by
three washes in 1xPBS. Fixed cells were blocked in 1% bovine serum albumin (BSA) (Merck,
Europe) with 0.25% TritonX-100 in PBS for 1 hour. The wells were then stained with different
primary antibodies, followed by conjugated secondary antibodies. DAPI was added to stain the
nucleus. Images were collected using a Zeiss Axioscope2plus (Carl Zeiss, United States) micro-
scope equipped with an AxioCam color Zeiss CCD camera. Images were processed using Ima-
geJ software. For P.berghei ookinete IFAs, purified ookinetes were allowed to settle on glass
slides pre-treated with poly-L-lysine for 1530 min. The following procedures were all per-
formed at RT. Samples were fixed in 4% paraformaldehyde (PFA) for 10 min and washed with
PBS. This was followed by permeabilisation in PBS, 0.1% triton-x100 (PBT) for 30 min. The
primary antibody directed against V5 was added, after 1 h followed by PBS washes and incuba-
tion with secondary antibody for 3045 min. Nuclei were stained with TO-PRO. Samples were
washed in PBS and mounted in Vectashield before being analyzed using a Leica TCS SP2 con-
focal laser scanning microscope. For mosquito midgut IFA, infected midguts were dissected in
ice-cold PBS, prefixed for 90 sec in ice-cold 4% PFA in PBS, transferred to ice-cold PBS and cut
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 6/23
open longitudinally. After removal of the midgut content the epithelium was fixed for another
4560 min in 4% PFA at RT. Samples were washed 3x10 min in PBS, blocked and permeabi-
lized for 1 h 30 min in 1% BSA in PBT at RT, followed by incubation overnight at 4°C with
anti-serpin6, followed by 3 washes with PBT and incubation with secondary antibodies for 1 h
at RT. Nuclei were stained with TO-PRO 3 (Molecular Probes). Midguts were washed three
times in PBT for 10 min, mounted in Vectashield and analyzed as described above.
In vitro inhibitory activity of transgenic parasite lysates
Increasing amounts of lysates (calculated as parasite number equivalents) prepared from RH
and NcPIS_6 were incubated with Bacillus licheniformis subtilisin A (Bl-subA) (3.6 nM) in re-
action buffer (100 mM TrisHCl, 150 mM NaCl, 10 mM CaCl
2
, Triton X-100 0.25% v/v,
0.05% sodium Deoxycholate, pH 8.0) for 15 min at 37°C followed by the addition of the sub-
strate CBZ-Gly-Gly-Leu-pNA (0.15 mM). Residual hydrolytic activity (R.H.A) was measured
as substrate conversion in a spectrophotometer. No enzymatic activity was detected when ly-
sates were incubated with the substrate alone (NcPIs_6/NP) or with Bl-subA in presence of
2mM PMSF (NcPIS_6/+PMSF).
Statistical analysis
Students t test using SPSS v.11.5 software was employed.
Results
NcPI-S expression in bacteria, NcPI-S modifications and their validation
The Kazal serine protease inhibitor NcPI-S is a 79 amino acid peptide with a theoretical mw of
6 kDa (Fig. 1)[33]. We first expressed NcPI-S in bacteria as a recombinant NcPI-S tagged with
V5 and 6xHis (r(His-V5)/NcPI-S) (Fig. 1A). Western blot experiments using a monoclonal Ab
recognizing anti-V5 revealed the presence of a predominant band of *17 kDa in our prepara-
tion (Fig. 1B, right) which is slightly bigger than the predicted 10.7 kDa of the r(His-V5)/
NcPI-S. This is consistent with previous studies showing that NcPI-S migrates slower on Tris-
tricine SDS-PAGE gels [34] and even slower on Tris SDS-PAGE gels [33,34]. The inhibitor r
(His-V5)/NcPI-S was tested against the Bl-subA using previously reported enzymatic assays
[33,34](Fig. 1B, left) indicating that the N-amino terminal extension does not interfere with
the inhibitory activity of NcPI-S.
In parallel, we used bacterial expression to validate an NcPI-S mutant variant. Although the
inhibitory specificity of Kazals is primarily dictated by the identity of the amino acid at the P1
position, other amino acid interactions between the Reactive Site Loop (RSL) and the substrate
binding cavities may equally influence the binding of Kazals to proteases [28]. As previously
shown, mutagenesis of the P1 Met to Ala of NcPI-S does not affect its inhibitory activity [34].
Thus, we decided to change a significant proportion of the (RSL). To achieve this, we have
substituted the NcPI-Ss P2-P4site with the sequence FASGKR (NcPI-Smut; Fig. 1A). This se-
quence corresponds to the non-prime site (P6-P1) of a cleavage site processed by the West Nile
Virus (WNV) trypsin-like serine protease [54]. Our hypothesis was to test the inhibitory profile
of a serine protease recognition site not present in P.berghei and T.gondii. The recombinant
mutant (His-V5)/NcPI-Smut, (r(His-V5)/NcPI-Smut), showed a substantial mobility shift in
respect to the r(His-V5)/NcPI-S, although the calculated size difference was only 0.2 kDa
(Fig. 1B). More importantly the NcPI-S mutant variant failed to inhibit Bl-subA in comparison
with the r(His-V5)/NcPI-S (Fig. 1C). At a high concentration of r(His-V5)/NcPI-Smut (750
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 7/23
fmol) a low degree of inhibition was detected, although we never detected more than 20% inhi-
bition, compared to the complete inhibition of r(His-V5)/NcPI-S.
Ectopic expression of NcPI-S in Toxoplasma gondii tachyzoites
As a proof of concept that ectopic expression of a PPI can affect an aspect of parasite infection,
we expressed NcPI-S and the inactive variant NcPI-Smut in Toxoplasma tachyzoites. NcPI-S
and NcPI-Smut were expressed as amino-terminal fusions of the PPI by replacing its own sig-
nal peptide with the ROP1 signal sequence [55] followed by a myc epitope tag under the con-
trol of the constitutive Gra1 gene promoter [56](Fig. 2A). Transfection of the recombinant
plasmids (pGra1/ssmycNcPIS or pGra1/ssmycNcPISmut) in the RHΔhxgprt recipient strain
[35] and clonal selection resulted in several stable clones in both cases. One clone in each case,
NcPIS_6 or NcPISmut_16 respectively, was selected for further studies.
Western blot analysis of NcPIS_6 tachyzoite extracts identified a predominant (myc)/
NcPI-S band of *17 kDa (Fig. 2B left, arrow). An additional fainter band at *24 kDa, possibly
corresponding to a (myc)/NcPI-S dimer, as has been previously described [34], was also appar-
ent (Fig. 2B, asterisk). The (myc)/NcPI-Smut variant was detected as a single faster migrating
band than (myc)/NcPI-S (Fig. 2C) suggesting that the NcPI-Smut dimer might be below the
threshold of detection.
Immunofluorescence analysis (IFA) was performed on tachyzoites using a mAb recognizing
the myc epitope. These experiments revealed that (myc)/NcPI-S was secreted into the parasito-
phorous vacuole (PV) when intracellular NcPIS_6 tachyzoites were labeled (Fig. 2B, right and
Fig 1. Bacterial expression of NcPI-S. (A) Amino acid sequence of NcPI-S. The deduced signal peptide
sequence is shaded in green and the signal peptidase cleavage site is underlined. An open box indicates
amino acid sequence of Kazal inhibitory domain. The methionine (M) in position P1 is shadedin red and the
conserved cysteine residues are in bold. Also depicted are the putative intra-domain disulfide bridges
between cysteine numbers 15, 24, 36. Native signal peptide of NcPI-S (without the peptidase cleavage
site) was replaced by a 6xHis and a V5 tag (in the case of bacterial expression), by the signal peptide of ROP-
1 and a myc-tag (T.gondii expression) and by the signal peptide of PbSUB2 and a V5-tag (P.berghei
expression). Ectopic expression of NcPI-Smut in all cases was accomplished by exchanging NcPI-S
sequence with that of NcPI-Smut, where residues P2-P4have been mutated. (B) Western blot analysis of
different amounts (110 μg) of r(His-V5)/NcPI-S and r(His-V5)/NcPI-Smut probed with anti-V5. (C) Activity
assay of recombinant NcPI-S/NcPI-Smut. Increasing amounts of r(His-V5)/NcPI-S and r(His-V5)/NcPI-Smut
were incubated with subtilisin A of Bacillus lechiniformis. Residual hydrolytic activity (R.H.A) was measured
through substrate conversion.
doi:10.1371/journal.pone.0121379.g001
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 8/23
Fig 2. NcPI-S expression in Toxoplasma tachyzoites. (A) Gra1/ssmycNcPIS WT or mutant construct is
shown indicating the signal sequence of ROP1, the myc-tag and the NcPIS variants. (B) (left) Western blot
analysis of lysates derived from extracellular parasites of transgenic NcPIs_6 and RHhxgprtstrains. An
arrow marks the predominant monomeric form of myc-tagged NcPI-S inhibitor (bottom), while asterisk
indicates the additional NcPIS dimeric form. SAG1 protein (top) was used as loading control. (Right)
Localization of NcPIS (red) and the DG protein GRA3 (green) in intracellular parasites, 24 h p.i. Nuclei
stained with DAPI (blue). Scale bars, 1μm. (C) (left) Western blot analysis of lysates derived from extracellular
parasites of transgenic NcPIsmut_16 (arrow) and RHhxgprt- strains. GRA1 protein (top) was used as loading
control. (right) Localization of mutant NcPI-S (green) and the chaperonin Bip protein (red) in intracellular
parasites, 24 h p.i. Nuclei stained with DAPI (blue). Scale bars, 1μm. (D) Inhibitory activity of tachyzoite
lysates against Bacillus lechiniformis (Bl) subtilisin. Tachyzoite lysates prepared from RH and NcPIS_6 were
incubated with Bl subtilisin in the presence of 0.15 mM CBZ-Gly-Gly-Leu-pNA substrate. Residual hydrolytic
activity (R.H.A) was measured. No activity was detected when lysates were incubated with the substrate
alone (NcPIs_6/NP) or in reactions in which lysates were incubated with the Bl subtilisin in the presence of
2mM PMSF (NcPIS_6/+PMSF). Error bars represent the means ±SEM. (n = 2 experiments per strain,
representative of triplicate samples). (E) Distribution of NcPI-S in soluble (S), membrane salt-eluted (SE) and
membrane (M) fractions derived from either extracellular (left) or intracellular (right) parasites. The monomeric
and dimeric forms of NcPIS (bottom) are marked with an arrow and asterisk, respectively. MIC4 micronemal
protein (upper) and GPI anchored SAG1 (middle) were used as indicators of soluble and membrane
associated proteins respectively.
doi:10.1371/journal.pone.0121379.g002
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 9/23
S1B Fig.), while in extracellular tachyzoites (myc)/NcPI-S exhibited a punctate staining pattern
(S1C Fig.). In contrast, no signal was detected in the intravacuolar space of the intracellular
growing NcPISmut_16 tachyzoites. Instead, IFA revealed retention of (myc)/NcPI-Smut within
the tachyzoites and specifically within the ER, which in tachyzoites is expanded around the cell
nucleus being more prominent posterior to the nucleus. The increased ER retention of (myc)/
NcPI-Smut was indicated by its co-localization with the ER-resident chaperone protein BiP
(Fig. 2C, right).
In order to verify that the myc-tagged NcPI-S retains its inhibitory potential we used the Bl-
SubA enzymatic assay to test extracts derived from either WT or NcPIS_6 extracellular tachy-
zoites. Only extracts derived from NcPIS_6 tachyzoites inhibited the Bl-SubA activity and
moreover this inhibition was directly proportional to the number of parasites that were used to
prepare the extracts (Fig. 2D).
Fractionation of extracts derived from extracellular tachyzoites (isolated from infected,
spontaneously-lysed cells) or from intracellular tachyzoites (isolated from vacuoles released by
force from infected cells 24 h post infection), revealed abundant presence of (myc)NcPI-S in
the soluble fraction (S) in both cases (Fig. 2E, lower panels). Association of the (myc)NcPI-S
with the membrane pellet (M), (insoluble after treatment with Na
2
CO
3
that removes mem-
brane associated proteins), was also observed but at a much lower degree (Fig. 2E, lower pan-
els). Importantly, both the *17 kDa (arrow) and the potential *24 kDa dimer (asterisk) of
the (myc)NcPI-S were detected in the S fractions of tachyzoites as has been previously observed
[34].
Expression of NcPI-S in T.gondii tachyzoites impairs parasite
replication
To study the effect of expressing NcPI-S and its mutated form in T.gondii tachyzoites we per-
formed phenotypic characterization. NcPIS_6 tachyzoites were compared to WT tachyzoites of
the RH and the parental RHΔhxgprt strains using well-established invasion [47], egress [48]
and growth assays. Neither invasion (Fig. 3A) nor egress assays (Fig. 3B) revealed any signifi-
cant phenotypic deviation of NcPIS_6 from the WT control RH strain (Fig. 3A and 3B). To as-
sess growth we carried out replication assays [57] where equal numbers of tachyzoites from
NcPIS_6 and WT control strains (RH and RHΔhxgprt), were used to infect HFF cells (Fig. 3C).
At 24 h p.i., vacuoles derived from control strains contained 8 tachyzoites in a percentage rang-
ing between 4070% of the total vacuoles, indicating 3 cell divisions as was expected. In con-
trast, only *17% of NcPIS_6 vacuoles contained 8 tachyzoites (Fig. 3C), indicating impaired
replication. A clone (NcPIS_8), derived from a second independent transformation experi-
ment, showed similar growth kinetics to that of the NcPIS_6 (S1A Fig.), verifying that a replica-
tion defect of T.gondii tachyzoites is reproducibly associated with the expression of (myc)
NcPI-S. In contrast NcPISmut_16 tachyzoites showed the same growth dynamics as the paren-
tal RHΔhxgprt strain (Fig. 3D). When combined, these results suggest that active NcPI-S is ex-
pressed in tachyzoites, and interfere with proliferation of the parasite, while the mutated form
has no effect on parasite growth.
Ectopic expression of NcPI-S and NcPI-Smut in Plasmodium berghei
zygotes/ookinetes
The results from the analysis of T.gondii encouraged us to test similar constructs in Plasmodi-
um, to determine the effect of NcPI-S in these parasites. To this end, recombinant P.berghei
parasites were produced using standard genetic transfection protocols. In brief, the constructs
were designed to express V5 epitope tagged NcPI-S and NcPI-Smut fused to the pbsub2 signal
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 10 / 23
peptide sequence [58], to ensure correct sorting in the secretory apparatus. The engineered
open reading frames (ORFs) were placed under the control of the 5and 3UTRs of ctrp and
p28 genes, respectively, which drive expression in the late zygote and ookinete stages [40]. The
construct was integrated in the c-ssu/d-ssu locus of the GFP507cl1 (GFPp) strain, constitutively
expressing GFP [37](S2A Fig.). We chose to restrict expression of NcPI-S and its inactive vari-
ant NcPI-Smut to the zygote/ookinete stages in P.berghei to avoid any toxic effects in the asex-
ual blood stages, in which transfection is carried out. Clonal lines were established and one
clone from each transfection (NcPIS_3 and NcPISmut) was selected for further analysis after
genotypic characterization (S2B and S2C Fig.). As expected asexual growth and gametocyto-
genesis were both unaffected in NcPIS_3 and NcPISmut parasites (data not shown). However,
when ookinete conversion was scored in in vitro cultures of the two strains, we found that the
majority of NcPIS_3 parasites were developmentally arrested after zygote formation. However,
*13% normal ookinetes were detected in each experiment. In contrast, the in vitro cultures of
the NcPISmut strain produced ookinetes at numbers comparable to the parental GFPp strain
(Fig. 4A).
Expression of (V5)/NcPI-S and (V5)/NcPI-Smut, was confirmed by Western blotting analy-
sis of in vitro cultured ookinetes. Consistent with the size of the bacterially expressed inhibitors,
the anti-V5 mAb detected a predominant *17 kDa band in extracts of the NcPIS_3 parasites,
while a *13 kDa band was detected in extracts of the NcPISmut strain (Fig. 4B). IFAs using
the V5 mAb revealed that in the developmentally arrested cells of the NcPIS_3 strain the PPI
was localized in an area proximal to the nucleus (Fig. 4C,NcPIS_3). In NcPISmut ookinetes the
inhibitor was detected throughout the entire cytosol including a perinuclear signal possibly co-
inciding with the ER, which in zygotes/ookinetes is composed of only a few tight stacks encircl-
ing the nucleus (Fig. 4D,NcPISmut). To further examine the potential localization of NcPI-S
Fig 3. Phenotypic characterization of NcPIS_6 strain. (A) Invasion competence of NcPIS_6 clone was
similar to the RH strain. BAPTA-AM treated parasites, which do not invade cells, were included as a control.
Error bars represent SEM (n = 3, each experiment was performed in duplicate and ten random fields were
screened blindly in each sample). (B) Percentage of lysed vacuoles of NcPIS_6 versus RH after inducing
egress with calcium ionophore A23187. Intracellular replicating tachyzoites (3640 h p.i.) were treated with
1μM A23187 or DMSO (solvent control) for 5min. Percentage of induced egress from host cells was
determined. Error bars, SEM (n = 3, each experiment was performed in duplicate and, 10 fields were blindly
screened per sample). (C) Replication assay of NcPIS_6, RH and RHΔhxgprt
-
(parental) and (D)
NcPISmut_16 and RHΔhxgprt
-
. Following a 24 h interval of intracellular replication, vacuoles from all the
above strains were scored according to their parasite context. Error bars, SEM (n = 3, representative of
duplicate samples); P<0.001, Students t-test.
doi:10.1371/journal.pone.0121379.g003
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 11 / 23
and NcPI-Smut we have performed double staining with the ER chaperone BiP (S3 Fig.). In the
case of NcPIS_3 parasites, some partial colocalization is seen. In these retort forms though and
Fig 4. Developmentally arrested NcPIS_3 zygotes/ookinetes show gross abnormalities. (A) Ookinete
conversion in NcPIS_3 and NcPISmut parasites versus GFPp (WT). The percentage of ookinetes formed in
NcPIS_3 was significantly lower compared to WT and NcPISmut parasites. Ookinetes, macrogametes and
zygotes were counted after labeling with an antibody recognizing the Pbs21 surface antigen. Error bars
represent ±SEM, n = 3. (B) Whole protein extracts from *1x10
6
ookinetes derived from in vitro cultured
NcPIS_3,NcPISmut or GFPp (WT) parasites were examined for the presence of NcPI-S or NcPI-Smut
respectively using anti-V5 mAb. In NcPIS_3 extracts a predominant *17 kDa band was detected while a
band of *14 kDa was detected in extracts of NcPISmut. P28 was used as a loading control. (C-D) Sub-
cellular distribution of NcPI-S and NcPI-Smut in ookinetes of transgenic P.berghei parasites. Confocal
microscopy using anti-V5 MAb (red) shows the different localization of NcPIS_3 and NcPISmut in parasites.
In NcPISmut there is dispersed and perinuclear staining pattern compared to the NcPIS_3 where expression
is in close proximity to the nucleus. Nucleus is stained with TO-PRO-3 (blue). Scale bar 5 μM. (E) Trans-
mission electron micrographs (TEM) of arrested NcPIS_3, zygotes/ookinetes show gross abnormalities. The
endomembrane system is dilated. Left image shows an arrested parasite with discontinuities in the inner
membrane (red arrowheads) while release of content from the nucleus (N) to the cytoplasm is manifested
(white arrowhead).
doi:10.1371/journal.pone.0121379.g004
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 12 / 23
based on the TEM experiments, the ER might be severely rearranged. In the NcPISmut strain
there is almost complete colocalization of NcPI-Smut with BiP as has been also the case in in-
tracellular T.gondii parasites expressing NcPI-Smut (Fig. 2C).
Transmission Electron Microscopy (TEM) was used to further examine the morphological
defects of NcPIS_3 developmentally arrested ookinetes. We detected a pronounced dilation of
the endomembrane system with electron dense aggregates being observed (Fig. 4E, middle and
right, S4A Fig.). Importantly, this dilation appeared also polarized, in accordance with the
(V5)/NcPI-S localization pattern (compare Fig. 4C left and Fig. 4E middle). Other develop-
mentally arrested ookinetes showed a progressive degeneration (Fig. 4E, left) manifesting a
range of ultrastructural defects such as incomplete formation of the inner membrane (Fig. 4E,
red arrowheads). In some extreme cases of terminally degenerated ookinetes the whole cyto-
plasm was replaced with vacuoles of different sizes (Fig. 4E, middle, S4B Fig.). In conclusion,
our analysis revealed that the expression of (V5)/NcPI-S during zygote to ookinete transition
resulted in a prompt and almost total block in development.
NcPIS_3 parasites are impaired in oocyst formation
To assess the ability of NcPIS expressing parasites to transmit through mosquitoes we fed A.
gambiae female mosquitoes on mice infected with the strain NcPIS_3 and the clone NcPIS_1
derived from an independent transfection using the ANKA reference strain as a recipient. The
result revealed a 9798% reduction of in oocyst numbers in mosquitoes infected with either
NcPIS_3 or NcPIS_1 parasites, compared to infection with the parental strains (Fig. 5A, left
and middle respectively) or in comparison to the NcPISmut strain (Fig. 5A, right). Midguts
were also dissected and stained 24 h after feeding with NcPIS_3 or NcPIS_1. For these IFAs,
ookinetes were labeled with the P28 antibody and with an antibody directed against the A.
gambiae serine protease inhibitor SRPN6 (AgSRPN6), which selectively labels invaded cells
[59]. Thus we confirmed that a reduced number of ookinetes invaded the midgut epithelium
(Fig. 5B). In addition we investigated the effect of NcPI-S on the sporogony of the few surviving
NcPIS_3 and NcPIS_1 oocysts (Fig. 5C). To determine if these oocysts develop normally, IFA
analysis was performed with the oocyst capsule protein PbCap380 [53] and with nuclear stain-
ing for the presence of sporozoites. From a total of 33 NcPIS_3 oocysts recorded in three inde-
pendent infection experiments only a single oocyst showed a normal morphology (budded
sporozoites), hence bite back experiments were not performed. The remaining oocysts fall into
two categories: smaller than the expected size, suggesting arrested growth and nuclear division
(minute oocysts, MO), or relatively normal in size with a collapsed capsule containing diffused
nuclear material (collapsed oocysts, CO) (Fig. 5C). Similarly, among twelve recorded NcPIS_1
oocysts, three had a normal morphology, four were MO and five were CO.
Microneme formation is severely reduced in NcPIS_3 ookinetes
Careful inspection of the TEM pictures revealed that the few mature NcPIS_3 ookinetes which
developed, suffered from ultra-structural malformations including a severe decrease in the
number of micronemes (Fig. 6A). To further investigate this we chose to study the micronemal
protein SOAP. We first verified that the soap transcript levels in the NcPIS_3 zygotes/ookinetes
were comparable to WT (Fig. 6B left). However, Western blot analysis showed that the micro-
nemal protein SOAP [60] was hardly detectable in NcPIS_3 zygote/ookinete extracts, com-
pared to GFPp or NcPISmut derived extracts where this protein was readily seen (Fig. 6B right).
Furthermore we repeated these experiments using the NcPIS_1 parasite clone to exclude any
effects deriving from the genetic background. The results were identical (S5 Fig.).
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 13 / 23
In apicomplexans, insults that perturb ER homeostasis result in repression of translation
which is mediated by the increased phosphorylation of the eukaryotic Initiation Factor 2a
(elF2a) [61,62]. To investigate if such an effect was caused by the expression of NcPI-S we ex-
amined the levels of elF2a phosphorylation by using a Phospho-eIF2α(Ser51) antibody, that
was previously shown to cross-react with the phosphorylated form of the P.falciparum eIF2a
[63], on Western blots containing extracts from NcPIS_3,NcPISmut and GFPp zygote/ookinete
cultures. A significant increase in PbeIF2a phosphorylation levels was revealed in NcPIS_3 ex-
tracts as compared to extracts derived from the GFPp and NcPISmut (Fig. 6C). Since we did
not reveal any significant alteration in the transcript levels of SOAP (as well as of CTRP) in the
Fig 5. NcPIS_3 Plasmodium ookinetes fail to produce normal oocysts. (A) Oocyst formation of NcPI-S
expressing clones (NcPIS_3 and NcPIS_1) compared to that of the parental WT strains (GFPp and ANKA
respectively) and of NcPISmut compared to NcPIS_3 710 days post infection. Four sets of paired infection
experiments were included in the case of NcPIS_3/GFPp, two in the case of NcPISmut/NcPIS_3 and three in
the case of NcPIS_1/ANKA 2.34 paired infections. Error bars; SEM; P<0.001, Mann-Whitney test, in all three
cases. (B) Confocal sections of representative mosquito midgut epithelial sheets infected with GFPp,
NcPIS_3, ANKA or NcPIS_1 parasites, and stained with antibodies against the mosquito serpin6 (SRPN6)
(red) and the ookinete surface protein P28 (green). Nuclei are stained with TO-PRO 3 (blue). Arrows indicate
the position of ookinetes. NcPIS_1 and 3ookinetes fail to induce an epithelial response, which in contrast is
revealed in the case of ANKA and GFPp infected midguts by SRPN6 specific antibody. Scalebar 10μM. (C)
Confocal microscopy of mature oocysts. The two types of NcPIS_3 oocysts (upper panel) are shown
alongside the single oocyst that showed a normal morphology 14 days post infection. Main oocyst
morphology includes: Normal sized oocysts with diffused nuclear material and a collapsed capsule (CO) and
oocysts arrested in an early developmental stage (minute oocysts: MO). Green corresponds to GFP
expression; nuclei are stained with TO-PRO 3. (Lower panel) NcPIS_1 abnormal oocysts labeled with the
antibody recognizing the oocyst capsule protein PbCap380. Scale bar 20μM.
doi:10.1371/journal.pone.0121379.g005
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 14 / 23
NcPIS_3 strain as compared to WT (Fig. 4B), the marked decrease in the SOAP protein levels
could probably reflect the impaired translation in NcPIS_3 parasites.
Discussion
In this study we investigated the use of serine protease inhibitors of the Kazal type to interfere
with T.gondii tachyzoite egress, invasion and growth and P.berghei zygote to ookinete transi-
tion. We chose NcPI-S, a Kazal inhibitor originating from the related apicomplexan parasite N.
caninum. Firstly, we expressed NcPI-S in T.gondii in an attempt to validate the PPI in a fast
and reliable manner. After initial validation we expanded our studies by designing a strategy
chosen to restrict the expression of the inhibitor in the P.berghei zygote to ookinete transitional
Fig 6. Mature NcPI-S_3 ookinetes contain fewer micronemes. (A) Selected longitudinal sections, across
the apical part of wild type (left image) and NcPIS_3 mature ookinetes (middle and right images). All are
surrounded by the characteristic subpellicular inner membrane complex. Scarcity of micronemes (white
arrows), is a common feature in NcPIS_3 ookinetes. The ookinete-specific characteristic structure of
crystalloid (cr) is clearly visible. (B) (Left panel) Semi-quantitative RT-PCR analysis of micronemal proteins
CTRP and SOAP in NcPIS_3 and GFPp ookinetes. No alteration was observed in transcript levels. p28 was
used as a control. (Right panel) Western analysis of soluble (SF) and membrane (MF) fraction of ookinete
extracts derived from in vitro cultured NcPIS_3,NcPISmut and GFPp ookinetes, with antibodies against the
micronemal protein SOAP. P28 was used as a sample normalization control. No SOAP is detected in the
NcPIS_3 line. (C) Western blot analysis of PbeIF2a phosphorylation. Western blots loaded with lysates from
GFPp,NcPIS_3 and NcPISmut ookinete cultures were probed with antibodies against phosphorylated eIF2α.
NcPIS_3 parasites show an increase in PbeIF2a phosphorylation. The endoplasmic reticulum (ER) marker,
BiP was used as the loading control.
doi:10.1371/journal.pone.0121379.g006
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 15 / 23
stage, a crucial time in the life cycle of malaria species as failure to successfully complete this
step blocks transmission. Importantly, this timing of expression was also chosen as it circum-
vented the possible detrimental effects on viability parasites of the blood stages, in which the
genetic transfection takes place.
All three characterized apicomplexan Kazal inhibitors (TgPI-1, TgPI-2 and NcPI-S) are
dense granule (DG) constituents. The ectopically expressed NcPI-S in Toxoplasma tachyzoites
was also trafficked in DGs and secreted in the parasitophorous vacuole, similarly to the endoge-
nously expressed NcPI-S in N.caninum tachyzoites. Strikingly though, the catalytically dead
variant NcPI-Smut was retained within the tachyzoite ER. This could be explained if NcPI-S
piggybacks on its potential targets through the secretory pathway, or if RSL mutagenesis dis-
rupted a sorting signal enabling NcPI-S anterograde trafficking. Additional studies will be nec-
essary to distinguish between these possibilities. Interestingly, there are conserved domains in
the RSLs prime site (P2to P5) of several Kazal domains including NcPI-S, but not in any of
the Kazal domains of TgPI-1 and TgPI-2 inhibitors. The biological role of these though, if any,
is yet to be defined.
In T.gondii the ectopically expressed NcPI-S in Toxoplasma tachyzoites was secreted in the
parasitophorous vacuole, presumably via the DGs, similar to the endogenously expressed
NcPI-S in N.caninum tachyzoites [34]. Strikingly though, a catalytically dead variant NcPI-S-
mut was retained within the ER of the tachyzoite. In contrast to the native NcPI-S no dimer
was observed for the mutant form. This could be attributed to structural changes in the mutant
preventing its efficient dimerization. For their ectopic expression in each case, NcPI-S and
NcPI-Smut were fused to the same signal peptide and epitope tag sequences, differing in 6
amino acids within the RSL including the P1-P1residues that are critical for the inhibitory ac-
tivity. Possible reasons firstly for this differential localization of NcPI-S and NcPI-Smut in
tachyzoites might be that either NcPI-S is trafficked within the secretory pathway piggybacked
on its potential targets, or that RSL mutagenesis disrupted the folding of a sorting signal
enabling NcPI-S anterograde trafficking as well as affected the ability of the protein to
homodimerize.
Fractionation of extracellular and 24 h intracellular tachyzoites expressing active NcPI-S re-
vealed its presence in the soluble fraction, with the *24 kDa dimer being more abundant in
the soluble fraction of the extracellular tachyzoites. In the study of Morris et al.[34] the endog-
enous NcPI-S in N.caninum was also shown to form a dimer, with anomalous migration
(*20 kDa) in extracellular tachyzoite extracts, which is in accordance with our observations.
Furthermore, our results imply that NcPI-S dimerization depends on the subcellular environ-
ment (dense granules versus vacuole). However, presently we do not know to which degree
dimerization influences the NcPI-S inhibitory capacity. Interestingly, phenotypic characteriza-
tion of NcPI-S and NcPI-Smut strains revealed that while the presence of NcPI-Smut in T.gon-
dii was not detrimental for tachyzoites, ectopic expression of NcPI-S significantly affected their
intracellular growth. Parasite replication was impaired, suggesting the possibility that NcPI-S
interfered with a proteolytic event during parasite replication.
Furthermore, ectopic expression of NcPI-S in P.berghei zygotes blocked their differentiation
to ookinetes. Consequently, oocyst production was severely impaired and the few surviving oo-
cysts were defective in sporulation. In contrast, ectopic expression of the inactive NcPI-Smut
did not have any effect on zygote to ookinete differentiation in vitro, and on mosquito virulence
in vivo. In TEM studies the majority of the developmentally arrested ookinetes showed a range
of morphological abnormalities indicative of a fatal toxic stress. The few morphologically ma-
ture ookinetes also exhibited severe defects with the most prominent being a drastic reduction
of the number of micronemes. In line with the reduced number of micronemes, the protein
level of the micronemal protein SOAP was significantly reduced even though the RNA levels of
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 16 / 23
this molecule were unaffected. Additional studies, showed elF2a increased phosphorylation in
NcPIS_3 zygotes/ookinetes that is indicative of translational repression. Studies in other organ-
isms (yeast, Toxoplasma), have shown that apart from the reduced global protein synthesis due
to the eIF2a phosphorylation, preferential translation of certain proteins helps the cells to cope
with the elevated stress [64]. If such is the case also in Plasmodium, it might not be surprising
that SOAP levels are decreased, in contrast to the BiP levels, (major ER chaperone in many or-
ganisms) which remain unaffected. It is noteworthy that inhibition of translation by the use of
cycloheximide results in a block in zygote to ookinete development (Siden-Kiamos and Deli-
gianni, unpublished).
Similarly to our study in T.gondii it was recently shown that elimination of one of the Kazal
inhibitors expressed in Toxoplasma, TgPI-1 (ΔTgPI1), results in higher parasite burden in vivo
suggesting that the endogenously expressed Kazal inhibitors also have a negative effect on
growth of the parasite [65]. Furthermore, ΔTgPI1 parasites exhibited additionally increased
bradyzoite gene expression and more efficient in vitro differentiation after stress of the tachy-
zoites in low CO
2
and alkaline pH [65]. Even though the target protease of TgPI-1 hasnt been
identified the construction of the ΔTgPI1 line led to the conclusion that PPIs can affect T.gon-
dii virulence and bradyzoite differentiation. The removal of a negative PPI regulator of prote-
ases in ΔTgPI1 parasites emphasizes the importance of the tight regulation of proteases in
apicomplexans. In contrast to TgPI-1, the function of NcPI-S in N.caninum is yet to be de-
fined. The notion, though, is that it might suppress proteolytic activity to protect resident PV
proteins from non-regulated proteolysis [34]. Similarly ectopic expression of active NcPI-S in
T.gondii, shows that addition of an exogenous PPI represents an insult against the balanced
regulation of protease activity, which is manifested by defects in parasite replication. More im-
portantly, the impaired oocyst development observed in P.berghei, the genome of which does
not encode a serine PPI, indicates that insults to the regulation of proteolysis cannot be tolerat-
ed in a system that has evolved to regulate proteolysis by PPI-independent mechanisms. Since
our efforts to generate transgenic P.berghei parasites expressing NcPI-S under a constitutive
promoter (eIF2a and hsp70) have so far been unsuccessful (Koussis and Loukeris, unpub-
lished), we assume that expression of NcPI-S in blood stages is probably detrimental for
the parasite.
A key question though that needs to be addressed in future studies is the identity of the tar-
get protease(s) of NcPI-S. The effects of NcPI-S in T.gondii tachyzoites and in P.berghei zy-
gotes/ookinetes are theoretically mediated through the disturbance or the complete inhibition
of processes involving subtilisin-like or rhomboid proteases. An impaired tachyzoite replica-
tion rate was previously reported as the main phenotypic manifestation conditionally depleting
a micronemal Toxoplasma rhomboid TgROM1, which shows residual Golgi localization in
tachyzoites [66]. Our notion though is that studies should also focus on other classes of mole-
cules. In Plasmodium spp and Toxoplasma genomes, proteins with a rhomboid fold have been
also identified (Derlins (Ders)). These are incapable of proteolysis but it is believed that they in-
teract with their client proteins in ways similar to the rhomboid proteases [67]. Plasmodium
spp. and T.gondii genomes encode for a yeast Der1p orthologue, hDer1-1, which is likely part
of the ER Associated Degradation (ERAD) machinery, and a second paralogue sDer1-1, which
is part of a duplicated ERAD-like translocon complex placed in the apicoplast [68]. This dupli-
cated ERAD-like translocon is possibly responsible for the transfer of apicoplast proteins across
the apicoplast plasma membranes. It would therefore be challenging to attempt silencing of
Ders at the zygote stage (by promoter swap) and investigate their effect on zygote to ookinete
transition and on the structural integrity of the zygote/ookinete, and compare it with the
NcPI-S line.
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 17 / 23
Apart from genetic studies, in vitro biochemical studies could also be implemented to find
interacting partners. The recombinantly expressed NcPI-S and its mutant form can be used for
pull down assays as has been previously done with a subtilisin Kazal inhibitor expressed by the
oomycete pathogen Phytophthora infestans [69]. Unfortunately, antibodies for serine proteases
in Apicomplexa are not available in many cases and therefore one possibility would be a com-
bination of pull down assays and a comparison of the whole proteome profiles of WT parasites
and those that express exogenous PPIs. Such an approach could help to delineate the role of ex-
ogenous PPI in both parasites and identify the target proteases of NcPI-S.
The results from this study support our initial hypothesis that NcPI-S (and potentially other
subtilisin specific Kazal inhibitors) may be used as a tool to interfere with the physiology of the
secretory apparatus of apicomplexan parasites, and in this way to manipulate their viability
and growth. With our study we exploited T.gondii parasites as a system for biochemical and
phenotypical validation of the chosen PPI. More importantly, since our main focus is P.berghei
mosquitos stages our interest is to develop tools that could lead to novel transmission blocking
strategies. Previously, studies have demonstrated the antiparasitic activity of antiretroviral pro-
tease inhibitors against P.falciparum [70], Leishmania [71] and T.gondii [72,73]. In addition a
native Kunitz-type serine protease inhibitor was recently proven to affect viability of T.cruzi
[74] and Leishmania [75]. The unique aspect of our study is that we chose to create transgenic
parasite lines with the PPI. We anticipate that by controlling the stage and the timing for
NcPI-S expression, as well as NcPI-Ss subcellular localization it may be possible to generate
parasitic strains growth attenuated in the vertebrate host, and at the same time incapable
of transmission.
Supporting Information
S1 Fig. Phenotypic analysis of NcPIS in T.gondii.A. Growth rate of NcPIs_8 clone compared
to that of RH. After 24 hrs of intracellular replication, vacuoles from the above strains were
scored according to their parasite context (1, 2, 4, 8, 16 and 32 parasites/vacuole). Error bars
represent the means ±S.E (n = 3 experiments per strain, duplicated samples/experiment); sta-
tistical significance was determined using t-test (p<0.001).B. 24hrs NcPIS_8 intracellular
tachyzoites, stained with a-myc antibody that detects NcPI-S (red) secreted into the parasito-
phorous vacuole partly co-localizing with the dense granule specific protein GRA3 (green).
Fluorescent images were collected at 100X with a Zeiss Axioscope2plus microscope equipped
with a CCD camera. Nuclei stained with DAPI (blue). Scale bars, 10μm. C. Extracellular freshly
lysed parasites 48 hrs p.i. were stained with-anti myc (red) and anti-ROM4 (Rhomboid prote-
ase 4) (green) antibodies. Nuclei were stained with DAPI (blue). Scale bars, 1μm.
(PDF)
S2 Fig. Strategy for the generation of transgenic NcPI-S expressing P.berghei parasites. A.
Schematic representation of the gene targeting strategy used for integrating NcPIS or NcPIs-
mut in the endogenous cssu-rrna locus. Double arrowhead line indicates the expected PCR
product for an intact locus. Primers used for diagnostic PCR (L665, L260, L270 and L740) are
indicated. B. PFIGE (Pulse-Field Inverted Gel Electrophoresis) analysis of transfected parasites.
(i) DNA from transfected parasites run on 1% agarose gel, showing chromosomes separated
according to their molecular size; Chromosomes 5/6 (which cant be separated from each
other) and 7 are indicated by arrows. (ii) The blot from the same gel hybridized with the 3
UTR sequences of Pbdhfr/ts. The probe detects the 3UTR of the endogenous dhfr gene on
chromosome 7, and the 3UTR of the selectable marker indicating correct integration on chro-
mosome 5. C. PCR genotypic analysis of the derived transgenic clones. Upper panel: Lane 3
corresponds to NcPIS_3 clone. The primer pair a (L665/L740) amplifies the integration
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 18 / 23
diagnostic 2.1kb band from genomic DNA isolated from all clones. Failure to amplify a 3kb
band using the primer pair c (L270/L740) from gDNA of the two clones indicates that integra-
tion took place at the cssu-rrna locus, while presence of a 3kb band amplified by the primer set
b (L260/L740) in WT control and the two NcPI-S expressing clones indicates the integrity of
dssu-rrna locus. (Lower panel) The same primer sets were used to confirm integration of
NcPISmut and NcPIS_1 in the specified locus.
(PDF)
S3 Fig. Localization studies of NcPI-S/NcPI-Smut and Bip in ookinetes. IFA analysis of
NcPI_S3 and NcPI_Smut lines, showing expression of the PPI (red) and the ER-chaperone BiP
(green) in P.berghei ookinetes. NcPI-Smut is expressed throughout the cytoplasm and the ER,
while NcPI-S has limited colocalisation with BiP. Scale bar: 5 μM.
(TIF)
S4 Fig. TEM images of arrested zygote/ookinetes. A. Developmentally arrested zygotes ex-
hibiting the characteristic dilation of the endomembrane system alongside extensive vacuola-
tion. B. Longitudinal sections of a sgIV ookinetes, in which the cytoplasm has been entirely
replaced by electron-lucent vacuoles. Some subcellular structures, however, are still identifiable,
such as subpellicular microtubules (arrowheads) and a well-structured collar with the charac-
teristic electron-dense depositions surrounding the apical split; notice vacuole excretion from
the apical split. A nucleus remnant (N) with a persistent electron dense area is defined by a
rough envelope (red arrow). Reduced number of micronemes is also shown in one ookinete
(white arrow).
(PDF)
S5 Fig. Western analysis of extracts derived from in vitro ookinete cultures of NcPIS_1
clone. Western analysis of extracts derived from in vitro ookinete cultures of NcPIS_1 and the
parental ANKA 15cy1A (ANKA) strain, with V5 Mab (revealing NcPI-S) and antibodies against
the micronemal protein SOAP, and the late zygote/ookinete specific P28 protein (sample nor-
malization control). While the levels of P28 are similar between the parental ANKA strain and
the NcPI-S clone, a reduction in the levels of SOAP is observed in the case of NcPIS_1 derived
extracts.
(PDF)
S1 Table. Primers used for DNA construct generation and genotype analysis.
(DOCX)
Acknowledgments
The authors thank Drs Chris J. Janse and Blandine Franke-Fayard for their help in establishing
P.berghei transgenic lines, Eva Papadogiorgaki for her excellent technical assistance with TEM,
Dr. Jean Francois Dubremetz, Dr Louis Weiss, Dr Ellen Knuepfer, Prof Dominique Soldati-
Favre and Dr. George Christofides for gifts of antibodies and Dr. Dina Vlachou for the plasmid
containing the CTRP promoter.
This manuscript is dedicated to the memory of Dr Zoi Tampaki and Dr. Thanasis Loukeris.
Author Contributions
Conceived and designed the experiments: TGL K. Kim VC APW. Performed the experiments:
ZT RSM EG SK K. Koussis. Analyzed the data: ZT RSM EG K. Koussis ISK TGL. Wrote the
paper: ZT RSM K. Koussis K. Kim VC APW ISK TGL.
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 19 / 23
References
1. Blackman MJ. Malarial proteases and host cell egress: an 'emerging' cascade. Cell Microbiol. 2008;
10: 19251934. doi: 10.1111/j.1462-5822.2008.01176.x PMID: 18503638
2. Kim K. Role of proteases in host cell invasion by Toxoplasma gondii and other Apicomplexa. Acta Trop.
2004; 91: 6981. PMID: 15158690
3. Li H, Child MA, Bogyo M. Proteases as regulators of pathogenesis: examples from the Apicomplexa.
Biochim Biophys Acta. 2012; 1824: 177185. doi: 10.1016/j.bbapap.2011.06.002 PMID: 21683169
4. Roiko MS, Carruthers VB. New roles for perforins and proteases in apicomplexan egress. Cell Micro-
biol. 2009; 11: 14441452. doi: 10.1111/j.1462-5822.2009.01357.x PMID: 19614666
5. McKerrow JH, Rosenthal PJ, Swenerton R, Doyle P. Development of protease inhibitors for protozoan
infections. Curr Opin Infect Dis. 2008; 21: 668672. doi: 10.1097/QCO.0b013e328315cca9 PMID:
18978536
6. Lagal V, Binder EM, Huynh MH, Kafsack BF, Harris PK, Diez R, et al. Toxoplasma gondii protease
TgSUB1 is required for cell surface processing of micronemal adhesive complexes and efficient adhe-
sion of tachyzoites. Cell Microbiol. 2010; 12: 17921808. doi: 10.1111/j.1462-5822.2010.01509.x
PMID: 20678172
7. Miller SA, Binder EM, Blackman MJ, Carruthers VB, Kim K. A conserved subtilisin-like protein TgSUB1
in microneme organelles of Toxoplasma gondii. J Biol Chem. 2001; 276: 4534145348. PMID:
11564738
8. Miller SA, Thathy V, Ajioka JW, Blackman MJ, Kim K. TgSUB2 is a Toxoplasma gondii rhoptry organ-
elle processing proteinase. Mol Microbiol. 2003; 49: 883894. PMID: 12890015
9. Binder EM, Kim K. Location, location, location: trafficking and function of secreted proteases of Toxo-
plasma and Plasmodium. Traffic. 2004; 5: 914924. PMID: 15522094
10. Alam A, Bhatnagar RK, Chauhan VS. Expression and characterization of catalytic domain of Plasmodi-
um falciparum subtilisin-like protease 3. Mol Biochem Parasitol. 2012; 183: 8489. doi: 10.1016/j.
molbiopara.2011.12.009 PMID: 22285468
11. Barale JC, Blisnick T, Fujioka H, Alzari PM, Aikawa M, Braun-Breton C, et al. Plasmodium falciparum
subtilisin-like protease 2, a merozoite candidate for the merozoite surface protein 142 maturase. Proc
Natl Acad Sci U S A. 1999; 96: 64456450. PMID: 10339607
12. Blackman MJ, Fujioka H, Stafford WH, Sajid M, Clough B, Fleck SL, et al. A subtilisin-like protein in se-
cretory organelles of Plasmodium falciparum merozoites. J Biol Chem. 1998; 273: 2339823409.
PMID: 9722575
13. Hackett F, Sajid M, Withers-Martinez C, Grainger M, Blackman MJ. PfSUB-2: a second subtilisin-like
protein in Plasmodium falciparum merozoites. Mol Biochem Parasitol. 1999; 103: 183195. PMID:
10551362
14. Dowse TJ, Pascall JC, Brown KD, Soldati D. Apicomplexan rhomboids have a potential role in micro-
neme protein cleavage during host cell invasion. Int J Parasitol. 2005; 35: 747756. PMID: 15913633
15. Dowse TJ, Soldati D. Rhomboid-like proteins in Apicomplexa: phylogeny and nomenclature. Trends
Parasitol. 2005; 21: 254258. PMID: 15922242
16. Koussis K, Withers-Martinez C, Yeoh S, Child M, Hackett F, Knuepfer E, et al. A multifunctional serine
protease primes the malaria parasite for red blood cell invasion. EMBO J. 2009; 28: 725735. doi: 10.
1038/emboj.2009.22 PMID: 19214190
17. Lin JW, Meireles P, Prudencio M, Engelmann S, Annoura T, Sajid M, et al. Loss-of-function analyses
defines vital and redundant functions of the Plasmodium rhomboid protease family. Mol Microbiol.
2013; 88: 318338. doi: 10.1111/mmi.12187 PMID: 23490234
18. Srinivasan P, Coppens I, Jacobs-Lorena M. Distinct roles of Plasmodium rhomboid 1 in parasite devel-
opment and malaria pathogenesis. PLoS Pathog. 2009; 5: e1000262. doi: 10.1371/journal.ppat.
1000262 PMID: 19148267
19. Yeoh S, O'Donnell RA, Koussis K, Dluzewski AR, Ansell KH, Osborne SA, et al. Subcellular discharge
of a serine protease mediates release of invasive malaria parasites from host erythrocytes. Cell. 2007;
131: 10721083. PMID: 18083098
20. Santos JM, Ferguson DJ, Blackman MJ, Soldati-Favre D. Intramembrane cleavageof AMA1 triggers
Toxoplasma to switch from an invasive to a replicative mode. Science. 2011; 331: 473477. doi: 10.
1126/science.1199284 PMID: 21205639
21. Blackman MJ, Corrie JE, Croney JC, Kelly G, Eccleston JF, Jameson DM. Structural and biochemical
characterization of a fluorogenic rhodamine-labeled malarial protease substrate. Biochemistry. 2002;
41: 1224412252. PMID: 12356327
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 20 / 23
22. J MS, Graindorge A, Soldati-Favre D. New insights into parasite rhomboid proteases. Mol Biochem
Parasitol. 2012; 182: 2736. doi: 10.1016/j.molbiopara.2011.11.010 PMID: 22173057
23. Banyal HS, Misra GC, Gupta CM, Dutta GP. Involvement of malarial proteases in the interaction be-
tween the parasite and host erythrocyte in Plasmodium knowlesi infections. J Parasitol. 1981; 67: 623
626. PMID: 7028937
24. Hadley T, Aikawa M, Miller LH. Plasmodium knowlesi: studies on invasion of rhesus erythrocytes by
merozoites in the presence of protease inhibitors. Exp Parasitol. 1983; 55: 306311. PMID: 6852169
25. Shaw MK, Roos DS, Tilney LG. Cysteine and serine protease inhibitors block intracellular development
and disrupt the secretory pathway of Toxoplasma gondii. Microbes Infect. 2002; 4: 119132. PMID:
11880042
26. Teo CF, Zhou XW, Bogyo M, Carruthers VB. Cysteine protease inhibitors block Toxoplasma gondii
microneme secretion and cell invasion. Antimicrob Agents Chemother. 2007; 51: 679688. PMID:
17145790
27. Krowarsch D, Cierpicki T, Jelen F, Otlewski J. Canonical protein inhibitors of serine proteases. Cell Mol
Life Sci. 2003; 60: 24272444. PMID: 14625687
28. Rimphanitchayakit V, Tassanakajon A. Structure and function of invertebrate Kazal-type serine protein-
ase inhibitors. Dev Comp Immunol. 2010; 34: 377386. doi: 10.1016/j.dci.2009.12.004 PMID:
19995574
29. Fenton JW 2nd. Leeches to hirulogs and other thrombin-directed antithrombotics. Hematol Oncol Clin
North Am. 1992; 6: 11211129. PMID: 1400076
30. Stubbs MT, Huber R, Bode W. Crystal structures of factor Xa specific inhibitors in complex with trypsin:
structural grounds for inhibition of factor Xa and selectivity against thrombin. FEBS Lett. 1995; 375:
103107. PMID: 7498454
31. Morris MT, Carruthers VB. Identification and partial characterization of a second Kazal inhibitor in Toxo-
plasma gondii. Mol Biochem Parasitol. 2003; 128: 119122. PMID: 12706808
32. Pszenny V, Angel SO, Duschak VG, Paulino M, Ledesma B, Yabo MI, et al. Molecular cloning, se-
quencing and expression of a serine proteinase inhibitor gene from Toxoplasma gondii. Mol Biochem
Parasitol. 2000; 107: 241249. PMID: 10779600
33. Bruno S, Duschak VG, Ledesma B, Ferella M, Andersson B, Guarnera EA, et al. Identification and char-
acterization of serine proteinase inhibitors from Neospora caninum. Mol Biochem Parasitol. 2004; 136:
101107. PMID: 15138071
34. Morris MT, Cheng WC, Zhou XW, Brydges SD, Carruthers VB. Neospora caninum expresses an un-
usual single-domain Kazal protease inhibitor that is discharged into the parasitophorous vacuole. Int J
Parasitol. 2004; 34: 693701. PMID: 15111091
35. Pfefferkorn ER, Borotz SE. Toxoplasma gondii: characterization of a mutant resistant to 6-thioxanthine.
Exp Parasitol. 1994; 79: 374382. PMID: 7957757
36. Roos DS, Donald RG, Morrissette NS, Moulton AL. Molecular tools for genetic dissection of the proto-
zoan parasite Toxoplasma gondii. Methods Cell Biol. 1994; 45: 2763. PMID: 7707991
37. Janse CJ, Franke-Fayard B, Waters AP. Selection by flow-sorting of genetically transformed, GFP-ex-
pressing blood stages of the rodent malaria parasite, Plasmodium berghei. Nat Protoc. 2006; 1: 614
623. PMID: 17406288
38. Soldati D, Lassen A, Dubremetz JF, Boothroyd JC. Processing of Toxoplasma ROP1 protein in nascent
rhoptries. Mol Biochem Parasitol. 1998; 96: 3748. PMID: 9851605
39. Kim K, Eaton MS, Schubert W, Wu S, Tang J. Optimized expression of green fluorescent protein in
Toxoplasma gondii using thermostable green fluorescent protein mutants. Mol Biochem Parasitol.
2001; 113: 309313. PMID: 11295185
40. Vlachou D, Zimmermann T, Cantera R, Janse CJ, Waters AP, Kafatos FC. Real-time, in vivo analysis
of malaria ookinete locomotion and mosquito midgut invasion. Cell Microbiol. 2004; 6: 671685. PMID:
15186403
41. Soldati D, Boothroyd JC. Transient transfection and expression in the obligate intracellular parasite
Toxoplasma gondii. Science. 1993; 260: 349352. PMID: 8469986
42. Black M, Seeber F, Soldati D, Kim K, Boothroyd JC. Restriction enzyme-mediated integration elevates
transformation frequency and enables co-transfection of Toxoplasma gondii. Mol Biochem Parasitol.
1995; 74: 5563. PMID: 8719245
43. Donald RG, Carter D, Ullman B, Roos DS. Insertional tagging, cloning, and expression of the Toxoplas-
ma gondii hypoxanthine-xanthine-guanine phosphoribosyltransferase gene. Use as a selectablemark-
er for stable transformation. J Biol Chem. 1996; 271: 1401014019. PMID: 8662859
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 21 / 23
44. Janse CJ, Ramesar J, Waters AP. High-efficiency transfection and drug selection of genetically trans-
formed blood stages of the rodent malaria parasite Plasmodium berghei. Nat Protoc. 2006; 1: 346
356. PMID: 17406255
45. Franke-Fayard B, Trueman H, Ramesar J, Mendoza J, van der Keur M, van der Linden R, et al. A Plas-
modium berghei reference line that constitutively expresses GFP at a high level throughout the com-
plete life cycle. Mol Biochem Parasitol. 2004; 137: 2333. PMID: 15279948
46. van Spaendonk RM, Ramesar J, van Wigcheren A, Eling W, Beetsma AL, van Gemert GJ, et al. Func-
tional equivalence of structurally distinct ribosomes in the malaria parasite, Plasmodium berghei. J Biol
Chem. 2001; 276: 2263822647. PMID: 11292830
47. Huynh MH, Rabenau KE, Harper JM, Beatty WL, Sibley LD, Carruthers VB. Rapid invasion of host cells
by Toxoplasma requires secretion of the MIC2-M2AP adhesive protein complex. EMBO J. 2003; 22:
20822090. PMID: 12727875
48. Black MW, Arrizabalaga G, Boothroyd JC. Ionophore-resistant mutants of Toxoplasma gondii reveal
host cell permeabilization as an early event in egress. Mol Cell Biol. 2000; 20: 93999408. PMID:
11094090
49. Sinden RE, Hartley RH, Winger L. The development of Plasmodium ookinetes in vitro: an ultrastructural
study including a description of meiotic division. Parasitology. 1985; 91 (Pt 2): 227244.
50. Reininger L, Billker O, Tewari R, Mukhopadhyay A, Fennell C, Dorin-Semblat D, et al. A NIMA-related
protein kinase is essential for completion of the sexual cycle of malaria parasites. J Biol Chem. 2005;
280: 3195731964. PMID: 15970588
51. Wan KL, Carruthers VB, Sibley LD, Ajioka JW. Molecular characterisation of an expressed sequence
tag locus of Toxoplasma gondii encoding the micronemal protein MIC2. Mol Biochem Parasitol. 1997;
84: 203214. PMID: 9084040
52. Winger LA, Tirawanchai N, Nicholas J, Carter HE, Smith JE, Sinden RE. Ookinete antigens of Plasmo-
dium berghei. Appearance on the zygote surface of an Mr 21 kD determinant identified by transmis-
sion-blocking monoclonal antibodies. Parasite Immunol. 1988; 10: 193207. PMID: 2453831
53. Srinivasan P, Fujioka H, Jacobs-Lorena M. PbCap380, a novel oocyst capsule protein, is essential for
malaria parasite survival in the mosquito. Cell Microbiol. 2008; 10: 13041312. doi: 10.1111/j.1462-
5822.2008.01127.x PMID: 18248630
54. Nall TA, Chappell KJ, Stoermer MJ, Fang NX, Tyndall JD, Young PR, et al. Enzymatic characterization
and homology model of a catalytically active recombinant West Nile virus NS3 protease. J Biol Chem.
2004; 279: 4853548542. PMID: 15322074
55. Ossorio PN, Schwartzman JD, Boothroyd JC. A Toxoplasma gondii rhoptry protein associated with
host cell penetration has unusual charge asymmetry. Mol Biochem Parasitol. 1992; 50: 115. PMID:
1542304
56. Cesbron-Delauw MF, Guy B, Torpier G, Pierce RJ, Lenzen G, Cesbron JY, et al. Molecular characteri-
zation of a 23-kilodalton major antigen secreted by Toxoplasma gondii. Proc Natl Acad Sci U S A.
1989; 86: 75377541. PMID: 2798425
57. Meissner M, Schluter D, Soldati D. Role of Toxoplasma gondii myosin A in powering parasite gliding
and host cell invasion. Science. 2002; 298: 837840. PMID: 12399593
58. Uzureau P, Barale JC, Janse CJ, Waters AP, Breton CB. Gene targeting demonstrates that the Plas-
modium berghei subtilisin PbSUB2 is essential for red cell invasion and reveals spontaneous genetic
recombination events. Cell Microbiol. 2004; 6: 6578. PMID: 14678331
59. Abraham EG, Pinto SB, Ghosh A, Vanlandingham DL, Budd A, Higgs S, et al. An immune-responsive
serpin, SRPN6, mediates mosquito defense against malaria parasites. Proc Natl Acad Sci U S A. 2005;
102: 1632716332. PMID: 16260729
60. Dessens JT, Siden-Kiamos I, Mendoza J, Mahairaki V, Khater E, Vlachou D, et al. SOAP, a novel ma-
laria ookinete protein involved in mosquito midgut invasion and oocyst development. Mol Microbiol.
2003; 49: 319329. PMID: 12828632
61. Narasimhan J, Joyce BR, Naguleswaran A, Smith AT, Livingston MR, Dixon SE, et al. Translation regu-
lation by eukaryotic initiation factor-2 kinases in the development of latent cysts in Toxoplasma gondii.
J Biol Chem. 2008; 283: 1659116601. doi: 10.1074/jbc.M800681200 PMID: 18420584
62. Zhang M, Fennell C, Ranford-Cartwright L, Sakthivel R, Gueirard P, Meister S, et al. The Plasmodium
eukaryotic initiation factor-2alpha kinase IK2 controls the latency of sporozoites in the mosquito salivary
glands. J Exp Med. 2010; 207: 14651474. doi: 10.1084/jem.20091975 PMID: 20584882
63. Fennell C, Babbitt S, Russo I, Wilkes J, Ranford-Cartwright L, Goldberg DE, et al. PfeIK1, a eukaryotic
initiation factor 2alpha kinase of the human malaria parasite Plasmodium falciparum, regulates stress-
response to amino-acid starvation. Malar J. 2009; 8: 99. doi: 10.1186/1475-2875-8-99 PMID:
19435497
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 22 / 23
64. Joyce BR, Tampaki Z, Kim K, Wek RC, Sullivan WJ Jr. The unfolded protein response in the protozoan
parasite Toxoplasma gondii features translational and transcriptional control. Eukaryot Cell. 2013; 12:
979989. doi: 10.1128/EC.00021-13 PMID: 23666622
65. Pszenny V, Davis PH, Zhou XW, Hunter CA, Carruthers VB, Roos DS. Targeted disruption of Toxoplas-
ma gondii serine protease inhibitor 1 increases bradyzoite cyst formation in vitro and parasite tissue
burden in mice. Infect Immun. 2012; 80: 11561165. doi: 10.1128/IAI.06167-11 PMID: 22202120
66. Brossier F, Starnes GL, Beatty WL, Sibley LD. Microneme rhomboid protease TgROM1 is required for
efficient intracellular growth of Toxoplasma gondii. Eukaryot Cell. 2008; 7: 664674. doi: 10.1128/EC.
00331-07 PMID: 18310358
67. Bergbold N, Lemberg MK. Emerging role of rhomboid family proteins in mammalian biology and dis-
ease. Biochim Biophys Acta. 2013; 1828: 28402848. doi: 10.1016/j.bbamem.2013.03.025 PMID:
23562403
68. Spork S, Hiss JA, Mandel K, Sommer M, Kooij TW, Chu T, et al. An unusual ERAD-like complex is tar-
geted to the apicoplast of Plasmodium falciparum. Eukaryot Cell. 2009; 8: 11341145. doi: 10.1128/
EC.00083-09 PMID: 19502583
69. Tian M, Huitema E, Da Cunha L, Torto-Alalibo T, Kamoun S. A Kazal-like extracellular serine protease
inhibitor from Phytophthora infestans targets the tomato pathogenesis-related protease P69B. J Biol
Chem. 2004; 279: 2637026377. PMID: 15096512
70. Parikh S, Gut J, Istvan E, Goldberg DE, Havlir DV, Rosenthal PJ. Antimalarial activity of human immu-
nodeficiency virus type 1 protease inhibitors. Antimicrob Agents Chemother. 2005; 49: 29832985.
PMID: 15980379
71. Savoia D, Allice T, Tovo PA. Antileishmanial activity of HIV protease inhibitors. Int J Antimicrob Agents.
2005; 26: 9294. PMID: 15955671
72. Derouin F. Anti-toxoplasmosis drugs. Curr Opin Investig Drugs. 2001; 2: 13681374. PMID: 11890349
73. Monzote L, Rodriguez M, Alfonso Y, Cox R. Antiretroviral activity of protease inhibitors against Toxo-
plasma gondii. Rev Inst Med Trop Sao Paulo. 2013; 55: 6567. PMID: 23328729
74. de Almeida Nogueira NP, Morgado-Diaz JA, Menna-Barreto RF, Paes MC, da Silva-Lopez RE. Effects
of a marine serine protease inhibitor on viability and morphology of Trypanosoma cruzi, the agent of
Chagas disease. Acta Trop. 2013; 128: 2735. doi: 10.1016/j.actatropica.2013.05.013 PMID:
23770204
75. Silva-Lopez RE, Morgado-Diaz JA, Chavez MA, Giovanni-De-Simone S. Effects of serine protease in-
hibitors on viability and morphology of Leishmania (Leishmania) amazonensis promastigotes. Parasitol
Res. 2007; 101: 16271635. PMID: 17726617
A Kazal Inhibitor Affects Apicomplexa Development
PLOS ONE | DOI:10.1371/journal.pone.0121379 March 24, 2015 23 / 23
... For Western-blot analysis, 10 6 cells (schizonts) were lysed in RIPA buffer (50 mM Tris-Cl pH 8.0, 150 mM NaCl, 2 mM EDTA, 1% NP-40, 1% Sodium Deoxycholate, 0.1% SDS) supplemented with Protease Inhibitor Cocktail (Sigma). Samples were processed for Western blot analysis as has been previously described [38] using rat α-HA antibody (3F10, Roche; 1:1,000) and goat anti-rat HRP conjugate as a secondary antibody (Invitrogen; 1:10,000). The blot was developed using the ECL system (SuperSignalWest Pico, Thermo Scientific) following manufacturer's instructions. ...
Article
Full-text available
Site-2 proteases (S2P) belong to the M50 family of metalloproteases, which typically perform essential roles by mediating activation of membrane–bound transcription factors through regulated intramembrane proteolysis (RIP). Protease-dependent liberation of dormant transcription factors triggers diverse cellular responses, such as sterol regulation, Notch signalling and the unfolded protein response. Plasmodium parasites rely on regulated proteolysis for controlling essential pathways throughout the life cycle. In this study we examine the Plasmodium-encoded S2P in a murine malaria model and show that it is expressed in all stages of Plasmodium development. Localisation studies by endogenous gene tagging revealed that in all invasive stages the protein is in close proximity to the nucleus. Ablation of PbS2P by reverse genetics leads to reduced growth rates during liver and blood infection and, hence, virulence attenuation. Strikingly, absence of PbS2P was compatible with parasite life cycle progression in the mosquito and mammalian hosts under physiological conditions, suggesting redundant or dispensable roles in vivo.
Article
Full-text available
BACKGROUND: Post-transcriptional control of gene expression is suspected to play an important role in malaria parasites. In yeast and metazoans, part of the stress response is mediated through phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha), which results in the selective translation of mRNAs encoding stress-response proteins. METHODS: The impact of starvation on the phosphorylation state of PfeIF2alpha was examined. Bioinformatic methods were used to identify plasmodial eIF2alpha kinases. The activity of one of these, PfeIK1, was investigated using recombinant protein with non-physiological substrates and recombinant PfeIF2alpha. Reverse genetic techniques were used to disrupt the pfeik1 gene. RESULTS: The data demonstrate that the Plasmodium falciparum eIF2alpha orthologue is phosphorylated in response to starvation, and provide bioinformatic evidence for the presence of three eIF2alpha kinases in P. falciparum, only one of which (PfPK4) had been described previously. Evidence is provided that one of the novel eIF2alpha kinases, PfeIK1, is able to phosphorylate the P. falciparum eIF2alpha orthologue in vitro. PfeIK1 is not required for asexual or sexual development of the parasite, as shown by the ability of pfeik1- parasites to develop into sporozoites. However, eIF2alpha phosphorylation in response to starvation is abolished in pfeik1- asexual parasites CONCLUSION: This study strongly suggests that a mechanism for versatile regulation of translation by several kinases with a similar catalytic domain but distinct regulatory domains, is conserved in P. falciparum.
Article
Full-text available
The Unfolded Protein Response (UPR) is an important regulatory network that responds to perturbations in protein homeostasis in the endoplasmic reticulum (ER). In mammalian cells, the UPR features translational and transcriptional mechanisms of gene expression aimed at restoring proteostatic control. A central feature of the UPR is phosphorylation of the α subunit of eIF2 by PERK (EIF2AK3/PEK), which reduces the influx of nascent proteins into the ER by lowering global protein synthesis, coincident with preferential translation of key transcription activators of genes that function to expand the processing capacity of this secretory organelle. Upon ER stress, the apicomplexan parasite Toxoplasma gondii is known to induce phosphorylation of Toxoplasma eIF2α and lower translation initiation. To characterize the nature of the ensuing UPR in this parasite, we carried out microarray analyses to measure the changes in the transcriptome and in translational control during ER stress. We determined that a collection of transcripts linked with the secretory process are induced in response to ER stress, supporting the idea that a transcriptional induction phase of the UPR occurs in Toxoplasma. Furthermore, we determined that about 500 gene transcripts showed enhanced association with translating ribosomes during ER stress. Many of these target genes are suggested to be involved in gene expression, including JmjC5, which continues to be actively translated during ER stress. This study indicates that Toxoplasma triggers a UPR during ER stress that features both translational and transcriptional regulatory mechanisms, which is likely to be important for parasite invasion and development.
Article
Full-text available
The introduction of highly active antiretroviral therapy (HAART) has caused a marked reduction in the occurrence and severity of parasitic infections, including the toxoplasmic encephalitis (TE). These changes have been attributed to the restoration of cell-mediated immunity. This study was developed to examine the activity of six antiretroviral protease inhibitors (API) on Toxoplasma gondii tachyzoites. The six API showed anti-Toxoplasma activity, with IC50 value between 1.4 and 6.6 µg/mL. Further studies at the molecular level should be performed to clarify if the use of API could be beneficial or not for AIDS patients with TE.
Article
Two cDNA clones obtained from the Neospora caninum Expressed Sequence Tag project were selected by their homology with the Toxoplasma gondii serine proteinase inhibitor (serpin) gene, TgPI-1 and TgPI-2. One of them, named NcPI-H, showed several premature stop codons. The other cDNA, named NcPI-S, encoded a 79 amino acid protein containing a putative signal peptide and only one non-classical Kazal domain. Two other N. caninum EST sequences (NcEST1 and NcEST2) and one from Eimeria tenella (EtPI-S) were retrieved from the database. Amino acid sequence analysis suggested that NcEST1 and NcEST2 might be the N. caninum counterparts of TgPI-1 and TgPI-2, respectively. EtEST-S, as NcPI-S, is a single domain serpin. The open reading frame encoding the mature version of NcPI-S was expressed as recombinant protein, fused to a 6 histidine tag in Escherichia coli. Specific rabbit antiserum generated against the recombinant NcPI-S was used in immunoblot assays. Bands of 20, 30, 40, and 66-kDa were detected by SDS-PAGE of whole parasite homogenate. In addition, when an anti-TgPI-1 serum was used, bands of 25 and 35-kDa were detected indicating that there is no cross-reactivity between both serpins, and showing as well, the presence of another putative serpin in N. caninum. The recombinant protein NcPI-S, inhibited bacterial subtilisin completely, and showed lower inhibitory capacity on human neutrophil elastase, animal trypsin, and chymotrypsin, suggesting differences in effectiveness.
Article
The genetic structure of Toxoplasma gondii is notable chiefly for being relatively conventional— similar to that of its mammalian host cells with respect to gene organization, codon usage, and nucleotide bias. These observations have led several investigators to examine the feasibility of molecular transformation in this parasite. This chapter outlines the use of several of the molecular genetic tools that have recently been developed for the T. gondii system. An introduction to parasite culture techniques is also provided. Recombinant molecules can be expressed either transiently or as stable transformants, as episomes or integrated into the parasite genome, and as single copy or multicopy transgenes. Stable integration can be produced by random nonhomologous recombination, single-site homologous recombination, or perfect gene replacement. Many of these outcomes can be selected specifically using appropriate vectors and transformation conditions. The extraordinarily high frequencies of stable transformation observed permit cloning by complementation, insertional mutagenesis/marker rescue, gene knock-outs, and allelic replacement. In combination with available classical and “cell-genetic” possibilities and physical and genetic mapping strategies, these tools provide a powerful arsenal for investigations into the biology of intracellular parasitism.
Article
The expressed sequence tag (EST) dataset of Toxoplasma gondii provides a wealth of information towards gene discovery. The complete cDNA and genomic sequence of EST tgc050 locus shows that it contains five copies of the conserved thrombospondin (TSP)-like motif present in a number of molecules with adhesive properties. A conserved region implicated with the adhesive characteristic of another group of proteins including several integrins, is also present in this molecule. The protein encoded by this sequence (rc50) is strongly recognised by monoclonal antibodies to MIC2. Affinity purified anti-rc50 antisera specifically reacted with a single protein of identical molecular mass as MIC2 and exclusively labeled the micronemes of T. gondii by cryo-immunoelectron microscopy. These results demonstrate that c50 encodes for MIC2, a previously characterised microneme protein of T. gondii. The extensive sequence similarity across multiple protein domains provides evidence that the protein encoded by this locus is the homologue to the Etp100 microneme protein of Eimeria tenella.
Article
It has been reported that serine peptidase activities of Trypanosoma cruzi play crucial roles in parasite dissemination and host cell invasion and therefore their inhibition could affect the progress of Chagas disease. The present study investigates the interference of the Stichodactyla helianthus Kunitz-type serine protease inhibitor (ShPI-I), a 55-amino acid peptide, in T. cruzi serine peptidase activities, parasite viability, and parasite morphology. The effect of this peptide was also studied in Leishmania amazonensis promastigotes and it was proved to be a powerful inhibitor of serine proteases activities and the parasite viability. The ultrastructural alterations caused by ShPI-I included vesiculation of the flagellar pocket membrane and the appearance of a cytoplasmic vesicle that resembles an autophagic vacuole. ShPI-I, which showed itself to be an important T. cruzi serine peptidase inhibitor, reduced the parasite viability, in a dose and time dependent manner. The maximum effect of peptide on T. cruzi viability was observed when ShPI-I at 1×10(-5)M was incubated for 24 and 48h which killed completely both metacyclic trypomastigote and epimastigote forms. At 1×10(-6)M ShPI-I, in the same periods of time, reduced parasite viability about 91 to 95% respectively. Ultrastructural analysis demonstrated the formation of concentric membranar structures especially in the cytosol, involving organelles and small vesicles. Profiles of endoplasmic reticulum were also detected, surrounding cytosolic vesicles that resembled autophagic vacuoles. These results suggest that serine peptidases are important in T. cruzi physiology since the inhibition of their activity killed parasites in vitro as well as inducing important morphological alterations. Protease inhibitors thus appear to have a potential role as anti-trypanosomatidal agents.
Article
From proteases that cleave peptide bonds in the plane of the membrane, rhomboids have evolved into a heterogeneous superfamily with a wide range of different mechanistic properties. In mammals 14 family members have been annotated based on a shared conserved membrane-integral rhomboid core domain, including intramembrane serine proteases and diverse proteolytically inactive homologues. While the function of rhomboid proteases is the proteolytic release of membrane-tethered factors, rhomboid pseudoproteases including iRhoms and derlins interact with their clients without cleaving them. It has become evident that specific recognition of membrane protein substrates and clients by the rhomboid fold reflects a spectrum of cellular functions ranging from growth factor activation, trafficking control to membrane protein degradation. This review summarizes recent progress on rhomboid family proteins in the mammalian secretory pathway and raises the question whether they can be seen as new drug targets for inflammatory diseases and cancer.
Article
Rhomboid-like proteases cleave membrane-anchored proteins within their transmembrane domains. In apicomplexan parasites substrates include molecules that function in parasite motility and host cell invasion. While two Plasmodium rhomboids, ROM1 and ROM4, have been examined, the roles of the remaining six rhomboids during the malaria parasite's life cycle are unknown. We present systematic gene deletion analyses of all eight Plasmodium rhomboid-like proteins as a means to discover stage-specific phenotypes and potential functions in the rodent malaria model, P. berghei. Four rhomboids (ROM4, 6, 7 and 8) are refractory to gene deletion, suggesting an essential role during asexual blood stage development. In contrast ROM1, 3, 9 and 10 were dispensable for blood stage development and exhibited no, subtle or severe defects in mosquito or liver development. Parasites lacking ROM9 and ROM10 showed no major phenotypic defects. Parasites lacking ROM1 presented a delay in blood stage patency following liver infection, but in contrast to a previous study blood stage parasites had similar growth and virulence characteristics as wild type parasites. Parasites lacking ROM3 in mosquitoes readily established oocysts but failed to produce sporozoites. ROM3 is the first apicomplexan rhomboid identified to play a vital role in sporogony.