ArticlePDF Available

A var gene promoter controls allelic exclusion of virulence genes in Plasmodium falciparum malaria

Authors:

Abstract and Figures

Mono-allelic expression of gene families is used by many organisms to mediate phenotypic variation of surface proteins. In the apicomplexan parasite Plasmodium falciparum, responsible for the severe form of malaria in humans, this is exemplified by antigenic variation of the highly polymorphic P. falciparum erythrocyte membrane protein 1 (PfEMP1). PfEMP1, encoded by the 60-member var gene family, represents a major virulence factor due to its central role in immune evasion and intravascular parasite sequestration. Mutually exclusive expression of PfEMP1 is controlled by epigenetic mechanisms involving chromatin modification and perinuclear var locus repositioning. Here we show that a var promoter mediates the nucleation and spreading of stably inherited silenced chromatin. Transcriptional activation of this promoter occurs at the nuclear periphery in association with chromosome-end clusters. Additionally, the var promoter sequence is sufficient to infiltrate a transgene into the allelic exclusion programme of var gene expression, as transcriptional activation of this transgene results in silencing of endogenous var gene transcription. These results show that a var promoter is sufficient for epigenetic silencing and mono-allelic transcription of this virulence gene family, and are fundamental for our understanding of antigenic variation in P. falciparum. Furthermore, the PfEMP1 knockdown parasites obtained in this study will be important tools to increase our understanding of P. falciparum-mediated virulence and immune evasion.
Content may be subject to copyright.
© 2006 Nature Publishing Group
Avar gene promoter controls allelic exclusion of
virulence genes in Plasmodium falciparum malaria
Till S. Voss
1
, Julie Healer
1
, Allison J. Marty
1,2
, Michael F. Duffy
3
, Jennifer K. Thompson
1
, James G. Beeson
1
,
John C. Reeder
4
, Brendan S. Crabb
1
& Alan F. Cowman
1
Mono-allelic expression of gene families is used by many organ-
isms to mediate phenotypic variation of surface proteins. In the
apicomplexan parasite Plasmodium falciparum, responsible for
the severe form of malaria in humans, this is exemplified by
antigenic variation of the highly polymorphic P. falciparum
erythrocyte membrane protein 1 (PfEMP1)
1,2
. PfEMP1, encoded
by the 60-member var gene family
3–6
, represents a major virulence
factor due to its central role in immune evasion and intravascular
parasite sequestration. Mutually exclusive expression of PfEMP1
is controlled by epigenetic mechanisms involving chromatin
modification and perinuclear var locus repositioning
7,8
. Here we
show that a var promoter mediates the nucleation and spreading
of stably inherited silenced chromatin. Transcriptional activation
of this promoter occurs at the nuclear periphery in association
with chromosome-end clusters. Additionally, the var promoter
sequence is sufficient to infiltrate a transgene into the allelic
exclusion programme of var gene expression, as transcriptional
activation of this transgene results in silencing of endogenous
var gene transcription. These results show that a var promoter
is sufficient for epigenetic silencing and mono-allelic transcrip-
tion of this virulence gene family, and are fundamental for our
understanding of antigenic variation in P. falciparum. Further-
more, the PfEMP1 knockdown parasites obtained in this study
will be important tools to increase our understanding of
P. falciparum-mediated virulence and immune evasion.
In individual P. falciparum parasites one var gene is expressed and
switching to an alternative occurs by in situ transcriptional acti-
vation
9,10
.var genes are flanked by one of three upstream sequences
(upsA,upsB and upsC)
6
, and differential expression of these subtypes
is linked to disease
11
.UpsA- and upsB-type var genes are located
subtelomerically, whereas upsC-type var genes are in chromosome-
internal clusters. UpsB and upsC sequences display promoter activi-
ties
12,13
and interact with DNA-binding proteins
14
. Cooperative
interactions in cis between upsC and the var intron have been
implicated in silencing
15–17
. A role for silent information regulator
2 (PfSIR2) in var silencing has been demonstrated
7,8
. These findings
suggest a vital function of var promoters in epigenetic regulation of
this family.
To test this hypothesis we transfected P. falciparum strain 3D7 with
plasmids pHBupsC, pHBupsC
R
and pHBupsC
RI
, each containing
blasticidin deaminase (bsd) and human dihydrofolate reductase
(hdhfr), to encode resistance to blasticidin-S and WR99210
(WR), respectively (Fig. 1a). Transfected parasites were grown on
blasticidin-S to obtain 3D7/upsC, 3D7/upsC
R
and 3D7/upsC
RI
carrying episomes. The hdhfr gene represented a tool to analyse var
promoter function, uncoupled from its chromosomal context, in its
natural (WR
2
) and activated (WR
þ
) state.
Growth assays showed that all three transfectants were sensitive to
WR (Fig. 1b). Parasites transfected with control constructs, where the
upsC promoter was replaced with the calmodulin (cam) promoter,
were WR resistant. After selection of 3D7/upsC, 3D7/upsC
R
and
3D7/upsC
RI
with WR, resistant populations were established after
six, four and ten generations, respectively. Similarly, the half-
maximal inhibitory concentration (IC
50
) for unselected populations
was the same as for the WR-sensitive parent 3D7, whereas after
selection they were resistant (Fig. 1c). This suggested that the
upsC-hdhfr cassette was silenced and WR challenge selected for
rare parasites where upsC was activated. To confirm this we mon-
itored hDHFR expression by immunofluorescence (Fig. 1d). A total
of 0.6% of parasites expressed hDHFR in WR-untreated parasites
whereas 80% showed detectable levels after selection. When the
WR-resistant population was maintained without WR for 40 genera-
tions the percentage of hDHFR-expressing cells dropped to 61%.
This demonstrated that upsC-mediated expression was variegated
and stably inherited with the same dynamics as var switching rates
18
.
In WR-untreated 3D7/upsC, 3D7/upsC
R
and 3D7/upsC
RI
parasites no hdhfr transcripts were observed, consistent with tran-
scriptional silencing (Fig. 2a and Supplementary Fig. 2). High hdhfr
transcript levels were present after WR selection, and the activated
episomal upsC promoter displayed temporal transcription similar to
endogenous counterparts
14
. Therefore the episomal promoter con-
tained sufficient regulatory information for upsC-mediated control
and was unaffected by the intron and/or the subtelomeric repeat
element rep20 (see below). Furthermore, the silencing emanating
from upsC had regional effects in cis because transcription of the bsd
gene was substantially reduced on all silenced episomes compared
to activated forms and the control plasmid pHBcam (Fig. 2a, c).
Alterations in transcriptional activity were specific to upsC as no
differences in hdhfr or bsd transcript abundance was evident in
3D7/cam parasites before and after WR selection (Fig. 2b). The var
intron and rep20 played no direct role in silencing because cam
promoter activity was unaffected by these elements (Supplementary
Fig. 3). Our analysis shows that the upsC promoter provides
sufficient information for epigenetic control to establish a silenced
or activated state.
We demonstrated that the P. falciparum nuclear periphery was
associated with transcriptional silencing
7
, and we expected that
targeting of pHBupsC
R
to chromosome-end clusters by inclusion
of rep20 (ref. 19) would result in increased silencing. However, rep20
had no effect on upsC-mediated silencing (Fig. 2a). In contrast, the
intron in 3D7/upsC
RI
antagonized epigenetic activation and spread-
ing of chromatin activation into the bsd locus (Fig. 2a). This agrees
with previous results suggesting that the upsC–intron interaction has
boundary function to protect var genes in chromosome-central
LETTERS
1
The Walter and Eliza Hall Institute of Medical Research, Parkville 3050, Australia.
2
Department of Microbiology, Monash University, Clayton 3800, Australia.
3
Department of
Medicine, The University of Melbourne, The Royal Melbourne Hospital, Parkville 3050, Australia.
4
Papua New Guinea Institute of Medical Research, Goroka EHP 441, Papua
New Guinea.
Vol 439|23 February 2006|doi:10.1038/nature04407
1004
© 2006 Nature Publishing Group
clusters from activation through nearby euchromatic regions and
neighbouring var genes
17
. In addition, the delayed resistance of
3D7/upsC
RI
parasites compared to 3D7/upsC and 3D7/upsC
R
para-
sites (Fig. 1b) indicated that activation of upsC paired with the intron
occurred less frequently than activation of the upsC promoter alone.
This is consistent with the role of the var intron in silencing
15–17
and
suggests that interplay between these elements provokes a solid
and stably inherited silenced chromatin environment. However, in
contrast to a recent study
16
we did not find that deletion of the var
intron was required to activate the upsC promoter (Supplementary
Discussion).
We showed previously that gene expression at the nuclear periph-
ery is restricted to a perinuclear region and activation and silencing
of var genes is linked to locus repositioning
7
. We determined the
nuclear position of episomal and integrated versions of active and
Figure 1 |WR sensitivity of upsC promoter transfectants. a, Vector maps.
hsp86 50, heat-shock protein 86 promoter; Pb T, P. berghei dhfr-thymidilate
synthase terminator; upsC,upsC upstream sequence
13
;PfT,P. falciparum
hrp2 30terminator; rep20, 0.5-kb rep20 repeats; intron, 0.6-kb var intron
sequence
17
. Scale bar, 1kb. b, Growth assay. Parasites were challenged with
WR at day 0. Assays were repeated twice with the same result. c,WR
sensitivities before (open) and after (filled) WR selection. d, hDHFR
expression visualized by indirect immunofluorescence assay. Percentage of
hDHFR-expressing parasites before (2WR, 2 of 361) and after (þWR, 125
of 157) WR treatment, and in WR-selected parasites maintained without
WR for 40 generations (þ/2WR, 269 of 422). Error bars are 95% confidence
intervals (P,0.001). Similar results were obtained for 3D7/upsC and
3D7/upsC
RI
(data not shown).
Figure 2 |Epigenetic regulation of upsC.a, Silencing and activation of
upsC-regulated transcription. Northern blots showing episomal hdhfr and
bsd transcription. Transcription of cam is a stage-specific loading control.
Densitometry compares relative hdhfr and bsd transcript per promoter
before and after selection on WR (see Supplementary Methods). b,hdhfr
and bsd transcription in 3D7/cam. c, Spreading of upsC-mediated silencing.
bsd transcription in WR-untreated 3D7/upsC and 3D7/cam parasites. The
densitometry chart compares relative bsd transcript production per hsp86
promoter. 1, 0–12h post-infection (h.p.i.); 2, 12–24 h.p.i.; 3, 24–36 h.p.i.;
4, 32–44 h.p.i.
NATURE|Vol 439|23 February 2006 LETTERS
1005
© 2006 Nature Publishing Group
silenced hdhfr transgenes by fluorescent in situ hybridization (FISH)
(Fig. 3 and Supplementary Fig. 4). In agreement with a previous
report
20
we found silenced and active chromosome-internal var loci
at the nuclear periphery. We also demonstrate that a chromosome-
central var locus preferentially co-localizes with telomeric clusters,
irrespective of upsC transcriptional state. This was not observed for
episomally maintained pHBcam, which, as expected for episomes
devoid of rep20 in P. falciparum
19
, showed no preferential co-
localization with telomere clusters (38 ^2.4% co-localization).
Therefore, as the parasites constitutively transcribed bsd,the
telomeric clusters identified by co-localization with integrated
pHBupsC probably represent the previously identified active
perinuclear zone
7
. Surprisingly, the activated pHBupsC episome
co-localized with chromosome-end clusters as frequently as
integrated versions, in contrast to the silenced episomes that
displayed no preferential co-localization (P¼0.01, paired one-tailed
t-test) (Fig. 3b). This indicates that activation of upsC is restricted to
a specific site associated with the active perinuclear zone. Addition-
ally, these findings may explain why targeting of pHBupsC
R
to
telomeric clusters resulted in faster acquisition of WR resistance
(Fig. 1b).
The above results suggested that upsC activation interferes with
endogenous var transcription. To test this we analysed var transcrip-
tion using three universal var probes and one probe specific to var
PFL1960w where integration of pHBupsC occurred (see Supplemen-
tary Methods and Supplementary Fig. 4). 3D7/upsC parasites
showed a var messenger RNA pattern similar to 3D7 when the
plasmid-encoded upsC promoter was silenced (Fig. 4a). Interestingly,
PFL1960w was highly transcribed in these parasites, probably due to
sequestration of this locus to the transcriptionally active perinuclear
zone as a result of bsd transcription. After activation of upsC none
of the probes detected any significant var transcription, but the
upsC-controlled hdhfr was transcribed. The observation that
Figure 3 |Nuclear localization of silenced and activated upsC loci.
a, Nuclear localization of plasmids in 3D7/upsC and 3D7/cam by FISH.
Epifluorescence images of nuclei (DAPI) and nuclei hybridized with TARE4
(red) and pGEM (green) to identify chromosome-end cluster and plasmid
positions, respectively. b, Quantification of co-localization. n, average
number of nuclei scored. Error bars are 95% confidence intervals. P-values
were generated using a paired one-tailed t-test comparing the per cent
co-localization in each of the 3D7/upsC lines to the control 3D7/cam.
S, silenced upsC promoter; A, activated upsC promoter.
Figure 4 |Effect of upsC activation on mono-allelic var transcription.
a, Northern blot analysis. var transcription was monitored by hybridization
with specific and universal var probes (Supplementary Methods). The short
transcripts above 2.37 kb detected with var exon 2 and varC represent
‘sterile’ exon 2 transcripts
3
.kahrp, knob-associated histidine-rich protein;
msp8, merozoite surface protein 8; S, silenced upsC; A, activated upsC;R,
ring stage; T, trophozoite stage; 2WR, WR-unselected parasites; 2/þWR,
WR-selected parasites grown in the absence of drug for 70 generations.
b, Binding of iRBCs to CD36. Graph showing iRBC binding to CD36 for
WR-selected parasites (þWR) and WR-selected parasites maintained
without drug for 70 generations (2/þWR) compared to WR-unselected
parasites (2WR). Values for WR-unselected and selected parasites are
means (^s.e.m.) of six experiments in duplicate with three independent
transfectants (3D7/upsC, 3D7/upsC
R
, 3D7/upsC
RI
); values for the 2/þWR
parasites are means of two experiments performed in duplicate with the
3D7/upsC transfectant. c, Binding of serum IgG to iRBCs. Bar graph shows
flow cytometry of IgG median binding for sera of 20 malaria-exposed adults,
and five non-exposed controls, to the surface of iRBCs for WR-unselected
(black) and WR-selected (white) 3D7/upsC parasites. Error bars are 95%
confidence intervals.
LETTERS NATURE|Vol 439|23 February 2006
1006
© 2006 Nature Publishing Group
PFL1960w transcripts were absent in this population demonstrates
allelic exclusion and promoter switching within a chromosome-
internal var cluster. This knockdown of var transcription occurred
in rings and trophozoites and was observed with 3D7/upsC
R
and
3D7/upsC
RI
carrying episomal or integrated plasmids (Fig. 4a,
Supplementary Fig. 5). var transcripts reappeared in WR-resistant
parasites after removal of WR, indicating that a subpopulation had
switched from transcribing hdhfr to endogenous var genes (Fig. 4a).
Therefore, var promoter activation is a crucial step in control of
allelic var exclusion because transcriptional activation of one var
promoter-associated locus inhibits transcription of the remaining
var loci.
To confirm these observations we tested the PfEMP1-mediated
binding of infected red blood cells (iRBCs) to CD36 and chondroitin
sulphate A (CSA). In WR-selected parasites binding of iRBCs
to CD36 was reduced to 14% of WR-untreated parasites, and
WR-resistant parasites cultured in the absence of WR had inter-
mediate binding, showing that var activation and PfEMP1
expression was returning as hdhfr was silenced (Fig. 4b). None of
the parasites tested bound to CSA (not shown). In addition, IgG
binding to the surface of infected erythrocytes in sera from malaria-
exposed donors was significantly higher for WR-unselected com-
pared to WR-selected 3D7/upsC parasites (median ^s.d. fluores-
cence 26.4 ^20.3 versus 9.0 ^7.2, respectively; P,0.00001,
Wilcoxons signed-rank sum test), and there was little binding of
IgG among samples from non-exposed donors (median fluorescence
0.9 ^0.9 versus 0 ^0.3) (Fig. 4c and Supplementary Fig. 6). These
findings are consistent with a substantial reduction or absence of
PfEMP1 on the iRBC surface due to silencing of endogenous var
transcription.
We have identified two important features of var promoters in
epigenetic regulation and mono-allelic expression. First, the upsC var
promoter mediates nucleation and spreading of stably inherited
silenced chromatin through interactions between unidentified
cis-acting promoter motifs and the P. falciparum silencing machin-
ery. We conclude that the upsC promoter is required to maintain
chromosome-internal var genes in their silenced default state, and
preliminary data indicate that upsB promoters have a similar role in
subtelomeric var silencing (T.S.V. et al., unpublished data). Second,
the information provided by the upsC promoter is sufficient to insert
a locus into the allelic exclusion programme of var transcription
independent of chromosomal context (Supplementary Discussion).
On the basis of our findings, the simplest model for mechanisms
underlying mono-allelic var expression is a unique perinuclear
compartment, associated with the active chromosome-end cluster,
responsible for transcription of a single var locus (Supplementary
Fig. 1). Switching in var transcription, and thus antigenic variation of
PfEMP1, would occur by competition of silenced var promoters for
occupancy of this compartment. A similar mechanism has been
identified to control mono-allelic expression of vsg genes in African
trypanosomes
21
, and has been proposed to control mono-allelic
transcription of the mammalian odorant receptor gene family
22
.
The results presented here are, to our knowledge, the first description
of transcriptional repression of a gene family and are fundamental
to understanding antigenic variation and epigenetic regulation in
P. falciparum and mono-allelic transcription and phenotypic varia-
tion in other systems. The var knockdown parasites obtained in this
study provide an invaluable tool to understand the role of PfEMP1
in parasite-mediated virulence, protective immunity and cyto-
adherence.
METHODS
Parasites, drug sensitivity and cyto-adherence. P. falciparum 3D7 and trans-
fected parasites were cultured as described previously
23
. Growth synchronization
was achieved using sorbitol. A total of 250
m
l of packed iRBCs (5–10% ring
parasites) were transfected by electroporation of 100
m
g of plasmid. Selection on
blasticidin-S (2
m
gml
21
) was 4–6 h after transfection. Selection on WR99210
was at 4 nM. Drug sensitivity assays were performed with standard techniques.
Cyto-adherence of parasitized erythrocytes to CD36 and CSA were as
described
24
.
Transfection constructs. The puromycin resistance cassette in pHH1/pac
25
was
excised with NotI/BglII. After ligation of adaptors the cassette was cloned into
SacII/NotI-digested pHHMC*/R0.5 (ref. 19) to generate pHH_VP, where a
0.5-kilobase (kb) rep20 fragment separates the head-to-tail expression cassettes.
The vector pHBcam
R
was obtained by replacing the puromycin resistance gene
with bsd amplified from pCBM
26
. pHBcam
RI
was generated by ligation of a
0.6-kb var intron fragment
17
into the EcoRI site of pHBcam
R
. pHBupsC
R
was
generated by replacing the BglII/BamHI cam promoter with the upsC promoter
(PFL1960w) amplified from pCAT5B1 (ref. 13). pHBupsC
RI
was derived in
thesamewayfrompHBcam
RI
. The 0.5-kb rep20 sequence in pHBcam
R
and pHBupsC
R
was deleted by digestion with PstI/BglII, end-polishing and
re-ligation to yield pHBcam and pHBupsC, respectively.
Southern and northern analysis. gDNA was digested with EcoRV and PvuII.
Southern blots were probed with hdhfr and pfsir2 fragments. Copy number was
determined by densitometry comparing signal intensities of EcoRV/PvuII frag-
ments representing hdhfr to the EcoRV fragment from the single copy pfsir2.
Plasmid integration was mapped by hybridization with hdhfr and a 735-base pair
fragment derived from the 5 0end of PFL1960w. Pulsed-field gel electrophoresis
(PFGE) was carried out as described
27
. Northern experiments are described in
detail in Supplementary Methods.
Indirect immunofluorescence assay and FISH. Indirect immunofluorescence
assays to detect hDHFR expression were performed as described
28
using a
monoclonal mouse anti-bovine dihydrofolate reductase antibody (BD
Biosciences) and anti-mouse Alexa-Fluor 488-nm (Molecular Probes). Parasite
DNA was visualized using DAPI and cells were counted for DAPI and hDHFR
positivity. FISH analysis was performed as presented elsewhere
7
using telomere-
associated repeat element 4 (TARE4) and pGEM probes for the detection of
telomere clusters and the plasmid backbone, respectively. Images were counted
by three independent scorers in a blinded design in two to three independent
FISH experiments.
Fluorescence-activated cell sorting. Serum samples were tested for IgG bound
to the surface of mature trophozoite-infected erythrocytes using flow cytome-
try
29
. Cells were sequentially incubated with test serum or plasma diluted 1/10,
rabbit anti-human IgG (Fc-specific, Dako; 1:100), Alexa-Fluor 488-conjugated
anti-rabbit Ig (Molecular Probes; 1:750) and ethidium bromide (10
m
gml
21
).
Samples were analysed using a FACSCalibur flow cytometer (Becton-Dickinson)
and Flowjo software (TreeStar). For each sample the mean fluorescence of
uninfected erythrocytes was deducted from the mean fluorescence of infected
erythrocytes. All samples were tested in duplicate. Serum samples were obtained
from malaria-exposed adults from Madang, Papua New Guinea (ten men, five
non-pregnant women, five pregnant women) following informed consent. Five
samples from non-exposed Australian residents were included as controls.
Ethical clearance was obtained from the Medical Research Advisory Committee,
Department of Health, Papua New Guinea.
Received 1 September; accepted 4 November 2005.
Published online 28 December 2005.
1. Biggs, B. A. et al. Antigenic variation in Plasmodium falciparum.Proc. Natl Acad.
Sci. USA 88, 9171–-9174 (1991).
2. Roberts, D. J. et al. Rapid switching to multiple antigenic and adhesive
phenotypes in malaria. Nature 357, 689–-692 (1992).
3. Su, X. Z. et al. The large diverse gene family var encodes proteins involved in
cytoadherence and antigenic variation of Plasmodium falciparum-infected
erythrocytes. Cell 82, 89–-100 (1995).
4. Smith, J. D. et al. Switches in expression of Plasmodium falciparum var genes
correlate with changes in antigenic and cytoadherent phenotypes of infected
erythrocytes. Cell 82, 101–-110 (1995).
5. Baruch, D. I. et al. Cloning the P. falciparum gene encoding PfEMP1, a malarial
variant antigen and adherence receptor on the surface of parasitized human
erythrocytes. Cell 82, 77–-87 (1995).
6. Gardner, M. J. et al. Genome sequence of the human malaria parasite
Plasmodium falciparum.Nature 419, 498–-511 (2002).
7. Duraisingh, M. T. et al. Heterochromatin silencing and locus repositioning
linked to regulation of virulence genes in Plasmodium falciparum.Cell 121, 13–-24
(2005).
8. Freitas-Junior, L. H. et al. Telomeric heterochromatin propagation and histone
acetylation control mutually exclusive expression of antigenic variation genes
in malaria parasites. Cell 121, 25–-36 (2005).
9. Chen, Q. et al. Developmental selection of var gene expression in Plasmodium
falciparum.Nature 394, 392–-395 (1998).
10. Scherf, A. et al. Antigenic variation in malaria: in situ switching, relaxed and
mutally exclusive transcription of var genes during intra-erythrocytic
development in Plasmodium falciparum.EMBO J. 17, 5418–-5426 (1998).
NATURE|Vol 439|23 February 2006 LETTERS
1007
© 2006 Nature Publishing Group
11. Jensen, A. T. et al. Plasmodium falciparum associated with severe childhood
malaria preferentially expresses PfEMP1 encoded by group A var genes. J. Exp.
Med. 199, 1179–-1190 (2004).
12. Deitsch, K. W., del Pinal, A. & Wellems, T. E. Intra-cluster recombination and
var transcription switches in the antigenic variation of Plasmodium falciparum.
Mol. Biochem. Parasitol. 101, 107–-116 (1999).
13. Voss, T. S. et al. Genomic distribution and functional characterisation of two
distinct and conserved Plasmodium falciparum var gene 5 0flanking sequences.
Mol. Biochem. Parasitol. 107, 103–-115 (2000).
14. Voss, T. S., Kaestli, M., Vogel, D., Bopp, S. & Beck, H. P. Identification of
nuclear proteins that interact differentially with Plasmodium falciparum var gene
promoters. Mol. Microbiol. 48, 1593–-1607 (2003).
15. Deitsch, K. W., Calderwood, M. S. & Wellems, T. E. Malaria. Cooperative
silencing elements in var genes. Nature 412, 875–-876 (2001).
16. Gannoun-Zaki, L., Jost, A., Mu, J., Deitsch, K. W. & Wellems, T. E. A silenced
Plasmodium falciparum var promoter can be activated in vivo through
spontaneous deletion of a silencing element in the intron. Eukaryot. Cell 4,
490–-492 (2005).
17. Calderwood, M. S., Gannoun-Zaki, L., Wellems, T. E. & Deitsch, K. W.
Plasmodium falciparum var genes are regulated by two regions with separate
promoters, one upstream of the coding region and a second within the intron.
J. Biol. Chem. 278, 34125–-34132 (2003).
18. Horrocks, P., Pinches, R., Christodoulou, Z., Kyes, S. A. & Newbold, C. I.
Variable var transition rates underlie antigenic variation in malaria. Proc. Natl
Acad. Sci. USA 101, 11129–-11134 (2004).
19. O’Donnell, R. A. et al. A genetic screen for improved plasmid segregation
reveals a role for Rep20 in the interaction of Plasmodium falciparum
chromosomes. EMBO J. 21, 1231–-1239 (2002).
20. Ralph, S. A., Scheidig-Benatar, C. & Scherf, A. Antigenic variation in Plasmodium
falciparum is associated with movement of var loci between subnuclear
locations. Proc. Natl Acad. Sci. USA 102, 5414–-5419 (2005).
21. Navarro, M. & Gull, K. A pol I transcriptional body associated with VSG mono-
allelic expression in Trypanosoma brucei.Nature 414, 759–-763 (2001).
22. Shykind, B. M. Regulation of odorant receptors: one allele at a time. Hum. Mol.
Genet. 14 (suppl. 1), R33–-R39 (2005).
23. Trager, W. & Jensen, J. B. Cultivation of malarial parasites. Nature 273,
621–-622 (1978).
24. Beeson, J. G. et al. Plasmodium falciparum isolates from infected pregnant
women and children are associated with distinct adhesive and antigenic
properties. J. Infect. Dis. 180, 464–-472 (1999).
25. de Koning-Ward, T. F., Waters, A. P. & Crabb, B. S. Puromycin-N-
acetyltransferase as a selectable marker for use in Plasmodium falciparum.Mol.
Biochem. Parasitol. 117, 155–-160 (2001).
26. Mamoun, C. B., Gluzman, I. Y., Goyard, S., Beverley, S. M. & Goldberg, D. E. A
set of independent selectable markers for transfection of the human malaria
parasite Plasmodium falciparum.Proc. Natl Acad. Sci. USA 96, 8716–-8720
(1999).
27. Rubio, J. P., Thompson, J. K. & Cowman, A. F. The var genes of Plasmodium
falciparum are located in the subtelomeric region of most chromosomes. EMBO
J. 15, 4069–-4077 (1996).
28. Tonkin, C. J. et al. Localization of organellar proteins in Plasmodium falciparum
using a novel set of transfection vectors and a new immunofluorescence
fixation method. Mol. Biochem. Parasitol. 137, 13–-21 (2004).
29. Beeson, J. G. et al. Antibodies to variant surface antigens of Plasmodium
falciparum-infected erythrocytes and adhesion inhibitory antibodies are
associated with placental malaria and have overlapping and distinct targets.
J. Infect. Dis. 189, 540–-551 (2004).
Supplementary Information is linked to the online version of the paper at
www.nature.com/nature. A summary figure is also included.
Acknowledgements We are grateful to A. Gemmill and J. Baum for statistical
advice. This work was supported by the National Health and Medical Research
Council and the Wellcome Trust. A.F.C. and B.S.C. are International Fellows of
the Howard Hughes Medical Institute. T.S.V. is supported by fellowships from
the Swiss National Science Foundation and the Roche Research Foundation.
A.J.M. is supported by an Australian Postgraduate Award. J.G.B. was supported
by the NHMRC and the Miller Fellowship of WEHI. We also thank G. Kelly and
A. Raiko for sample collection and processing in Papua New Guinea.
Author Information Reprints and permissions information is available at
npg.nature.com/reprintsandpermissions. The authors declare no competing
financial interests. Correspondence and requests for materials should be
addressed to A.F.C. (cowman@wehi.edu.au).
LETTERS NATURE|Vol 439|23 February 2006
1008
... The Pf EMP1 family is encoded by about 45-90 var genes per parasite genome [12]. Expression of the var genes is mutually exclusive in ring-stage parasites, such that only a single Pf EMP1 variant is present on the surface of trophozoite-or schizont-stage Pf IEs at any given time [13,14] for review [10]. Mutually exclusive expression relies on very complex mechanisms. ...
... The var genes and their encoding Pf EMP1s vary greatly from parasite to parasite, and recombination constantly generates new variants, so there is an enormous repertoire of var genes in nature [13][14][15][16]. The molecular masses of Pf EMP1s range from 150 to 400 kDa. ...
Article
Full-text available
Plasmodium falciparum-infected erythrocytes (PfIEs) present P. falciparum erythrocyte membrane protein 1 proteins (PfEMP1s) on the cell surface, via which they cytoadhere to various endothelial cell receptors (ECRs) on the walls of human blood vessels. This prevents the parasite from passing through the spleen, which would lead to its elimination. Each P. falciparum isolate has about 60 different PfEMP1s acting as ligands, and at least 24 ECRs have been identified as interaction partners. Interestingly, in every parasite genome sequenced to date, at least 75% of the encoded PfEMP1s have a binding domain for the scavenger receptor CD36 widely distributed on host endothelial cells and many other cell types. Here, we discuss why the interaction between PfIEs and CD36 is optimal to maintain a finely regulated equilibrium that allows the parasite to multiply and spread while causing minimal harm to the host in most infections.
... P. falciparum parasites of strains F12, NF54, and K1 were transfected using standard procedures. 55 Two different plasmids with identical functions, p626-P230p (for F12) or pHF-gC-P230p (for NF54 and K1) (containing the Cas9 and the guideRNA expression cassettes), 44 were used in combination with the rescue plasmid P230p-lacZ (based on P230p-sfGFP) 44 for editing of the P230p locus. The LacZ sequence from E. coli was codonoptimized for P. falciparum (Genewiz). ...
Article
Full-text available
Malaria is caused by parasites of the Plasmodium genus and remains one of the most pressing human health problems. The spread of parasites resistant to or partially resistant to single or multiple drugs, including frontline antimalarial artemisinin and its derivatives, poses a serious threat to current and future malaria control efforts. In vitro drug assays are important for identifying new antimalarial compounds and monitoring drug resistance. Due to its robustness and ease of use, the [³H]-hypoxanthine incorporation assay is still considered a gold standard and is widely applied, despite limited sensitivity and the dependence on radioactive material. Here, we present a first-of-its-kind chemiluminescence-based antimalarial drug screening assay. The effect of compounds on P. falciparum is monitored by using a dioxetane-based substrate (AquaSpark β-D-galactoside) that emits high-intensity luminescence upon removal of a protective group (β-D-galactoside) by a transgenic β-galactosidase reporter enzyme. This biosensor enables highly sensitive, robust, and cost-effective detection of asexual, intraerythrocytic P. falciparum parasites without the need for parasite enrichment, washing, or purification steps. We are convinced that the ultralow detection limit of less than 100 parasites of the presented biosensor system will become instrumental in malaria research, including but not limited to drug screening.
... Finally, the rh3 5 and 3' homology target was ampli ed using primers FLP05 and FLP06 respectively and cloned into the plasmid using restriction enzymes PacI, NotI, and AvrII. The nal construct was sequenced and linearized with StuI, PvuI and NotI and then transfected into NF54 P. falciparum parasites following standard procedures 71 . Parasites that had correctly integrated the repair template were selected for after 10 days of incubation with 4 nM WR99210 (WR). ...
Preprint
Full-text available
Plasmodium falciparum assembles a protein translocon (PTEX) at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported. The preceding Plasmodium liver stage develops in hepatocytes within a PVM; however, the importance of PTEX and identification of exported proteins in P. falciparum liver stages remains unexplored. Here, we apply the FlpL/ FRT system to P. falciparum NF54 to conditionally excise genes in sporozoites, enabling studies at the liver stage. Conditional disruption of PTEX components PTEX150 and EXP2 in sporozoites does not affect their development or infectivity but attenuates liver stage growth. While PTEX150-deficiency significantly reduces liver load in humanized mice, EXP2-deficiency conferred a severe fitness cost, demonstrating that PTEX is essential for P. falciparum liver stage development. We show that liver specific protein 2 (LISP2) and circumsporozoite protein (CSP) contain putative PEXEL sequences cleaved by plasmepsin V, yet they localize to the PVM of infected hepatocytes. The abundance of LISP2 is reduced in PTEX-deficient liver stages, suggesting this protein is degraded in the absence of a functioning PTEX complex. This study employs the FlpL/ FRT system for functional analysis of P. falciparum pre-erythrocytic biology, revealing that the protein export translocon required for growth in erythrocytes is essential for P. falciparum development in hepatocytes and normal LISP2 expression. It also describes two P. falciparum proteins that contain putative PEXEL motifs that are targeted to the PVM.
... Even though var gene loci are spread across 13 out 14 chromosomes, DNA FISH showed that subtelomeric and central var genes form four to seven clusters at the transcriptionally repressive nuclear periphery (Lopez- Rubio et al., 2009;Ralph et al., 2005), with the single active var gene spatially separated from those repressive clusters (Mok et al., 2008;Ralph et al., 2005) (Fig. 1.16). DNA and RNA FISH demonstrated that, after switching, the newly active var gene relocates to an as-yet undefined transcriptionally competent site at the nuclear periphery (Freitas et al., 2005;Lopez-Rubio et al., 2009;Ralph et al., 2005;Voss et al., 2006). ...
Thesis
The most virulent human malaria parasite, Plasmodium falciparum, has a complex life cycle between its human host and mosquito vector. Each stage is driven by a specific transcriptional program, but with a relatively high ratio of genes to specific transcription factors, it is unclear how genes are activated or silenced at specific times. The P. falciparum genome is relatively euchromatic compared to the mammalian genome, except for specific genes that are uniquely heterochromatinized via HP1. There seems to be an association between gene activity and spatial organization; however, the molecular mechanisms behind genome organization are unclear. While P. falciparum lacks key genome-organizing proteins found in metazoans, it does have all core components of the cohesin complex. In other eukaryotes, cohesin is involved in sister chromatid cohesion, transcription, and genome organization. To investigate the role of cohesin in P. falciparum, we combined genome editing, mass spectrometry, chromatin immunoprecipitation and sequencing (ChIP-seq), and RNA sequencing to functionally characterize the cohesin subunit Structural Maintenance of Chromosomes protein 3 (SMC3). SMC3 knockdown in early stages of the intraerythrocytic developmental cycle (IDC) resulted in significant upregulation of a subset of genes involved in erythrocyte egress and invasion, which are normally expressed at later stages. ChIP-seq of SMC3 revealed that over the IDC, enrichment at the promoter regions of these genes inversely correlates with their expression and chromatin accessibility levels. These data suggest that SMC3 binding helps to repress specific genes until their appropriate time of expression, revealing a new mode of stage-specific, HP1-independent gene repression in P. falciparum.
... This approach failed probably because of the high homology among var genes at the 3' ATS segment. Therefore, we used an alternative approach by taking advantage of the fact that members of the PfEMP1 undergo allelic exclusive expression 40 . We tagged membrane-associated histidine rich protein 1 (MAHRP1, PF3D7_1370300), which is required for PfEMP1 surface expression but not required for export of STEVOR and other parasite proteins to the erythrocyte surface 41,42 , with the TetR-DOZI system to obtain NF54CR AS mahrp1 conditional knockdown parasites ( Supplementary Fig. 4c). ...
Article
Full-text available
Plasmodium falciparum has developed extensive mechanisms to evade host immune clearance. Currently, most of our understanding is based on in vitro studies of individual parasite variant surface antigens and how this relates to the processes in vivo is not well-understood. Here, we have used a humanized mouse model to identify parasite factors important for in vivo growth. We show that upregulation of the specific PfEMP1, VAR2CSA, provides the parasite with protection from macrophage phagocytosis and clearance in the humanized mice. Furthermore, parasites adapted to thrive in the humanized mice show reduced NK cell-mediated killing through interaction with the immune inhibitory receptor, LILRB1. Taken together, these findings reveal new insights into the molecular and cellular mechanisms that the parasite utilizes to coordinate immune escape in vivo. Identification and targeting of these specific parasite variant surface antigens crucial for immune evasion provides a unique approach for therapy. During the erythrocyte (RBC) stage of P. falciparum infection variant surface antigens (VSAs) such as PfEMP1s and RIFINs expressed on RBCs are important for infection and evasion of host innate immune system. Here, Chew et al. use a NSG mouse model, which is deficient in B, T and NK cells but retains macrophages, to show that PfEMP1 surface expression is required for in vivo adaptation as well as in vitro evasion of macrophage phagocytosis.
Preprint
Introduction Malaria remains a significant global health problem, particularly due to the human malaria parasite Plasmodium falciparum , which is responsible for most fatal infections. Infected red blood cells (iRBCs) evade spleen clearance by adhering to endothelial cells (ECs), triggering capillary blockage, inflammatory cytokine release, endothelial dysfunction, and altered vascular permeability, prompting an endothelial transcriptional response. Methods The iRBCIT4var04/HBEC-5i model, where iRBCs present IT4var04 (VAR2CSA) on their surface was employed to analyse the effects of iRBC binding on ECs. We used this model to investigate how cytoadhesion of iRBCs to ECs influences their expression profile depending on the temperature (37°C vs 40°C). Results Binding of non-infected RBCs (niRBCs) and fever alone significantly changes expression of hundreds of genes in ECs. Comparing the expression profile of HBEC-5i cultured either in the presence of iRBCs or in the presence of niRBCs, genes encoding proteins assigned to the GO terms immune response, nucleosome assembly, NF-kappa B signaling, angiogenesis, and antiviral immune response/interferon-alpha/beta signaling pathway were significantly up-regulated. If the cultivation temperature is increased from 37°C to 40°C, which simulates fever, a further significant increase in expression can be observed for most regulated genes, especially for genes coding for cytokines and proteins involved in angiogenesis. Conclusion The presence of iRBCs leads to the stimulation of ECs, activating several immunological signaling pathways and affecting antiviral (-parasitic) mechanisms and angiogenesis. Furthermore, to our knowledge, the induction of the interferon-alpha/beta signaling pathway in ECs in response to iRBCs has been described for the first time.
Article
Antigenic variation as a strategy to evade the host adaptive immune response has evolved in divergent pathogens. Antigenic variation involves restricted, and often mutually exclusive, expression of dominant antigens and a periodic switch in antigen expression during infection. In eukaryotes, nuclear compartmentalization, including three-dimensional folding of the genome and physical separation of proteins in compartments or condensates, regulates mutually exclusive gene expression and chromosomal translocations. In this Review, we discuss the impact of nuclear organization on antigenic variation in the protozoan pathogens Trypanosoma brucei and Plasmodium falciparum. In particular, we highlight the relevance of nuclear organization in both mutually exclusive antigen expression and genome stability, which underlie antigenic variation.
Chapter
Malaria infection triggers immune responses, critical for limiting Plasmodium parasite infections and severity of disease manifestation. Albeit repetitive exposures to the parasite lead to the development of nonsterile natural immunity to asexual forms of Plasmodium, only partial protection of the host against disease manifestation results. Thus, naturally acquired immunity against malaria occurs inefficiently, and protection is relatively short-lived. Moreover, the intensity of the immune response to Plasmodium parasite is stage-specific. However, the stages comprise of a wide array of effector mechanisms that can be exploited to achieve efficient immunity against malaria infection. Understanding the humoral and cellular adaptive immune responses against liver stages of malaria infection is crucial in the development of vaccines that can generate desired vaccine-induced responses. Optimal vaccine-induced responses will aid in the achievement of sterilizing immunity. Likewise, it is imperative to fully elucidate immune effector mechanisms involved in the development of immunity against erythrocytic forms of Plasmodium. Mimicking this natural immunity to blood stages has been the strategy with some malaria vaccines being investigated for effectiveness in the reduction of malaria-related morbidity and mortality. This chapter focuses on protective immune responses to malaria infection and also highlights the areas of immense relevance to the development of malaria vaccines. Here, we review recent advances and explore emerging hypotheses regarding the molecular and cellular pathways that regulate Plasmodium parasite-specific humoral immunity.KeywordsMalaria Plasmodium Erythrocytic stagePreerythrocytic stageCerebral malariaHumoralMalaria vaccine
Chapter
One of the key mechanisms contributing to the virulence of Plasmodium falciparum is its ability to undergo antigenic switching among antigenically distinct variants of the PfEMP1 adhesive proteins, encoded by the var gene family. To avoid premature exposure of its antigenic repertoire, the parasite transcribes its var genes in a mutually exclusive manner, and switch expression at a very slow rate. This process is epigenetically regulated and it relies on “epigenetic memory,” which imprints the single active var gene to remain active for multiple replication cycles. Erasing this epigenetic memory in parasites grown in culture resembles parasites, which egress from the liver. It could therefore be of interest for investigating var switching patterns at the onset of malaria infections. In addition, this procedure could be used for creating heterogeneity of var expression among parasite populations. The methodology described here for resetting of var gene expression is based on promoter titration, also known as molecular sponging.
Article
Full-text available
Adhesion of parasitized erythrocytes to post-capillary venular endothelium or uninfected red cells is strongly implicated in the pathogenesis of severe Plasmodium falciparum malaria. Neoantigens at the infected red-cell surface adhere to a variety of host receptors, demonstrate serological diversity in field isolates and may also be a target of the host-protective immune response. Here we use sequential cloning of P. falciparum by micromanipulation to investigate the ability of a parasite to switch antigenic and cytoadherence phenotypes. Our data show that antigens at the parasitized cell surface undergo clonal variation in vitro in the absence of immune pressure at the rate of 2% per generation with concomitant modulations of the adhesive phenotype. A clone has the potential to switch at high frequency to a variety of antigenic and adhesive phenotypes, including a new type of cytoadherence behaviour, 'auto-agglutination' of infected erythrocytes. This rapid appearance of antigenic and functional heterogeneity has important implications for pathogenesis and acquired immunity.
Article
Full-text available
Antigenic variation of infectious organisms is a major factor in evasion of the host immune response. However, there has been no definitive demonstration of this phenomenon in the malaria parasite Plasmodium falciparum. In this study, cloned parasites were examined serologically and biochemically for the expression of erythrocyte surface antigens. A cloned line of P. falciparum gave rise to progeny that expressed antigenically distinct forms of an erythrocyte surface antigen but were otherwise identical. This demonstrates that antigenic differences on the surface of P. falciparum-infected erythrocytes can arise by antigenic variation of clonal parasite populations. The antigenic differences were shown to result from antigenic variation of the parasite-encoded protein, the P. falciparum erythrocyte membrane protein 1.
Article
Full-text available
Plasmodium falciparum expresses on the host erythrocyte surface clonally variant antigens and ligands that mediate adherence to endothelial receptors. Both are central to pathogenesis, since they allow chronicity of infection and lead to concentration of infected erythrocytes in cerebral vessels. Here we show that expression of variant antigenic determinants is correlated with expression of individual members of a large, multigene family named var. Each var gene contains copies of a motif that has been previously shown to bind diverse host receptors; expression of a specific var gene correlated with binding to ICAM-1. Thus, our findings are consistent with the involvement of var genes in antigenic variation and binding to endothelium.
Article
The limited number of selectable markers available for malaria transfection has hindered extensive manipulation of the Plasmodium falciparum genome and subsequently thorough genetic analysis of this organism. In this paper, we demonstrate that P. falciparum is highly sensitive to the drug puromycin, but that transgenic expression of the puromycin-N-acetyltransferase (PAC) gene from Streptomyces alboninger confers resistance to this drug with the IC50 and IC90 values increasing ≈3- and 7-fold, respectively in PAC-expressing parasites. Despite this relatively low level of resistance, parasite populations transfected with the PAC selectable marker and selected directly on puromycin emerged at the same rate post-transfection as human dihydrofolate reductase (hDHFR)-expressing parasites, selected independently with the anti-folate drug WR99210. Transfected parasites generally maintained the PAC expression plasmid episomally at between two and six copies per parasite. We also demonstrate by cycling transfected parasites in the presence and absence of puromycin for several weeks, that the PAC selectable marker can be used for gene-targeting. Since the mode of action of puromycin is distinct from other drugs currently used for the stable transfection of P. falciparum, the PAC selectable marker should also have applicability for use in conjunction with other positive selectable markers, thereby increasing the possibilities for more complex functional studies of this organism.
Article
Approximately 50 highly diverse var genes distributed throughout the haploid genome of the malaria parasite Plasmodiumfalciparum code for PfEMP1 variants located on the surface of infected erythrocytes. PfEMP1 is involved in cytoadherence of parasitised red blood cells and undergoes antigenic variation through differential expression of var genes. Members of the var gene family are located in chromosome-internal positions on chromosomes 4, 7, 8 and 12, and in subtelomeric regions of all chromosomes. Here we show that there are two distinct and conserved types of 5′ upstream regions (var17-type and 5B1-type) of var genes, and suggest that most subtelomeric var genes are flanked by a var17-type 5′ upstream sequence. In contrast, 5B1-type 5′ upstream are localised to chromosomes that have been shown to contain var genes within chromosome-internal regions. Transcriptional analysis using RT-PCR revealed that var genes flanked by either type of 5′ upstream sequence are transcribed in in vitro cultured trophozoite stage parasites. In addition, we have shown that the 5′ flanking sequences of four different var genes are able to drive transient expression of the cat reporter gene. Our results suggest that at least the minimal regulatory sequences required for transcription of var genes are conserved among both subgroups of the var gene family. Furthermore, these sequences provide new markers for the investigation of the chromosomal organisation of var genes.
Article
The method for continuous cultivation of Plasmodium falciparum has now been successfully applied to several strains from different geographical areas. It has been used for tests of antimalarial drugs, for studies of parasite-host cell interactions with special reference to sickle haemoglobin, and for the production of amounts of parasite sufficient for experimental immunisation of Aotus trivirgatus monkeys.
Article
Plasmodium falciparum-infected human erythrocytes evade host immunity by expression of a cell-surface variant antigen and receptors for adherence to endothelial cells. These properties have been ascribed to P. falciparum erythrocyte membrane protein 1 (PfEMP1), an antigenically diverse malarial protein of 200-350 kDa on the surface of parasitized erythrocytes (PEs). We describe the cloning of two related PfEMP1 genes from the Malayan Camp (MC) parasite strain. Antibodies generated against recombinant protein fragments of the genes were specific for MC strain PfEMP1 protein. These antibodies reacted only with the surface of MC strain PEs and blocked adherence of these cells to CD36 but without effect on adherence to thrombospondin. Multiple forms of the PfEMP1 gene are apparent in MC parasites. The molecular basis for antigenic variation in malaria and adherence of infected erythrocytes to host cells can now be pursued.
Article
The human malaria parasite Plasmodium falciparum evades host immunity by varying the antigenic and adhesive character of infected erythrocytes. We describe a large and extremely diverse family of P. falciparum genes (var) that encode 200-350 kDa proteins having the expected properties of antigenically variant adhesion molecules. Predicted amino acid sequences of var genes show a variable extracellular segment with domains having receptor-binding features, a transmembrane sequence, and a terminal segment that is a probable submembrane anchor. There are 50-150 var genes on multiple parasite chromosomes, and some are in clustered arrangements. var probes detect two classes of transcripts in steady-state RNA: 7-9 kb var transcripts, and an unusual family of 1.8-2.4 kb transcripts that may be involved in expression or rearrangements of var genes.