ArticlePDF Available

Correctly folded Pfs48/45 protein of Plasmodium falciparum elicits malaria transmission-blocking immunity in mice

Authors:

Abstract and Figures

Malaria kills >1 million people each year, in particular in sub-Saharan Africa. Although asexual forms are directly responsible for disease and death, sexual stages account for the transmission of Plasmodium parasites from human to the mosquito vector and therefore the spread of the parasite in the population. Development of a malaria vaccine is urgently needed to reduce morbidity and mortality. Vaccines against sexual stages of Plasmodium falciparum are meant to decrease the force of transmission and consequently reduce malaria burden. Pfs48/45 is specifically expressed in sexual stages and is a well established transmission-blocking (TB) vaccine candidate. However, production of correctly folded recombinant Pfs48/45 protein with display of its TB epitopes has been a major challenge. Here, we show the production of a properly folded Pfs48/45 C-terminal fragment by simultaneous coexpression with four periplasmic folding catalysts in Escherichia coli. This C-terminal fragment fused to maltose binding protein was produced at medium scale with >90% purity and a stability over at least a 9-month period. It induces uniform and high antibody titers in mice and elicits functional TB antibodies in standard membrane feeding assays in 90% of the immunized mice. Our data provide a clear perspective on the clinical development of a Pfs48/45-based TB malaria vaccine.
Content may be subject to copyright.
Correctly folded Pfs48/45 protein of
Plasmodium
falciparum
elicits malaria transmission-blocking
immunity in mice
Nikolay S. Outchkourov*
, Will Roeffen
, Anita Kaan*, Josephine Jansen*, Adrian Luty
, Danielle Schuiffel
,
Geert Jan van Gemert
, Marga van de Vegte-Bolmer
, Robert W. Sauerwein
, and Hendrik G. Stunnenberg*
§
Departments of *Molecular Biology and Medical Microbiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Center,
P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
Communicated by Louis H. Miller, National Institutes of Health, Rockville, MD, January 16, 2008 (received for review December 21, 2007)
Malaria kills >1 million people each year, in particular in sub-
Saharan Africa. Although asexual forms are directly responsible for
disease and death, sexual stages account for the transmission of
Plasmodium parasites from human to the mosquito vector and
therefore the spread of the parasite in the population. Develop-
ment of a malaria vaccine is urgently needed to reduce morbidity
and mortality. Vaccines against sexual stages of Plasmodium fal-
ciparum are meant to decrease the force of transmission and
consequently reduce malaria burden. Pfs48/45 is specifically ex-
pressed in sexual stages and is a well established transmission-
blocking (TB) vaccine candidate. However, production of correctly
folded recombinant Pfs48/45 protein with display of its TB epitopes
has been a major challenge. Here, we show the production of a
properly folded Pfs48/45 C-terminal fragment by simultaneous
coexpression with four periplasmic folding catalysts in Escherichia
coli. This C-terminal fragment fused to maltose binding protein
was produced at medium scale with >90% purity and a stability
over at least a 9-month period. It induces uniform and high
antibody titers in mice and elicits functional TB antibodies in
standard membrane feeding assays in 90% of the immunized mice.
Our data provide a clear perspective on the clinical development of
a Pfs48/45-based TB malaria vaccine.
Malaria parasites are spread in the population by Plasmo-
dium-infected Anopheles mosquitoes. Successful transmis-
sion of malarial parasites from humans to mosquitoes depends
on the presence and infectiousness of gametocytes in the pe-
ripheral blood and the number of Anopheles mosquitoes in the
area. Transmission of Plasmodium falciparum can be blocked
inside the mosquito by antibodies that have been ingested
together with the gametocytes as part of a blood meal, inter-
rupting the sporogonic cycle inside the mosquito (1).
Pfs48/45 is a transmission-blocking (TB) target protein ex-
pressed by gametocytes (2–4) and present on the surface of the
sporogonic (macrogametes) stages of the malaria parasites.
Pfs48/45 plays a key role in parasite fertilization (5) and anti-
bodies that exclusively target conformational epitopes of
Pfs48/45 protein prevent fertilization (6, 7). Furthermore, anti-
Pfs48/45 antibodies are present in human sera from endemic
areas (8) and correlate with TB activity (8 –10). The induction of
antibodies after natural infection as observed in the field creates
the highly beneficial potential of vaccine boosting in the endemic
setting. TB vaccines might be applied alone or more likely as part
of a combination vaccine or package of control measures
depending on the intensity of malaria transmission (11).
A strategy for vaccine development requires the production of
correctly folded recombinant Pfs48/45 protein. Proper folding of
many cysteine-rich proteins, including Pfs48/45, depends on
correct formation of disulphide bridges. In eukar yotes the
oxidizing environment of the endoplasmic reticulum (ER) pro-
vides a milieu for disulphide bonds formation. Plasmodium
parasites are one of the few eukaryotes that lack the N-linked
glycosylation machinery, and many Plasmodium proteins contain
multiple potential glycosylation sites that are aberrantly glyco-
sylated when expressed in any of the available eukaryotic hosts.
On the other hand, prokaryotic expression systems such as
Escherichia coli, which lacks N-glycosylation also lack the so-
phisticated ER machinery of disulphide bond formation. In E.
coli correct disulphide bonds are formed in the periplasmic space
and catalyzed by a set of periplasmic oxidoreductases, termed
Dsb (12, 13). These proteins function in two separate pathways:
(i) oxidation by DsbA/DsbB, responsible for introducing S-S
bonds, and (ii) reduction and isomerization of aberrant disulfide
bonds by DsbC/DsbD. Previous studies (12) have shown that
overproduction of the enzymes DsbA and DsbC greatly improve
proper disulfide bond formation in cysteine-rich proteins.
Another well known rate-limiting step of the folding of
proteins in vivo is the cis/trans isomerization of prolyl-
iminopeptide bonds that is catalyzed by peptidyl-prolyl cis/
transisomerases (PPIases). The actions of PPIases such as FkpA
and SurA have already been shown to improve the production of
recombinant proteins in the periplasm of E. coli. Previously, the
genes for the oxidoreductase, PPIase, and the general chaperone
activities of DsbA, DsbC, FkpA, and SurA have been combined
on a expression plasmid called pTUM4 (14). Coexpression of the
periplasmic folding catalysts was shown to improve the folding of
two recombinant proteins carrying several disulfide bonds and
showing poor folding efficiency in the periplasm of E. coli.
In this study, we investigated the effect of coexpression of the
four periplasmic folding catalysts on the folding, yield, and
immunogenicity of the recombinant Pfs48/45 protein and frag-
ments thereof. Our results demonstrate that the yield of recom-
binant Pfs48/45 protein is significantly improved as compared
with the recombinant 10C as described (15). A Pfs48/45 fragment
of 10C cysteines retained the highest stability in terms of
conformation and resistance to proteases and elicited high titers
of functional TB antibodies. Our data provide an efficient
production and rapid purification of properly folded Pfs48/45–
10C and a clear perspective on the clinical development of a
Pfs48/45-based TB malaria vaccine.
Results and Discussion
Effect of Folding Catalysts on the Yield of Correctly Folded Pfs48/45.
We investigated the effect of coexpression of the protein folding
catalysts DsbA, DsbC, FkpA, and SurA on the yield and con-
Author contributions: N.S.O. and W.R. contributed equally to this work; N.S.O., W.R.,
R.W.S., and H.G.S. designed research; N.S.O., W.R., A.K., J.J., D.S., G.J.v.G., M.v.d.V.-B., and
R.W.S. performed research; N.S.O., W.R., A.L., R.W.S., and H.G.S. analyzed data; and N.S.O.,
W.R., R.W.S., and H.G.S. wrote the paper.
The authors declare no conflict of interest.
Freely available online through the PNAS open access option.
Present address: Department of Physiological Chemistry, University Medical Center, 3584
CG, Utrecht, The Netherlands.
§To whom correspondence should be addressed. E-mail: h.stunnenberg@ncmls.ru.nl.
© 2008 by The National Academy of Sciences of the USA
www.pnas.orgcgidoi10.1073pnas.0800459105 PNAS
March 18, 2008
vol. 105
no. 11
4301–4305
MEDICAL SCIENCES
formation of recombinant Pfs48/45 protein and fragments
thereof in the periplasm of E. coli. Previous unpublished results
had indicated that Pfs48/45 fused to the PelB leader peptide
could not be detected in E. coli periplasm and there was little
effect of the coexpression of chaperones. To attain periplasmic
localization Pfs48/45 full-length (16C) or C-terminal (10C)
(residues 26– 428 and 159 428, respectively) were fused to a
periplasmic maltose binding protein (MBP) as a carrier mole-
cule. As shown in Fig. 1B, both proteins M-Pfs16C and M-Pfs10C
were detected in the periplasmic extracts of E. coli at low levels.
Thus, MBP was an efficient vehicle in targeting Pfs48/45 to the
periplasm. Furthermore, pTUM4 encoded periplasmic chaper-
ones accumulated at high levels in the periplasmic fraction and
had a profound effect of at least 10-fold enhancement on protein
accumulation and epitope recognition of both M-Pfs16C and
M-Pfs10C. Note that in addition to the full-length M-Pfs16C and
M-Pfs10C we observed a degradation product with apparent
mobility of 43–45 kDa (43 kDa in the case of M-Pfs16C and
45 kDa for the M-Pfs10C) that reacted to the MBP antibody
(data not shown) in the Coomassie-stained SDS/PAGE. Thus
the Pfs48/45 part of the MBP fusion degraded rapidly in the E.
coli periplasm, and the protease-resistant MBP part accumulated
as a prominent product. Coexpression of the chaperones in-
creased significantly the amount of full-length M-Pfs16C and
M-Pfs10C with an concomitant increase of reactivity with the
conformation-dependent mAb (Fig. 1B). Apparently, proper
folding of the Pfs48/45 protein is essential for its stability and
accumulation.
Purification and Primary Characterization of M-Pfs16C and M-Pfs10C.
Both M-Pfs16C and M-Pfs10C were extracted from the E. coli
periplasm and purified on a DEAE FF column. Although the
M-Pfs10C remained soluble after purification, the M-Pfs16C
showed a strong tendency to aggregate upon storage for 1 week
at 4°C especially at protein concentrations of 0.25 mg/ml or
more (Fig. 2A). Further purification of the M-Pfs16C and
M-Pfs10C on Superdex 75 yielded 95% pure protein prepa-
rations as judged from a nonreduced SDS/PAGE (Figs. 2 Aand
3A). In a two-site capture ELISA, purified M-Pfs10C showed
similar epitope recognition as native gametocyte-derived
Pfs48/45 protein (Fig. 3 Band C), whereas the recognition of the
M-Pfs16C was much weaker (Fig. 2 Band C). Different com-
binations of Pfs48/45-specific mAbs for capture and detection
gave similar results (data not shown). Thus, we obtained a 90%
properly folded homogeneous M-Pfs10C preparation and a less
well folded M-Pfs16C preparation. Because of poor solubility
and much weaker epitope recognition of the M-Pfs16C prepa-
rations, we focused for further analysis mainly at the M-Pfs10C
protein preparation. The final yield of this correctly folded
M-Pfs10C protein was 1 mg/liter. Purified M-Pfs10C protein
was stable at 4°C for at least 9 months, i.e., repetitive confor-
mation-dependent two-site capture ELISA and mobility on a
nonreduced SDS/PAGE yielded nearly identical results (data
not shown). A second batch of M-Pfs10C was produced with
similar yields and ELISA values.
Immunogenicity of Recombinant M-Pfs10C. Immunogenicity of two
independently produced batches of the purified M-Pfs10C pro-
tein with 90% proper folding was assessed in BALB/c mice.
A
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
Domain III
(ep. I)
Domain II
(ep. IIb and III)
Domain I
(ep. V)
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
10C
16C
mAb IIb
mAb III
mAb V
mAb I
Coomassie
M-Pfs16C, pTUM4
M-Pfs16C
M-Pfs10C,pTUM4
M-Pfs10C
16C
10C
B
82
64
49
37
26
19
**
KDa
Fig. 1. Effect of chaperones expression from the pTUM4 plasmid on the
MBP-fused Pfs48/45 full-length (M-Pfs16C) and 10C fragment (M-Pfs10C). (A)
A schematic representation of the Pfs48/45 protein with three putative do-
mains recognized by different TB mAb as described (15). The coding sequence
contains a total of 448 aa. Bars indicate the relative position of cysteine
residues. (B) Periplasmic E. coli extracts were screened with mAbs against
epitope I, IIb, III, and V or Coomassie blue. Arrows indicate the position of
M-Pfs16C (16C) and M-Pfs10C (10C) proteins. Samples were mixed with SDS
sample buffer without reducing agent and separated on SDS/PAGE. Note that
the blot with the mAb against epitope I was overexposed to show the
expression in the lanes without chaperones.
A C
B
0
0.5
1
1.5
2
2.5
3
0.1 1 10 100 1000
ng Pfs 48/45 prote in or M-Pfs 16C / we ll
OD 450 nm
M-Pf s 1 6C
gct
0
0.5
1
1.5
2
2.5
3
0.1 1 10 100 1000
ng Pfs48/45 protein or M -Pfs16c / well
OD 450 nm
M-Pf s 1 6C
gct
1 2 3 4
KDa
82
64
49
37
26
19
Fig. 2. Purification and characterization of M-Pfs16C. (A) Coomassie-stained SDS/PAGE analysis after DEAE column purification of the M-Pfs16C protein. Lane
1, aggregated insoluble M-Pfs16C after 1 week at 4°C; lane 2, broad-range protein marker; lane 3, remaining soluble M-Pfs16C; lane 4, soluble M-Pfs16C purified
over Superdex 75 column. (Band C) Two-site ELISA experiment using rat mAbs against epitopes III for capture and peroxidase-conjugated mAb against epitope
I(B) or IIb (C) for detection. Gametocyte extracts (gct) with known concentration of the Pfs48/45 protein were used as a positive control.
4302
www.pnas.orgcgidoi10.1073pnas.0800459105 Outchkourov et al.
Fig. 4 shows that serum of M-Pfs10C (batch 1)-immunized mice
showed antibody reactivity in an ELISA using whole gametocyte
extract with increasing titers after each subsequent boost. Re-
activity against the M-Pfs10C was already apparent after the first
immunization. In the final bleed (S5), the M-Pfs10C fragment
induced titers of 1/165,000 (SD 1/101,143; range 1/30,000 to
1/300,000) when tested in the Gametocyte-ELISA using en-
riched gametocyte extracts and up to 1/1,000,000 in the
M-Pfs10C ELISA. Control sera from the two protein batches
were negative in the Gametocyte-ELISA. Sera tested in an
ELISA using MBP gave similar results as compared with the
M-Pfs10C ELISA. Sera from the immunization experiment with
batch 2 of M-Pfs10C yielded similar results (data not shown).
The ability of the antiserum to recognize native sexual stage
protein was further assessed by immunofluorescence assays
(IFAs) of air-dried sexual stage parasites and live intact mac-
rogametes/zygotes in suspension IFA (SIFA). All sera reacted
specifically with the antigen present in air-dried gametocytes
(Fig. 5A) and on the surface of live intact gametes and zygotes
of P. falciparum (Fig. 5C) but not against Pfs48/45 knockout
parasites (Fig. 5B) or asexual stage parasites.
To test for recognition of TB epitopes, M-Pfs10C immune sera
(batches 1 and 2) were used in a two-site competition ELISA
with a fixed amount of peroxide-conjugated rat anti-epitope I
and III mAbs for binding to native Pfs48/45 (Fig. 6 and data not
shown).Sera from batch 1 competed effectively for epitope I and
III of Pfs48/45 at serum dilutions from 1/20 to 1/160. Ten of 12
sera of M-Pfs10C-immunized mice competed 50% with con-
jugated anti-epitope I and III mAbs, whereas no competition was
found with adjuvant-immunized control sera or preimmune sera.
Competition with nonconjugated anti-epitope I or III Pfs48/45
mAb was used as positive control and for quantification (data not
A
C
B
0
0.5
1
1.5
2
2.5
3
0.1 1 10 100 1000
ng Pfs48/45 protein or M-Pfs10C/well
OD 450 nm
M-Pf s 1 0 C
gct
0
0.5
1
1.5
2
2.5
3
0.1 1 10 100 1000
ng Pfs48/45 protein or M -Pfs10C/well
OD 450 nm
M-Pf s 1 0 C
gct
KDa
82
64
49
37
26
19
Fig. 3. Immunocharacterization of M-Pfs10C. (A) Coomassie-stained SDS/PAGE analysis of the purified M-Pfs10C protein. (Band C) Two-site ELISA experiment
using rat mAbs against epitopes III for capture and peroxidase-conjugated mAb against epitope I (B) or IIb (C) for detection. Gametocyte extracts with known
concentration of the Pfs48/45 protein were used as a positive control.
Parasite M-Pfs10C
Fig. 4. Immunogenicity of sera from mice immunized with M-Pfs10C. Anti-
body reactivity of sera from six mice immunized with recombinant M-Pfs10C
from the first immunization experiment in the gametocyte-ELISA (Left) and
M-Pfs10C ELISA (Right). S0, S1, S3, and S5 indicate preimmune sera, day 14, day
56, and day 98 of the time course of the immunization, respectively; see
Materials and Methods. Boxes extend from the 25th and 75th percentiles with
the median value of the six mice sera.
B
B
A
C
Fig. 5. IFA and SIFA analysis of sera from mice immunized with M-Pfs10C. (A
and B) Immunofluorescence microscopy with anti-M-Pfs10C mouse sera on P.
falciparum gametocytes air-dried on a multispot slide (IFA) with wild-type
parasites (A) and Pfs48/45 knockout parasites (B). (C) SIFA using live intact
macrogametes/zygotes. (Magnification: 400.)
Outchkourov et al. PNAS
March 18, 2008
vol. 105
no. 11
4303
MEDICAL SCIENCES
shown). Estimates based on unlabeled mAb added to the assay
revealed concentrations of anti-epitope I and anti-epitope III
antibodies in the sera of the M-Pfs10C-immunized mice between
20 and 100
g/ml, which is in the range in which TB activity can
be expected (15). Briefly, a fixed amount of labeled mAb
competed for Pfs48/45 binding with either test serum or a known
concentration range of the same but unlabeled mAb. An esti-
mate of anti-epitope-specific antibodies in the test serum was
made based on comparison of OD values between test serum and
labeled mAb.
TB Activity Induced by M-Pfs10C. Individual sera of mice immu-
nized with M-Pfs10C (batches 1 and 2) after bleed 5 (S5) were
analyzed for TB activity in the standard membrane-feeding assay
(SMFA). Table 1 shows a near-complete blockade of transmis-
sion in 11/12 sera at a standard dilution of 1:10 (Mann–Whitney
test P0.0001). Sera of control mice immunized with PBS and
MBP alone showed no reducing activity in the SMFA. Finally, a
clear correlation was observed between TB activity of the sera
from M-Pfs10C-immunized mice including results obtained with
a recombinant 10C fragment without the MBP fusion (15) and
the percentage of competition in the epitope I competition
ELISA (r
2
0.873) (Fig. 7). The curve shows that 90%
inhibition in the SMFA correlated with 40% competition for
epitope I.
Concluding Remarks. Our data show that an N-terminally trun-
cated Pfs48/45 protein fused to MBP, coined M-Pfs10C, and
coexpressed with four periplasmic folding catalysts in the
periplasm of E. coli yielded a properly folded homogeneous
Pfs48/45 protein that is stable over for at least 9 months. The two
independent batches of M-Pfs10C protein elicited functional TB
antibodies (transmission reducing activity 90%) in 90% of
the mice.
Clinical development of malaria vaccines has accelerated over
recent years with a clear increase in the portfolio of candidates.
Although most efforts are concentrated on preeythrocytic and
blood-stage vaccines, progress in TB vaccine development has
lagged behind and was basically limited to two postfertilization
proteins, P25 and P28 (16).
From a biological perspective and supported by experimental
data, Pfs48/45 has been for a long time considered as an
attractive prefertilization target for inhibition of sporogonic
development. However, technological constraints related to
protein folding have hampered its development for many years.
The high TB activity of the C-terminal fragment of Pfs48/45
described here provides a solid basis for the clinical development
of a prefertilization TB malaria vaccine.
Materials and Methods
Construct Preparation. A synthetic Pfs48/45 gene optimized for expression in
E. coli (GenBank accession no. EU366251) encoding wild-type Pfs48/45 protein
(17) was designed and used throughout this study. Fragments corresponding
to processed Pfs48/45 protein residues 26428 (16C) and N-terminally trun-
0.1
20.1
40.1
60.1
80.1
5 2.5 1.25 0.61
% v/v dilution of the mice sera
Percentage Competition
Fig. 6. Two-site competition ELISAs. mAb against epitope IIb (85RF45.2b)
was used to capture Pfs48/45 molecule from gametocyte extract. Percentage
competition of sera from mice immunized with the M-Pfs10C protein (batch
1) was calculated by using a fixed amount of peroxidase-conjugated mAb
anti-epitope I (Ep I) or anti-epitope III (Ep III) at serum dilutions [ranging from
1/20 to 1/160 (5–0.62% vol/vol)]. Data are presented as mean percentage
competition 1 SD of the six mice.
Table 1. TB activity of sera from mice immunized with M-Pfs10C
Sera AM, mean no. TN, nTBA, %
Immunization batch 1
1A1 0 0 100
1A2 0.05 1 99.6
1A3 0.30 6 97.4
1A4 0 0 100
1A5 0.75 15 94.5
1A6 0 0 100
Control 13.6 263
Immunization batch 2
2A1 30.7 614 27.1
2A2 0 0 100
2A3 5.1 91 88.0
2A4 0.05 1 99.9
2A5 0.15 3 99.6
2A6 0 0 100
Control 42 843
Sera (1:10 diluted) were tested in the SMFA. Seven days after membrane
feed, the number of infected mosquitoes and oocyst density were deter-
mined. Sera, mouse serum from M-Pfs10C-immunized group; Control, mean
of five feeders with serum from control immunization; AM, arrhythmic mean
oocyst number in 20 dissected mosquitoes; TN, total number of oocysts in 20
dissected mosquitoes; TBA, percentage of TB activity (19).
-20.0
0.0
20.0
40.0
60.0
80.0
100.0
120.0
0.0 20.0 40.0 60.0 80.0 100.0
Percentage competition against epitope I
Percentage inhibition of oocysts
-20.0
0.0
20.0
40.0
60.0
80.0
100.0
120.0
0.0 20.0 40.0 60.0 80.0 100.0
Fig. 7. TB activity correlates with the concentration of the anti-epitope I
antibodies. Percentage inhibition of oocysts are related to the percentage
competition of the anti-epitope I antibodies in the sera from mice after five
immunizations with the rec10C (
) (15) and M-Pfs10C (}) protein as calculated
by using a fixed amount of peroxidase-conjugated mAb anti-epitope I
(85RF45.1) at a dilution of 1/20 of the serum. The line represents the regression
of results by use of a polygon equation (y⫽⫺1E 06x50.0003x40.0322x3
1.2884x217.369x43.997; Rvalue of 0.93).
4304
www.pnas.orgcgidoi10.1073pnas.0800459105 Outchkourov et al.
cated (10C) residues 159428 were cloned into pMAL-p2x (New England
Biolabs), resulting in N-terminal MBP fusions. The pTUM4 plasmid has been
described (14).
Expression and Purification of M-Pfs10C. Expression and preparations of
periplasmic fractions were performed essentially as described (14). The re-
combinant proteins were purified on a DEAE FF (Amersham Biosciences)
column equilibrated in 20 mM TrisHCl, pH 8.6. The column was washed with
50 mM NaCl, and bound proteins were eluted with a linear gradient of 50 –400
mM NaCl in the same buffer. Fractions containing the M-Pfs10C protein as
analyzed by Coomassie staining and ELISA were concentrated on a Vivaspin
20, 30-kDa molecular mass cut-off utrafiltration unit (Vivascience). M-Pfs10C
protein was further purified over a Superdex 75 HR 10/30 column (Amersham
Biosciences) in PBS that contained 0.01% N-dodecyl-N-N-dimethyl-3-
ammonio-1-propane sulfonate.
Parasites. Mature P. falciparum gametocytes (NF54 strain) were produced in
an automated static culture system as described (7, 18), isolated (19), and
stored at 70°C until used. NF54 gametocytes were extracted in 25 mM
TrisHCl (pH 8.0) supplemented with 150 mM NaCl, 1.0% sodium desoxy-
cholate, and 1 mM phenylmethylsulphonyl fluoride. Insoluble debris was
pelleted by centrifugation (13,000 gfor 5 min at room temperature); the
supernatant provided antigen for Western blot analysis and ELISAs.
Antibodies. To detect epitopes of Pfs48/45, various mAbs of mouse origin, 32F5
(epitope I) and 32F1 (epitope IIb) (6, 20), or rat origin, 85RF45.1 (epitope I),
85RF45.2b (epitope IIb), 85RF45.3 (epitope III), and 85RF45.5 (epitope V) (7,
19), were used. HRP-conjugated anti-mouse IgG was purchased from Dako
(P0161). Alexa Fluor488 anti-mouse IgG (A21200) was purchased from Molec-
ular Probes.
Immunization of BALB/c Mice. Groups (n6) of female BALB/c mice were
immunized s.c. with 50
g of recombinant M-Pfs10C fragment per mouse or
with adjuvant alone (control, n5) as described (15). Briefly, mice were
immunized with a total volume of 0.1 ml per mouse emulsified in complete
Freund’s adjuvant on day 1 and boosted with the same amount of antigen on
21, 42, 63, and 84 days with incomplete Freund’s adjuvant. Blood was taken on
days 0 (preimmune serum), 14, 35, 56, 77, and 98 and tested for specific
antibody reactivity and TB activity in the SMFA.
IFA. An indirect IFA was done with cultured sexual-stage parasites (NF54
isolate of P. falciparum) air-dried on multispot slides as described (7, 8). Briefly,
parasites were incubated with a 1:100 dilution of the test sera in PBS, rinsed
with PBS, and incubated with Alexa-conjugated goat-anti-mouse Ig (IgG). The
cells were examined for clear sexual-stage parasite-specific green fluores-
cence. For SIFA analysis, gametocytes were allowed to undergo gametogen-
esis as described (7, 8, 19), and the cells were examined as above under IFA.
ELISAs. All incubation steps, except antigen coating, were done at room
temperature.
Detection of Pfs48/45. Pfs48/45-specific ELISA was performed by a two-site
ELISA as described (7, 8, 15). Briefly, 96-well microtiter plates were coated with
rat mAbs recognizing particular epitopes of Pfs48/45 (19). After blocking with
5% milk in PBS, the antigen (starting with 250,000 parasites per well or 20
g/ml M-Pfs10C) was captured by incubation in a serial dilution with PBS, and
the bound antigen was detected by HRP-conjugated rat mAbs.
Gametocyte and M-Pfs10C ELISA. Serum antibody analysis was conducted by
ELISA with whole gametocyte extracts or M-Pfs10C ELISA as described (15).
Briefly, ELISA plates were coated with enriched gametocyte antigen extract
(250,000 parasites per well) or 2
g/ml M-Pfs10C diluted in PBS, stored at 4°C
overnight, and then blocked with 5% skimmed milk/PBS. Diluted sera were
added and incubated for 2 h, and bound antibodies were detected by HRP-
conjugated goat anti-Mouse IgG.
Competition ELISA. The competition ELISA was performed by a two-site ELISA
as described (7, 8, 15, 19). Briefly, gametocyte extract was captured essentially
as for the Pfs48/45-specific ELISA by using mAb 85RF45.2b. Competition of
HRP-labeled mAb 85RF45.1 or 85RF.45.3 with sera antibodies from M-Pfs10C-
immunized mice was performed by incubation of 30
l of HRP-labeled mAb
(2.5
g/ml, diluted with PBS containing 1% milk) and 30
l of mice serum
(dilutions ranging from 1/20 to 1/160) or 30
l of unlabeled mAb (5-fold serially
diluted started at 5
g/ml) for 2 h.
TB Assay. Antisera obtained from mice immunized with the M-Pfs10C frag-
ment were tested for their TB activities in a SMFA as described (15, 21, 22).
Briefly, 27
l of the mice sera was mixed with 63
lofnaı¨ve human serum and
180
lofin vitro gametocyte culture of P. falciparum (NF54 line). This mixture
was fed to Anopheles stephensi (Nijmegen strain) mosquitoes through a
membrane feeding apparatus, and 7 days later at least 20 mosquitoes (90%
survival) were dissected and oocysts were counted from extracted midguts of
test and control mosquitoes.
ACKNOWLEDGMENTS. We thank Dr. Arne Skerra (Technische Universitat,
Munich) for pTUM4 plasmid, Karina Teelen for technical assistance, and
Rumyana Karlova for stimulating discussions. This work was supported by
European Malaria Vaccine Initiative Grant 041222 and BioMalPar.
1. Bousema JT, Drakeley CJ, Sauerwein RW (2006) Sexual-stage antibody responses to P.
falciparum in endemic populations. Curr Mol Med 6:223–229.
2. Carter R, Mendis KN, Miller LH, Molineaux L, Saul A (2000) Malaria transmission-
blocking vaccines: How can their development be supported? Nat Med 6:241–244.
3. Stowers A, Carter R (2001) Current developments in malaria transmission-blocking
vaccines. Exp Opin Biol Ther 1:619– 628.
4. Kaslow DC (2002) Transmission-blocking vaccines. Chem Immunol 80:287–307.
5. van Dijk MR, et al. (2001) A central role for P48/45 in malaria parasite male gamete
fertility. Cell 104:153–164.
6. Vermeulen AN, et al. (1985) Plasmodium falciparum transmission blocking monoclonal
antibodies recognize monovalently expressed epitopes. Dev Biol Stand 62:91–97.
7. Roeffen W, et al. (2001) Plasmodium falciparum: Production and characterization of
rat monoclonal antibodies specific for the sexual-stage Pfs48/45 antigen. Exp Parasitol
97:45–49.
8. Roeffen W, et al. (1995) A comparison of transmission-blocking activity with reactivity
inaPlasmodium falciparum 48/45-kDa molecule-specific competition enzyme-linked
immunosorbent assay. Am J Trop Med Hyg 52:60– 65.
9. Bousema JT, et al. (2006) Rapid onset of transmission-reducing antibodies in javanese
migrants exposed to malaria in Papua, Indonesia. Am J Trop Med Hyg 74:425–431.
10. Bousema JT, et al. (2007) A longitudinal study of immune responses to Plasmodium
falciparum sexual stage antigens in Tanzanian adults. Parasite Immunol 29:309–317.
11. Sauerwein RW (2007) Malaria transmission-blocking vaccines: The bonus of effective
malaria control. Microbes Infect 9:792–795.
12. Frand AR, Cuozzo JW, Kaiser CA (2000) Pathways for protein disulphide bond forma-
tion. Trends Cell Biol 10:203–210.
13. Baneyx F, Mujacic M (2004) Recombinant protein folding and misfolding in Escherichia
coli.Nat Biotechnol 22:1399–1408.
14. Schlapschy M, Grimm S, Skerra A (2006) A system for concomitant overexpression of
four periplasmic folding catalysts to improve secretory protein production in Esche-
richia coli.Protein Eng Des Sel 19:385–390.
15. Outchkourov N, et al. (2007) Epitope analysis of the malaria surface antigen pfs48/45
identifies a subdomain that elicits transmission-blocking antibodies. J Biol Chem
282:17148–17156.
16. Epstein JE, Giersing B, Mullen G, Moorthy V, Richie TL (2007) Malaria vaccines: Are we
getting closer? Curr Opin Mol Ther 9:12–24.
17. Kocken CH, et al. (1993) Cloning and expression of the gene coding for the transmission
blocking target antigen Pfs48/45 of Plasmodium falciparum.Mol Biochem Parasitol
61:59– 68.
18. Ponnudurai T, Lensen AH, Leeuwenberg AD, Meuwissen JH (1982) Cultivation of fertile
Plasmodium falciparum gametocytes in semiautomated systems. 1. Static cultures.
Trans R Soc Trop Med Hyg 76:812–818.
19. Roeffen WF, et al. (2001) Recombinant human antibodies specific for the Pfs48/
45 protein of the malaria parasite Plasmodium falciparum.J Biol Chem
276:19807–19811.
20. Vermeulen AN, et al. (1985) Sequential expression of antigens on sexual stages of
Plasmodium falciparum accessible to transmission-blocking antibodies in the mos-
quito. J Exp Med 162:1460–1476.
21. van der Kolk M, et al. (2005) Evaluation of the standard membrane feeding assay
(SMFA) for the determination of malaria transmission-reducing activity using empirical
data. Parasitology 130:13–22.
22. Lensen A, et al. (1996) Measurement by membrane feeding of reduction in Plasmo-
dium falciparum transmission induced by endemic sera. Trans R Soc Trop Med Hyg
90:20–22.
Outchkourov et al. PNAS
March 18, 2008
vol. 105
no. 11
4305
MEDICAL SCIENCES

Supplementary resource (1)

... Pfs48/45 is a relatively conserved glycophosphatidylinositol (GPI)-linked protein expressed on the surface of gametes and is assumed to be essential for male gametocyte fertility (van Dijk et al., 2001). Developing a Pfs48/45-based TBV has proven challenging, largely due to the difficulties of recombinantly expressing Pfs48/45 in sufficient quality and quantity (Outchkourov et al., 2008). The most advanced Pfs48/45 antigen is genetically fused to the R0 protein for enhanced expression, folding, and stabilization of the Pfs48/45-6C domain (Outchkourov et al., 2008;Acquah et al., 2017;Singh et al., 2021). ...
... Developing a Pfs48/45-based TBV has proven challenging, largely due to the difficulties of recombinantly expressing Pfs48/45 in sufficient quality and quantity (Outchkourov et al., 2008). The most advanced Pfs48/45 antigen is genetically fused to the R0 protein for enhanced expression, folding, and stabilization of the Pfs48/45-6C domain (Outchkourov et al., 2008;Acquah et al., 2017;Singh et al., 2021). Although this antigen recently entered a phase 1 clinical trial (NCT04862416), it remains unclear whether stabilization of Pfs48/45 through fusion to another protein can be sufficient to elicit the most potent transmission-blocking activity against this target. ...
... Despite being the target of the most potent anti-malarial transmission-blocking mAb yet reported, Pfs48/45 has largely remained a challenging target for vaccine development. Its low recombinant expression and intrinsic instability have been the focus of various rescue strategies, including exploration of alternative expression systems, fusion proteins, and multimerization methods (Outchkourov et al., 2008;Acquah et al., 2017;Singh et al., 2021), all with mixed success. Structure-guided protein engineering has emerged as a viable strategy to stabilize antigens, most notably the fusion proteins of enveloped viruses. ...
Article
Full-text available
Malaria transmission-blocking vaccines (TBVs) aim to elicit human antibodies that inhibit sporogonic development of Plasmodium falciparum in mosquitoes, thereby preventing onward transmission. Pfs48/45 is a leading clinical TBV candidate antigen and is recognized by the most potent transmission-blocking monoclonal antibody (mAb) yet described; still, clinical development of Pfs48/45 antigens has been hindered, largely by its poor biochemical characteristics. Here, we used structure-based computational approaches to design Pfs48/45 antigens stabilized in the conformation recognized by the most potently inhibitory mAb, achieving >25°C higher thermostability compared with the wild-type protein. Antibodies elicited in mice immunized with these engineered antigens displayed on liposome-based or protein nanoparticle-based vaccine platforms exhibited 1–2 orders of magnitude superior transmission-reducing activity, compared with immunogens bearing the wild-type antigen, driven by improved antibody quality. Our data provide the founding principles for using molecular stabilization solely from antibody structure-function information to drive improved immune responses against a parasitic vaccine target.
... It remains unclear, however, whether this effect is due to the loss of Pfs48/45 itself or due to an absence of Pfs230 from the parasite surface, which is anchored to the membrane by its interaction with Pfs48/45 . Recognition of Pfs48/45 by human sera correlates with the ability of sera to block parasite transmission (Graves et al., 1998;Mulder et al., 1999;van der Kolk et al., 2006;Bousema et al., 2010;Ouedraogo et al., 2011;Stone et al., 2018) and antibodies against Pfs48/45 have transmission-blocking activity (Targett, 1988;Targett et al., 1990;Roeffen et al., 2001;Outchkourov et al., 2008;Lennartz et al., 2018;Stone et al., 2018). Pfs48/45-based transmissionblocking vaccines are under development against both P. falciparum (Theisen et al., 2017;Mistarz et al., 2017;Singh et al., 2017a;Singh et al., 2017b;Mamedov et al., 2019;Lee et al., 2020;Singh et al., 2021a) and P. vivax (Arevalo-Herrera et al., 2015;Tachibana et al., 2015;Cao et al., 2016;Arevalo-Herrera et al., 2021;Arevalo-Herrera et al., 2022). ...
... MBP was used for the early expression of Pfs230 fragments (Williamson et al., 1993;Riley et al., 1995;Williamson et al., 1995;Milek et al., 1998;Bustamante et al., 2000) and GST for Pfs48/45 fragments (Kocken et al., 1993;Milek et al., 1998;Outchkourov et al., 2007). Co-expression of an MBP-tagged Pfs48/45 fragment (10C, aa 159-428) with periplasmic folding catalysts produced a properly folded protein with an increased yield relative to the GST fusion of the same fragment (Outchkourov et al., 2007;Outchkourov et al., 2008). The full ectodomain and single domain constructs of Pfs47 have been expressed as a fusion to E. coli protein thioredoxin. ...
... Anti-Pfs48/45 antibodies with transmission-blocking activity target at least four epitope groups (epitopes I, IIb, III, and V) that span all three 6-cysteine domains of Pfs48/45 (N. Targett, 1988;Targett et al., 1990;Roeffen et al., 2001b;Outchkourov et al., 2007;Outchkourov et al., 2008;Lennartz et al., 2018) (Supplementary Table 2). The disulfide bonds within the central and C-terminal 6-cysteine domains are critical for the presentation of the transmission-blocking epitopes, but dispensable for epitope presentation on the N-terminal domain of Pfs48/45 (Outchkourov et al., 2007). ...
Article
Full-text available
The 6-cysteine protein family is one of the most abundant surface antigens that are expressed throughout the Plasmodium falciparum life cycle. Many members of the 6-cysteine family have critical roles in parasite development across the life cycle in parasite transmission, evasion of the host immune response and host cell invasion. The common feature of the family is the 6-cysteine domain, also referred to as s48/45 domain, which is conserved across Aconoidasida. This review summarizes the current approaches for recombinant expression for 6-cysteine proteins, monoclonal antibodies against 6-cysteine proteins that block transmission and the growing collection of crystal structures that provide insights into the functional domains of this protein family.
... The C-terminal domain containing six cysteines and epitope I is still the target of the most potent transmissionblocking mAb: 85RF45.1. 16 A fully humanized version of mAb 85RF45.1(TB31F) has been manufactured for clinical development and is currently being tested in a clinical phase I study (https://clinicaltrials.gov/ct2/show/NCT04238689) for safety and tolerability. ...
... The exact and expected pairing of cysteines for Pfs48/45 was resolved by structural analysis of the 6-Cys domain, where Cys1-Cys2 (Cys298-Cys327, as noted here), Cys3-Cys6 (Cys344-Cys412, as noted here), and Cys4-Cys5 (Cys352-410, as noted here) form the cysteine pairs. 16 become R0.6C. Although various recombinant subdomains of the native Pfs48/45 have been shown to elicit functional antibodies in multiple animal species, R0.6C was selected as a prime candidate for downstream clinical development. ...
Article
Full-text available
In the early 1980s, Richard Carter was among the first researchers to identify the sexual stage-specific Pfs48/45 protein, leading to the identification of target epitopes. Carter predicted its tertiary conformation while involved in a number of studies on naturally acquired sexual stage-specific antibodies. Pfs48/45 is a cysteine-rich surface protein of sexual stages of Plasmodium falciparum that plays a critical role in male gamete fertility. Antibodies against Pfs48/45 prevent parasite development in the mosquito vector, and therefore prevent the spread of malaria in the population. Since the gene was sequenced in the early 1990s, Pfs48/45 has been considered a prime target candidate for a malaria transmission-blocking vaccine. However, major manufacturing challenges—in particular, difficulty realizing satisfactory yields of a properly folded protein for the induction of functional antibodies—delayed clinical development significantly. These challenges were met roughly 20 years later. The first clinical trial with a Pfs48/45 subunit vaccine (R0.6C) was started in the Netherlands in early 2021. The excellent contributions to the long and winding path of Pfs48/45 research by Richard Carter are well recognized and are an integrated part of his seminal contributions to unraveling Plasmodium sexual stage biology.
... In previous work, we reported the functional and genetic details of naturally acquired human mAbs targeting D1, D2, and D3 of Pfs48/45 derived from two donors 27 . Notably, the D1-directed mAbs were found to possess TRA that was comparable to those directed to D3, while D2-directed mAbs possessed markedly lower TRA compared to D3 or D1, as previously described 21,23,28 . Here, we extend the functional and biophysical characterization of several of these mAbs against D1 and D2. ...
Preprint
Full-text available
Biomedical interventions capable of preventing the transmission of malaria-causing Plasmodium falciparum (Pf) between the human host and mosquito vector could prove a valuable tool in malaria elimination efforts. Pfs48/45, a gamete-surface protein essential for Pf development in the mosquito midgut, is a key component of clinical-stage transmission-blocking vaccines. Antibodies against this antigen have been demonstrated to efficiently reduce Pf transmission from humans to mosquitoes. Potent human monoclonal antibodies (mAbs) against Domain 3 (D3) of Pfs48/45 have been structurally and functionally described; however, in-depth information about other inhibitory epitopes on Pfs48/45 is currently limited. Here, we present a 3.3 Å resolution cryo-electron microscopy structure of full-length Pfs48/45 in complex with potent mAbs targeting Domain 1 (D1) and D3, and a moderately potent mAb targeting Domain 2 (D2). Our data indicate that while Pfs48/45 D1 and D2 are rigidly coupled, there is substantial conformational flexibility between D2 and D3. Characterization of mAbs against D1 revealed the presence of a conformational epitope class that is largely conserved across Pf field isolates and is associated with recognition by highly potent antibodies. Our study provides comprehensive insights into epitopes across full-length Pfs48/45 and has implications for the design of next-generation malaria transmission-blocking vaccines and antibodies.
... Pfs48/45 is present on the surface of sexual stage parasites transferred from human to mosquito during a blood meal and in the mosquito prior to fertilization, with peak surface expression on gametes and zygotes. Pfs48/45 is required for efficient parasite fertilization and oocyst formation in the mosquito, and serum from animals immunized with Pfs48/45 blocks oocyst development [6][7][8][9][10] . Human infection elicits antibodies to Pfs48/45, suggesting vaccine-induced immunity could be boosted by natural infection 11 . ...
Article
Full-text available
A malaria vaccine that blocks parasite transmission from human to mosquito would be a powerful method of disrupting the parasite lifecycle and reducing the incidence of disease in humans. Pfs48/45 is a promising antigen in development as a transmission blocking vaccine (TBV) against the deadliest malaria parasite Plasmodium falciparum. The third domain of Pfs48/45 (D3) is an established TBV candidate, but production challenges have hampered development. For example, to date, a non-native N-glycan is required to stabilize the domain when produced in eukaryotic systems. Here, we implement a SPEEDesign computational design and in vitro screening pipeline that retains the potent transmission blocking epitope in Pfs48/45 while creating a stabilized non-glycosylated Pfs48/45 D3 antigen with improved characteristics for vaccine manufacture. This antigen can be genetically fused to a self-assembling single-component nanoparticle, resulting in a vaccine that elicits potent transmission-reducing activity in rodents at low doses. The enhanced Pfs48/45 antigen enables many new and powerful approaches to TBV development, and this antigen design method can be broadly applied towards the design of other vaccine antigens and therapeutics without interfering glycans.
... This is essential for gamete fusion, as male Plasmodium berghei gametes that lack the homologous protein, Pbs48/45, are unable to penetrate female gametes to form zygotes 6 . In addition, antibodies induced by immunising animals with Pfs48/45 block the sexual development of the parasite within infected mosquitos [7][8][9][10][11][12] . Pfs48/45 is expressed on gametocytes found in human blood and is therefore exposed to the human immune system. ...
Article
Full-text available
An effective malaria vaccine remains a global health priority and vaccine immunogens which prevent transmission of the parasite will have important roles in multi-component vaccines. One of the most promising candidates for inclusion in a transmission-blocking malaria vaccine is the gamete surface protein Pfs48/45, which is essential for development of the parasite in the mosquito midgut. Indeed, antibodies which bind Pfs48/45 can prevent transmission if ingested with the parasite as part of the mosquito bloodmeal. Here we present the structure of full-length Pfs48/45, showing its three domains to form a dynamic, planar, triangular arrangement. We reveal where transmission-blocking and non-blocking antibodies bind on Pfs48/45. Finally, we demonstrate that antibodies which bind across this molecule can be transmission-blocking. These studies will guide the development of future Pfs48/45-based vaccine immunogens. Pfs48/45, a surface protein of Plasmodium falciparum, is a promising anti-malarial vaccine candidate whose structure is not entirely resolved. Here, the authors present the structure of the full-length molecule, and characterise the binding and activity of transmission blocking antibodies.
... A recombinant construct representing Pfs48/45 of P. falciparum has induced antibodies that almost consistently reduce infectivity to mosquitoes by 99-100%; it is not clear whether complement was active or inactive in the tests. 33 A construct representing the Pvs48/45 of P. vivax has induced antibodies that totally suppressed infectivity of P. vivax to mosquitoes in membrane feeding tests with active complement present. Unfortunately, no controls were included for the effect of the antibodies when complement is inactivated. ...
... This is essential for gamete fusion, as male Plasmodium berghei gametes that lack the homologous protein, Pbs48/45, are unable to penetrate female gametes to form zygotes [6]. In addition, antibodies induced by immunising animals with Pfs48/45 block the sexual development of the parasite within infected mosquitos [7][8][9][10][11][12]. Pfs48/45 is expressed on gametocytes found in human blood and is therefore exposed to the human immune system. ...
Preprint
Full-text available
An effective malaria vaccine remains a global health priority and vaccine immunogens which prevent transmission of the parasite will have important roles in multi-component vaccines. One of the most promising candidates for inclusion in a transmission-blocking malaria vaccine is the gamete surface protein Pfs48/45, which is essential for development of the parasite in the mosquito midgut. Indeed, antibodies which bind Pfs48/45 can prevent transmission if ingested with the parasite as part of the mosquito bloodmeal. Here we present the first structure of full-length Pfs48/45, revealing its three domains to form a dynamic, planar, triangular arrangement. From this, we show where transmission-blocking and non-blocking antibodies bind on Pfs48/45. Finally, we demonstrate that antibodies which bind across this molecule can be transmission-blocking. These studies will guide the development of future Pfs48/45-based vaccine immunogens.
Article
Two malaria transmission-blocking vaccine (TBV) candidates, R0.6C and ProC6C, have completed preclinical development including the selection of adjuvants, Alhydrogel® with or without the saponin based adjuvant Matrix-M™. Here, we report on the final drug product (formulation) design of R0.6C and ProC6C and evaluate their safety and biochemical stability in preparation for preclinical and clinical pharmacy handling. The point-ofinjection stability studies demonstrated that both the R0.6C and ProC6C antigens are stable on Alhydrogel in the presence or absence of Matrix-M for up to 24 h at room temperature. As this is the first study to combine Alhydrogel and Matrix-M for clinical use, we also evaluated their potential interactions. Matrix-M adsorbs to Alhydrogel, while not displacing the > 95 % adsorbed protein. The R0.6C and ProC6C formulations were found to be safe and well tolerated in repeated dose toxicity studies in rabbits generating high levels of functional antibodies that blocked infection of mosquitoes. Further, the R0.6C and ProC6C drug products were found to be stable for minimally 24 months when stored at 2–8 ◦C, with studies ongoing through 36 months. Together, this data demonstrates the safety and suitability of the L. lactis expression system as well as supports the clinical testing of the R0.6C and ProC6C malaria vaccine candidates in First-In-Human clinical trials.
Article
Pfs25, a vaccine candidate, expressed on the surface of the malarial parasite, plays an important role in the development of Plasmodium falciparum. 1269, a monoclonal antibody targeting the epidermal growth factor like-1 and epidermal growth factor like-3 domains of Pfs25, blocks the transmission of parasites in mosquitoes. In this study, we refolded a dimeric antibody fragment referred as diabody designed from 1269 (1269-Db), with a yield of 3 mg/litre of bacterial culture. Structural integrity of the protein was validated with thermal stability, disulphide bond analysis and glutaraldehyde crosslinking experiments. To evaluate the functionality of 1269-Db, recombinant monomeric MBP-Pfs25 was produced from bacteria. Qualitative binding assays demonstrated that 1269-Db recognized the epitopes on Pfs25 in its native, but not the denatured state. An apparent KD of 2.6 nM was determined for 1269-Db with monomeric MBP-Pfs25 using isothermal titration calorimetry. 1269-Db recognized the periphery of zygotes/Ookinetes, demonstrating recognition of Pfs25, expressed on the surface of the parasite. As the established refolding method resulted in a functional diabody, the optimized method pipeline for 1269-Db can potentially facilitate engineering of antibody fragments with desired properties.
Article
Full-text available
Energetic particle instrumentation on the Polar satellite has discovered that significant fluxes of energetic particles are continuously present in the region of the dayside mag-netosphere where they cannot be stably trapped. This region is associated with either open magnetic field lines or a magnetic topology associated with pseudo-trapping. Two distinct features [Time-Energy Dispersion (TED) sig-natures and Cusp Energetic Particle (CEP) events] are observed in these energetic particle fluxes that strongly suggest a local acceleration of mostly shocked solar wind particles. These thermalized fluxes form diamagnetic cav-ities in the cusp with strong turbulence that apparently energize a fraction of their number to energies of 100s and 1000s of kiloelectronvolts. The decay of these cavities forms a layer of energetic particles on the magnetopause as well as permits such particles to enter the equatorial nightside magnetosphere to distances as close as six earth radii due to drift caused by gradient and curvature effects in the local magnetic field. The fluxes of these particles have all of the properties associated with the ring current and can probably supply the magnitude of the cross tail current required. Because the CEP event process is impul-sive and time variable the charge separation produced by the drifting electrons (eastward) and ions (westward) may be responsible for the cross tail electric field that has been ascribed to the convection process. Introduction The NASA GGS Polar satellite has discovered two features of the energetic particle fluxes within the dayside magnetosphere that appear to be associated with the high altitude cusp that may well lead to a new paradigm for the way we view the coupling of the magnetosphere with the solar wind and interplanetary medium. These two fea-tures are Time-Energy-Dispersion (TED) signatures that are observed throughout the dayside high altitude magne-tosphere and the Cusp Energetic Particle (CEP) events observed in a region of the cusp populated by significant fluxes of thermalized solar wind particles. These observa-tions coupled with subsequent tracing of particle trajecto-ries in a realistic geomagnetic model and a revisit to an older energetic particle data set from the NASA ISEE-1 satellite produce a very plausible picture that energetic particle fluxes are produced in a continuous but impulsive manner in diamagnetic cavities craved out of the cusp within the magnetosphere by the entry of shocked solar wind plasma. Once produced these particles move in response to their gradient and curvature drifts, electrons drifting east from the dusk side of the magnetosphere and ions drifting west in through and from the dawn side of the magnetosphere. These particles have access to the entire nightside magnetosphere and geomagnetic tail, where it is suggested, they are responsible for the ring current and possibly the cross-tail current. It is suggested that their flux variations in time and the resulting charge separation of the two particle species drifting in opposite directions may give rise to a cross-tail electric field that has been ascribed to the existence of a convection process and the imposition of some fraction of the imposed solar wind -VXB electric field.
Article
Full-text available
Pfs48/45, a member of a Plasmodium-specific protein family, displays conformation-dependent epitopes and is an important target for malaria transmission-blocking (TB) immunity. To design a recombinant Pfs48/45-based TB vaccine, we analyzed the conformational TB epitopes of Pfs48/45. The Pfs48/45 protein was found to consist of a C-terminal six-cysteine module recognized by anti-epitope I antibodies, a middle four-cysteine module recognized by anti-epitopes IIb and III, and an N-terminal module recognized by anti-epitope V antibodies. Refolding assays identified that a fragment of 10 cysteines (10C), comprising the middle four-cysteine and the C-terminal six-cysteine modules, possesses superior refolding capacity. The refolded and partially purified 10C conformer elicited antibodies in mice that targeted at least two of the TB epitopes (I and III). The induced antibodies could block the fertilization of Plasmodium falciparum gametes in vivo in a concentration-dependent manner. Our results provide important insight into the structural organization of the Pfs48/45 protein and experimental support for a Pfs48/45-based subunit vaccine.
Article
Full-text available
Plasmodium falciparum gametocytes contain specific antigens, some of which (Mr 230,000, 48,000, 45,000) are expressed on the surface of the newly emerged macrogamete. A different antigen (Mr 25,000) surrounds the surface of the ookinete and, although present to some extent in the developing gametocyte, is synthesized in high quantities by the macrogamete/zygote and expressed progressively on the transforming zygote surface. These antigens are targets of transmission blocking antibodies that are effective at two distinct points after gametogenesis: fertilization of the macrogamete and ookinete to oocyst development. The antigens involved in the fertilization blockade are the Mr 48 and 45 proteins, which are expressed on the macrogamete surface. The Mr 230 K coprecipitating protein probably plays no part in transmission block. mAb directed against the Mr 25 K ookinete surface protein blocked transmission without inhibiting ookinete formation, indicating that this protein has an important role in the transformation of ookinete into oocyst. A combination of mAb recognizing different epitopes on the same protein molecule acted synergistically in inhibiting oocyst formation. Using a mixture of two blocking mAb reacting against the Mr 48/45 and 25 K proteins, respectively, an additive blocking effect could be demonstrated.
Article
Plasmodium falciparum gametocytes contain specific antigens, some of which (Mr 230,000, 48,000, 45,000) are expressed on the surface of the newly emerged macrogamete. A different antigen (Mr 25,000) surrounds the surface of the ookinete and, although present to some extent in the developing gametocyte, is synthesized in high quantities by the macrogamete/zygote and expressed progressively on the transforming zygote surface. These antigens are targets of transmission blocking antibodies that are effective at two distinct points after gametogenesis: fertilization of the macrogamete and ookinete to oocyst development. The antigens involved in the fertilization blockade are the Mr 48 and 45 proteins, which are expressed on the macrogamete surface. The Mr 230 K coprecipitating protein probably plays no part in transmission block. mAb directed against the Mr 25 K ookinete surface protein blocked transmission without inhibiting ookinete formation, indicating that this protein has an important role in the transformation of ookinete into oocyst. A combination of mAb recognizing different epitopes on the same protein molecule acted synergistically in inhibiting oocyst formation. Using a mixture of two blocking mAb reacting against the Mr 48/45 and 25 K proteins, respectively, an additive blocking effect could be demonstrated.
Article
Host responses to the transmittable stages of the malaria parasite may reduce transmission effectively. Transmission-reducing activity (TRA) of human serum can be determined as a percentage, using the Standard Membrane Feeding Assay (SMFA). This laboratory assay was evaluated using the results of 121 experiments with malaria-endemic sera among which many repeated measurements were obtained. The assay consists of the feeding of Anopheles stephensi mosquitoes with cultured Plasmodium falciparum gametocytes, mixed with human red blood cells, and control and experimental sera. The TRA of individual sera was determined by the comparison of oocyst densities between these sera. Bootstrap data on oocyst densities in individual mosquitoes in control feeds were used to construct confidence limits for TRA percentages of serum feeds. Low (<20%) and high TRA (>90%) values for individual sera were usually reproduced in a second experiment, whereas this was more difficult for values between 20% and 90%. The observed variability of TRA values is explained in part by the variability in oocyst density per mosquito. Oocyst densities in control feeds varied more between experiments than within experiments and showed a slight decline over the 3 years of experiments. Reproducibility of TRA of field sera was low (20%) between experiments, but much higher (61%) within experiments. A minimum of 35 oocysts per mosquito in control feeds gave optimal reproducibility (44%) between experiments. We recommend that (1) sera are compared within an experiment, or (2) assays are only analysed where controls have at least 35 oocysts per mosquito. The SMFA is under the recommended conditions appropriate for the study of factors that may influence TRA, e.g. transmission blocking vaccines.
Article
Target proteins of transmission blocking monoclonal antibodies (MoAbs) are present on the surface of Plasmodium falciparum macrogametes with Mr of 48,000 and 45,000 and on the surface of developing ookinetes with Mr of 25,000. Other MoAbs directed against the same proteins were not able to reduce the number of oocysts in mosquitoes. A combination of a blocking MoAb with a non-blocking one potentiated the transmission blocking effect. This implies that at least two different epitopes are present on the target antigens. This was confirmed using a sandwich immunoradiometric assay. The results demonstrated that on the 48/45 kD proteins as well as on the 25 kD protein a "blocking" and a "non-blocking" epitope exist which are not repeated anywhere else in the molecule.
Article
A semi-automated cultivation apparatus for the in vitro culture of Plasmodium falciparum gametocytes is described. This apparatus has been designed to produce large numbers of fertile sexual stages for use in the development of a gamete vaccine or for the infection of suitable mosquitoes. These mosquitoes in turn may be used for the development of a possible sporozoite vaccine. Loss of red cells during medium change has been eliminated and the addition of warmed fresh medium simplified compared to similar systems described previously. Material harvested from this apparatus has been used for infecting mosquitoes. Up to 98% of Anopheles stephensi were infected with a mean oocyst count of 24 per positive gut (range one to 109). The importance of satisfactory presentation of gametocytes for mosquito infection is stressed. The possible presence of substances in normal human sera which inhibits exflagellation to a variable degree and reduces mosquito infectivity is also discussed.
Article
Monoclonal antibodies (MAbs) 32F1 and 32F3 react with two independent epitopes of a protein doublet with molecular weights of 48 and 45 kilodaltons (kD) expressed on the surface of Plasmodium falciparum (Pfs48/45) macrogametes and zygotes; only 32F3 blocks transmission. These MAbs were used to develop a Pfs48/45-specific competition enzyme-linked immunosorbent assay (ELISA) using 32F1 to capture antigen and labeled 32F3 for quantification and analysis of the contribution of antibodies in human serum to transmission-blocking activity. A comparison analysis was used to determine agreement of competition ELISA titers and transmission-blocking activity as observed in the bioassay in three groups of serum samples: 37 from European travelers with previous exposure to malaria, 56 from gametocyte carriers, and 66 from schoolchildren from a malaria-endemic area in Cameroon. The index of agreement between outcomes of the ELISA and transmission-blocking assay in gametocyte carriers and in travelers was specifically defined as fair-to-moderate; in schoolchildren the agreement was not significant. The combined analysis of all sera showed a significant and fair-to-moderate agreement between the results of the competition ELISA and the transmission-blocking assay, with a relative specificity of 94% (of 105 cases negative in the transmission-blocking assay, 99 were also negative in the competition ELISA) and a relative sensitivity of 44% (of 54 cases positive in the transmission-blocking assay, 24 were also positive in the competition ELISA). This study shows that a positive C48/45-ELISA is indicative for transmission-blocking activity in the mosquito assay, while a negative result does not exclude transmission-blocking activity.
Article
The gene encoding the gametocyte/gamete-specific membrane protein Pfs48/45 of Plasmodium falciparum has been cloned. The Pfs48/45 gene is a non-interrupted, single copy gene that codes for a hydrophobic, non-repetitive protein of 448 amino acid residues containing a putative signal peptide at the N-terminus, a hydrophobic C-terminus and 7 potential N-glycosylation sites. Antibodies directed against a Pfs48/45-glutathione-S-transferase fusion protein reacted with both the 45-kDa and 48-kDa proteins of gametocytes. When Pfs48/45 is expressed in the baculovirus-insect cell system the recombinant Pfs48/45 protein is targeted and exposed to the insect cell surface in such a configuration that it is recognized by transmission-blocking anti-45/48-kDa monoclonal antibodies.
Article
The standard laboratory test for reduction in malaria transmission is based on the measurement of oocyst numbers in mosquitoes fed on blood meals containing test and control sera. Interpretation of the results, however, is often hampered by the large variation in numbers of infected mosquitoes and oocysts. The objective of this study was to compare 3 measures for the assessment of transmission reduction (so-called R values) and to define the experimental criteria that allow interpretation of the results. To determine variability in R values of control sera, a replicate experiment was performed with 10 non-endemic sera of Dutch blood donors. Furthermore, 2 measures for calculation of transmission reduction were compared in a triplicate experiment using Plasmodium falciparum, Anopheles gambiae and malaria endemic sera. Calculations using the geometric mean of Williams are currently used to identify blocking and non-blocking sera. However, calculations using log-transformed data could distinguish more gradual levels of transmission reduction activity by endemic sera--i.e. blocking, reducing and non-blocking activity. Grading of transmission reduction activity is important for epidemiological studies on transmission immunity and for validation of future transmission-blocking vaccines.