ArticlePDF Available

High frequency of malaria-specific T cells in non-exposed humans

Authors:

Abstract and Figures

A major goal of current candidate malaria vaccines is to stimulate the expansion of clones of malaria-specific lymphocytes. We have examined the in vitro T cell responses of a group of malaria exposed and non-exposed adult Caucasian donors to recombinant circumsporozoite (CS) proteins, one of which is undergoing clinical trials, to blood-stage parasites, and to synthetic peptides copying the CS protein and defined blood-stage proteins. In nearly all individuals tested, CD4 T cell proliferation or lymphokine production occurred in response to whole parasite or CS protein stimulation, and T cells from many individuals responded to synthetic peptides. T cell responses were major histocompatibility complex-restricted, and stimulation of T cells with malaria parasites or CS protein did not appear to expand a population of T cell receptor gamma/delta cells. Malaria-specific responses were independent of prior malaria exposure, and in some cases exceeded the magnitude of response to tetanus toxoid. Specific T cells are present in high frequency in the peripheral blood of many donors who have never been exposed to malaria. Although malaria-specific CD4 T cells play an important role in immunity, these data question whether vaccines need to stimulate such cells, and focus attention on other aspects of malaria immunity which may be more critical to a successful vaccine.
Content may be subject to copyright.
Eur.
J.
Immunol.
1992.
22:
689-696
Malaria-specific responses in non-exposed humans
689
Yinka Zevering.,
Fiona Amante.,
Anne Smillie.,
Jeff Currier.,
Gale Smithv,
Richard A. HoughtenA and
Michael F. Good.
Tropical Health Program.,
Queensland Institute
of
Medical
Research, Brisbane. MicroGeneSys
Inc.v, Meriden, Torrey Pines
Institute
for
Molecular StudiesA.,
San Diego
High frequency of malaria-specific
T
cells in
non-exposed humans
A major goal of current candidate malaria vaccines is to stimulate the expansion
of clones of malaria-specific lymphocytes. We have examined the
in vitro
T
cell
responses of a group of malaria exposed and non-exposed adult Caucasian donors
to recombinant circumsporozoite (CS) proteins, one of which is undergoing
clinical trials, to blood-stage parasites, and to synthetic peptides copying the CS
protein and defined blood-stage proteins. In nearly all individuals tested, CD4
T
cell proliferation or lymphokine production occurred in response to whole
parasite or CS protein stimulation, and T cells from many individuals responded
to synthetic peptides.
T
cell responses were major histocompatibility complex-
restricted, and stimulation of T cells with malaria parasites or CS protein did not
appear to expand a population of Tcell receptor
$6
cells. Malaria-specific
responses were independent of prior malaria exposure, and in some cases
exceeded the magnitude of response to tetanus toxoid. SpecificT cells are present
in high frequency in the peripheral blood of many donors who have never been
exposed to malaria. Although malaria-specific CD4
T
cells play an important role
in immunity, these data question whether vaccines need to stimulate such cells,
and focus attention on other aspects of malaria immunity which may be more
critical to a successful vaccine.
1
Introduction
Tcells develop in the thymus from fetal liver- or bone
marrow-derived hematogenous progenitor cells
[
11.
Fol-
lowing positive and negative selection events in the thymus,
during which rearrangement of the TcR genes occurs, a
population of clonally distributed precursor cells is
exported from the thymic medulla to seed the peripheral
lymph organs. Classical activation of T cells occurs when
theTcR engages a processed peptide from a foreign antigen
(epitope) bound to an MHC molecule on the surface of an
antigen-presenting cell
[2].
While MHC antigen-specific
(allo-specific)
T
cells from unprimed animals can readily be
grown
in vitro
with allogeneic stimulation, onlyT cells from
specifically primed individuals respond
in vitro
to
nominal
antigens
[3].
T cells are central to malaria immunity in that they function
as helper cells for B cells or effector cells, in their own right.
Animals which lack antibody (p-suppressed mice) can, in
fact, be protectively immunized with blood-stage malaria
parasites
[4]
or sporozoites
[5].
CD4 T cells can transfer
immunity in mice to blood-stage parasites
[6],
as well as to
sporozoites
[7-81,
and in the case of sporozoite immunity,
the circumsporozoite (CS) protein is an important antigen.
[I
96471
*
This work was Suported by UNDPlWorld Bank/WHO Special
Programme for Research and Training in Tropical Diseases,
and
by
the National Health and Medical Research Council
(Australia).
Correspondence:
Michael
F.
Good,
Tropical Health Program,
Queensland Institute
of
Medical Research, Herston Road, Bris-
bane
4029, QLD,
Australia
Abbreviations:
CS:
Circumsporozoite
Thus, central to the development
of
a malaria vaccine
aimed at cellular immune defense, is the principle of
stimulating a specific population of precursor T lympho-
cytes that will expand clonally, then recognize and destroy
the parasite encountered following natural challenge.
Recent studies, however, have shown that malaria antigen-
specific and MHC-restricted
T
cell clones could be gene-
rated from humans with no history of malaria exposure
[9-111.
This suggests that malaria-specific memory
T
cells
are present in non-exposed individuals. The frequency of
such cells is not known, but a basic premise of many current
vaccine programs (clonal expansion of a population of
malaria-specific precursor T lymphocytes) is questioned
if
memory
T
cells from non-exposed individuals which can
respond to malaria antigens are present at high frequency.
Because of the potential importance of these reports, we
have investigated the human T cell immune response to
purified bacuolovirus-expressed
F1
fakiparum
recombinant
CS protein
[12, 131
currently in use in human trials, as well
as to a yeast recombinant CS protein and to blood-stage
malaria parasites. Also, a group of synthetic peptides
copying the CS protein sequence and known to be immu-
nodominant for this group
of
donors was tested
[14].
2
Materials and methods
2.1
Donors
Half of the donors were adult Caucasian residents of
Brisbane who have lived in malaria endemic regions,
mainly Papua New Guinea, for varying periods
of
time (see
Sect.
3.1,
Table
1)
and come from a previously described
group
of
subjects
[14].
These donors are described in the
Tables as
‘E’
(exposed). The other donors
(‘C,
control)
were adult Caucasian residents
of
Brisbane with no history
of
malaria exposure.
0
VCH Verlagsgesellschaft mbH,
D-6940
Weinheim,
1YY2
0014-2980/Y2/0303-0689$3.50
+
,2510
690
Y.
Zevering,
E
Amante,
A.
Smillie et al.
Eur.
J.
Immunol. 1992.
22:
689-696
2.2 ELISA
Antibodies binding the (NANP), repeat motif of the
recombinant construct, R32tet32, were detected by ELISA
as previously described
[15,
161. Titer is defined as the
greatest dilution of test serum giving an absorbance >3 SD
above the mean of a panel of normal sera at the same
dilution.
2.3 Antigens
The baculovirus-expressed recombinant CS protein, cur-
rently in clinical studies, has previously been described
[12, 131. Its characteristics and purity are described in
Fig.
1.
A yeast-expressed recombinant
CS
protein was also
used in some studies, and has previously been described
[17]. It contains residues 43-348, and was kindly provided
by Dr. Philip Barr, Chiron.The following synthetic peptides
were used: PfCS proteinl-20 (peptide
1,
MMRKLAILSVS-
SFLFVEALF);
PfCS
protein296-315 (peptide 16, QGHNM-
PDPNRNVDENANAN); PfCS protein331-350 (peptide 21,
IEQYLKKIKNSISTEWSPCS);
PfCS
protein386-405 (pep-
tide 28, MEKCSSVFNVVNSSIGLIMV); PfMSAl (YKL-
NFYFDLLRAKL); PvMSAl (HVINFHYDLL
RANV).
The peptides are described further in listed references
[14,
18, 191.
2.4 Lymphocyte proliferation assays
Human PBL were obtained by density centrifugation
of
peripheral blood over Ficoll Hypaque. PBL were added to
round-bottom microtiter plates at 2
x
lo5
cells per well
(quadruplicate) in Hepes-buffered Eagle's minimum essen-
tial medium containing
10
%
pooled normal human serum
and antigen at various concentrations. Six or seven days
later wells were pulsed with
0.5
pCi
=
18,5 kBq of
'[Hlthymidine and incorporation of label was determined
18 h later by liquid scintillation spectroscopy. Specific
0.20
-I
cs
t
Mobility
Figure
I.
Analysis of purity
of
rCS protein. A recombinant
baculovirus expression vector containing a full-lenght
Rfulciparurn
CS
gene was used to produce
CS
protein in insect cells
(Spodoptera
frugiperdu).
The
rCs
polypeptide (66000
kDa)
was purified. Ten
micrograms
of
purified
CS
antigen was electrophoresed on
an
11.5
%
polyacrylamide gel, stained with coomassie brilliant blue,
and scanned on
an
LKB
UltroScan
XL
laser densitometer. The
arrows indicate minor contaminants which are present in the
CS
preparation.
rCS
protein represents
299
%
of the purified protein.
Amino acid sequencing revealed cleavage
of
a
signal peptide
between residues
65
and
66
of
the deduced amino acid
sequence.
proliferation was calculated as a stimulation index, defined
as (cpm in presence of antigen)/(cpm in absence of
antigen), or simply by listing cpm. Assays were performed
in quadruplicate.
2.5
Generation of
T
cell clones
The method of Sinigaglia and Pink [20] was used. Briefly,
PBL (2
x
106/well, in 24-well plates) were stimulated
in
vim with
19
fakiparum
(FCR3 clone)-infected RBC (lo6/
ml) for
7
days. Blast cells were enriched
on
Ficoll-Paque
and cultured with IL 2
(50
unitslml) for
7
days and then
plated at limiting dilution (0.3,
1
cell/well) with irradiated
allogeneic PBL (4
x
lo5
cells/well, flat-bottom 96-well
plates), I1 2 and PHA at
5
pg/ml. Medium was changed
regularly and clones were obvious by 2-3 weeks. Clones
were expanded with allogeneic filler cells, but tested with
autologous APC.
2.6
Generation of short- and long-term
T
cell lines
For short-term lines, PBL (2
x
lo5
cells/well) were
stimulated in 96-well round-bottom microwells with CS
protein at
5
pg/ml for
1
week. Cells were then washed in the
wells (X3), split into replicates, and restimulated with
either CS protein
(5
pg/ml) or peptide (30 pg/ml) in the
presence
of
fresh irradiated autologous PBM as APC. After
a further week, wells were pulsed with 3[H]thymidine and
incorporation measured after
18
h. For long-term lines,
PBL were stimulated in 24-well plates (2
x
lo6
cells/well)
with antigen and after a week the cells were washed and
rested with fresh irradiated autologous filler cells and IL 2
(50
U/ml) for 2-3 weeks. Following this, blasts were
prepared over Ficoll-Paque. Cells were then tested by
adding
5
x
lo4
cells to
lo5
irradiated APC and antigen and
measuring proliferation after
4
days.
2.7
Limiting dilution assays
T cells were purified from the lymphocytes by 2-amino-
ethyl-isothiouronium bromide-treated sheep red blood cell
rosette sedimentation [lo, 21, 221. Purified T cells were
plated out in round-bottom microtiter plates at cell con-
centrations ranging from
lo2
to 2
x
lo5.
For each concen-
tration, 24 wells with antigen (parasite or peptide), and 12
or more wells without antigen were used. Irradiated
non-T
cells were added at
lo4
cells/well, as a source of APC.
Proliferation was determined a week later by liquid scintil-
lation counting of incorporated [3H]thymidine. Antigen-
specific responses were determined to be those whose cpm
exceeded the average cpm of the negative control wells by
23
SD. Precursor frequencies of peptide-specific T cells
were determined from the slope of the line of best fit (least
squares) plotting cell number
(X
axis)
and the log
of
the
fraction
of
non-responding wells (LnF,)
(Y
axis).
2.8 Cytokine assays
IFN-y (Commonwealth Serum Laboratories, Melbourne,
Australia), and I1 6 (Genzyme, Boston, MA) were assayed
by ELISA as described in the manufacturer's kits.
Eur.
J.
Immunol.
1992.
22:
689-696
Malaria-specific responses in non-exposed humans
691
fluorescinated sheep anti-mouse IgG (Silenus Labs., Haw-
thorn, Australia) for
1
h at
4
"C. Cells were then washed and
analyzed on a Becton Dickinson FACScan (Mountainview,
CA). For T cell receptor usage studies, cloned
T
cells or
PBM gated for lymphocytes by forward and side scatter,
were incubated with directly fluorescinated anti-a@ TcR
antibody (TCR-1, Becton Dickinson) or anti-6 antibody
(TCR
61,
T Cell Sciences, Boston, MA) prior to washing
and analysis.
2.9
Depletion
of
lymphocytes with magnetic beads
PBM were incubated with optimal concentrations of either
antLCD4 mAb (ATCC CRL 8002) or anti-CD8 mAb
(ATCC CRL
8014),
washed and then incubated with mag-
netic Dynabeads (M-450; Dynal A.S., Oslo) coated with
sheep anti-mouse IgG at a ratio of 10 beads per cell for CD8
and a ratio
of
5
beads per cell for CD4. The beads were
mixed with rocking for 30 min at
4
"C before removal with a
magnet.
2.10
FCM
analysis
Briefly, cells were incubated with mouse mAb against
either human CD3, CD4, or CD8 (ATCC CRL 8001,8002
and 8014, respectively) at optimal concentration for
1
h at
4°C. Cells were then washed twice and incubated with a
3
Results
3.1
CS
protein-specific responses
The donors are described
in
Table
1
and come from a group
of previously described donors [14]. Sera from seven
Table
1.
Stimulation indeces from
20
malaria-exposed and
20
non-exposed individuals to
rCS
protein (CSP), the known immunodom-
inant
T
cell epitopes in the protein
[14],
and tetanus toxoid")
Subject
El*
E2*
E3
E7*
E8*
E9*
El0
E 18
E25
E31*
E32
E40*
E42*
E43
E47*
E56"
E65
*
E66*
E67*
E68
c1
c2
c3
C8
c10
c12
C13
C15
C19
c20
c21
c22
C23
c24
C25
C26
C27
C28
C29
C30
PeDtide number
16 21
1.1
1.1
2.4 2.8
1.6 1.9
2.6 2.6
NT NT
NT NT
1.5 31.5
4.3 3.1
1.4 1.6
1.3 2.1
1.4 1.6
0.6 2.1
5.2 2.3
4.3 7.8
0.8 0.9
1.1 1.8
2.1 4.5
1.6 1.5
1.0 1.4
9.9 4.8
2.6 5.6
0.7 1.3
2.2 1.9
1.6 13.4
4.7 12.4
6.3 8.8
3.0 3.6
8.5 26.5
2.5 2.5
12.2 18.9
2.5 6.7
1.2
1.5
0.9 2.2
7.5 6.1
0.7 0.9
1.7 4.9
3.2 7.8
1.7 2.1
1.5 1.9
2.4 4.6
28
3.5
1.3
1.9
1.8
NT
NT
2.5
4.0
1.7
2.4
3.3
3.0
4.2
9.2
0.9
1.7
6.9
1.4
2.1
26.8
2.2
4.0
2.2
20.6
13.3
5.4
4.4
20.0
12.6
38.2
6.0
4.2
36.6
43.2
2.4
3.0
2.5
5.1
2.6
3.0
Stimulation index
CSP
0.5
pg/ml
0.2
0.6
0.8
3.5
0.8
1.6
1.5
8.2
0.3
0.5
6.7
1.5
1 .9
0.1
5.6
0.7
5.2
5.4
1.6
1.2
3.1
0.9
0.0
6.0
24.0
2.2
0.6
4.1
1.1
1.7
3.7
2.2
0.8
4.0
3.1
1.3
15.0
2.2
2.4
0.9
5
pg/ml
4.1
5.1
1.1
5.5
5.5
8.6
9.9
7.5
4.0
0.2
24.0
35.0
6.4
2.5
45.2
6.3
22.3
27.0
2.3
48.7
13.9
4.8
3.3
11.2
27.6
10.9
21.6
52.2
5.4
44.2
14.2
2.4
38.0
45.0
66.7
4.6
27.5
1.6
5.2
4.5
Tetanus
toxoid
4.4
13.4
3.3
10.4
NT
NT
30.8
5.4
4.8
3.5
11.7
16.6
24.5
14.6
0.5
5.7
260
1 .5
3.0
220
56.9
41.8
6.4
27.8
36.0
96.0
0.3
199.7
141.5
88.9
62.5
557
21.6
65.0
16.8
15.6
8.7
10.8
12.4
82.0
Anti-
NANP
Ab
titer
>64
21024
<16
46
<16
<16
<16
46
<16
<I6
<16
>64
<16
<16
>64
>64
>2048
46
46
>2048
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
NT
Years
of
exposure
4
7
10
14
14
16
3
3
4
months
10
23
19
10
9
7
14
6
6
2
months
9
a)
All
'E'
subjects have had
sporozoite exposure as
listed, but only
'E*'
subjects
have had clinical malaria.
Peptides were tested at their
optimal concentration
(30
kg/ml).
Mean cpm
of
background (no antigen):
1411.
692
Y.
Zevering,
F.
Amante, A. Smillie et
al.
malaria-exposed individuals contained antibodies against
the major
B
cell epitope of the protein ([NANP],). This
suggests that their degree of exposure to sporozoites was
less than that for lifelong residents of malaria-endemic
villages in Gambia where we have previously found that
58
Yo
-66
YO
of sera contain such antibodies [15, 161. Lym-
phocytes from 40 donors (half of whom had a history
of
sporozoite exposure) were tested for proliferation to the
recombinant baculovirus-expressed CS protein, to the
three most immunogenic synthetic peptides chosen from a
panel of 29 overlapping peptides spanning the entire
protein [14], and to tetanus toxoid.The extent of lympho-
cyte proliferation to these antigens is given inTable
1
where
it can be seen that T cells from 35 of the 40 donors (17
of
the
20 malaria-exposed and
18
of the 20 non-exposed donors)
responded by proliferation (stimulation index >3) to
recombinant CS protein at the optimal concentration
(5 yg/ml), that T cells from many donors responded to one
or more of the synthetic peptides, and that
T
cells from 35
of
38
donors tested responded to tetanus toxoid.There was
no evidence for increased affinity of the
T
cells for the CS
protein from the malaria-exposed group since
T
cells from
both groups responded comparably at the lower concentra-
tion of
0.5
yglml (Table 1). Some individuals responded
poorly to the protein but, nevertheless, did respond to one
or more of the peptides (E43, C22, C28).This may indicate
that inadequate antigen processing with respect to those
peptides was occurring for this protein, as previously
suggested [13, 23].Two individuals (E3, E67) responded to
neither the
CS
protein nor the synthetic peptides. There
was no correlation between response to tetanus toxoid and
response to CS protein.The cells responsible for the prolife-
rative response to CS protein could be removed by
antibodies to CD4 and magnetic beads but not by antibod-
ies to CD8 and magnetic beads.
Eleven short-term T cell lines generated by baculovirus-
expressed CS protein stimulation were challenged with
synthetic peptides as well as (in some cases) with recombi-
nant protein. Four of these lines proliferated in response to
the synthetic peptides, indicating that the synthetic pep-
tides can represent actual epitopes within the recombinant
protein (Table 2). The response of the lines to the peptides
does not represent a primary response to the peptides from
cells carried over in culture from the initial bleed since cells
carried in culture without CS protein stimulation did not
respond to any of the peptides. It is possible, however, that
some of the peptides that are stimulatory for a given
individual's peripheral blood
T
cells do not represent actual
epitopes present in the protein for that individual, due to
difficulties
in
antigen processing (see above, for E43, C22,
C28).
Using the yeast-expressed recombinant CS protein, we have
tested whether specific T cell responses from non-exposed
individuals are genetically restricted.Three long-termT cell
lines were generated from different donors and restimu-
lated with CS protein in the presence of irradiated PBM
from different donors. As shown in Fig.
2,
proliferation was
significantly greater
in
the presence of autologous APC for
both CS protein-specific responses and for control tetanus
toxoid-specific responses. The moderate response from
both
T
cell lines from donor C15 in the presence
of
APC
from donor C21 may result from the fact that they share
MHC class
I1
alleles (DR2 and DQwl).
Eur.
J.
Immunol.
1992. 22: 689-696
Table
2.
Proliferation of short-term
CS
protein-specificT
cell
lines
to rCs protein
and
the three immunodominant peptide epitopes
present
in
the recombinant protein")
Stimulation index
Subject
16 21 28 5
pglrnl
Peptide number
CSP
E2
E7
E8
E9
E40
E47
c12
C23
C24
c28
c29
0.9 0.9
1 .0 0.9
3.9 9.3
0.5 5.7
1.2 2.0
2.1 0.5
1.9 0.4
6.0 1.4
1.6 3.3
0.5 1.4
2.0 0.6
1.5
0.4
5.2
15.8
1.1
1.6
0.5
22.0
0.7
1 .0
0.7
a)
Mean
cpm
of
background
(no
antigen):
1497
CSP-specific
lines
tested with CSP
11.9
3.5
33.6
24.5
NT
NT
NT
NT
NT
NT
6.0
TT-apecific
line
teated with
ll
C15
C24
C25
Figure
2.
Proliferation of long-term
T
cell
lines specific for
CS
protein for
CS
protein (yeast-expressed)
and
tetanus toxoid
(TT)
in
the presence
of
autologous
and
allogeneic
APC.
3.2
Precursor frequency
of
CS
peptide-specific responses
The frequency of Tcells reactive with one of the most
immunodominant peptides from the molecule, peptide 21
(residues 331-350 [14], was then determined. For six
Eur.
J.
Immunol. 1992.
22:
689-696
..
posed donors and three non-
exposed
donors
responding
to
individuals, the T cell precursor frequencies ranged from
1/11
000
to
1/850
000
(Fig. 3).This compares with precursor
frequencies to tetanus toxoid in two individuals of
1/11
000
and
1/3500
as we previously defined using the same
methodologies as here
1241.
Due to limitations of material,
it was not possible to determine the precursor frequencies
to recombinant CS protein. However, we compared the
precursor frequencies of
T
cells specific for all the major
epitopes of the protein for a large group of malaria-exposed
and non-exposed individuals. At the same time as setting
up the limiting dilution assay for each indivudual, we also
set up a standard bulk culture. We were thus able to
compare directly the precursor frequency for each do-
nodpeptide combination with the stimulation index for
that same combination. This data (comparing
31
separate
donorlpeptide combinations) demonstrated that there was
a close correlation between the two measurements, and
that for the precursor frequencies measured, there was
no
obvious difference between the exposed and non-exposed
groups as a whole.The highest frequency we recorded from
a non-exposed individual was
286
T
cells responding to
peptide
#1
per
lo6
peripheral blood
T
cells, and the highest
response from exposed individual was
74
T
cells respond-
ing to peptide
#21
per
lo6
T
cells. Since the stimulation
indeces of T cells from both groups of donors reactive to the
recombinant protein were similar (Tables
1
and
3),
we
predict that malaria-exposed and non-exposed donors
T
CELLS
PER
WELL
(x
loa)
0
50
100
150
200
I
Donor:
El0
Frequency:
92
x
rod
Y
4
I
Donor:
€32
Frequency: 2.4
x
lod
100
30
Donor:
€43
Frequency: 1.2
x
1
0'
7
0
Malaria-specific responses in non-exposed humans
6Y3
contain comparable numbers of
T
cells specific for the CS
protein.
3.3
Cytokine production following CS protein
stimulation
T
cell cultures from malaria-exposed and non-exposed
individuals produced IFN-y and IL
6
(cytokines specifically
involved in sporozoite immunity
[25, 261)
following
CS
protein stimulation (Table
3).
T cells from two individuals
tested in this study did not proliferate to rCS protein
(C31,
C33);
however, significant quantities of lymphokines were
produced by
T
cells from both. There was
no
significant
difference in lymphokine production in response
to
CS
protein between the malaria-exposed and non-exposed
individuals, and
T
cells from two individuals tested who
had high antibody titers
(E2, E65)
produced less lympho-
kines than
T
cells from some non-exposed donors.
3.4
Blood-stage-specific responses
We have also been able to stimulate blood-stage-specific
T cells and readily generate clones from non-exposed
humans (Table
4).
In
vitro,
T
cell responses peaked at
6-7
days, and precursor frequencies ranged from
1/500-1/2000
50
100
150
100
I
I
I
I
100
I
I
I
1-
Donor:
c10
Frequency: 2.2
x
lod
50
Donor:
c10
Frequency: 2.2
x
lod
+
50
Figure
3.
Limiting dilution
analysis
of
peripheral
blood
T
cells from three malaria ex-
30
Donor:
C12
Frequency:
4.8
x
10'
?
3.
Limiting dilution
;is
of
peripheral
blood
j
from three malaria ex-
694
Eur.
J.
Immunol.
1992.
22:
689-696
Table
3.
Stimulation indeces and lymphokine production
on
following stimulation
of T
cells from malaria-exposed and non-exposed
individuals with recombinant CS protein
Y.
Zevering,
F.
Amante,
A.
Smillie et al.
Subjcct
E2
E9
E65
Eh9
E70
c3
C4
c10
C3 1
C32
c33
Stimulation index Lymphokine production following CS
protein stimulation
CSP”)
Day
1
Day
4
Tetanus
(5
pg/ml) IFN-yb)
IL
6c)
IFN-yb)
IL
64
toxoid
53.8 7.6 0.0 4.2 0.6
32.8 10.8 0.0 2.7 0.44
176 44.4 0.74 8.3 0.8
36.7 5.2 0.0 6.0 0.86
60.0 6.1 0.0 2.6 0.0
7.5 3.6 0.0 3.9 0.78
39.5 3.2 0.0 3.5 0.0
41.3 13.8 0.6 0.7
0.0
5.8 1.2
0.0
2.7 0.68
8.0 3.5 0.78 12.8 1.3
39.6 1.4 0.0 4.4 0.54
peripheral blood
T
cells. All of
10
T
cell clones examined
have stained positive for CD3, CD4 and TcR
a@,
and
negative for CD8 and TcR
y/6.
The responses of four clones
are listed in Table 4. Certain clones were specific in that
they could discriminate
P
vivax
from
l?
falciparum
(clones
C4 and C5,Table 4),whereas some other clones responded
equally well to both parasites (not shown), presumably
because they recognized shared antigens. Proliferation of
these clones was genetically restricted by APC expressing
MHC class
I1
genes in common with the clone, as has
previously been demonstrated for malaria gamete-specific
T
clones
[lo].
Following antigenic stimulation, clones
secreted
IFN-y
a lymphokine known to mediate malaria
parasite killing
[27,
281.
Clones could also be generated
from non-exposed individuals to synthetic peptides from
the MSAl proteins of
I?
falciparum
and
P
vivax
(see Sect.
2.3,
data not shown).
1.7
0.9
7.2
3.4
0.7
1.3
1.9
0.0
0.7
8.1
1.1
b) I.U./ml.
a)
CS
protein tested at
5
and
1
pg/ml(5 pglml data
only given as this was optimal in all cases). Mean
cpm of backgraound
(no
antigen):
1515.
c) ng/ml.
3.5
TcR usage
of
malaria-specific T cells
The type of
T
cell receptors used in the response to the
recombinant CS protein and whole parasite was deter-
mined. Lymphocytes from five individuals (one exposed,
four non-exposed) were analyzed by FCM to determine the
fraction of cells expressing TcR
alp
and TcR
y/6.
Lympho-
cytes were then stimulated with the yeast-expressed CS
protein or whole parasite and proliferated; thereafter the
cells were re-examined.
As
shown in Table
5,
there was no
evidence that TcR
y/6
cells were preferentially expanded in
these experiments, although it cannot be excluded that for
other individuals or for certain malaria antigens TcR
y/6
cells can be activated. There may have been some stimula-
tion of TcR
y/6
cells in our experiments, which was not
detected since we were not studying purified populations of
such cells.
Table
4.
Proliferation and specificity
of
peripheral blood lymphocytes and
T
cells clones to
R
falciparum-infected
RBC
(Pf. pRBC).”)
Subject
E42
E7 1
E72
C13
c14
c15
C16
C17
c24
C31
c34
c35
Clone
C1
Clone
C3
CloneC4
Clone
C5
PHA
60
620
34 213
34 701
70 106
32 654
13 290
36 489
25 815
53 444
64 848
77 207
59 272
Blank
204
571
284
368
102
118
264
108
95
104
272
463
CPm
Pf.
pRBC (FCR-3)lml
106 105
104
55 462
5753
3319
30 577
8363
15 877
25 455
5 078
21 214
37 209
33 303
11 515
-
44 330
2552
17 870
14 457
1412
9634
5259
654
9088
27 581
7214
277 1
8730
9424
13 192
2491
(58)
(131)
34 330
2748
11 069
745
370
1699
745
944
1903
9198
2815
2436
402
1
2176
263
308
(68)
(41)
Blank
(Day
7)
3193
2969
5398
1880
1883
943
2241
1697
33
1
1974
1569
3002
817
129
160
148
a) “E” subjects
in
this table have experienced
clinical
R
falciparum
infections;
“C”
subjects
have neither had clinical malaria nor been
exposed to sporozoites. Proliferations in re-
sponse to
PHA
was measured after
48
h, and
proliferation in response to
l?
falciparum
was
measured after
6
days. The proliferative re-
sponses of the clones
C4
and
C5
to
R
vivax
parasites is given in parentheses:
().
None
of
the
clones proliferated in the presence
of
uninfected
RBC.
Eur. J. Immunol.
1992.
22:
689-696
Table
5.
TcR analysis
of
lymphocytes before and after exposure to
CS protein or malaria parasites
Donor
El4
C4
C15
C24
C31
Percentage
of
lymphocytes
Antigen TcR
al$
rCS
protein
Tetanus toxoid
Nil
rCS
protein
Tetanus toxoid
Nil
rCS
protein
Tetanus toxoid
Nil
pRBC
PMC
Pre
70.7
70.7
70.7
NT
NT
NT
NT
NT
NT
73.1
67.6
post
85.9
86.1
88.9
NT
NT
NT
NT
NT
NT
92.2
88.8
TcR
yl6
Pre
5.1
5.1
5.1
4.6
4.6
4.6
2.5
2.5
2.5
15.6
9.6
post
1.6
1.7
2.2
2.3
1.9
2.6
2.3
3.0
1.9
10.3
7.8
4
Discussion
Malaria-specific CD4 T cells bearing TcR
alp
chains and
capable of proliferation and lymphokine production are
present
in
non-exposed non-vaccinated individuals at high
frequency. They are genetically restricted, apparently by
MHC genes. It is likely that all individuals have Tcells
which will either proliferate or secrete parasiticidal lym-
phokines (or both) in response to CS protein or whole
parasite stimulation. While previous malaria exposure does
not appear to expand this population of cells (at least to the
extent that the donors of this study were exposed), our
results would not preclude the possibility that immuniza-
tion with a sporozoite or blood-stage ‘vaccine’could expand
further the already existent population of specific
T
cells.
However, the proliferative response of some non-exposed
individuals to the CS protein was comparable to or even
greater than their response to tetanus toxoid (Tables
1
and
3).
These responses may arise through cross-reactivity [29]
since a peptide 21-specific T cell clone from a BCG-
vaccinated individual reacted strongly with
PPD
[30].
Peptide 21-specific clones from other individuals, however,
have not responded to PPD.
The antigens recognized by the blood-stage-specific T cells
have not been identified but different clones clearly
recognize different antigens based on different patterns of
reactivities to a panel of different
f?
fulcipurum
clones.
Although T cells from approximately
90
%
of individuals
responded to the CS protein by proliferation, we do not
believe that this response rcpresents a mitogenic response
since: (a) the peak of the response to both CS protein and
tetanus toxoid occurred on days
5
and
6
compared to a peak
of days 1-3 for PHA and Con
A
stimulation;
(b)
the
response
of
CS protein-specific T cell lines to synthetic
peptides from the protein suggests that the induction of the
response is not due to a mitogen or a superantigen; (c) the
response was not associated with CDS T cells and (d) the
responses of three T cell lines specific to the CS protein
were MHC restricted.
Malaria-specific
responses
in non-exposed humans
695
Trial human vaccine candidates have been designed to
expand a population
of
specific lymphocyte precursors.
While our data in no way denies an important role for CD4
T cells in malaria immunity, in pre-vaccinated adult
humans, at least, many malaria-specific precursor lympho-
cytes appear to have been already stimulated and many
apparent memory cells are already present in the circula-
tion. It is also possible that actual malaria-specific precur-
sor cells are present in the periphery at very high frequency
(comparable to the frequency of tetanus toxoid-specific
memory Tcells) and that
in
vitro
these are capable of
responding to malaria peptides or parasites. Although
non-exposed humans are not immune to malaria, it cannot
be argued that their malaria-specific T cells could play no
role in anti-parasite immunity. Other factors acting in
concert with these T cells, but not present in non-exposed
individuals, may be critical for full expression
of
cellular
immunity to malaria. One such factor could be a suitably
modified spleen
[31].
Furthermore, malaria-specific T cells
in non-exposed individuals could partially contribute to the
varying degrees of innate resistance of adult humans to
malaria parasites [32].
We
sincerely thank the donors for making this study possible, Dr.
Phil Burr, Chiron, for supplying the yeast-expressed recombinant
CS
protein and Dr. Mitch Gross,
SKB,
for supplying R32tet32.
Received June
19, 1991;
in revised form October
29, 1991.
5
References
1
Miller,
J.
F.
A.
P.,
Lancet
1961.
ii:
748.
2
Bjorkman,
P.
J.,
Saper, M. A., Samraoui,
B.,
Bennett, W.
S.,
Strominger, J. L. and Wiley,
D.
C.,
Nature
1987.
329:
506.
3
Corradin, G., Etlinger,
H.
M. and Chiller,
J.
M.,
J.
Immunol.
1977.
119:
1048.
4
Grun, J.
L.
and Weidanz, W.
P.,
Infect. lmmun.
1983.
41:
1197.
5
Chen, D. H., Tigelaar, R.
E.
and Weinbaum,
F.
I.,
J.
Immu-
nol.
1977.
118:
1322.
6
Brake,
D.
A.,
Long, C.
A.
and Weidanz, W. P.,
J.
Immunol.
1988.
140:
1989.
7
Del
Giudice, G.. Grillot,
D.,
Renia. L., Muller,
I.,
Corra-
din, G., Louis,
J.
A.,
Mazier, D., Lambert, P-H.,
Immunol.
Lett.
1990.
25:
59.
8
Tsuji, M., Romero,
P.,
Nussenzweig, R. S.. and Zavala,
F.,
J.
Exp. Med.
1990.
172:
1353.
9
Chizzolini, C. and Perrin, L.,
J.
Immunol.
1986.
137:
1022.
10
Good, M.
F.,
Quakyi,
I.
A., Saul,
A.,
Berzofsky,
J.
A., Car-
ter. R., and Miller, L.
H.,
J.
Immunol.
1987.
138:
306.
11
Sinigaglia,
F.,
Guttinger, M., Gillessen,
D.,
Doran, Dm M.,
Takacs, B., Matile, H., Trzeciak, A., Gillessen,
D.,
and
Pink,
J.
R.
L,
Eur.
J.
Immunol.
1988.
18:
633.
12
Dontfraid,
F.,
Cochran,
M.
A.,
Pombo,
D.,
Knell,
J.
D.,
Qua-
kyi,
1.
A.,
Kumar,
S..
Houghten, R. A., Berzofsky,
J.
A,,
Miller,
L.
H., and Good,
M.
F.,
Mol.
Biol. Med.
1988.
5:
18.5.
13
Good, M.
F.,
Branigan,
J.,
Smith, G.,
and
Houghten, R.
A,,
Peptide Res.
1990.
3:
110.
14
Zevering,
Y.,
Houghten,
R.
A.,
and Good, M.
F.,
Inte. Immu-
nol.
1990.
2:
945.
1.5
Good, M.
F.,
Pombo.
D.,
Quakyi,
I..
Riley,
E.,
Houghten, R.,
Menon,
A.,
Alling,
D.
W., Berzofsky,
J.
A.,
andMiller,
L.
H.,
Proc. Natl. Acad. Sci.
USA
1988.
85:
1199.
16 De
Groot,
A.
S., Johnson,
A.
H., Maloy, W. L., Quakyi,
I.
A.,
Riley,
E.
M.,
Menon,
A., Banks,
S.
M.,
Berzofsky,
J.
A,,
and Good, M.
F.,
J.
Immunol.
1989.
142:
4000.
696
Y.
Zevering,
F.
Amante, A. Smillie et al.
Eur.
J.
Immunol.
1992. 22: 689-696
17
Nardin, E. H., Herrington, D.
A.,
Davis,
J.,
Levine, M., Stu-
ber, D., Takacs, B., Caspers,
P,
Barr,
P,
Altszuler, R., Clavi-
jo,
P.,
and Nussenzweig, R.
S.,
Science
1989. 246: 1603.
18
Del Portillo,
H.
A.,
Gysin,
J.,
Mattei, D. M., Khouri, E.,
Udagama,
P
V,
Mendis, K.
N.
and David,
P
H.,
Exp. Para-
sitol.
1988.
67:
346.
19
Crisanti,
A,,
Muller,
H.
M., Hilbich, C., Sinigaglia,
F.,
Matile,
H., McKay,
M.,
Scaife,
J.,
Beyreuther,
K.
and Bujard, H.,
Science
1988. 240: 1324.
20
Sinigaglia,
F.
and Pink,
J.
R.
L.,
EMBO
J.
1985. 4: 3819
21
Lefkovits,
I.,
and Waldman, H.,
Limiting Dilution AnaZysis
of
Cells in the Immune System,
Cambridge University Press,
London
1979.
22
Van Oers, M. H.
J.,
Zeijlemaker, W.
P.
and Schellekens,
P
Th.
A.,
Eur.
J.
Immunol.
1977.
7:
143.
23
Link, H. T., Ballou,
W.
R. and Krzych,
U.,
Am.
J.
Trop. Med.
Hyg.
1990. 43: 452.
24
Good, M. F., Chapman, D. E., Powell, L. W., and Halli-
day,
J.
W.,
Clin. Exp.
Immunol.
1986.
66:
340.
25
Ferreria, A., Schofield, L., Enea,
V,
Schellekens,
H.,
van der
Meide,
I?,
Collins, W. E., Nussenzweig,
R.
S.,
and Nussen-
zweig,
V.,
Science
1986. 232: 881.
26
Pied,
S.,
Renia, L., Nussler,
A.,
Miltgen,
F.,
and Mazier,
D.,
Parasite Immunol.
1991.
13:
211.
27
Clark,
I.
A.,
Hunt, N. H., Butcher, G.
A.
andCowden,W. B.,
J.
Immunol.
1987. 139: 3493.
28
Stevenson, M. M.,Tam, M.
F.
and Nowotarski, M.,
Immunol.
Lett.
1990. 25: 115.
29
Jones, K. R., Hickling,
J.
K.,
Target, G.
A.
T.,
and Playfair,
J.
H.
L.,
Eur.
J.
Immunol.
1990. 20: 307.
30
Fern,
J.,
and Good, M.
F.,
J.
Immunol.
1992,
in press.
31
Winkel, K.
D.,
and Good, M.
F.,
Parasite Immunol.
1991.13:
32
Powell, R.
D.,
McNamara,
J.
V.,
and Rieckmann, K. H.,
517.
Proc. Helminth.
SOC.
Wash.
1972. 39: 51.
... In a study addressing this issue, 68 of exposure to organisms containing cross-reactive epitopes. 69,70 It is likely that there are many potential T-cell epitopes in Plasmodium but that few can be processed for presentation to T cells, and that different individuals will process parasite proteins differently. This argues against a strategy of making a polyvalent vaccine by combining multiple defined T-cell epitopes. ...
Article
Full-text available
After many decades of research, an effective vaccine for malaria is still not available. Most research efforts have focused on identifying a key target antigen and then using powerful adjuvants to generate specific antibodies that can block parasites from entering host cells (hepatocytes, red blood cells). However, the inability to generate sufficiently potent antibody responses has led to significant disappointment with current vaccine programs. An additional challenge for sub‐unit vaccines is that key vaccine antigens are highly polymorphic. These challenges have spurred radically different approaches to malaria vaccine development. Many of these involve the use of “whole parasites”—either extracted from mosquitoes or cultured. With these, every parasite molecule for that particular strain is included in the vaccine. This strategy is showing great promise following several clinical trials with irradiated sporozoites. However, a whole‐parasite approach to a blood stage vaccine has not advanced as quickly. This article outlines the history, the different approaches that are being taken and the challenges associated with whole parasite blood stage vaccines and discusses recent exciting developments as these vaccines now move into the clinic.
... Previous studies have shown that PBMCs from naive individuals stimulated with P. falciparum parasitised RBC [80] or stimulated with malaria antigens did produce IFN-γ [81], mainly innate cells, such as natural killer cells. PBMCs from non-exposed individuals in another study produced IFN-γ and IL-6 after stimulation with P. falciparum CS protein [82]. Accordingly, higher IFN-γ and IL-6 production from control group PBMCs in this study suggested that innate cells, innate lymphoid cells and naive T-cells recognised lysate and produced cytokines [83]. ...
Article
Full-text available
Malaria is an infectious disease caused by parasites from the genus Plasmodium (P. falciparum and P. vivax are responsible for 90% of all clinical cases); it is widely distributed throughout the world’s tropical and subtropical regions. The P. vivax Pv12 protein is involved in invasion, is expressed on merozoite surface and has been recognised by antibodies from individuals exposed to the disease. In this study, B- and T-cell epitopes from Pv12 were predicted and characterised to advance in the design of a peptide-based vaccine against malaria. For evaluating the humoral response of individuals exposed to natural P. vivax infection from two endemic areas in Colombia, BepiPred-1.0 software was used for selecting B-cell epitopes. B-cell epitope 39038 displayed the greatest recognition by naturally-acquired antibodies and induced an IgG2/IgG4 response. NetMHCIIpan-3.1 prediction software was used for selecting peptides having high affinity binding for HLA-DRβ1* allele lineages and this was confirmed by in-vitro binding assays. T-epitopes 39113 and 39117 triggered a memory T-cell response (Stimulation Index≥2) and significant cytokine production. Combining in-silico, in-vitro and functional assays, two Pv12 protein regions (containing peptides 39038, 39040, 39113 and 39117) have thus been characterised as promising vaccine candidates against P. vivax malaria.
... Antigenicity of iRBCs was examined using PBMCs from malaria non-exposed Japanese donors. CD4 + T cells in PBMCs proliferated and PBMCs produced IFN-γ in response to iRBCs and not to uRBC, in line with the previous reports (data not shown) [31,32]. ...
Article
Individuals living in malaria endemic areas become clinically immune after multiple re-infections over time and remain infected without apparent symptoms. However, it is unclear why a long period is required to gain clinical immunity to malaria, and how such immunity is maintained. Although malaria infection is reported to induce inhibition of immune responses, studies on asymptomatic individuals living in endemic regions of malaria are relatively scarce. We conducted a cross-sectional study of immune responses in asymptomatic school children aged 4-16years living in an area where Plasmodium falciparum and Schistosoma mansoni infections are co-endemic in Kenya. Peripheral blood mononuclear cells were subjected to flow cytometric analysis and cultured to determine proliferative responses and cytokine production. The proportions of cellular subsets in children positive for P. falciparum infection at the level of microscopy were comparable to the negative children, except for a reduction in central memory-phenotype CD8+ T cells and natural killer cells. In functional studies, the production of cytokines by peripheral blood mononuclear cells in response to P. falciparum crude antigens exhibited strong heterogeneity among children. In addition, production of IL-2 in response to anti-CD3 and anti-CD28 monoclonal antibodies was significantly reduced in children with P. falciparum-positive as compared to -negative children, suggesting a state of unresponsiveness. These data suggest that the quality of T cell immune responses is heterogeneous among asymptomatic children living in the endemic region of P. falciparum, and that the responses are generally suppressed by active infection with Plasmodium parasites.
... It seems unusual that malaria-naïve donors present large numbers of T cells specific for P. falciparum antigens. However, high frequencies of T cells specific for P. falciparum antigens in nonexposed donors has been reported before (14,(64)(65)(66). Because malaria is considered the strongest evolutionary-selective force in recent human history (67), it is possible that having a large number of T cells specific for this parasite may be under positive selection. ...
Article
Significance With more than 200 million cases per year worldwide and more than 400,000 deaths, mostly affecting children in sub-Saharan Africa, malaria is still one of the most prevalent infectious diseases. Infection with the malaria parasite Plasmodium falciparum is characterized by high inflammation but also the failure of the immune system to form efficient memory, leading to recurring infections. No efficient vaccine is available to date. Here we have studied the response of dendritic cells (DCs), an essential cell type in the orchestration of immune and vaccine responses. We found that P. falciparum induces a distinct transcriptional profile compared with a classical inflammatory stimulus in primary human DCs, leading to a highly atypical response, which may contribute to parasite immune evasion during malaria.
... Despite many reports of γδ T cells in malaria patients, it is unclear whether γδ T cells are induced during malarial infection (Ho et al., 1990;Roussilhon et al., 1990;Goerlich et al., 1991;Currier et al., 1992;Goodier et al., 1992;Zevering et al., 1992;Dick et al., 1996;Hviid et al., 1996Hviid et al., , 2001Rzepczyk et al., 1996;Goodier and Targett, 1997;Worku et al., 1997;Waterfall et al., 1998). To investigate this phenomenon in more detail, PBMCs and plasma obtained from UMPs and NCs living in malaria-endemic areas of Attapeu Province, Lao PDR and from NECs in Vientiane, the country's capital, were analyzed during physical examination of the subjects. ...
Article
Full-text available
Although expansions in γδ T cell populations are known to occur in the peripheral blood of patients infected with Plasmodium falciparum, the role of these cells in people with naturally acquired immunity against P. falciparum who live in malaria-endemic areas is poorly understood. We used a cross-sectional survey to investigate the role of peripheral blood γδ T cells in people living in Lao People’s Democratic Republic, a malaria-endemic area. We found that the proportion of non-Vγ9 γδ T cells was higher in non-hospitalized uncomplicated falciparum malaria patients (UMPs) from this region. Notably, we found that the non-Vγ9 γδ T cells in the peripheral blood of UMPs and negative controls from this region had the potential to expand and produce IL-10 and interferon-γ when cultured in the presence of IL-2 and/or crude P. falciparum antigens for 10 days. Furthermore, these cells were associated with plasma interleukin 10 (IL-10), which was elevated in UMPs. This is the first report demonstrating that, in UMPs living in a malaria-endemic area, a γδ T cell subset, the non-Vγ9 γδT cells, expands and produces IL-10. These results contribute to understanding of the mechanisms of naturally acquired immunity against P. falciparum.
... Although two subjects had transient measurable antibody levels to CSP, neither maintained a detectable response at six months post-challenge. A third subject had pre-existing humoral and cellular responses to CSP that persisted at all timepoints, did not markedly increase after infection and remains unexplained; anomalous cross-reactive responses to CSP have also been reported by others [26]. ...
Article
Full-text available
Background: Controlled human malaria infection (CHMI) studies which recapitulate mosquito-borne infection are a critical tool to identify protective vaccine and drug candidates for advancement to field trials. In partnership with the Walter Reed Army Institute of Research, the CHMI model was established at the Seattle Biomedical Research Institute's Malaria Clinical Trials Center (MCTC). Activities and reagents at both centers were aligned to ensure comparability and continued safety of the model. To demonstrate successful implementation, CHMI was performed in six healthy malaria-naïve volunteers. Methods: All volunteers received NF54 strain Plasmodium falciparum by the bite of five infected Anopheles stephensi mosquitoes under controlled conditions and were monitored for signs and symptoms of malaria and for parasitemia by peripheral blood smear. Subjects were treated upon diagnosis with chloroquine by directly observed therapy. Immunological (T cell and antibody) and molecular diagnostic (real-time quantitative reverse transcriptase polymerase chain reaction [qRT-PCR]) assessments were also performed. Results: All six volunteers developed patent parasitemia and clinical malaria. No serious adverse events occurred during the study period or for six months post-infection. The mean prepatent period was 11.2 days (range 9-14 days), and geometric mean parasitemia upon diagnosis was 10.8 parasites/µL (range 2-69) by microscopy. qRT-PCR detected parasites an average of 3.7 days (range 2-4 days) earlier than blood smears. All volunteers developed antibodies to the blood-stage antigen merozoite surface protein 1 (MSP-1), which persisted up to six months. Humoral and cellular responses to pre-erythrocytic antigens circumsporozoite protein (CSP) and liver-stage antigen 1 (LSA-1) were limited. Conclusion: The CHMI model was safe, well tolerated and characterized by consistent prepatent periods, pre-symptomatic diagnosis in 3/6 subjects and adverse event profiles as reported at established centers. The MCTC can now evaluate candidates in the increasingly diverse vaccine and drug pipeline using the CHMI model. Trial registration: ClinicalTrials.gov NCT01058226.
... Whether or not this response really represents an in vitro priming of CD4 cells needs to be further investigated. Indeed, CD4 cells cross-reactive to malarial antigens have been described by numerous authors (32)(33)(34). Dendritic cells could be involved in Treg cell generation either through the presentation of ligands which preferentially expand Treg cell populations or through mechanisms independent of specific TCR activation based on the production of "suppressive" cytokines. In our experimental conditions, the functional phenotype of MDDC acquired in the presence of parasitic extracts resembles that of semimature dendritic cells with prevalent tolerogenic functions (35), and the maximum expansion of Treg cell population induced is achieved by MDDC pulsed with relatively small amounts of PfSE (1 to 10 g/ml, corresponding to 10 4 to 10 5 iRBC), indicating that, under conditions of low antigenic concentrations, MDDC may present high-affinity ligands to T cells which primarily promote Treg induction (25). ...
Article
Full-text available
The optimal immune response to malaria infection comprises rapid induction of inflammatory responses promptly counteracted by regulatory mechanisms to prevent immunopathology. To evaluate the role of dendritic cells (DC) in the balance of parasite-induced inflammatory/anti-inflammatory mechanisms, we studied the activity of monocyte-derived dendritic cells (MDDC), previously exposed to soluble extracts of Plasmodium falciparum-infected red blood cells (PfSE), in the differentiation of CD4 cells isolated from donors never exposed to malaria infection. We show that MDDC exposed to PfSE are extremely efficient to induce a contemporary differentiation of TH1 effector cells and T regulatory (Treg) cells in CD4 T cells even when exposed to low concentrations of parasitic extracts. Treg cells induced by MDDC infected with PfSE (MDDC-PfSE) produce transforming growth factor beta (TGF-β) and interleukin 10 (IL-10) and are endowed with strong suppressive properties. They also show phenotypical and functional peculiarities, such as the contemporary expression of markers of Treg and TH1 differentiation and higher sensitivity to TLR4 ligands both inducing an increasing production of suppressive cytokines. On the whole, our data indicate that MDDC exposed to PfSE orchestrate a well-balanced immune response with timely differentiation of TH1 and Treg cells in CD4 cells from nonimmune donors and suggest that, during the infection, the role of MDCC could be particularly relevant in low-parasitemia conditions.
Article
Regulatory T (Treg) cells have been shown in some cases to limit vaccine-specific immune responses and impact efficacy. Very little is known about the regulatory responses to the leading malaria vaccine candidate, RTS,S. The goal of this study was to begin to characterize the regulatory responses to the RTS,S vaccine. Using multi-parameter flow cytometry, we examined responses in 13 malaria naïve adult volunteers who received 2 doses of RTS,S given eight weeks apart. Five of these volunteers had previously received 3 doses of a candidate DNA-CSP vaccine, with the final dose given approximately one year prior to the first dose of the RTS,S vaccine. We found that the frequency of CD25hiFoxp3⁺ Treg cells decreased following administration of RTS,S (p = 0.0195), with no differences based on vaccine regimen. There was a concomitant decrease in CTLA-4 expression on CD25hiFoxp3⁺ Treg cells (p = 0.0093) and PD-1 levels on CD8⁺ T cells (p = 0.0002). Additionally, the frequency of anergic CTLA-4⁺CCR7⁺ T cells decreased following vaccination. An inverse correlation was observed between the frequency of Plasmodium falciparum circumsporozoite protein (PfCSP)-specific IFN-γ and PfCSP-specific IL-10, as well as an inverse correlation between IL-10 induced by Hepatitis B surface antigen, the carrier of RTS,S, and PfCSP-specific IFN-γ, suggesting that immunity against the vaccine backbone could impact vaccine immunogenicity. These results have implications for future malaria vaccine design.
Article
Full-text available
Frequencies and absolute numbers of peripheral gamma/delta T cells have been reported to increase after episodes of Plasmodium falciparum malaria in adults with limited or no previous malaria exposure. In contrast, little is known about the gamma/delta T-cell response to malaria in children from areas where malaria is endemic, who bear the burden of malaria-related morbidity and mortality. We investigated the gamma/delta T-cell response in 19 Ghanaian children from an area of hyperendemic, seasonal malaria transmission. The children presented with cerebral malaria (n = 7), severe malarial anemia (n = 5), or uncomplicated malaria (n = 7) and were monitored from admission until 4 weeks later. We found no evidence of increased frequencies of gamma/delta T cells in any of the patient groups, whereas one adult expatriate studied in Ghana and three adults admitted to the hospital in Copenhagen, Denmark, all with uncomplicated, primary P. falciparum malaria, showed increased gamma/delta T-cell frequencies similar to those previously reported. All patients had lowered absolute numbers of peripheral gamma/delta T cells at admission, changing to increased numbers by days 7 to 14 and then returning to normal levels. The study raises questions regarding age and degree of previous exposure as determinants of malaria-induced gamma/delta T-cell responses.
Article
It is now generally accepted that peripheral blood of humans not exposed previously to malaria contains T cells which proliferate vigorously in response to malaria parasites and antigens. Although it has been claimed that these cells express a memory phenotype, their origin is uncertain. We have examined the phenotype and immunological responses of such cells. We confirm that these cells do express the 'memory phenotype', CD45Ro, in that depletion of such cells, but not of CD45Ra (virgin) cells, abrogates the immune response to malaria parasites. In an effort to define the genesis of these responses, numerous malaria-specific T cell clones have been generated from non-exposed individuals. These were tested for reactivity to a large panel of common bacterial, viral, and fungal pathogenic and non-pathogenic organisms. Most clones proliferated vigorously in response to one or more such organisms, while many clones demonstrated smaller but significant degrees of proliferation in response to many different organisms. Our data offers insights into the maintenance of immunological memory. All clones examined were CD3+, CD4+, CD8-, TCRalphabeta+, and TCRdelta-. The ratio of TCRalphabeta+ to TCRdelta+ cells among peripheral blood lymphocytes increased during polyclonal culture in the presence of parasite. The high frequency of such cells in peripheral blood (1/800 - 1/9000), and their response to a wide range of geographically different Plasmodium falciparum isolates and clones by both proliferation and lymphokine secretion (predominantly IFN-gamma) with a high degree of sensitivity (<1 parasite/mul blood in some cases) suggests that these cells must be quickly activated following malaria infection. Their contribution to the outcome of the disease (protection/immunopathology) may be significant.
Article
The cellular basis of immunity to sporozoites was investigated by examining the effect of immunization of T and B cell-deficient C57BL/6N × BALB/c AnN F1 (BLCF1) mice compared to immunocompetent controls. Immunization of T cell-deficient (ATX-BM-ATS) BLCF1 mice with x-irradiated sporozoites did not result in the generation of protective immunity. The same immunization protocols protected all immunocompetent controls. In contrast, B cell-deficient (μ-suppressed) BLCF1 mice were protected by immunization in the majority of cases. The absence of detectable serum circumsporozoite precipitins or sporozoite neutralizing activity in the μ-suppressed mice that resisted a sporozoite challenge suggests a minor role for these humoral factors in protection. These data demonstrate a preeminent role for T cells in the induction of protective immunity in BLCF1 mice against a P. berghei sporozoite infection.
Article
An in vitro assay which measures antigen-induced proliferation of primed murine lymph node cells is described. The response is mediated by T cells since it can be obtained by using nylon wool-passed lymphocytes (< 1% Ig+ cells) and it can be abolished by treatment with anti-Thy 1.2 and C. Furthermore, LN cells from nu/nu mice injected with antigen do not demonstrate antigen-induced proliferation in contrast to the response observed in euthymic littermate controls. Other relevant parameters of this proliferative assay include the observations that the response is highly antigen specific, can be seen as early as 4 days and as late as 60 days after in vivo priming, is restricted to the use of certain sets of LN when animals are injected subcutaneously at the base of the tail, and can be seen with LN cells from mice primed with antigen in either CFA or ICFA. The ease of the assay coupled with its specificity and quantitative dimensions provides a direct and simple method to evaluate processes involved in antigen-induced murine T lymphocyte activation.
Article
A CD4+ T cell clone (A1.6) was derived from spleen cells of mice immunized with irradiated sporozoites. This T cell clone recognizes an antigen that is shared by sporozoites and blood forms of Plasmodium berghei and differs from the circumsporozoite protein. Clone A1.6 displays cytotoxic activity, produces IFN-gamma and IL-2 in vitro, and recognizes the plasmodial antigen in the context of the class II I-Ed molecule. Passive transfer of this CD4+ clone into naive mice resulted in a high degree of protection against sporozoite challenge.
Article
Summary Addition of recombinant inlcrleukin-6 (IL-6) to Plasmodium yoelii hepatic cultures resulted in a specific dose-dependent inhibition of parasite development. Time course experiments showed that, without any direct effect on free sporozoites, IL-6 exerts its action during both the early phase of infection and during the subsequent maturation of the schizonts. Elicitation of the oxidative burst appears lo be one mechanism by which IL-6 interferes with the development of hepatic phase. Catalase and superoxide dismutase, two scavengers of hydrogen peroxide and superoxide anions, reversed the IL-6 mediated parasiticidal activity.
Article
The effect of iron (Fe3+) and normal human liver ferritin on the proliferative response of normal human lymphocytes to tetanus toxoid was examined. This proliferative response involved memory T4+ lymphocytes as shown by a selective depletion study. Limit dilution analysis revealed that iron, present as ferric citrate, affected the initiation of clone development, and that concentrations of ferric citrate from 30 microM to 1 nM were able to reduce significantly the cloning efficiency of precursor T cells (up to 90% reduction). The reduced cloning frequency was not due to immunological suppression. Clone size was also reduced when iron was present during culture. In contrast, the presence of normal human liver ferritin during culture (concentration range: 300 micrograms/1-10,000 micrograms/1) had no effect on lymphocyte proliferation. The data indicate that low molecular weight iron (as ferric citrate) in concentrations similar to those which have been reported in the serum of patients with iron overload diseases, can interfere with antigen-specific lymphocyte responses and this may have implications for the development of infections and neoplasia in diseases of iron-overload.
Article
An in vitro assay which measures antigen-induced proliferation of primed murine lymph node cells is described. The response is mediated by T eclls since it can be obtained by using nylon wool-passed lymphocytes (less than 1% Ig+ cells) and it can be abolished by treatment with anti-Thy 1.2 and C. Furthermore, LN cells from nu/nu mice injected with antigen do not demonstrate antigen-induced proliferation in contrast to the response observed in euthymic littermate controls. Other relevant parameters of this proliferative assay include the observations that the response is highly antigen specific, can be seen as early as 4 days and as late as 60 days after in vivo priming, is restricted to the use of certain sets of LN when animals are injected subcutaneously at the base of the tail, and can be seen with LN cells from mice primed with antigen in either CFA or ICFA. The ease of the assay coupled with its specificity and quantitative dimensions provides a direct and simple method to evaluate processes involved in antigen-induced murine T lymphocyte activation.
Article
The cellular basis of immunity to sporozoites was investigated by examing the effect of immunization of T and B cell-deficient C57BL/6N X BALB/c AnN F1 (BLCF1) mice compared to immunocompetent controls. Immunization of T cell-deficient (ATX-BM-ATS) BLCF1 mice with x-irradiated sporozoites did not result in the generation of protective immunity. The same immunization protocols protected all immunocompetent controls. In contrast, B cell-deficient (micron-suppressed) BLCF1 mice were protected by immunization in the majority of cases. The absence of detectable serum circumsporozoite precipitins or sporozoite neutralizing activity in the micron-suppressed mice that resisted a sporozoite challenge suggests a minor role for these humoral factors in protection. These data demonstrate a preeminent role for T cells in the induction of protective immunity in BLCF 1 mice against a P. berghei sporozoite infection.
Article
Human peripheral blood lymphocytes were separated into subpopulations enriched or depleted with respect to B lymphocytes (Ig-bearing cells), T lymphocytes, (cells forming rosettes with sheep erythrocytes: E-RFC) and Fc receptor-bearing lymphocytes (EA-RFC). From the distributions and recoveries of the various cell types it could be concluded that there was very little overlap between Ig-bearing lymphocytes and EA-RFC. The latter cells partly belonged to “null” (non-T, non-B) cells; it was however demonstrated that 30 % of the EA-RFC were T cells (E-RFC). The lytic capacity in antibody-dependent lymphocytotoxicity (ADL) was shown to correspond with the proportions of EA-RFC in the various fractions. Non-T cells showed enhanced ADL activity when compared to the unseparated cells. Purified T cell populations also displayed ADL activity. Since the latter could not be due to contaminating non-T cells, this activity was ascribed to Fc receptor-bearing T lymphocytes.
Article
Mice can be immunized to Plasmodium vinckei by repeated infections followed by cure. Such immunity is dependent on CD4 T cells and an architecturally modified spleen, but has little requirement for antibody. Thus, athymic mice can be exposed to P. vinckei and cured, but do not develop immunity. They are resistant to challenge with parasites, however, if they are then given spleen cells from euthymic immunized animals. Such immune spleen cells, however, cannot transfer resistance to normal mice which have been exposed to BCG, Salmonella typhimurium, or vaccinia virus, and are only partially effective in transferring resistance to mice which have been previously immunized with heterologous plasmodia, P. yoelii, P. chabaudi and P. berghei. Mice exposed to varying numbers of irradiated P. vinckei-pRBC do not develop immunity and nor are such animals protected following adoptive transfer of immune spleen cells. Cellular immunity to malaria may not only be dependent on a population of immune CD4 T cells, but may require a specifically architecturally modified spleen which may not occur following either exposure to candidate vaccine vectors, heterologous plasmodia or non-viable homologous plasmodia.