ArticlePDF Available

Cross-Recognition of Avian H5N1 Influenza Virus by Human Cytotoxic T-Lymphocyte Populations Directed to Human Influenza A Virus

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

Since the number of human cases of infection with avian H5N1 influenza viruses is ever increasing, a pandemic outbreak caused by these viruses is feared. Therefore, in addition to virus-specific antibodies, there is considerable interest in immune correlates of protection against these viruses, which could be a target for the development of more universal vaccines. After infection with seasonal influenza A viruses of the H3N2 and H1N1 subtypes, individuals develop virus-specific cytotoxic T-lymphocyte responses, which are mainly directed against the relatively conserved internal proteins of the virus, like the nucleoprotein (NP). Virus-specific cytotoxic T lymphocytes (CTL) are known to contribute to protective immunity against infection, but knowledge about the extent of cross-reactivity with avian H5N1 influenza viruses is sparse. In the present study, we evaluated the cross-reactivity with H5N1 influenza viruses of polyclonal CTL obtained from a group of well-defined HLA-typed study subjects. To this end, the recognition of synthetic peptides representing H5N1 analogues of known CTL epitopes was studied. In addition, the ability of CTL specific for seasonal H3N2 influenza virus to recognize the NP of H5N1 influenza virus or H5N1 virus-infected cells was tested. It was concluded that, apart from some individual epitopes that displayed amino acid variation between H3N2 and H5N1 influenza viruses, considerable cross-reactivity exists with H5N1 viruses. This preexisting cross-reactive T-cell immunity in the human population may dampen the impact of a next pandemic.
Content may be subject to copyright.
JOURNAL OF VIROLOGY, June 2008, p. 5161–5166 Vol. 82, No. 11
0022-538X/08/$08.000 doi:10.1128/JVI.02694-07
Copyright © 2008, American Society for Microbiology. All Rights Reserved.
Cross-Recognition of Avian H5N1 Influenza Virus by Human
Cytotoxic T-Lymphocyte Populations Directed to Human
Influenza A Virus
J. H. C. M. Kreijtz, G. de Mutsert, C. A. van Baalen, R. A. M. Fouchier,
A. D. M. E. Osterhaus, and G. F. Rimmelzwaan*
Department of Virology, Erasmus Medical Center, P.O. Box 2040, Rotterdam, The Netherlands
Received 19 December 2007/Accepted 4 March 2008
Since the number of human cases of infection with avian H5N1 influenza viruses is ever increasing, a
pandemic outbreak caused by these viruses is feared. Therefore, in addition to virus-specific antibodies, there
is considerable interest in immune correlates of protection against these viruses, which could be a target for
the development of more universal vaccines. After infection with seasonal influenza A viruses of the H3N2 and
H1N1 subtypes, individuals develop virus-specific cytotoxic T-lymphocyte responses, which are mainly directed
against the relatively conserved internal proteins of the virus, like the nucleoprotein (NP). Virus-specific
cytotoxic T lymphocytes (CTL) are known to contribute to protective immunity against infection, but knowledge
about the extent of cross-reactivity with avian H5N1 influenza viruses is sparse. In the present study, we
evaluated the cross-reactivity with H5N1 influenza viruses of polyclonal CTL obtained from a group of
well-defined HLA-typed study subjects. To this end, the recognition of synthetic peptides representing H5N1
analogues of known CTL epitopes was studied. In addition, the ability of CTL specific for seasonal H3N2
influenza virus to recognize the NP of H5N1 influenza virus or H5N1 virus-infected cells was tested. It was
concluded that, apart from some individual epitopes that displayed amino acid variation between H3N2 and
H5N1 influenza viruses, considerable cross-reactivity exists with H5N1 viruses. This preexisting cross-reactive
T-cell immunity in the human population may dampen the impact of a next pandemic.
Since the first documentation of bird-to-human transmis-
sions of highly pathogenic avian H5N1 influenza viruses, these
viruses have spread from Southeast Asia to other regions of the
world (2, 3, 22, 27). Since 2003 the number of human cases has
continued to increase; as of 28 February 2008, 369 human cases
have been reported, of which 234 were fatal (28). It is feared
that an H5N1 virus may cause the next influenza pandemic
when it is able to replicate in mammalian species by adaptation
through genetic reassortment or accumulation of point muta-
tions in relevant gene segments (11). Although neuraminidase
subtype 1 cross-reactive antibodies have been demonstrated in
human subjects, antibodies to H5 molecules are hardly existent
in the human population as a result of limited exposure to
H5N1 viruses, which contributes to a scenario for these viruses
to become pandemic (20). In general, the exposure history and
the immune status of the human population will influence the
size and the severity of pandemics (4, 8, 9, 14). The presence of
T-cell immunity induced by infection with human influenza
virus strains may provide some degree of cross-protective im-
munity against the H5N1 viruses. Cytotoxic T-lymphocyte
(CTL) responses are predominantly directed to internal viral
proteins, the nucleoprotein (NP) in particular (23, 29),
which is much more conserved than the surface hemagglu-
tinin and neuraminidase glycoproteins (5, 13, 15, 29). It has
been suggested that cross-reactive CD8
T cells may temper
the impact a pandemic potentially could have on the human
population (9, 14, 19). In humans, the presence of cross-
reactive CTL responses inversely correlated with the
amount of shedding of a heterosubtypic strain that was used
for experimental infection of study subjects (14). Although
preexisting CTL immunity against influenza virus may be of
importance in the face of the current H5N1 pandemic
threat, our knowledge of the cross-reactive nature of the
human CTL response is limited (9).
In the present study we tested the cross-reactivity of poly-
clonal virus-specific CD8
T-cell populations obtained from
well-defined HLA-typed study subjects with H5N1 virus. The
recognition of target cells pulsed with peptide variants, trans-
fected with the NP gene from a human or an avian influenza
virus, or infected with viruses of the H3N2 or H5N1 subtype
was tested.
It was concluded that the human CTL response displays a
high degree of cross-reactivity with avian H5N1 influenza vi-
ruses and could reduce morbidity and mortality during a pan-
demic caused by these H5N1 strains.
MATERIALS AND METHODS
Cells. Peripheral blood mononuclear cells (PBMC) were isolated from hepa-
rinized blood obtained from fifteen HLA-typed healthy blood donors (Sanquin
Bloodbank, Rotterdam, The Netherlands) by density gradient centrifugation
using lymphoprep (Axis-Shield PoC AS, Oslo, Norway) and then cryo-preserved
at 135°C. Genetic subtyping was performed in the laboratory for Histocom-
patibility and Immunogenetics at the Sanquin Bloodbank using a commercial
typing system (Genovision, Vienna, Austria). Three groups of study subjects
were selected on the basis of their major histocompatibility complex class I
(MHC-I) alleles, for which influenza CTL epitopes were identified. Within
groups the subjects shared identical HLA-A and -B alleles; between groups there
* Corresponding author. Mailing address: Department of Virol-
ogy, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam,
The Netherlands. Phone: 31 10 7044243. Fax: 31 10 7044760. E-
mail: g.rimmelzwaan@erasmusmc.nl.
Published ahead of print on 19 March 2008.
5161
were differences of one or two alleles. The groups were as follows: group I, HLA
A*0101, A*0201, B*0801, and B*3501; group II, HLA A*0101, A*0201, B*0801,
and B*2705 (2702); and group III, HLA A*0101, A*0301, B*0801, and B*3501
(3503) (1). Subject 15 was not tested since PBMC of this donor were no longer
available.
Peptides. Amino acid sequences of all known human influenza A virus CTL
epitopes were compared with their counterparts in H5N1 influenza viruses iso-
lated since 2003, which were obtained from the influenza sequence database (12).
All possible variants that could be identified in the H5N1 sequences are listed in
Table 1. A set of immunograde peptides representing immunodominant CTL
epitopes and the most prevalent analogues in H5N1 strains were synthesized and
analyzed by mass spectrometry and were found to be 70% pure (Eurogentec,
Seraing, Belgium). Variant peptide analogues from the NP, which is the main
target for CTL responses, were synthesized when they had a prevalence in H5N1
strains of 0.25%, with the exception of the NP
383-391
epitope since the G384K
mutation observed in H5N1 viruses was known to abrogate recognition by spe-
cific CTLs completely (26). For the remaining viral proteins, all variants with a
prevalence of 2.25% were synthesized and tested. Only peptides that matched
the HLA alleles of the study subjects were considered.
Target cells. B-lymphoblastoid cell lines (BLCL) were established as described
previously (18) and used as target or stimulator cells. A total of 30,000 cells were
incubated in the absence or presence of 10 M peptide for1hat37°C, washed
once, and resuspended in RPMI 1640 medium (Cambrex, East Rutherford, NJ)
containing antibiotics, L-glutamine, and 10% fetal calf serum (R10F medium).
Cells of the BLCL were also infected at a multiplicity of infection of five 50%
tissue culture infective doses/cell (17) with influenza viruses A/Netherlands/18/94
(A/NL/18/94) (H3N2) or A/Vietnam/1194/04 (A/VN/1194/04) (H5N1), which
were propagated and titrated in MDCK cells using standard procedures. After an
incubation period of1hat37°C, the cells were washed and resuspended in R10F
medium and incubated for 16 to 18 h at 37°C prior to their use for the stimulation
of CD8
T cells. Infection rates were determined by an immunofluorescence
assay and were similar for both viruses (data not shown). The human influenza
virus A/NL/18/94 (H3N2) was used as a representative of seasonal influenza
viruses, while influenza virus A/VN/1194/04 was used as an example for H5N1
influenza virus.
T-cell clones. CD8
T-cell clones directed against the HLA-A1-restricted
NP
44–52
(CTELKLSDY) epitope, HLA-A3-restricted NP
265–273
(ILRGSVAHK)
epitope, HLA-B27-restricted NP
174–184
(RRSGAAGAAVK) epitope, and the
HLA-B*3501-restricted NP
418–426
(LPFEKSTVM) epitope were generated as
described previously (26).
In vitro expansion of influenza A virus-specific T-cell populations. PBMC
were stimulated with influenza virus A/NL/18/94-infected cells as previously
described (1). Eight days after stimulation, cells were harvested and used as
effector cells in an enzyme-linked immunospot (ELISPOT) or fluorescent anti-
gen-transfected target cell (FATT)-CTL assay. For the ELISPOT assays, CD8
T cells were purified from the in vitro expanded PBMC by magnetic bead cell
sorting (Miltenyi Biotec, Bergisch Gladbach, Germany). Typically, a purity of
96% was obtained.
FATT-CTL assay. The NP genes of influenza viruses A/NL/18/94 and A/VN/
1194/04 without their stop codons were cloned into the plasmid pEGFP-N1
(Becton Dickinson, Alphen a/d Rijn, The Netherlands) in frame with the
open reading frame of the green fluorescent protein (GFP) as previously
described (25). Plasmid DNA was purified using a Genopure plasmid midi kit
(Roche, Woerden, The Netherlands). Nucleotide sequences of the recombi-
nant plasmids were confirmed using a Big Dye Terminator, version 3.1, cycle
sequencing kit (Applied Biosystems, Foster City, CA) and an ABI Prism 3100
Genetic Analyzer (Applied Biosystems). Primer and plasmid sequences are
available on request.
The plasmids were used in the FATT-CTL assay for the detection of lytic
activity of virus-specific CTLs as described previously (25). In brief, BLCL were
nucleofected using cell line nucleofector kit V (Amaxa Biosystems, Cologne,
Germany) with program T16 and subsequently incubated in R10F medium for
4 h at 37°C. Then, they were cocultured for another4hintriplicate with PBMC
cultures and in vitro expanded after stimulation with influenza virus A/NL/18/94
at various effector-to target (E:T) cell ratios. The number of viable GFP-positive
cells was measured using a FACSCalibur (Becton Dickinson). The percent nu-
cleoprotein-specific lysis was then calculated by the following formula: 100
[(number of viable GFP-positive cells in the sample without effector number
of viable GFP-positive cells in the sample with effector)/number of viable GFP-
positive cells in the sample without effector].
Gamma interferon (IFN-) assay. ELISPOT assays were performed with in
vitro expanded CD8
T cells as effector cells and peptide-pulsed or virus-
infected HLA-matched BLCL as stimulator cells as described previously (1). The
number of spots was determined using an ELISPOT reader and image analysis
software (Aelvis, Sanquin Reagents, Amsterdam, The Netherlands), and the
average number was calculated of triplicate wells.
RESULTS
Comparison of amino acid sequences of known influenza A
virus CTL epitopes. The amino acid sequences of known hu-
man influenza A virus CTL epitopes were compared with the
corresponding sequences in approximately 900 H5N1 viruses
obtained from the influenza sequence database (12). As shown
in Fig. 1A, the epitope sequences were identical in 95% of
the H5N1 viruses for the majority of the known epitopes an-
alyzed including PB1
591–599
,M1
13–21
,M1
128–135
, NS1
158–166
,
NP
44–52
,NP
146–154
,NP
174–184
,NP
265–273
,NP
380–388,
NP
381–388
,
and NP
383–391
. For some of the other epitopes the percentage
TABLE 1. Variant sequences of known CTL epitopes in H5N1 viruses
Sequence
name
Variant sequence of the CTL epitope at the indicated HLA allele
a
HLA-A1 PB1
(591–599)
HLA-A3 M1
(13–21)
HLA-A*0201
M1 (58–66)
HLA-B*3501
M1 (128–135)
HLA-A*0201
NS1 (122–130)
HLA-A*0201
NS1 (123–132)
HLA-B*44
NS1 (158–166)
HLA-A1 NP
(44–52)
HLA-A*6801
NP (91–99)
Epitope VSDGGPNLY SIIPSGPLK GILGFVFTL ASCMGLIY AIMDKNIIL IMDKNIILKA GEISPLPSL CTELKLSDY KTGGPIYKR
Variant
1I-------- -V------- -M------- ------S- -----T--- ----T----- ------H-- --------H -------R-
2-------P- --V------ --W------ -----V--- ----V----- -------F- --------Q -----V---
3------IP- F-------- -------T- ------T--- ---L----- ------N-- -----F-RG
4G-------- -T------- -----TV-- ----TV---- --------I ------T-- --------G
5-A------- -L------- -----A--- ----A----- -----S--- ---F-----
6ATS------ -T-----T- T-----T--- -------Y-
7--VR----Q -----D--- ----D----- A--------
8--VL----- --V--T--- -V--T----- -----I---
9S----T--- ----T-S---
10 -----T-S- --N-------
11 ---N----- -V--T-T-- -
12 --V- -T-T- -V----T---
13 --V- -- -T- -- --T-L- --
14 -----T-L- ----I-----
15 -----I--- -V--TV----
16 --V- -TV-- - -- -T--W- -
17 -----T--W ----T----G
a
Variant sequences of known CTL epitopes were ranked according to their relative prevalence. Anchor residues of the epitopes are underlined. Amino acid residue
positions are given in parentheses.
5162 KREIJTZ ET AL. J. VIROL.
of H5N1 viruses with identical sequences was variable and
ranged from 79% for epitope M1
58–66
to 4% for epitope
NS1
122–130
. For the epitopes NP
91–99
,NP
188–198
,NP
339–347
,
and NP
418–426
, no identical sequences were found in the H5N1
viruses. In order to identify the most prevalent variant se-
quences in H5N1 viruses, the number of individual variants
was analyzed (Fig. 1B). In some cases a single variant was
identified that accounted for almost all variant sequences ob-
served in H5N1 viruses (Table 1 and Fig. 1B). For other
epitopes multiple variants were identified, although for some
of these the number was low, and the number of major variants
was limited (12).
The recognition of known CTL epitopes and their avian
analogues. All subjects in group I (HLA A*0101, A*0201,
B*0801, and B*3501) displayed T-cell reactivity with the
epitopes NP
44–52
, NS1
122–130
,NP
418–426
, and M1
58–66
as they
are present in human influenza A viruses although the fre-
quency of specific CTLs varied between study subjects and the
peptides tested (Fig. 2A). In none of the subjects of this group
was reactivity observed with the peptide variants of epitopes
NP
44–52
and NS1
122–130
, obtained from H5N1 influenza vi-
ruses. Three out of four subjects responded to the NP
418–426
variant LPFERSTIM, and all subjects responded to the
M1
58–66
variant GMLGFVFTL. Of group II (HLA A*0101,
A*0201, B*0801, and B*2705 [2702]), most subjects responded
to the peptides representing epitopes from human influenza A
viruses (Fig. 2B) although the magnitudes of the responses
varied considerably. Only one subject in this group had an
appreciable response to the H5N1 analogue sequence of the
NS1
122–130
epitope. Four out of five subjects responded to the
H5N1 variants of the HLA B*2705-restricted NP
174–184
epitope whereas all five responded to the M1
58–66
variant. The
subjects of group III (HLA A*0101, A*0301, B*0801, and
B*3501 [3503]) responded to the original epitopes to various
extents. Some subjects were poor responders and hardly dis-
played CTL reactivity with some of these epitopes (Fig. 2C).
However, the in vitro expanded PBMC of subject 14 responded
strongly to the NP
418–426
and also reacted with both epitope
variants from H5N1 viruses, indicating that at least a fraction
of the CTL population was capable of cross-recognizing these
analogues. The same holds true for the NP
44–52
- and NP
265–273
-
specific CTL responses in this study subject. Clones were used
as positive and negative controls, and the clonal responses
supported the results obtained with the polyclonal populations
(data not shown).
FIG. 1. The presence of known CTL epitopes in H5N1 strains. The
percentage of H5N1 viruses with an epitope sequence identical to
human influenza viruses (white bars) is shown. The black bars indicate
the percentage of H5N1 viruses with one or more amino acid substi-
tutions in the epitope sequence. The absolute numbers of each variant
of an epitope are shown in panel B, where each color represents a
single variant (sequences can be found in Table 1). For this analysis
almost 900 H5N1 viruses for which sequence information was available
in the influenza sequence database (12) were analyzed.
TABLE 1—Continued
Variant sequence of the CTL epitope at the indicated HLA allele
a
HLA-B*1402
NP (146–154)
HLA-B*2705
NP (174–184)
HLA-A3
NP (188–198)
HLA-A3
NP (265–273)
HLA-B*3701
NP (339–347)
HLA-B*08
NP (380–388)
HLA-B*2702
NP (381–388)
HLA-B*2705
NP (383–391)
HLA-B*3501
NP (418–426)
TTYQRTRAL RRSGAAGAAVK TMVMELVRMIK ILRGSVAH EDLRVLSFI ELRSRYWAI LRSRYWAI SRYWAIRTR LPFEKSTVM
S-------- ---------I- ------I---- -----I--- -----S--- ----K---- ---K---- -K------- ----RA-I-
A-------- -I------L-- --A-------- -----M--- ----IS--- D-LGK---K -LGK---K ------K-- ----R--I-
AA------- --F-------- -----QI---- V-------- -----H--- ----H--- -------P- ----RAAI-
A--H----- Q----VI---- --------K ----RV-I-
AP------- --------P ----RA---
A-------V GK-- -KWMI
A-SQ----- --H------
--------G
VOL. 82, 2008 CROSS-RECOGNITION OF AVIAN H5N1 VIRUS BY HUMAN CTLs 5163
Cross-recognition of the NP derived from influenza virus
A/VN/1194/04. The capacity of polyclonal T-cell populations
directed to the human influenza virus A/NL/18/94 to cross-
react with the NP of influenza virus A/VN/1194/04 was as-
sessed in the FATT-CTL assay. PBMC from all study subjects
were stimulated with influenza virus A/NL/18/94 and allowed
to proliferate. As shown in Fig. 3, 2 out of the 15 subjects tested
(subjects 5 and 8) were low responders or nonresponders (Fig.
3) since no NP-specific lytic activity could be demonstrated. In
the remaining 13 subjects, lytic activity was observed against the
homologous NP. In most cases the PBMC cross-reacted with the
NP of influenza virus A/VN/1194/04 to a considerable extend
(Fig. 3). Only for subject 3 (group I) and subject 9 (group III) did
the influenza virus A/NL/18/94 NP-specific CTLs fail to recognize
the NP of influenza virus A/VN/1194/04 (Fig. 3).
Cross-recognition of BLCL infected with influenza virus
A/VN/1194/04. Next, we wished to assess the cross-reactive
nature of the whole repertoire of CD8
T lymphocytes specific
for the human influenza virus A/NL/18/94. To this end, PBMC
were stimulated with this virus, and after 8 days the CD8
cells
were isolated to obtain virus-specific polyclonal CTL populations.
These cells were used as effector cells in an IFN-ELISPOT
assay using MHC-I-matched BLCL infected with influenza virus
A/NL/18/94 or A/VN/1194/04 as stimulator cells.
As shown in Fig. 4, the in vitro expanded PBMC population
that recognized cells infected with influenza virus A/NL/18/94
also recognized cells infected with influenza virus A/VN/1194/
04. The average number of IFN--positive spots per 10
4
cells
observed after stimulation with A/NL/18/94-infected cells was
151 (standard deviation, 58), and the number observed after
stimulation with A/VN/1194/04 was even slightly higher at 192
(standard deviation, 65) although this difference was not sta-
tistically significant.
DISCUSSION
In the present paper the cross-reactive nature of the human
influenza virus-specific CTL response was investigated. It was
concluded that a considerable portion of CTL populations
specific for the H3N2 influenza virus A/NL/18/94 cross-reacted
with the H5N1 strain A/VN/1194/04.
For most CTL epitopes, it was found that a vast majority of
the H5N1 strains contained epitope sequences identical to
those present in human influenza A viruses. This conservation
of epitopes is responsible for the cross-reactive nature of CTL
responses in humans against seasonal influenza A viruses of
the H3N2 and H1N1 subtypes. However, some variation in
these epitopes was observed also, and for a number of CTL
epitopes the H5N1 strains did not contain identical sequences.
Apart from the NP
174–184
and M1
58–66
epitope restricted by
HLA-B*2705 and HLA-A*0201, respectively, very little cross-
reaction was observed of polyclonal CTL populations with
variant peptides derived from H5N1 viruses.
However, as indicated above, most epitopes are relatively
conserved, including those located in the NP, which contrib-
uted to the cross-reactive nature of the NP-specific CTL re-
sponse. Most of the study subjects that responded to NP de-
rived from seasonal H3N2 influenza viruses also responded to
the NP derived from influenza virus A/VN/1194/04 (H5N1).
The polyclonal virus-specific T-cell populations of two of these
subjects failed, however, to cross-react with the NP of influenza
virus A/VN/1194/04 for reasons that are unclear. Possibly, the
most immunodominant responses in these subjects were di-
rected to CTL epitopes in the NP that were not conserved.
To account for the full repertoire of virus-specific CD8
T
lymphocytes, the reactivity with MHC-I-matched cells infected
with influenza virus A/NL/18/94 or A/VN/1194/04 was also
analyzed. In all cases A/VN/1194/04-infected target cells were
recognized to a similar extent as A/NL/18/94-infected cells,
indicating that the level of cross-reactivity of human CTL re-
sponses to seasonal H3N2 influenza viruses with H5N1 strains
is substantial.
Thus, apart from some individual epitopes that display
amino acid sequence variation between H3N2 and H5N1 in-
fluenza A viruses, the level of cross-reactivity is considerable
FIG. 2. Epitope-specific IFN-production by CTLs after stimulation with peptide-pulsed BLCL. The number of IFN--producing cells per
10,000 CD8
T cells (5,000 cells for subject 3) from subjects from HLA groups I (A), II (B), and III (C) were measured by ELISPOT assay. CD8
T cells were isolated from PBMC populations expanded in vitro with influenza virus A/NL/18/94 and subsequently stimulated with peptide variants
as indicated. (*, peptide sequence of the known human influenza virus CTL epitopes).
5164 KREIJTZ ET AL. J. VIROL.
and does not seem to be influenced by the HLA phenotype of
the study subjects. The NP
383–391
epitope is present in H5N1
viruses but has disappeared from human H3N2 viruses during
decades of virus evolution (7, 26). Since a virus was used for
stimulation of the PBMC that does not contain this epitope,
this does not give a false sense of cross-reactivity in HLA-
B*2705-positive subjects. It is unknown whether the presence
of the NP
383–391
epitope interferes with the presentation of
other HLA-B*2705-restricted epitopes. Interference has been
observed only with the overlapping HLA-B*0801-restricted
NP
380–388
epitope (1, 24).
Although it is unknown to what extent preexisting T-cell
immunity can dampen the impact of a next influenza pan-
demic, it is speculated that the protective effect of cross-reac-
tive CTL responses has a beneficial effect on the outcome of
infection with new pandemic influenza virus strains. This spec-
ulation is supported by a number of different observations.
First, in animal models it has been shown that virus-specific
CTLs contribute to heterosubtypic immunity (6, 10, 16); sec-
ondly, it was found in 1957 that individuals that had experi-
enced documented infections with H1N1 influenza A viruses
were less likely to develop severe disease or succumb to infec-
tion with the pandemic strain of the H2N2 subtype (4). In this
respect it is of interest that during the current outbreak of
H5N1 infections in humans, younger individuals are especially
at risk for severe disease and a fatal outcome of infection (21).
It can be hypothesized that younger individuals are less likely
to have been exposed to seasonal influenza A viruses of the
H3N2 and H1N1 subtypes and thus have not mounted a (cross-
reactive) CTL response to an alternative subtype. However, it
cannot be excluded that confounding factors play a role in the
observed disproportionate age distribution of severe H5N1
human cases. Last but not least, the human CTL response
against epidemic strains is largely cross-reactive with H5N1
influenza virus strains, as was demonstrated in the present
study. Although these cross-reactive CTL populations may not
prevent infection with pandemic strains, they may contribute to
a certain degree of heterosubtypic immunity and facilitate a
more rapid clearance of the infection than in immunologically
naı¨ve individuals who lack cross-reactive T-cell populations.
This may determine the difference between life and death
during a pandemic outbreak. In addition, the induction of
cross-reactive CTL responses may be an attractive target for
the development of universal vaccines that could confer
broadly protective immunity against influenza viruses of vari-
ous subtypes.
FIG. 3. Recognition of NP derived from H3N2 and H5N1 influ-
enza virus by in vitro expanded PBMC specific for influenza virus
A/NL/18/94 (H3N2). The lytic activity of in vitro expanded PBMC was
tested with MHC-I matched BLCL nucleofected with NP-GFP coding
plasmid (NP of either influenza virus A/NL/18/94 [black circles] or
A/VN/1194/04 [open circles]) or empty control GFP plasmid (gray
circles). The NP-specific lytic activity was tested with PBMC of the
subjects from group I (subjects 1 to 4), II (subjects 5 to 9), and III
(subjects 10 to 14 and 16). E:T ratios were 0, 3.125, 6.25, 12.5, 25, and
50 for all subjects except numbers 13 and 14, for whom E:T ratios were
0, 0.3, 1, 3, 10 and 30. The lytic activities against control plasmid-
transfected target cells are not visible for subjects 13 and 14 as a result
of negative values for the percentages of specific lysis, which were
caused by slight increases in the number of GFP-positive viable cells.
Standard deviation of the means was 10%.
FIG. 4. Recognition of influenza virus-infected BLCL by CTLs.
The number of IFN--producing cells per 10,000 CD8
T cells was
measured by ELISPOT assay after stimulation with BLCL infected
with influenza virus A/NL/18/94 or A/VN/1194/04. Each symbol rep-
resents an individual subject from group I (A), II (B), or III (C).
Uninfected BLCL were used as negative controls. The horizontal
bars represent the average responses of all study subjects in groups
I, II, and III.
VOL. 82, 2008 CROSS-RECOGNITION OF AVIAN H5N1 VIRUS BY HUMAN CTLs 5165
ACKNOWLEDGMENTS
This study was conducted under the auspices of The Netherlands
Influenza Vaccine Research Center and financially supported in part
by The Netherlands Organization for Health Research and Develop-
ment (ZonMW; grant 91402008).
We thank T. Bestebroer and C. Baas for outstanding technical
assistance.
REFERENCES
1. Boon, A. C., G. de Mutsert, Y. M. Graus, R. A. Fouchier, K. Sintnicolaas,
A. D. Osterhaus, and G. F. Rimmelzwaan. 2002. The magnitude and speci-
ficity of influenza A virus-specific cytotoxic T-lymphocyte responses in hu-
mans is related to HLA-A and -B phenotype. J. Virol. 76:582–590.
2. Claas, E. C., A. D. Osterhaus, R. van Beek, J. C. De Jong, G. F. Rimmel-
zwaan, D. A. Senne, S. Krauss, K. F. Shortridge, and R. G. Webster. 1998.
Human influenza A H5N1 virus related to a highly pathogenic avian influ-
enza virus. Lancet 351:472–477.
3. de Jong, J. C., E. C. Claas, A. D. Osterhaus, R. G. Webster, and W. L. Lim.
1997. A pandemic warning? Nature 389:554.
4. Epstein, S. L. 2006. Prior H1N1 influenza infection and susceptibility of
Cleveland Family Study participants during the H2N2 pandemic of 1957: an
experiment of nature. J. Infect. Dis. 193:49–53.
5. Fleischer, B., H. Becht, and R. Rott. 1985. Recognition of viral antigens by
human influenza A virus-specific T lymphocyte clones. J. Immunol. 135:
2800–2804.
6. Flynn, K. J., G. T. Belz, J. D. Altman, R. Ahmed, D. L. Woodland, and P. C.
Doherty. 1998. Virus-specific CD8
T cells in primary and secondary influ-
enza pneumonia. Immunity 8:683–691.
7. Gog, J. R., G. F. Rimmelzwaan, A. D. Osterhaus, and B. T. Grenfell. 2003.
Population dynamics of rapid fixation in cytotoxic T lymphocyte escape
mutants of influenza A. Proc. Natl. Acad. Sci. USA 100:11143–11147.
8. Jameson, J., J. Cruz, and F. A. Ennis. 1998. Human cytotoxic T-lymphocyte
repertoire to influenza A viruses. J. Virol. 72:8682–8689.
9. Jameson, J., J. Cruz, M. Terajima, and F. A. Ennis. 1999. Human CD8
and
CD4
T lymphocyte memory to influenza A viruses of swine and avian
species. J. Immunol. 162:7578–7583.
10. Kreijtz, J. H., R. Bodewes, G. van Amerongen, T. Kuiken, R. A. Fouchier,
A. D. Osterhaus, and G. F. Rimmelzwaan. 2007. Primary influenza A virus
infection induces cross-protective immunity against a lethal infection with a
heterosubtypic virus strain in mice. Vaccine 25:612–620.
11. Kuiken, T., E. C. Holmes, J. McCauley, G. F. Rimmelzwaan, C. S. Williams,
and B. T. Grenfell. 2006. Host species barriers to influenza virus infections.
Science 312:394–397.
12. Macken, C., H. Lu, J. Goodman, and L. Boykin. 2001. The value of a
database in surveillance and vaccine selection. Elsevier Science, Amsterdam,
The Netherlands.
13. McMichael, A. 1994. Cytotoxic T lymphocytes specific for influenza virus.
Curr. Top. Microbiol. Immunol. 189:75–91.
14. McMichael, A. J., F. M. Gotch, G. R. Noble, and P. A. Beare. 1983. Cytotoxic
T-cell immunity to influenza. N. Engl. J. Med. 309:13–17.
15. McMichael, A. J., C. A. Michie, F. M. Gotch, G. L. Smith, and B. Moss. 1986.
Recognition of influenza A virus nucleoprotein by human cytotoxic T lym-
phocytes. J. Gen. Virol. 67:719–726.
16. O’Neill, E., S. L. Krauss, J. M. Riberdy, R. G. Webster, and D. L. Woodland.
2000. Heterologous protection against lethal A/HongKong/156/97 (H5N1)
influenza virus infection in C57BL/6 mice. J. Gen. Virol. 81:2689–2696.
17. Rimmelzwaan, G. F., M. Baars, E. C. Claas, and A. D. Osterhaus. 1998.
Comparison of RNA hybridization, hemagglutination assay, titration of in-
fectious virus and immunofluorescence as methods for monitoring influenza
virus replication in vitro. J. Virol. Methods 74:57–66.
18. Rimmelzwaan, G. F., N. Nieuwkoop, A. Brandenburg, G. Sutter, W. E. Beyer,
D. Maher, J. Bates, and A. D. Osterhaus. 2000. A randomized, double blind
study in young healthy adults comparing cell mediated and humoral immune
responses induced by influenza ISCOM vaccines and conventional vaccines.
Vaccine 19:1180–1187.
19. Rimmelzwaan, G. F., and A. D. Osterhaus. 1995. Cytotoxic T lymphocyte
memory: role in cross-protective immunity against influenza? Vaccine 13:
703–705.
20. Sandbulte, M. R., G. S. Jimenez, A. C. Boon, L. R. Smith, J. J. Treanor, and
R. J. Webby. 2007. Cross-reactive neuraminidase antibodies afford partial
protection against H5N1 in mice and are present in unexposed humans.
PLoS Med. 4:e59.
21. Smallman-Raynor, M., and A. D. Cliff. 2007. Avian influenza A (H5N1) age
distribution in humans. Emerg. Infect. Dis. 13:510–512.
22. Subbarao, K., A. Klimov, J. Katz, H. Regnery, W. Lim, H. Hall, M. Perdue,
D. Swayne, C. Bender, J. Huang, M. Hemphill, T. Rowe, M. Shaw, X. Xu, K.
Fukuda, and N. Cox. 1998. Characterization of an avian influenza A (H5N1)
virus isolated from a child with a fatal respiratory illness. Science 279:393–
396.
23. Townsend, A. R., and J. J. Skehel. 1984. The influenza A virus nucleoprotein
gene controls the induction of both subtype specific and cross-reactive cyto-
toxic T cells. J. Exp. Med. 160:552–563.
24. Tussey, L. G., S. Rowland-Jones, T. S. Zheng, M. J. Androlewicz, P. Cress-
well, J. A. Frelinger, and A. J. McMichael. 1995. Different MHC class I
alleles compete for presentation of overlapping viral epitopes. Immunity
3:65–77.
25. van Baalen, C. A., D. Kwa, E. J. Verschuren, M. L. Reedijk, A. C. Boon, G.
de Mutsert, G. F. Rimmelzwaan, A. D. Osterhaus, and R. A. Gruters. 2005.
Fluorescent antigen-transfected target cell cytotoxic T lymphocyte assay for
ex vivo detection of antigen-specific cell-mediated cytotoxicity. J. Infect. Dis.
192:1183–1190.
26. Voeten, J. T., T. M. Bestebroer, N. J. Nieuwkoop, R. A. Fouchier, A. D.
Osterhaus, and G. F. Rimmelzwaan. 2000. Antigenic drift in the influenza A
virus (H3N2) nucleoprotein and escape from recognition by cytotoxic T
lymphocytes. J. Virol. 74:6800–6807.
27. World Health Organization. 2007. Affected areas with confirmed cases of H5N1
avian influenza since 2003, status as of 17.10.2007. http://gamapserver.who.int
/mapLibrary/Files/Maps/Global_H5N1inHumanCUMULATIVE_FIMS_2007
1017.png. Accessed 26 October 2007.
28. World Health Organization. 2007. Cumulative number of confirmed human
cases of avian influenza A/(H5N1) reported to WHO. http://www.who.int/csr
/disease/avian_influenza/country/cases_table_2008_02_28/en/index.html. Ac-
cessed 3 March 2008.
29. Yewdell, J. W., J. R. Bennink, G. L. Smith, and B. Moss. 1985. Influenza A
virus nucleoprotein is a major target antigen for cross-reactive anti-influenza
A virus cytotoxic T lymphocytes. Proc. Natl. Acad. Sci. USA 82:1785–1789.
5166 KREIJTZ ET AL. J. VIROL.
... 21 Thirdly, the response elicited should activate cross-reactive CD8 + T cells that may offer superior protection against mutations and distinct influenza virus strains. 21,[31][32][33][34][35][36][37] CD8 + T cell cross-reactivity offers an advantage against viral mutation within the same strain of a virus, but also can provide immunity across strains. For example, several studies have shown that T cells specific for seasonal commoncold causing coronaviruses could provide a level of pre-existing immunity towards SARS-CoV-2, the causative agent of the ongoing COVID-19 pandemic. ...
... CD8 + T cells capable of cross-reacting towards homologous peptides could provide a level of protection towards distinct virus strains. Indeed, cross-reactive CD8 + T cells have been shown to recognise and respond to distinct influenza virus strains 21,28,[31][32][33][34][35][36][37] further supporting that these homologous peptides, and perhaps specifically the NP 265-IAV peptide, could be a good target in vaccination strategies. ...
Article
Full-text available
Objective: Influenza A, B and C viruses (IAV, IBV and ICV, respectively) circulate globally, infecting humans and causing widespread morbidity and mortality. Here, we investigate the T cell response towards an immunodominant IAV epitope, NP265-273, and its IBV and ICV homologues, presented by HLA-A*03:01 molecule expressed in ~ 4% of the global population (~ 300 million people). Methods: We assessed the magnitude (tetramer staining) and quality of the CD8+ T cell response (intracellular cytokine staining) towards NP265-IAV and described the T cell receptor (TCR) repertoire used to recognise this immunodominant epitope. We next assessed the immunogenicity of NP265-IAV homologue peptides from IBV and ICV and the ability of CD8+ T cells to cross-react towards these homologous peptides. Furthermore, we determined the structures of NP265-IAV and NP323-IBV peptides in complex with HLA-A*03:01 by X-ray crystallography. Results: Our study provides a detailed characterisation of the CD8+ T cell response towards NP265-IAV and its IBV and ICV homologues. The data revealed a diverse repertoire for NP265-IAV that is associated with superior anti-viral protection. Evidence of cross-reactivity between the three different influenza virus strain-derived epitopes was observed, indicating the discovery of a potential vaccination target that is broad enough to cover all three influenza strains. Conclusion: We show that while there is a potential to cross-protect against distinct influenza virus lineages, the T cell response was stronger against the IAV peptide than IBV or ICV, which is an important consideration when choosing targets for future vaccine design.
... Targeting more conserved sections of the influenza virus that can induce CD8 + T-cell responses towards antigenically distinct strains, along with B-cell and antibody responses, may be one way to achieve this. 21,77 Following activation by a pathogen-derived peptide, CD8 + T cells can produce cytolytic molecules such as granzymes and perforins that can directly destroy infected cells. 78,79 Furthermore, CD8 + T cells also produce cytokines such as IFNc and TNF, which recruit neighbouring immune cells to the site of infection to assist in viral clearance. ...
Article
Full-text available
Seasonal influenza viruses continue to cause severe medical and financial complications annually. Although there are many licenced influenza vaccines, there are billions of cases of influenza infection every year, resulting in the death of over half a million individuals. Furthermore, these figures can rise in the event of a pandemic, as seen throughout history, like the 1918 Spanish influenza pandemic (50 million deaths) and the 1968 Hong Kong influenza pandemic (~4 million deaths). In this review, we have summarised many of the currently licenced influenza vaccines available across the world and current vaccines in clinical trials. We then briefly discuss the important role of CD8 ⁺ T cells during influenza infection and why future influenza vaccines should consider targeting CD8 ⁺ T cells. Finally, we assess the current landscape of known immunogenic CD8 ⁺ T‐cell epitopes and highlight the knowledge gaps required to be filled for the design of rational future influenza vaccines that incorporate CD8 ⁺ T cells.
... The predominant H3N2 NP-specific T cell responses during seasonal and pandemic flu outbreaks during 2006 to 2010 were associated with robust cross-protection in the absence of protective antibody responses [3]. A cross-reactive cluster of differentiation 8 positive T-lymphocyte (CD8 + T cell) response has a significant protective role in heterologous clinical and preclinical prime/challenge studies between H1N1, H7N7, H5N1, and H3N2 influenza viruses [5][6][7][8]. Furthermore, a universal influenza vaccine candidate in pigs mitigated the lung pathology and reduced the nasal and lung viral load of homologous and heterologous challenge viruses in the absence of neutralizing antibodies [9]. ...
Article
Full-text available
Swine influenza A viruses (SwIAVs) are pathogens of both veterinary and medical significance. Intranasal (IN) vaccination has the potential to reduce flu infection. We investigated the efficacy of split SwIAV H1N2 antigens adsorbed with a plant origin nanoparticle adjuvant [Nano11–SwIAV] or in combination with a STING agonist ADU-S100 [NanoS100–SwIAV]. Conventional pigs were vaccinated via IN and challenged with a heterologous SwIAV H1N1-OH7 or 2009 H1N1 pandemic virus. Immunologically, in NanoS100–SwIAV vaccinates, we observed enhanced frequencies of activated monocytes in the blood of the pandemic virus challenged animals and in tracheobronchial lymph nodes (TBLN) of H1N1-OH7 challenged animals. In both groups of the virus challenged pigs, increased frequencies of IL-17A+ and CD49d+IL-17A+ cytotoxic lymphocytes were observed in Nano11–SwIAV vaccinates in the draining TBLN. Enhanced frequency of CD49d+IFNγ+ CTLs in the TBLN and blood of both the Nano11-based SwIAV vaccinates was observed. Animals vaccinated with both Nano11-based vaccines had upregulated cross-reactive secretory IgA in the lungs and serum IgG against heterologous and heterosubtypic viruses. However, in NanoS100–SwIAV vaccinates, a slight early reduction in the H1N1 pandemic virus and a late reduction in the SwIAV H1N1-OH7 load in the nasal passages were detected. Hence, despite vast genetic differences between the vaccine and both the challenge viruses, IN vaccination with NanoS100–SwIAV induced antigen-specific moderate levels of cross-protective immune responses.
... In addition, the flu watch cohort study also observed a positive correlation between pre-existing IAV-specific CD8 + T cells and less symptomatic IAV infections during three seasonal epidemics and the 2009 H1N1 pandemic, which was independent of baseline antibodies, confirming the heterosubtypic protection delivered by CD8 + T cells in influenza virus infection [19]. Many other studies have also described the potential cross-reactive response of the CD8 + T cells to different influenza virus strains, including the highly pathogenic H7N9 and H5N1 avian influenza viruses [18,56,82,[91][92][93][94][95]. Indeed, H7N9-infected patients with early H7N9-specific, IFNγ-producing CD8 + T cell populations recovered more rapidly from severe infection. ...
Article
Full-text available
Influenza A virus is a respiratory pathogen that is responsible for regular epidemics and occasional pandemics that result in substantial damage to life and the economy. The yearly reformulation of trivalent or quadrivalent flu vaccines encompassing surface glycoproteins derived from the current circulating strains of the virus does not provide sufficient cross-protection against mismatched strains. Unlike the current vaccines that elicit a predominant humoral response, vaccines that induce CD8+ T cells have demonstrated a capacity to provide cross-protection against different influenza strains, including novel influenza viruses. Immunopeptidomics, the mass spectrometric identification of human-leukocyte-antigen (HLA)-bound peptides isolated from infected cells, has recently provided key insights into viral peptides that can serve as potential T cell epitopes. The critical elements required for a strong and long-living CD8+ T cell response are related to both HLA restriction and the immunogenicity of the viral peptide. This review examines the importance of HLA and the viral immunopeptidome for the design of a universal influenza T-cell-based vaccine.
... Sherritt et al. (2000) have used peptide vaccines in the preexisting multiple CD8+ T-cell epitopes delivered in mice with CD8+ T-cells specific for one of these epitopes and showed that it induced CTL with additional specificity. In addition to data from animals supporting a role for CD8+ T-cell responses in viral clearance and survival (Talker et al., 2016;McMahon et al., 2019), there are also relevant data showing the presence of cross-reactive influenza virus-specific CD8+ memory T cells in humans (Boon et al., 2004;Kreijtz et al., 2008;Duan and Thomas, 2016;Grant et al., 2016). A separate study by McMichael et al. (1983) showed that high levels of CD8+ T cells were associated with reduced viral shedding following experimental infection in individuals lacking specific antibodies. ...
Article
Full-text available
Vaccination is the most effective means of protecting people from influenza virus infection. The effectiveness of existing vaccines is very limited due to antigenic drift of the influenza virus. Therefore, there is a requirement to develop a universal vaccine that provides broad and long-lasting protection against influenza. CD8+ T-cell response played a vital role in controlling influenza virus infection, reducing viral load, and less clinical syndrome. In this study, we optimized the HA sequences of human seasonal influenza viruses (H1N1, H3N2, Victoria, and Yamagata) by designing multivalent vaccine antigen sets using a mosaic vaccine design strategy and genetic algorithms, and designed an HA mosaic cocktail containing the most potential CTL epitopes of seasonal influenza viruses. We then tested the recombinant mosaic antigen, which has a significant number of potential T-cell epitopes. Results from genetic evolutionary analyses and 3D structural simulations demonstrated its potential to be an effective immunogen. In addition, we have modified an existing neutralizing antibody-based seasonal influenza virus vaccine to include a component that activates cross-protective T cells, which would provide an attractive strategy for improving human protection against seasonal influenza virus drift and mutation and provide an idea for the development of a rationally designed influenza vaccine targeting T lymphocyte immunity.
... Influenza virus is another representative virus that escapes from antibodies via mutations. Notably, CD8 + T cells induced by seasonal influenza can cross-recognize the 2009 pandemic H1N1 virus (23,24) and the pre-pandemic avian influenza A (H5N1) virus (25,26), both of which resulted from antigenic shifts. Importantly, higher ratios of pre-existing T cells to conserved CD8 + T cell epitopes were found in individuals who developed less-severe illness due to the H1N1 virus (27), consistent with an observation that individuals unexposed to SARS-CoV-2 but having CD8 + T cells specific to conserved epitopes among seasonal human coronaviruses tend to show mild illness (28). ...
Article
Full-text available
The primary goal of vaccines that protect against respiratory viruses appears to be the induction of neutralizing antibodies for a long period. Although this goal need not be changed, recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have drawn strong attention to another arm of acquired immunity, CD8⁺ T cells, which are also called killer T cells. Recent evidence accumulated during the coronavirus disease 2019 (COVID-19) pandemic has revealed that even variants of SARS-CoV-2 that escaped from neutralizing-antibodies that were induced by either infection or vaccination could not escape from CD8⁺ T cell-mediated immunity. In addition, although traditional vaccine platforms, such as inactivated virus and subunit vaccines, are less efficient in inducing CD8⁺ T cells, newly introduced platforms for SARS-CoV-2, namely, mRNA and adenoviral vector vaccines, can induce strong CD8⁺ T cell-mediated immunity in addition to inducing neutralizing antibodies. However, CD8⁺ T cells function locally and need to be at the site of infection to control it. To fully utilize the protective performance of CD8⁺ T cells, it would be insufficient to induce only memory cells circulating in blood, using injectable vaccines; mucosal immunization could be required to set up CD8⁺ T cells for the optimal protection. CD8⁺ T cells might also contribute to the pathology of the infection, change their function with age and respond differently to booster vaccines in comparison with antibodies. Herein, we overview cutting-edge ideas on CD8⁺ T cell-mediated immunity that can enable the rational design of vaccines for respiratory viruses.
... This protective immune correlate could guide universal influenza vaccine development [166]. Furthermore, CD8+ T cells activated upon infection with seasonal influenza type A strains can cross-react with pH1N1 [167,168], highly pathogenic avian H5N1viruses [169,170] and H7N9 variants [171]. Similar finding was published by Wang et al. who showed that early effective CD8+ T cell response (most likely recalled from the memory pool) was associated with less cytokine/chemokine-driven inflammatory disease and better recovery of hospitalized patients with H7N9 [172]. ...
Article
Full-text available
Negative-stranded RNA viruses (NSVs) are important human pathogens, including emerging and reemerging viruses that cause respiratory, hemorrhagic and other severe illnesses. Vaccine design traditionally relies on the viral surface glycoproteins. However, surface glycoproteins rarely elicit effective long-term immunity due to high variability. Therefore, an alternative approach is to include conserved structural proteins such as nucleoprotein (NP). NP is engaged in myriad processes in the viral life cycle: coating and protection of viral RNA, regulation of transcription/replication processes and induction of immunosuppression of the host. A broad heterosubtypic T-cellular protection was ascribed very early to this protein. In contrast, the understanding of the humoral immunity to NP is very limited in spite of the high titer of non-neutralizing NP-specific antibodies raised upon natural infection or immunization. In this review, the data with important implications for the understanding of the role of NP in the immune response to human NSVs are revisited. Major implications of the elicited T-cell immune responses to NP are evaluated, and the possible multiple mechanisms of the neglected humoral response to NP are discussed. The intention of this review is to remind that NP is a very promising target for the development of future vaccines.
Article
Full-text available
Objectives Seasonal influenza viruses cause roughly 650 000 deaths annually despite available vaccines. CD8⁺ T cells typically recognise influenza‐derived peptides from internal structural and non‐structural influenza proteins and are an attractive avenue for future vaccine design as they could reduce the severity of disease following infection with diverse influenza strains. CD8⁺ T cells recognise peptides presented by the highly polymorphic Human Leukocyte Antigens class I molecules (HLA‐I). Each HLA‐I variant has distinct peptide binding preferences, representing a significant obstacle for designing vaccines that elicit CD8⁺ T cell responses across broad populations. Consequently, the rational design of a CD8⁺ T cell‐mediated vaccine would require the identification of highly immunogenic peptides restricted to a range of different HLA molecules. Methods Here, we assessed the immunogenicity of six recently published novel influenza‐derived peptides identified by mass‐spectrometry and predicted to bind to the prevalent HLA‐B*18:01 molecule. Results Using CD8⁺ T cell activation assays and protein biochemistry, we showed that 3/6 of the novel peptides were immunogenic in several HLA‐B*18:01⁺ individuals and confirmed their HLA‐B*18:01 restriction. We subsequently compared CD8⁺ T cell responses towards the previously identified highly immunogenic HLA‐B*18:01‐restricted NP219 peptide. Using X‐ray crystallography, we solved the first crystal structures of HLA‐B*18:01 presenting immunogenic influenza‐derived peptides. Finally, we dissected the first TCR repertoires specific for HLA‐B*18:01 restricted pathogen‐derived peptides, identifying private and restricted repertoires against each of the four peptides. Conclusion Overall the characterisation of these novel immunogenic peptides provides additional HLA‐B*18:01‐restricted vaccine targets derived from the Matrix protein 1 and potentially the non‐structural protein and the RNA polymerase catalytic subunit of influenza viruses.
Article
Full-text available
Influenza (flu) infection is a leading cause of respiratory disease and death worldwide. While seasonal flu vaccines are effective at reducing morbidity and mortality, such effects rely on the odds of successful prediction of the upcoming viral strains. Additional threats from emerging flu viruses that we cannot predict and avian flu viruses that can be directly transmitted to humans, urge the strategic development of universal vaccinations that can protect against flu viruses of different subtypes and across species. Annual flu vaccines elicit mainly humoral responses. Under circumstances when antibodies induced by vaccination fail to recognize and neutralize the emerging virus adequately, virus‐specific cytotoxic T lymphocytes (CTLs) are the major contributors to the control of viral replication and elimination of infected cells. Our studies exploited the evolutionary conservation of influenza A nucleoprotein (NP) and the fact that NP‐specific CTL responses pose a constant selecting pressure on functional CTL epitopes, to screen for NP epitopes that are highly conserved among heterosubtypes but are subjected to positive selection historically. We identified a region on NP that is evolutionarily conserved and historically positively selected (NP137‐182) and validated that it contains an epitope that is functional in eliciting NP‐specific CTL responses and immunity that can partially protect immunized mice against lethal dose infection of a heterosubtypic influenza A virus. Our proof‐of‐concept study supports the hypothesis that evolutionary conservation and positive selection of influenza nucleoprotein can be exploited to identify functional CTL epitope to elicit cross protection against different heterosubtypes, therefore, to help develop strategies to modify flu vaccine formula for a broader and more durable protective immunity. This article is protected by copyright. All rights reserved.
Article
Full-text available
The Influenza Sequence Database (ISD) is a curated, specialized database of influenza genomic and protein sequences, and influenza-specific sequence analysis tools that have a web interface available to the public http://www.flu.lanl.gov). Contents are primarily those influenza sequences that have been deposited in GenBank. A growing number of sequences are deposited directly into the ISD by WHO surveillance sites around the world. The core function of the interface is a database search. Analysis tools are a combination of developments by ISD staff and adaptation of existing, freely available software. Future developments of the database and its web site will be driven by user needs and by output from our in-house research. Projects underway include a layered, secure interface with the database to allow variable access privileges to sensitive data, and clickable mapping of a homology model if Influenza B hemagglutinin.
Article
Full-text available
A recombinant vaccinia virus (NP-VAC) containing cDNA corresponding to segment 5, the nucleoprotein (NP) gene of influenza A/PR/8/34 virus was used to examine the specificity of human influenza virus immune cytotoxic T lymphocytes (CTL). Effector cell preparations from two donors recognized autologous lymphocytes that had been infected with NP-VAC. Lysis was specific because cells infected with vaccinia virus were not killed and recognition was HLA-restricted. In one donor, the influenza virus-specific CTL response changed with time so that his effector cells no longer recognized autologous lymphocytes infected with NP-VAC. However, a component that was NP-specific remained because these CTL lysed the more sensitive autologous B lymphoblastoid cells that had been infected with NP-VAC. In four other donors, no NP-specific CTL response could be detected using autologous lymphocyte targets. Thus NP, an internal virus protein, is one antigen that is recognized by human influenza A virus-specific CTL, but it is likely that other individual virus components contribute to the total CTL response.
Article
Full-text available
Influenza A virus-specific cytotoxic T lymphocytes (CTL) capable of lysing cells infected with any influenza A virus ("cross-reactive CTL") constitute a major portion of the host CTL response to influenza. The viral nucleoprotein (NP), a major internal virion structural protein, has been implicated as a possible target antigen for cross-reactive CTL. To directly examine CTL recognition of NP, a vaccinia virus recombinant containing a DNA copy of an influenza A virus NP gene was constructed. We found that murine cells infected with this virus were efficiently lysed in a major histocompatibility complex-restricted manner by cross-reactive CTL populations obtained by immunization with a variety of influenza A virus subtypes. In addition, the recombinant vaccinia virus containing the PR8 NP gene was able to both stimulate and prime for a vigorous secondary cross-reactive CTL response. Significantly, splenocytes from mice primed by inoculation with the recombinant vaccinia virus containing the PR8 NP gene could be stimulated by influenza A viruses of all three major human subtypes. Finally, unlabeled target competition experiments suggest that NP is a major, but not the sole, viral target antigen recognized by cross-reactive CTL.
Article
Full-text available
Using genetically typed recombinant influenza A viruses that differ only in their genes for nucleoprotein, we have demonstrated that repeated stimulation in vitro of C57BL/6 spleen cells primed in vivo with E61-13-H17 (H3N2) virus results in the selection of a population of cytotoxic T lymphocytes (CTL) whose recognition of infected target cells maps to the gene for nucleoprotein of the 1968 virus. Influenza A viruses isolated between 1934 and 1979 fall into two groups defined by their ability to sensitize target cells for lysis by these CTL: 1934-1943 form one group (A/PR/8/34 related) and 1946-1979 form the second group (A/HK/8/68 related). These findings complement and extend our previous results with an isolated CTL clone with specificity for the 1934 nucleoprotein (27, 28). It is also shown that the same spleen cells derived from mice primed with E61-13-H17 virus in vivo, but maintained in identical conditions by stimulation with X31 virus (which differs from the former only in the origin of its gene for NP) in vitro, results in the selection of CTL that cross-react on target cells infected with A/PR/8/1934 (H1N1) or A/Aichi/1968 (H3N2). These results show that the influenza A virus gene for NP can play a role in selecting CTL with different specificities and implicate the NP molecule as a candidate for a target structure recognized by both subtype-directed and cross-reactive influenza A-specific cytotoxic T cells.
Article
Full-text available
We previously identified an HLA-B8+ donor, NW, whose lymphoblastoid cells failed to present a B8-restricted epitope from the influenza A nucleoprotein following viral infection, although added peptide could still be presented. The failure to present through HLA-B8 following viral infection appears to be specific for the NP epitope. Here, we report that donor NW makes an HLA-B2702-restricted influenza-specific CTL response to an epitope in the nucleoprotein that overlaps the B8-restricted epitope by 8 aa. Two mechanisms for the failure of this cell line to present the B8-restricted epitope following viral infection are investigated. One is that there is an antigen processing polymorphism specific to the NW cell line, so that there is either preferential generation or preferential transport of the B2702 epitope. The other is that B8 and B2702 compete for a common peptide fragment in the ER and this leads to suboptimal loading of HLA-B8.
Article
The nature of the viral antigens recognized by influenza A virus-immune cytotoxic T lymphocytes (CTL) is still a matter of debate. We have used four human influenza A virus-specific T lymphocyte clones with antigen-specific cytotoxic and proliferative activity to investigate the requirements for recognition of viral antigens on infected cells. One clone recognized a cross-reactive determinant on the viral hemagglutinin, and two clones were specific for different epitopes on the viral nucleoprotein (NP). A fourth clone seemed to be specific for the viral M protein. Target cell recognition was abrogated by the addition, during infection, of the lysosomotropic drug chloroquine, known to inhibit antigen processing. Furthermore, target cells that had been pulsed with soluble purified NP were recognized and were lysed by the NP-specific clone. This reaction could also be abrogated by the addition of chloroquine during pulsing. These results were obtained irrespective of whether EBV-transformed B lymphoblastoid cells or Ia antigen-expressing T cell blasts were used as target cells. It is concluded that CTL can recognize internal viral proteins that are actively presented at the surface of the target cell. These data indicate that probably every viral protein can function as a target molecule for virus-immune CTL.
Article
We have studied five cases suggesting a relation between Epstein-Barr virus infection and primary lymphoma of the central nervous system. A 48-year-old man had primary lymphoma of the central nervous system in the absence of systemic lymphoma or immunosuppression. Development of the tumor was associated with serologic evidence suggesting a recent primary infection with Epstein-Barr virus. DNA preparations from tumor tissue, but not from adjacent normal brain tissue, contained Epstein-Barr virus genomes when hybridized with a probe consisting of the BamHI K fragment of Epstein-Barr virus strain FF41. Evaluation of serum samples from four additional patients with central-nervous-system lymphoma revealed patterns of Epstein-Barr virus-specific antibody that were suggestive of an ongoing infection with EBV. Our results suggest induction of the lymphoma by Epstein-Barr virus.
Article
In a study designed to determine whether cytotoxic T lymphocytes contribute to immunity against influenza virus infection, we inoculated 63 volunteers intranasally with live unattenuated influenza A/Munich/1/79 virus. Over the next seven days clinical observations were made, and the amount of virus shed was measured. The protective effects of preinfection serum antibody and of cytotoxic T-cell immunity against influenza A virus were assessed for each participant. All subjects with demonstrable T-cell responses cleared virus effectively. This response was observed in volunteers in all age groups, including those born after 1956, who did not have specific antibody and hence had probably not been exposed to this subtype of influenza A virus before. Cytotoxic T cells show cross-reactivity in their recognition of the different subtypes of influenza A virus, in contrast to the antibody response that is specific for each virus subtype. We conclude that cytotoxic T cells play a part in recovery from influenza virus infection.
Article
Influenza virus has proved an excellent model for the study of the specificity of cytotoxic T lymphocytes (CTL) and of the processing of viral antigens. Fundamental information about these processes has been gained in both mice and humans. At the same time, a considerable amount has been learned about the role of CTL in influenza viral infections, particularly in mice. The virus has proved to be a malleable tool because it can be readily grown, the full nucleotide structure has been determined (in the 1970s; reviewed in Lamb 1983), there are multiple antigenic variants, many of which have been sequenced, the three-dimensional structures of haemagglutinin and neuraminidase are known (Varghese et al. 1983; Wiley et al. 1981) and, for human studies, it has the advantage that it has infected a very high percentage of the population. All of these features combine to make this a safe and invaluable model for the study of theoretical aspects of T cell function as well as investigating the practical issues of how T cells control virus infections. Studies with influenza virus answered long-standing questions about the interaction between viral antigens and class I molecules of the major histocompatibility complex (MHC). Along the way came information about mutant and variant MHC molecules and the demonstration that peptide epitopes presented by class I MHC molecules are recognised by CTL. More recently, the virus has been invaluable for the elucidation of the antigenprocessing pathways.