1 Institute of Virology and Immunoprophylaxis (IVI), Postfach, CH-3147 Mittelhäusern, Switzerland
2 Animal Sciences Group, 8200 Lelystad, The Netherlands
3 Organic Chemistry, Auf der Morgenstelle 18, D-72076 Tübingen, Germany
4 Friedrich-Loeffler-Institut, Paul-Ehrlich-Straße 28, D-72076 Tübingen, Germany
Correspondence
A. Summerfield
artur.summerfield{at}ivi.admin.ch
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Present address: Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France.
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
Consequently, the localization of T-cell epitopes in particular, epitopes recognized by cytotoxic T lymphocytes (CTLs) is important for new vaccine development and improvement of existing vaccines, with respect to inducing early protective immunity. Viral proteins capable of inducing virus-specific T-cell responses have been examined previously, and a nonapeptide CTL epitope was identified on NS4A (Pauly et al., 1995). More recently, 26 different pentadecapeptides capable of inducing virus-specific T-cell responses have been found, distributed over different viral proteins (Armengol et al., 2002
). One of these peptides present on the NS2 protein was shown to carry a CTL epitope (Armengol et al., 2002
). In contrast, no CTL epitopes have been identified on the CSFV structural proteins.
T-lymphocyte epitopes have to be recognized in the context of major histocompatibility complex (MHC) molecules, which imparts an influence of the MHC haplotypes on epitope recognition. Considering that the above CTL epitopes were detected in d/d-haplotype inbred animals, it was necessary to verify that CTL epitopes on CSFV proteins would be recognized in outbred animals. To this end, we employed a novel methodology based on mRNA-transfected antigen-presenting cells (APCs) both dendritic cells (DCs) and fibrocytes were employed. DCs can be loaded efficiently with mRNA molecules, leading to induction of antigen-specific T-cell responses (Ponsaerts et al., 2003). DCs transfected with mRNA encoding viral antigens can induce strong, virus-specific CTL responses (Strobel et al., 2000
; Weissman et al., 2000
; Zarei et al., 2003
; Ueno et al., 2004
). Unlike the peptide-antigen approach, this methodology permits the identification of T-cell epitopes arising naturally from the antigen processing by DCs and is therefore not influenced by the MHC haplotype. The procedure is therefore particularly applicable to use with outbred animals, and does not require the establishment of target-cell lines for CTL assays. By using this approach of mRNA-transfected DCs, the existence of T-cell epitopes recognized by T lymphocytes from outbred animals was determined. The mRNA-transfected DCs induced CSFV-specific responses efficiently, showing that CTL epitopes were present on both non-structural proteins and the E2 structural protein.
![]() |
METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Immunization of pigs against CSFV.
Specific pathogen-free, 3-month-old pigs (Swiss Land race), bred at the IVI, were infected intranasally with 103 TCID50 vA187-1, a recombinant CSFV derived from the Alfort/187 strain (Ruggli et al., 1996). Peripheral blood mononuclear cells (PBMCs) from these outbred animals were used 36 months after infection for the antigen-presentation assays using transfected APCs.
For the CTL analyses using the MAX.b2 cells (see below), three miniature pigs inbred for swine leukocyte-antigen complex (haplotype d/d) were used (kindly obtained from Dr Henry Salmon, INRA, Tours-Nouzilly, France). Three pigs were vaccinated intramuscularly with 104 immunofluorescence-stained foci of vFlc23 complemented on SK-6 cells expressing Erns (Widjojoatmodjo et al., 2000) at 2 months of age. After 4 weeks, the pigs were challenged intranasally with 103·2 TCID50 CSFV strain Brescia 456610, corresponding to 100 LD50 (Terpstra & Wensvoort, 1988
), followed by a second challenge after 8 weeks. This protocol was selected to induce a strong cellular immune response with sufficient virus-specific T lymphocytes in the circulation. Unvaccinated pigs were used as controls to determine the specifity of the antigen-presentation assays.
Plasmids and mRNA molecules encoding CSFV-derived genes.
The E2 gene, including its signal sequence (sigE2), and the sequences encoding NS3NS4A of the CSFV genome (strain Alfort/187; Fig. 1) were cloned into the pCR-XL-TOPO cloning vector (Invitrogen). The sigE2 sequence, including the E2 gene and the 3'-terminal 48 nt of the E1 gene, encoding the E2 signal sequence, was amplified with primers sigE2_BamHI_L3 and E2_XbaI_R2; the NS3NS4A sequence was amplified with primers NS3_BglII_L and NS4A_XbaI_R3. Subsequent cloning in the T7TSbetaglobinA70 vector (Hoerr et al., 2000
; Carralot et al., 2004
) (CureVac) was via the BglII and the SpeI restriction sites, and the resulting vectors were linearized by PstI followed by in vitro transcription using a T7-Opti-mRNA transcription kit (CureVac). The 5'-capped and polyadenylated (A70), in vitro-transcribed mRNA molecules were purified by LiCl precipitation, quantified, extracted with phenol/chloroform, precipitated with ethanol/NaCl, washed with 75 % (v/v) ethanol and air-dried. All constructs were verified by sequencing on a LI-COR 4200 sequencer (LI-COR Biosciences). The sequences of the oligonucleotide primers used were as follows [polarity of the primers: (+), sense orientation; (), antisense orientation]: sigE2_BamHI_L3(+), 5'-CCGGATCCGCCACCATGATCGTGCAAGGTGTGATATG-3'; E2_XbaI_R2(), 5'-GGTCTAGATTAACCAGCGGCGAGTTGTTCTGT-3'; NS3_BglII_L(+), 5'-GAAACGCATAGATCTGCCGCCACCGCCATGGGGCCTGCCGTTTGC-3'; NS4A_XbaI_R3(), 5'-GACGGCTCTAGATTATAGCTCTTTCAACTCTCT-3' (restriction sites are shown in italics and start and stop codons are underlined). Stabilized, capped and polyadenylated, in vitro-transcribed enhanced green fluorescent protein (eGFP) mRNA molecules (produced from the T7TSbetaglobinA70 vector) were purchased from CureVac.
For the construction of the pcDNA-E2/NS3 plasmid, viral RNA isolated from CSFV strain Glentorf (GenBank accession no. U45478)-infected cells was amplified by using primers with a BglII restriction site at the 5' end (5'-TTTTTTAGATCTATCGTGCAAGGTGTGATATGGCTG-3') and a NotI restriction site at the 3' end (5'-AAAAAAGCGGCCGCTCATAGACCAACTACTTGTTTTAGTGC-3') (restriction sites are shown in bold). To fuse the E2 and NS3 sequences, overlapping primers were designed (forward primer E2NS3, 5'-ACAGAACAACTCGCCGCTGGTGGTGGTATGCCAAGGGGCACC-3'; reverse primer E2NS3, 5'-GGTGCCCCTTGGCATAACACCACCAGCGGCGAGTTGTTCTGT-3'). After cDNA synthesis with avian myeloblastosis virus reverse transcriptase (Boehringer Mannheim), the E2NS3 gene was amplified by PCR using the TripleMaster PCR system (Eppendorf). The PCR product was purified, digested with BglII and NotI and cloned into the pcDNA4 vector (Invitrogen), and the inserted sequence was verified by sequencing.
In vitro translation of CSFV-specific mRNA and protein detection by Western blotting.
Proteins were expressed in vitro by using the rabbit reticulocyte lysate system, using 1 µg RNA translated in a 25 µl reaction without radioactive labelling, following the manufacturer's instructions (Promega). Proteins were separated by SDS-PAGE under reducing conditions according to standard protocols and analysed by Western blotting. Gels were equilibrated for 30 min in SDS blotting buffer (12·5 mM Tris/HCl, 125 mM glycine, 0·05 % SDS, 20 % methanol, pH 8·3) prior to transfer to a Trans-Blot nitrocellulose membrane (Bio-Rad) at 15 V for 30 min using a Trans-Blot SemiDry transfer device (Bio-Rad). The membranes were blocked for 1 h at room temperature with Odyssey blocking reagent (LI-COR Biosciences) diluted 1 : 1 with PBS. Immunodetection was performed for 30 min at room temperature with a mixture of the eGFP-specific mAb JL8 (BD Biosciences) and a rabbit antiserum against NS3 (J. D. Tratschin, unpublished data), followed by Alexa Fluor 680 goat anti-mouse and anti-rabbit IgG (Molecular Probes) secondary antibody, using standard protocols. Image acquisition used the Odyssey Infrared Imaging system (LI-COR).
PBMC, DC and fibrocyte preparation.
PBMCs were purified from citrated blood by density-gradient centrifugation over Ficoll-Paque (1·077 g l1; Amersham Biosciences) (McCullough et al., 1993). For generation of DCs, monocytes were enriched from the PBMCs by magnetic cell sorting using the magnetic-activated cell-sorting (MACS) system (Miltenyi Biotec) and an anti-CD172a mAb (74-22-15, kindly provided by Dr J. K. Lunney, USDA, Beltsville, MD, USA) (Alvarez et al., 2000
). DCs were cultured for 5 days at 39 °C, 6 % (v/v) CO2 in Dulbecco's modified Eagle's medium (DMEM; Invitrogen) supplemented with 10 % (v/v) porcine serum (Sigma-Aldrich) and recombinant porcine (rp) granulocytemacrophage colony-stimulating factor (150 ng ml1, kindly provided by Dr S. Inumaru, Institute for Animal Health, Ibaraki, Japan) and rp interleukin 4 (100 U ml1) (Carrasco et al., 2001
).
Porcine fibrocytes were derived and cultured as described previously (Balmelli et al., 2005). Briefly, PBMCs were plated at 4x106 cells ml1 in fibrocyte medium [DMEM, 20 % (v/v) fetal bovine serum (FBS), 1 % (v/v) penicillin/streptomycin (Invitrogen)]. After 24 h culture at 39 °C, 6 % (v/v) CO2, non-adherent cells were removed by a single, gentle aspiration. After 1012 days further culture, the loosely adherent, growing fibrocytes were further passaged or used as antigen-presenting cells (Balmelli et al., 2005
).
Transfection of SK-6 cells, DCs and fibrocytes.
At 16 h before transfection, SK-6 cells were detached by using trypsin/EDTA solution and plated in six-well plates with 105 cells per well. Prior to transfection, the cells were rinsed twice with PBS. For the transfection, 2 µg mRNA (or 4 µg plasmid DNA) diluted in 1 ml Opti-MEM was mixed with 8 µl TransFast lipofection reagent (Promega) at a lipid : mRNA ratio of 4 : 1 (or 2 : 1 lipid : DNA ratio). After 15 min incubation at room temperature, the transfection mix was added to the cells and incubated for 1 h at 37 °C. Following transfection, 4 ml pre-warmed Earl's medium containing 7 % (v/v) horse serum was added to the cells. At 24 h post-transfection, the cells were analysed for protein expression.
DCs were transfected by mRNA electroporation as described previously (Ceppi et al., 2005). Briefly, DCs were washed twice and adjusted to 5x106 cells ml1 in Opti-MEM (Invitrogen) in a 0·2 cm gap electroporation cuvette (Bio-Rad). After 5 min on ice, 5 µg mRNA was added and the cells were pulsed for 24 ms by using a Gene Pulser II apparatus (300V, 125 µF; Bio-Rad) and cultured for another 48 h. Fibrocytes were transfected by mRNA lipofection. At 4 h before transfection, fibrocytes were plated in six-well plates with 0·5x1051·0x105 cells per well. Then, the cells were rinsed twice with PBS, and 3 µg mRNA diluted in 1 ml Opti-MEM was mixed with 6 µl TransFast (Promega) at a lipid : mRNA ratio of 2 : 1. After 15 min at room temperature, the transfection mix was added to the cells and incubated for 1 h at 39 °C. Following transfection, 4 ml fibrocyte medium was added and, 24 h later, the cells were employed.
Establishment of MAX cell lines expressing CSFV E2.
MAX cells, an SV40-immortalized swine kidney-cell line of MHC class I haplotype d/d (Pauly et al., 1995), were grown in DMEM containing 5 % (v/v) FBS, glutamine (0·3 mg ml1), penicillin (200 U ml1), streptomycin (0·2 mg ml1) and mycostatin (100 U ml1). The MAX cells were transfected with the pPRb2 plasmid containing the E2 gene of CSFV strain Brescia (Widjojoatmodjo et al., 2000
) under control of the transcription and translation signals of the pEVhisD12 vector (Peeters et al., 1992
). For transfection, 2 µg lipofectamine (Invitrogen) was diluted in 50 µl Opti-MEM I (Invitrogen), mixed with plasmid DNA (1 µg) in 50 µl medium and the mixture was incubated for 15 min at room temperature. MAX cells grown in 10 cm2 tissue-culture plates were washed with Opti-MEM I and cultured for 20 h in 0·5 ml medium. Then, the cells were harvested for limiting-dilution cultures in microtitre plates in medium containing 7·5 mM histidinol, which was replaced every 34 days until single colonies were visible. After 2 weeks, surviving colonies were recloned by limiting dilution and screened for E2 expression by immunostaining with mAbs b3 and b6, directed respectively against the conserved domain A and non-conserved domain B of E2 from CSFV strain Brescia (Wensvoort et al., 1989
). The two domains were recognized by the mAbs in 7080 % of the cells, referred to as MAX.b2.
Confocal-microscopy analysis.
For CSFV-specific protein expression, transfected or infected cells were washed twice in PBS, fixed in 4 % (w/v) paraformaldehyde for 15 min and washed again. Then, the cells were stained for 20 min at 4 °C with the anti-E2 mAb HC26 (Greiser-Wilke et al., 1990) (kindly provided by Bommeli AG) or the anti-NS3 mAb C16 (Greiser-Wilke et al., 1992
) (kindly provided by Dr Greiser-Wilke, Hannover, Germany), diluted in 0·3 % (w/v) saponin (S4521; Sigma-Aldrich) permeabilization solution. After washing with PBS containing 0·1 % (w/v) saponin, Alexa-488 or Alexa-546 fluorochrome-labelled anti-mouse secondary antibody (Molecular Probes) diluted in the permeabilizing solution was added. Following incubation for 20 min at 4 °C, washing and mounting in Mowiol mounting solution, the cells were analysed by using a Leica TCS-SL spectral confocal microscope (CFM) and Leica LCS software (Leica Microsystems AG).
Antigen-presentation assays.
Purified T lymphocytes were sorted by MACS using an anti-CD6 mAb, as described previously (Carrasco et al., 2001). Purity of the positive fraction was >95 %. Either autologous DCs or fibrocytes (transfected or infected) were employed as APCs at a ratio of 1 : 20 or 1 : 400, respectively. Where indicated, CSFV strain Eystrup was added to the co-cultures at an m.o.i. of 0·01 TCID50 per cell. These cultures were incubated for 5 days and then tested for the presence of activated T cells.
IFN- ELISPOT.
The ELISPOT assay was performed as described previously (Armengol et al., 2002) with minor modifications. Stimulated lymphocytes were added to duplicate wells on nitrocellulose plates (Nunc) coated with mouse anti-pig IFN-
mAb (MP700, 5 µg ml1; Pierce Biotechnology). After 48 h incubation, IFN-
spots were visualized with rabbit anti-pig IFN-
polyclonal antibody (PP700, 2·5 µg ml1; Pierce Biotechnology) followed by peroxidase-conjugated goat anti-rabbit antibody (Jackson Immunoresearch Laboratories) and developed with 3,3'-diaminobenzidine (Sigma) for 20 min at room temperature in the dark. The number of spots was determined with a computer-assisted video-image analyser (AID ELISPOT reader; AID GmbH).
Cytotoxic assay.
Autologous fibrocytes infected with CSFV were used as target cells in a flow-cytometry assay for cytotoxicity (Balmelli et al., 2005). Briefly, fibrocytes were detached, stained with PKH26 (Sigma) and plated at 10 000 cells per well in 96-well flat-bottomed plates. The fibrocytes were infected with CSFV or treated with mock antigen for 24 h at an m.o.i. of 2 TCID50 per cell. Restimulated lymphocytes (see above) were then added to the target fibrocytes at various effector : target-cell (E : T) ratios for 16 h at 39 °C. Then, the percentage of dead cells was quantified by using propidium iodide (Sigma).
For the experiments using d/d-haplotype MAX cells as target cells, miniature pig-derived PBMCs were restimulated with CSFV strain C1.1.1 (5x106 p.f.u. ml1) for 7 days and CTL activity was tested in a 51Cr-release assay, as described previously (de Bruin et al., 2000). Briefly, MAX.b2 cells, MAX cells transfected with an empty control plasmid or infected MAX cells were labelled with 51Cr [400 µCi (14·8 MBq); Amersham Biosciences, catalogue no. CJS4] and cultured with restimulated PBMCs in 96-well V-bottomed microtitre plates (Nunc) at E : T ratios decreasing from 50 to 6·25 for 5 h. The supernatant was then tested for radioactivity in a Wallac Microbetaplus 1450 scintillation counter (EG&G Instruments). Maximal release of 51Cr was induced by adding 50 µl 20 % Triton X-100. Spontaneous release was determined in wells that did not contain effector cells. The percentage of specific lysis was calculated as (c.p.m. experimental releasec.p.m. spontaneous release)/(c.p.m. maximal releasec.p.m. spontaneous release) x100.
Statistical analysis of data.
Statistical evaluation of the experimental data was performed by using Student's t-test. A value of P<0·05 was considered statistically significant.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
As a first step, it was considered important to verify the translation of the mRNA molecules. To this end, in vitro translation in the rabbit reticulocyte lysate system followed by Western blotting was used. Both eGFP and NS3 were detectable with the expected molecular masses, indicating that both proteins are translated, although only NS3 at lower levels (Fig. 1c). This technique was not used for the E2 mRNA, due to the lack of a suitable antibody.
Expression of the E2 and NS3 proteins in transfected cells
The protein-expression capacity of the CSFV-derived mRNAs was tested in transfected cells and compared with the expression obtained with plasmid DNA transfection or CSFV infection. Protein expression was first tested in SK-6 cells, an established porcine cell line that is permissive for CSFV infection and replication. At 24 h after lipofection of pcDNA-E2/NS3 (for construct, see Fig. 1d), both E2 and NS3 were detectable (Fig. 2a
, top panels). The proteins were not detectable in all cells due to the limited transfection efficiency of SK-6 cells (25 %, relative to an eGFP reporter plasmid). Moreover, in some cells, E2 and NS3 could not be detected simultaneously, probably because of differences in the sensitivity of their detection. Lipofection of SK-6 cells with the sigE2 mRNA also resulted in E2 protein expression (Fig. 2a
, bottom panels). No signal for NS3 was detected with the sigE2-transfected cells, as expected.
|
mRNA-transfected DCs induce CSFV-specific CTL responses
Although only the E2 mRNA produced detectable protein in the transfected DCs, it was necessary to determine whether this had any immunological relevance. Consequently, DCs transfected with the mRNA molecules encoding E2 and NS3NS4 were tested for their capacity to induce virus-specific T-cell responses in vitro. The mRNA-transfected DCs were co-cultured with CSFV-immune lymphocytes at a ratio of 1 : 20, whilst CSFV-infected DCs (m.o.i. of 0·01 TCID50 per cell) were employed as a positive control. Mock-treated DCs were the negative control. After 5 days DC/lymphocyte co-culture, the presence of activated T cells was determined by using an IFN- ELISPOT assay. As expected, the co-cultures using virus-infected DCs yielded significantly higher numbers of IFN-
SFCs than the mock-treated controls (Fig. 3a
, CSFV compared with Mock; *P<0·05). The co-cultures using DCs transfected with the different CSFV protein-encoding sigE2 and NS3NS4A mRNAs resulted in significantly higher numbers of IFN-
SFCs compared with co-cultures employing eGFP-transfected DCs (Fig. 3a
, sigE2 or NS3NS4A compared with eGFP; *P<0·05).
|
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Whilst DCs represent a well-established and potent APC, a relatively new cell type has been identified with a propensity for inducing CTL activity. This is the fibrocyte, an APC derived from blood (Balmelli et al., 2005). Fibrocytes have the additional advantage that they can be cultured easily in the absence of cytokines and frozen as primary cell lines for future use. This makes them a particularly flexible alternative to monocyte-derived DCs for antigen-processing/presentation studies.
Both DCs and fibrocytes were employed for the transfection studies. The mRNA used encoded the structural E2 protein or non-structural proteins NS3NS4A of CSFV. Transfection of APCs with the mRNA encoding E2 was revealed easily, in terms of detecting the E2 protein by immunofluorescent staining. In contrast, the NS3 protein could not be detected by immunofluorescence in transfected cells, but was identified in immunoblots after in vitro translation, albeit in low quantities. Furthermore, APCs transfected with the mRNA encoding the non-structural proteins were capable of restimulating T lymphocytes from immune animals, although any quantitative comparison between the different mRNA-transfected APCs is difficult. The difficulty in detecting the viral protein in situ could be the result of viral or cellular protease digestion. Furthermore, the formation of defective ribosomal products (DRiPs) resulting from translational and post-translational errors could have been involved (Schubert et al., 2000). There is evidence showing that DRiPs are a major source of peptides presented in the context of MHC class I (Yewdell et al., 2001
) and could therefore play a role in the antigen processing of mRNA-loaded APCs.
This is the first report demonstrating CTL epitopes on CSFV E2. Flavivirus-specific CTL epitopes have mainly been identified on the viral non-structural proteins (Roehrig, 2003); nevertheless, reports demonstrating the presence of CTL epitopes on the structural proteins of Hepatitis C virus, including E2 (Koziel et al., 1993
; Sarobe et al., 2001
), are related to our study, considering that both viruses are in the family Flaviviridae.
These findings could be of importance for future vaccine design, considering the immunology of CSFV. Although the presence of neutralizing anti-E2 antibodies can be related to protective immunity against CSFV, pigs can be protected in the absence of such antibodies, such as 13 weeks after vaccination (Terpstra & Wensvoort, 1988; Dewulf et al., 2004
). Cellular immunity has been identified as early as 1 week post-vaccination, in the form of IFN-
SFCs (Suradhat et al., 2001
). The present work demonstrates that E2-specific CTL activity is particularly evident 13 weeks after challenge. With CSFV being non-cytopathogenic and the inaccessibility of the cell-associated virus to neutralizing antibodies, CTL-based immune defences have an important role to play.
Both the CTL epitopes and major epitopes for inducing neutralizing antibodies should be targeted in novel CSFV vaccines. The application of mRNA vaccines or self-replicating pestivirus replicons are known to induce potent CTL responses (Hoerr et al., 2000; Racanelli et al., 2004
). Such replicon-based vaccines have been described for classical swine fever (Widjojoatmodjo et al., 2000
; van Gennip et al., 2002
; Maurer et al., 2005
) and represent promising vaccine candidates. However, such vaccines could possibly lose immunogenicity if E2 is deleted or replaced by a heterologous E2 from another pestivirus. On the other hand, for subunit protein vaccines based on E2, the present study would indicate that protein-delivery systems suitable for the induction of both neutralizing antibodies and CTL response have to be investigated. Therein, those systems known to mediate cross-presentation, such as liposomes, bacterial toxins, heat-shock proteins and particles, are particularly interesting (Bungener et al., 2002
; Chikh & Schutze-Redelmeier, 2002
; Morón et al., 2004
).
![]() |
ACKNOWLEDGEMENTS |
---|
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Armengol, E., Wiesmüller, K.-H., Wienhold, D., Büttner, M., Pfaff, E., Jung, G. & Saalmüller, A. (2002). Identification of T-cell epitopes in the structural and non-structural proteins of classical swine fever virus. J Gen Virol 83, 551560.
Balmelli, C., Ruggli, N., McCullough, K. & Summerfield, A. (2005). Fibrocytes are potent stimulators of anti-virus cytotoxic T cells. J Leukoc Biol 77, 923933.
Bungener, L., Huckriede, A., Wilschut, J. & Daemen, T. (2002). Delivery of protein antigens to the immune system by fusion-active virosomes: a comparison with liposomes and ISCOMs. Biosci Rep 22, 323338.[CrossRef][Medline]
Carralot, J.-P., Probst, J., Hoerr, I., Scheel, B., Teufel, R., Jung, G., Rammensee, H.-G. & Pascolo, S. (2004). Polarization of immunity induced by direct injection of naked sequence-stabilized mRNA vaccines. Cell Mol Life Sci 61, 24182424.[Medline]
Carrasco, C. P., Rigden, R. C., Schaffner, R. & 7 other authors (2001). Porcine dendritic cells generated in vitro: morphological, phenotypic and functional properties. Immunology 104, 175184.[CrossRef][Medline]
Ceppi, M., Ruggli, N., Tache, V., Gerber, H., McCullough, K. C. & Summerfield, A. (2005). Double-stranded secondary structures on mRNA induce type I interferon (IFN /
) production and maturation of mRNA-transfected monocyte-derived dendritic cells. J Gene Med 7, 452465.[CrossRef][Medline]
Chikh, G. & Schutze-Redelmeier, M.-P. (2002). Liposomal delivery of CTL epitopes to dendritic cells. Biosci Rep 22, 339353.[CrossRef][Medline]
Dahle, J. & Liess, B. (1995). Assessment of safety and protective value of a cell culture modified strain "C" vaccine of hog cholera/classical swine fever virus. Berl Munch Tierarztl Wochenschr 108, 2025.[Medline]
de Bruin, T. G. M., van Rooij, E. M. A., de Visser, Y. E., Voermans, J. J. M., Samsom, J. N., Kimman, T. G. & Bianchi, A. T. J. (2000). Discrimination of different subsets of cytolytic cells in pseudorabies virus-immune and naive pigs. J Gen Virol 81, 15291537.
Dewulf, J., Laevens, H., Koenen, F., Mintiens, K. & de Kruif, A. (2004). Efficacy of E2-sub-unit marker and C-strain vaccines in reducing horizontal transmission of classical swine fever virus in weaner pigs. Prev Vet Med 65, 121133.[CrossRef][Medline]
Greiser-Wilke, I., Moennig, V., Coulibaly, C. O. Z., Dahle, J., Leder, L. & Liess, B. (1990). Identification of conserved epitopes on a hog cholera virus protein. Arch Virol 111, 213225.[CrossRef][Medline]
Greiser-Wilke, I., Dittmar, K. E., Liess, B. & Moennig, V. (1992). Heterogeneous expression of the non-structural protein p80/p125 in cells infected with different pestiviruses. J Gen Virol 73, 4752.[Abstract]
Hoerr, I., Obst, R., Rammensee, H.-G. & Jung, G. (2000). In vivo application of RNA leads to induction of specific cytotoxic T lymphocytes and antibodies. Eur J Immunol 30, 17.[CrossRef][Medline]
Knoetig, S. M., Summerfield, A., Spagnuolo-Weaver, M. & McCullough, K. C. (1999). Immunopathogenesis of classical swine fever: role of monocytic cells. Immunology 97, 359366.[CrossRef][Medline]
Koziel, M. J., Dudley, D., Afdhal, N., Choo, Q.-L., Houghton, M., Ralston, R. & Walker, B. D. (1993). Hepatitis C virus (HCV)-specific cytotoxic T lymphocytes recognize epitopes in the core and envelope proteins of HCV. J Virol 67, 75227532.[Abstract]
Maurer, R., Stettler, P., Ruggli, N., Hofmann, M. A. & Tratschin, J. D. (2005). Oronasal vaccination with classical swine fever virus (CSFV) replicon particles with either partial or complete deletion of the E2 gene induces partial protection against lethal challenge with highly virulent CSFV. Vaccine 23, 33183328.[CrossRef][Medline]
McCullough, K. C., Schaffner, R., Fraefel, W. & Kihm, U. (1993). The relative density of CD44-positive porcine monocytic cell populations varies between isolations and upon culture and influences susceptibility to infection by African swine fever virus. Immunol Lett 37, 8390.[CrossRef][Medline]
Meyers, G. & Thiel, H.-J. (1996). Molecular characterization of pestiviruses. Adv Virus Res 47, 53118.[Medline]
Meyers, G., Rümenapf, T. & Thiel, H.-J. (1989). Molecular cloning and nucleotide sequence of the genome of hog cholera virus. Virology 171, 555567.[CrossRef][Medline]
Morón, G., Dadaglio, G. & Leclerc, C. (2004). New tools for antigen delivery to the MHC class I pathway. Trends Immunol 25, 9297.[CrossRef][Medline]
Pauly, T., Elbers, K., König, M., Lengsfeld, T., Saalmüller, A. & Thiel, H.-J. (1995). Classical swine fever virus-specific cytotoxic T lymphocytes and identification of a T cell epitope. J Gen Virol 76, 30393049.[Abstract]
Peeters, B., de Wind, N., Broer, R., Gielkens, A. & Moormann, R. (1992). Glycoprotein H of pseudorabies virus is essential for entry and cell-to-cell spread of the virus. J Virol 66, 38883892.[Abstract]
Ponsaerts, P., Van Tendeloo, V. F. I. & Berneman, Z. N. (2003). Cancer immunotherapy using RNA-loaded dendritic cells. Clin Exp Immunol 134, 378384.[CrossRef][Medline]
Racanelli, V., Behrens, S.-E., Aliberti, J. & Rehermann, B. (2004). Dendritic cells transfected with cytopathic self-replicating RNA induce crosspriming of CD8+ T cells and antiviral immunity. Immunity 20, 4758.[CrossRef][Medline]
Roehrig, J. T. (2003). Antigenic structure of flavivirus proteins. Adv Virus Res 59, 141175.[CrossRef][Medline]
Ruggli, N., Tratschin, J. D., Mittelholzer, C. & Hofmann, M. A. (1996). Nucleotide sequence of classical swine fever virus strain Alfort/187 and transcription of infectious RNA from stably cloned full-length cDNA. J Virol 70, 34783487.[Abstract]
Sarobe, P., Huarte, E., Lasarte, J. J., López-Díaz de Cerio, A., García, N., Borrás-Cuesta, F. & Prieto, J. (2001). Characterization of an immunologically conserved epitope from hepatitis C virus E2 glycoprotein recognized by HLA-A2 restricted cytotoxic T lymphocytes. J Hepatol 34, 321329.[CrossRef][Medline]
Schubert, U., Antón, L. C., Gibbs, J., Norbury, C. C., Yewdell, J. W. & Bennink, J. R. (2000). Rapid degradation of a large fraction of newly synthesized proteins by proteasomes. Nature 404, 770774.[CrossRef][Medline]
Strobel, I., Berchtold, S., Götze, A., Schulze, U., Schuler, G. & Steinkasserer, A. (2000). Human dendritic cells transfected with either RNA or DNA encoding influenza matrix protein M1 differ in their ability to stimulate cytotoxic T lymphocytes. Gene Ther 7, 20282035.[CrossRef][Medline]
Suradhat, S., Intrakamhaeng, M. & Damrongwatanapokin, S. (2001). The correlation of virus-specific interferon-gamma production and protection against classical swine fever virus infection. Vet Immunol Immunopathol 83, 177189.[CrossRef][Medline]
Terpstra, C. & Wensvoort, G. (1988). The protective value of vaccine-induced neutralising antibody titres in swine fever. Vet Microbiol 16, 123128.[CrossRef][Medline]
Terpstra, C., Woortmeyer, R. & Barteling, S. J. (1990). Development and properties of a cell culture produced vaccine for hog cholera based on the Chinese strain. Dtsch Tierarztl Wochenschr 97, 7779.[Medline]
Thornburg, C., Boczkowski, D., Gilboa, E. & Nair, S. K. (2000). Induction of cytotoxic T lymphocytes with dendritic cells transfected with human papillomavirus E6 and E7 RNA: implications for cervical cancer immunotherapy. J Immunother 23, 412418.[CrossRef][Medline]
Tuyaerts, S., Michiels, A., Corthals, J., Bonehill, A., Heirman, C., De Greef, C., Noppe, S. M. & Thielemans, K. (2003). Induction of influenza matrix protein 1 and MelanA-specific T lymphocytes in vitro using mRNA-electroporated dendritic cells. Cancer Gene Ther 10, 696706.[CrossRef][Medline]
Ueno, H., Tcherepanova, I., Reygrobellet, O., Laughner, E., Ventura, C., Palucka, A. K. & Banchereau, J. (2004). Dendritic cell subsets generated from CD34+ hematopoietic progenitors can be transfected with mRNA and induce antigen-specific cytotoxic T cell responses. J Immunol Methods 285, 171180.[CrossRef][Medline]
van Gennip, H. G. P., Bouma, A., van Rijn, P. A., Widjojoatmodjo, M. N. & Moormann, R. J. M. (2002). Experimental non-transmissible marker vaccines for classical swine fever (CSF) by trans-complementation of Erns or E2 of CSFV. Vaccine 20, 15441556.[CrossRef][Medline]
van Oirschot, J. T. (2003). Vaccinology of classical swine fever: from lab to field. Vet Microbiol 96, 367384.[CrossRef][Medline]
Van Tendeloo, V. F. I., Ponsaerts, P., Lardon, F., Nijs, G., Lenjou, M., Van Broekhoven, C., Van Bockstaele, D. R. & Berneman, Z. N. (2001). Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells: superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. Blood 98, 4956.
Weissman, D., Ni, H., Scales, D. & 7 other authors (2000). HIV gag mRNA transfection of dendritic cells (DC) delivers encoded antigen to MHC class I and II molecules, causes DC maturation, and induces a potent human in vitro primary immune response. J Immunol 165, 47104717.
Wensvoort, G., Terpstra, C., De Kluijver, E. P., Kragten, C. & Warnaar, J. C. (1989). Antigenic differentiation of pestivirus strains with monoclonal antibodies against hog cholera virus. Vet Microbiol 21, 920.[CrossRef][Medline]
Widjojoatmodjo, M. N., van Gennip, H. G. P., Bouma, A., van Rijn, P. A. & Moormann, R. J. M. (2000). Classical swine fever virus Erns deletion mutants: trans-complementation and potential use as nontransmissible, modified, live-attenuated marker vaccines. J Virol 74, 29732980.
Yewdell, J. W., Schubert, U. & Bennink, J. R. (2001). At the crossroads of cell biology and immunology: DRiPs and other sources of peptide ligands for MHC class I molecules. J Cell Sci 114, 845851.
Zarei, S., Arrighi, J.-F., Ongaro, G., Calzascia, T., Haller, O., Frossard, C., Piguet, V., Walker, P. R. & Hauser, C. (2003). Efficient induction of CD8 T-associated immune protection by vaccination with mRNA transfected dendritic cells. J Invest Dermatol 121, 745750.[CrossRef][Medline]
Received 20 January 2005;
accepted 3 June 2005.
HOME | HELP | FEEDBACK | SUBSCRIPTIONS | ARCHIVE | SEARCH | TABLE OF CONTENTS |
INT J SYST EVOL MICROBIOL | MICROBIOLOGY | J GEN VIROL |
J MED MICROBIOL | ALL SGM JOURNALS |