Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Virol J ; 21(1): 57, 2024 03 06.
Article in English | MEDLINE | ID: mdl-38448981

ABSTRACT

BACKGROUND: Non-pharmaceutical interventions implemented during the COVID-19 pandemic resulted in a marked reduction in influenza infections globally. The absence of influenza has raised concerns of waning immunity, and potentially more severe influenza seasons after the pandemic. METHODS: To evaluate immunity towards influenza post-COVID-19 pandemic we have assessed influenza A epidemics in Norway from October 2016 to June 2023 and measured antibodies against circulating strains of influenza A(H1N1)pdm09 and A(H3N2) in different age groups by hemagglutination inhibition (HAI) assays in a total of 3364 serum samples collected in 2019, 2021, 2022 and 2023. RESULTS: Influenza epidemics in Norway from October 2016 until June 2023 were predominately influenza As, with a mixture of A(H1N1)pdm09 and A(H3N2) subtype predominance. We did not observe higher numbers of infections during the influenza epidemics following the COVID-19 pandemic than in pre-COVID-19 seasons. Frequencies of protective HAI titers against A(H1N1)pdm09 and A(H3N2) viruses were reduced in sera collected in 2021 and 2022, compared to sera collected in 2019. The reduction could, however, largely be explained by antigenic drift of new virus strains, as protective HAI titers remained stable against the same strain from one season to the next. However, we observed the development of an immunity gap in the youngest children during the pandemic which resulted in a prominent reduction in HAI titers against A(H1N1)pdm09 in 2021 and 2022. The immunity gap was partially closed in sera collected in 2023 following the A(H1N1)pdm09-dominated influenza seasons of 2022/2023. During the 2022/2023 epidemic, drift variants of A(H1N1)pdm09 belonging to the 5a.2a.1 clade emerged, and pre-season HAI titers were significantly lower against this clade compared to the ancestral 5a.2 clade. CONCLUSION: The observed reduction in protective antibodies against A(H1N1)pdm09 and A(H3N2) viruses post COVID-19 is best explained by antigenic drift of emerging viruses, and not waning of antibody responses in the general population. However, the absence of influenza during the pandemic resulted in an immunity gap in the youngest children. While this immunity gap was partially closed following the 2022/2023 influenza season, children with elevated risk of severe infection should be prioritized for vaccination.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Child , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Cross-Sectional Studies , Antigenic Drift and Shift , Influenza A Virus, H3N2 Subtype , COVID-19/epidemiology , Pandemics
3.
PNAS Nexus ; 2(12): pgad403, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38077689

ABSTRACT

Immunocompromised patients often fail to raise protective vaccine-induced immunity against the global emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Although monoclonal antibodies have been authorized for clinical use, most have lost their ability to potently neutralize the evolving Omicron subvariants. Thus, there is an urgent need for treatment strategies that can provide protection against these and emerging SARS-CoV-2 variants to prevent the development of severe coronavirus disease 2019. Here, we report on the design and characterization of a long-acting viral entry-blocking angiotensin-converting enzyme 2 (ACE2) dimeric fusion molecule. Specifically, a soluble truncated human dimeric ACE2 variant, engineered for improved binding to the receptor-binding domain of SARS-CoV-2, was fused with human albumin tailored for favorable engagement of the neonatal fragment crystallizable receptor (FcRn), which resulted in enhanced plasma half-life and allowed for needle-free transmucosal delivery upon nasal administration in human FcRn-expressing transgenic mice. Importantly, the dimeric ACE2-fused albumin demonstrated potent neutralization of SARS-CoV-2 immune escape variants.

4.
J Immunother Cancer ; 11(8)2023 08.
Article in English | MEDLINE | ID: mdl-37607769

ABSTRACT

BACKGROUND: Multiple myeloma (MM) cancers originate from plasma cells that have passed through the germinal center reaction where somatic hypermutation of Ig V regions takes place. Myeloma protein V regions often express many mutations and are thus a rich source of neoantigens (traditionally called idiotopes (Id)). Therefore, these are highly tumor-specific and excellent targets for immunotherapy. METHODS: We have developed a DNA Id vaccine which as translated protein targets conventional dendritic cells (cDC) for CCL3-mediated delivery of myeloma protein V regions in a single-chain fragment variable (scFv) format. Vaccine efficacy was studied in the mouse MM model, mineral oil-induced plasmacytoma 315.BM. RESULTS: The Id vaccine protected mice against a challenge with MM cells. Moreover, the vaccine had a therapeutic effect. However, in some of the vaccinated mice, MM cells not producing H chains escaped rejection, resulting in free light chain (FLC) MM. Depletion of CD8+ T cells abrogated vaccine efficacy, and protection was observed to be dependent on cDC1s, using Batf3-/- mice. Modifications of scFv in the vaccine demonstrated that CD8+ T cells were specific for two mutated VH sequences. CONCLUSIONS: VH neoantigen-specific CD8+ T cells elicited by CCL3-containing Id vaccines had a therapeutic effect against MM in a mouse model. MM cells could escape rejection by losing expression of the H chain, thus giving rise to FLC MM.


Subject(s)
Multiple Myeloma , Vaccines, DNA , Animals , Mice , Multiple Myeloma/therapy , CD8-Positive T-Lymphocytes , Immunotherapy , Dendritic Cells
5.
Influenza Other Respir Viruses ; 17(6): e13144, 2023 06.
Article in English | MEDLINE | ID: mdl-37273461

ABSTRACT

New immune evasive variants of SARS-CoV-2 continue to emerge, potentially causing new waves of covid-19 disease. Here, we evaluate levels of neutralizing antibodies against isolates of Omicron variants, including BQ.1.1 and XBB, in sera harvested 3-4 weeks after vaccination or breakthrough infections. In addition, we evaluate neutralizing antibodies in 32 sera from October 2022, to evaluate immunity in Norwegian donors prior to the winter season. Most serum samples harvested in October 2022 had low levels of neutralizing antibodies against BQ.1.1 and especially XBB, explaining why these variants and their descendants have dominated in Norway during the 2022 and 2023 winter season.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/epidemiology , Norway/epidemiology , Antibodies, Neutralizing , Antibodies, Viral
6.
Mol Ther ; 31(7): 2188-2205, 2023 07 05.
Article in English | MEDLINE | ID: mdl-36926694

ABSTRACT

Conventional influenza vaccines focus on hemagglutinin (HA). However, antibody responses to neuraminidase (NA) have been established as an independent correlate of protection. Here, we introduced the ectodomain of NA into DNA vaccines that, as translated dimeric vaccine proteins, target antigen-presenting cells (APCs). The targeting was mediated by an single-chain variable fragment specific for major histocompatibility complex (MHC) class II, which is genetically linked to NA via a dimerization motif. A single immunization of BALB/c mice elicited strong and long-lasting NA-specific antibodies that inhibited NA enzymatic activity and reduced viral replication. Vaccine-induced NA immunity completely protected against a homologous influenza virus and out-competed NA immunity induced by a conventional inactivated virus vaccine. The protection was mainly mediated by antibodies, although NA-specific T cells also contributed. APC-targeting and antigen bivalency were crucial for vaccine efficacy. The APC-targeted vaccine was potent at low doses of DNA, indicating a dose-sparing effect. Similar results were obtained with NA vaccines that targeted different surface molecules on dendritic cells. Interestingly, the protective efficacy of NA as antigen compared favorably with HA and therefore ought to receive more attention in influenza vaccine research.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Vaccines, DNA , Animals , Mice , Humans , Influenza, Human/prevention & control , Neuraminidase/genetics , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Histocompatibility Antigens Class II , DNA , Mice, Inbred BALB C
7.
Cell Rep ; 42(2): 112123, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36795562

ABSTRACT

The contribution of cross-presenting XCR1+ dendritic cells (DCs) and SIRPα+ DCs in maintaining T cell function during exhaustion and immunotherapeutic interventions of chronic infections remains poorly characterized. Using the mouse model of chronic LCMV infection, we found that XCR1+ DCs are more resistant to infection and highly activated compared with SIRPα+ DCs. Exploiting XCR1+ DCs via Flt3L-mediated expansion or XCR1-targeted vaccination notably reinvigorates CD8+ T cells and improves virus control. Upon PD-L1 blockade, XCR1+ DCs are not required for the proliferative burst of progenitor exhausted CD8+ T (TPEX) cells but are indispensable to sustain the functionality of exhausted CD8+ T (TEX) cells. Combining anti-PD-L1 therapy with increased frequency of XCR1+ DCs improves functionality of TPEX and TEX subsets, while increase of SIRPα+ DCs dampened their proliferation. Together, this demonstrates that XCR1+ DCs are crucial for the success of checkpoint inhibitor-based therapies through differential activation of exhausted CD8+ T cell subsets.


Subject(s)
Cross-Priming , Virus Diseases , Mice , Animals , Dendritic Cells , CD8-Positive T-Lymphocytes , Immunotherapy
8.
Cell Rep ; 39(9): 110901, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35649357

ABSTRACT

Antibodies are important for vaccine efficacy. Targeting antigens to antigen-presenting cells (APCs) increases antibody levels. Here, we explore the role of antigen valency in MHC class II (MHCII)-targeted vaccines delivered as DNA. We design heterodimeric proteins that carry either two identical (bivalent vaccines), or two different antigens (monovalent vaccines). Bivalent vaccines with two identical influenza hemagglutinins (HA) elicit higher amounts of anti-HA antibodies in mice than monovalent versions with two different HAs. Bivalent vaccines increase the levels of germinal center (GC) B cells and long-lived plasma cells. Only HA-bivalent vaccines completely protect mice against challenge with homologous influenza virus. Similar results are obtained with other antigens by targeting CD11c and Xcr1 on dendritic cells (DCs) or when administering the vaccine as protein with adjuvant. Bivalency probably increases B cell responses by cross-linking BCRs in readily observable DC-B cell synapses. These results are important for generating potent APC-targeted vaccines.


Subject(s)
Cancer Vaccines , Influenza Vaccines , Vaccines, DNA , Animals , Antibodies, Viral , Antigen-Presenting Cells , Hemagglutinins , Mice , Vaccines, Combined , Vaccines, DNA/genetics
9.
Front Immunol ; 13: 752714, 2022.
Article in English | MEDLINE | ID: mdl-35296089

ABSTRACT

Targeting antigen to conventional dendritic cells (cDCs) can improve antigen-specific immune responses and additionally be used to influence the polarization of the immune responses. However, the mechanisms by which this is achieved are less clear. To improve our understanding, we here evaluate molecular and cellular requirements for CD4+ T cell and antibody polarization after immunization with Xcl1-fusion vaccines that specifically target cDC1s. Xcl1-fusion vaccines induced an IgG2a/IgG2b-dominated antibody response and rapid polarization of Th1 cells both in vitro and in vivo. For comparison, we included fliC-fusion vaccines that almost exclusively induced IgG1, despite inducing a more mixed polarization of T cells. Th1 polarization and IgG2a induction with Xcl1-fusion vaccines required IL-12 secretion but were nevertheless maintained in BATF3-/- mice which lack IL-12-secreting migratory DCs. Interestingly, induction of IgG2a-dominated responses was highly dependent on the early kinetics of Th1 induction and was important for optimal protection in an influenza infection model. Early Th1 induction was dominant, since a combined Xcl1- and fliC-fusion vaccine induced IgG2a/IgG2b polarized antibody responses similar to Xcl1-fusion vaccines alone. In summary, our results demonstrate that targeting antigen to Xcr1+ cDC1s is an efficient strategy for enhancing IgG2a antibody responses through rapid Th1 induction, which can be utilized for improved vaccine design.


Subject(s)
Influenza Vaccines , Influenza, Human , Animals , Antibody Formation , Antigens , Dendritic Cells , Humans , Immunoglobulin G , Interleukin-12 , Mice
10.
Scand J Immunol ; 95(3): e13128, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34923667

ABSTRACT

Targeting antigens to dendritic cells represent a promising method for enhancing immune responses against specific antigens. However, many studies have focused on systemic delivery (intravenous or intraperitoneally) of targeted antigen, approaches that are not easily transferable to humans. Here we evaluate the efficacy of an influenza vaccine targeting Xcr1+ cDC1 administered by intranasal immunization. Intranasal delivery of antigen fused to the chemokine Xcl1, the ligand of Xcr1, resulted in specific uptake by lung CD103+ cDC1. Interestingly, intranasal immunization with influenza A/PR/8/34 haemagglutinin (HA) fused to Xcl1, formulated with poly(I:C), resulted in enhanced induction of antigen-specific IFNγ+ CD4+ and IFNγ+ CD8+ T cell responses in lung compared non-targeted anti-NIP-HA (αNIP-HA). Induction of antibody responses was, however, similar in Xcl1-HA and αNIP-HA immunized mice, but significantly higher than in mice immunized with monomeric HA. Both Xcl1-HA and αNIP-HA vaccines induced full protection when mice were challenged with a lethal dose of influenza PR8 virus, reflecting the strong induction of HA-specific antibodies. Our results demonstrate that i.n. delivery of Xcl1-HA is a promising vaccine strategy for enhancing T cell responses in addition to inducing strong antibody responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, C/metabolism , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Poly I-C/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Antigens/immunology , Antigens, CD/immunology , Cell Line , Dendritic Cells/immunology , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Integrin alpha Chains/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C
11.
EMBO Rep ; 22(4): e52196, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33719206

ABSTRACT

T and B cells continually recirculate between blood and secondary lymphoid organs. To promote their trans-endothelial migration (TEM), chemokine receptors control the activity of RHO family small GTPases in part via GTPase-activating proteins (GAPs). T and B cells express several RHO-GAPs, the function of most of which remains unknown. The ARHGAP45 GAP is predominantly expressed in hematopoietic cells. To define its in vivo function, we describe two mouse models where ARHGAP45 is ablated systemically or selectively in T cells. We combine their analysis with affinity purification coupled to mass spectrometry to determine the ARHGAP45 interactome in T cells and with time-lapse and reflection interference contrast microscopy to assess the role of ARGHAP45 in T-cell polarization and motility. We demonstrate that ARHGAP45 regulates naïve T-cell deformability and motility. Under physiological conditions, ARHGAP45 controls the entry of naïve T and B cells into lymph nodes whereas under competitive repopulation it further regulates hematopoietic progenitor cell engraftment in the bone marrow, and T-cell progenitor thymus seeding. Therefore, the ARGHAP45 GAP controls multiple key steps in the life of T and B cells.


Subject(s)
T-Lymphocytes , Virus Internalization , Animals , B-Lymphocytes , Cell Movement , GTPase-Activating Proteins/genetics , Lymph Nodes , Mice , Thymus Gland
12.
J Immunol ; 205(3): 661-673, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591401

ABSTRACT

Targeting Ag to surface receptors on conventional type 1 dendritic cells can enhance induction of Ab and T cell responses. However, it is unclear to what extent the targeted receptor influences the resulting responses. In this study, we target Ag to Xcr1, Clec9A, or DEC-205, surface receptors that are expressed on conventional type 1 dendritic cells, and compare immune responses in BALB/c and C57BL/6 mice in vitro and in vivo after intradermal DNA vaccination. Targeting hemagglutinin from influenza A to Clec9A induced Ab responses with higher avidity that more efficiently neutralized influenza virus compared with Xcr1 and DEC-205 targeting. In contrast, targeting Xcr1 resulted in higher IFN-γ+CD8+ T cell responses in spleen and lung and stronger cytotoxicity. Both Clec9A and Xcr1 targeting induced Th1-polarized Ab responses, although the Th1 polarization of CD4+ T cells was more pronounced after Xcr1 targeting. Targeting DEC-205 resulted in poor Ab responses in BALB/c mice and a more mixed Th response. In an influenza challenge model, targeting either Xcr1 or Clec9A induced full and long-term protection against influenza infection, whereas only partial short-term protection was obtained when targeting DEC-205. In summary, the choice of targeting receptor, even on the same dendritic cell subpopulation, may strongly influence the resulting immune response, suggesting that different targeting strategies should be considered depending on the pathogen.


Subject(s)
Antigens, CD/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Receptors, Chemokine/immunology , Receptors, Immunologic/immunology , Th1 Cells/immunology , Animals , Female , HEK293 Cells , Humans , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C
13.
Sci Rep ; 10(1): 5944, 2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32231213

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Mol Ther Methods Clin Dev ; 17: 378-392, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32128342

ABSTRACT

Immunogenicity of DNA vaccines can be increased by constructing the DNA in such a way that it encodes secreted homodimeric fusion proteins that target antigen-presenting cells (APCs). In this study, we have developed novel APC-targeting vaccine molecules with an increased flexibility due to introduction of a heterodimerization motif. The heterodimeric proteins permit four different fusions within a single molecule, thus allowing expression of two different APC-targeting moieties and two different antigens. Two types of heterodimeric fusion proteins were developed that employed either the ACID/BASE or the Barnase/Barstar motifs, respectively. The ACID/BASE heterodimeric vaccines conferred protection against challenges with either influenza virus or tumor cells in separate preclinical models. The ACID/BASE motif was flexible since a large number of different targeting moieties and antigens could be introduced with maintenance of specificity, antigenicity, and secretion. APC-targeting ACID/BASE vaccines expressing two different antigens induced antibody and T cell responses against either of the two antigens. Heterodimeric ACID/BASE DNA vaccines were of approximately the same potency as previously reported homodimeric DNA vaccines. The flexibility and potency of the ACID/BASE format suggest that it could be a useful platform for DNA vaccines that encode APC-targeting fusion proteins.

15.
Vaccine ; 38(6): 1280-1285, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31836256

ABSTRACT

Efficient influenza vaccination of pigs can reduce disease burdens for the swine industry, but also represents an important measure for reducing the risk from novel viral reassortments that pose pandemic threats to the human population. Here, we have vaccinated pigs with a DNA vaccine encoding influenza virus hemagglutinin (HA) linked to the chemokine MIP1α that bind chemokine receptors 1, 3, and 5 expressed on antigen presenting cells (APC). Such MIP1α targeting of HA to APC enhanced induction of HA reactive antibodies, particularly IgG2. In addition, the MIP1α- HA vaccine induced strong T cell responses that could cross-react with different influenza subtypes. Thus, the strategy of targeting HA to chemokine receptors could be important for inducing broad protection against antigenically diverse influenza strains in pigs.


Subject(s)
Cross Protection , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Viral/immunology , Antigen-Presenting Cells/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Swine , Vaccines, DNA/immunology
16.
Front Immunol ; 10: 1529, 2019.
Article in English | MEDLINE | ID: mdl-31333661

ABSTRACT

Dendritic cells (DCs) facilitate cross talk between the innate and adaptive immune system. They sense and phagocytose invading pathogens, and are not only capable of activating naïve T cells, but can also determine the polarization of T cell responses into different effector subtypes. Polarized T cells in turn have a crucial role in antibody class switching and affinity maturation, and consequently the quality of the resulting humoral immunity. Targeting vaccines to DCs thus provides a great deal of opportunities for influencing the humoral immune responses, by fine-tuning the T cell response as well as regulating antigen availability for B cells. In this review we aim to outline how different DC targeted vaccination strategies can be utilized to induce a desired humoral immune response. A range of factors, including route of vaccine administration, use of adjuvants, choice of DC subset and surface receptor to target have been reported to influence the resulting immune response and will be reviewed herein. Finally, we will discuss opportunities for designing improved vaccines and challenges with translating this knowledge into clinical or veterinary medicine.


Subject(s)
Antibody Formation/drug effects , Dendritic Cells/immunology , Drug Delivery Systems , Immunity, Humoral/drug effects , Vaccination , Vaccines , Animals , B-Lymphocytes/immunology , Gene Rearrangement, B-Lymphocyte/drug effects , Gene Rearrangement, B-Lymphocyte/immunology , Humans , T-Lymphocytes/immunology , Vaccines/immunology , Vaccines/therapeutic use
17.
Front Immunol ; 10: 1086, 2019.
Article in English | MEDLINE | ID: mdl-31156636

ABSTRACT

Targeting antigen to surface receptors on dendritic cells (DCs) can improve antibody response against subunit vaccines. We have previously observed that human XCL1-fusion vaccines target murine Xcr1+ DCs without actively inducing endocytosis of the antigen, resulting in enhanced antibody responses in mice. However, the use of foreign chemokines for targeting is undesirable when translating this observation to human or veterinary medicine due to potential cross-reactive responses against the endogenous chemokine. Here we have identified a mutant version of murine Xcl1, labeled Xcl1(Δ1) owing to removal of a conserved valine in position 1 of the mature chemokine, that retains specific binding to Xcr1+ DCs without inducing endocytosis of the receptor. DNA immunization with Xcl1(Δ1) conjugated to influenza hemagglutinin (HA) induced improved antibody responses, with higher end point titers of IgG compared to WT Xcl1-HA. The Xcl1(Δ1) fusion vaccine also resulted in an increased number of HA reactive germinal center B cells with higher avidity toward the antigen, and serum transfer experiments show that Xcl1(Δ1)-HA induced antibody responses provided better protection against influenza infection as compared to WT Xcl1-HA. In summary, our observations indicate that targeting antigen to Xcr1+ DCs in an endocytosis deficient manner enhances antibody responses. This effect was obtained by introducing a single mutation to Xcl1, suggesting our strategy may easily be translated to human or veterinary vaccine settings.


Subject(s)
Antibodies, Viral/metabolism , Chemokines, C/metabolism , Dendritic Cells/immunology , Influenza Vaccines/metabolism , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Recombinant Fusion Proteins/metabolism , Animals , Antibody Formation , Chemokines, C/chemistry , Chemokines, C/genetics , Endocytosis , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Influenza Vaccines/chemistry , Influenza Vaccines/genetics , Mice , Mutation/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Vaccines, Subunit
18.
Sci Rep ; 9(1): 1820, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755656

ABSTRACT

Fusing antigens to chemokines to target antigen presenting cells (APC) is a promising method for enhancing immunogenicity of DNA vaccines. However, it is unclear how different chemokines compare in terms of immune potentiating effects. Here we compare Ccl3- and Xcl1-fusion vaccines containing hemagglutinin (HA) from influenza A delivered by intramuscular (i.m.) or intradermal (i.d.) DNA vaccination. Xcl1 fusion vaccines target cDC1s, and enhance proliferation of CD4+ and CD8+ T cells in vitro. In contrast, Ccl3 target both cDC1 and cDC2, but only enhance CD4+ T cell proliferation in combination with cDC2. When Ccl3- or Xcl1-HA fusion vaccines were administered by i.m. DNA immunization, both vaccines induced Th1-polarized immune responses with antibodies of the IgG2a/IgG2b subclass and IFNγ-secreting T cells. After i.d. DNA vaccination, however, only Xcl1-HA maintained a Th1 polarized response and induced even higher numbers of IFNγ-secreting T cells. Consequently, Xcl1-HA induced superior protection against influenza infection compared to Ccl3-HA after i.d. immunization. Interestingly, i.m. immunization with Ccl3-HA induced the strongest overall in vivo cytotoxicity, despite not inducing OT-I proliferation in vitro. In summary, our results highlight important differences between Ccl3- and Xcl1- targeted DNA vaccines suggesting that chemokine fusion vaccines can be tailor-made for different diseases.


Subject(s)
Chemokine CCL3/metabolism , Chemokines, C/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Vaccines, DNA/immunology , Animals , Female , Immunoglobulin G/metabolism , Mice , Mice, Inbred BALB C
19.
Immunohorizons ; 2(1): 38-53, 2018 01 18.
Article in English | MEDLINE | ID: mdl-31022690

ABSTRACT

Upon APC-targeted DNA vaccination, transfected cells secrete fusion proteins with targeting units specific for surface molecules on APC. In this study, we have tested several different targeting units for their ability to influence the magnitude and subclass of Ab responses to hemagglutinin from influenza A virus. The experiments employed bivalent homodimeric Ig-based molecules (vaccibodies). The overall efficiency in BALB/c mice depended on the targeting units in the following order: αMHC class II > αCD11c > αCD40 > Xcl-1 = MIP-1α > FliC > GM-CSF > Flt-3L > αDEC205. GM-CSF induced mainly IgG1, whereas Xcl1, MIP-1α, αCD40, and αDEC205 induced predominantly IgG2a. A more balanced mixture of IgG1 and IgG2a was observed with αCD11c, αMHC class II, Flt-3L, and FliC. Similar results of IgG subclass-skewing were obtained in Th1-prone C57BL/6 mice with a more limited panel of vaccines. IgG1 responses in BALB/c occurred early after immunization but declined relatively rapidly over time. IgG2a responses appeared later but lasted longer (>252 d) than IgG1 responses. The most efficient targeting units elicited short- and long-term protection against PR8 influenza (H1N1) virus in BALB/c mice. The results suggest that targeting of Xcr1+ conventional type 1 dendritic cells preferentially induces IgG2a responses, whereas simultaneous targeting of several dendritic cell subtypes also induces IgG1 responses. The induction of distinct subclass profiles by different surface molecules supports the APC-B cell synapse hypothesis. The results may contribute to generation of more potent DNA vaccines that elicit high levels of Abs with desired biologic effector functions.


Subject(s)
Antigen-Presenting Cells/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/biosynthesis , Antibody Formation , Cell Line , Dendritic Cells/immunology , HEK293 Cells , Hemagglutinins/immunology , Humans , Influenza Vaccines/genetics , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transfection
20.
Sci Rep ; 7(1): 7639, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28794452

ABSTRACT

XCR1 is selectively expressed on a conventional dendritic cell subset, the cDC1 subset, through phylogenetically distant species. The outcome of antigen-targeting to XCR1 may therefore be similar across species, permitting the translation of results from experimental models to human and veterinary applications. Here we evaluated in pigs the immunogenicity of bivalent protein structures made of XCL1 fused to the external portion of the influenza virus M2 proton pump, which is conserved through strains and a candidate for universal influenza vaccines. Pigs represent a relevant target of such universal vaccines as pigs can be infected by swine, human and avian strains. We found that cDC1 were the only cell type labeled by XCR1-targeted mCherry upon intradermal injection in pig skin. XCR1-targeted M2e induced higher IgG responses in seronegative and seropositive pigs as compared to non-targeted M2e. The IgG response was less significantly enhanced by CpG than by XCR1 targeting, and CpG did not further increase the response elicited by XCR1 targeting. Monophosphoryl lipid A with neutral liposomes did not have significant effect. Thus altogether M2e-targeting to XCR1 shows promises for a trans-species universal influenza vaccine strategy, possibly avoiding the use of classical adjuvants.


Subject(s)
Antibody Formation , Chemokines, C/metabolism , Dendritic Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Recombinant Fusion Proteins/immunology , Skin/immunology , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Chemokines, C/administration & dosage , Chemokines, C/genetics , Dendritic Cells/metabolism , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Oligodeoxyribonucleotides/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Skin/metabolism , Swine , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...