1
|
LENG D, YAMADA S, CHIBA Y, YONEYAMA S, SAKAI Y, HIKONO H, MURAKAMI K. Co-administration of a plasmid encoding CD40 or CD63 enhances the immune responses to a DNA vaccine against bovine viral diarrhea virus in mice. J Vet Med Sci 2022; 84:1175-1184. [PMID: 35793950 PMCID: PMC9523294 DOI: 10.1292/jvms.22-0085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/26/2022] [Indexed: 11/30/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) causes substantial economic losses in the livestock industry worldwide. Plasmids encoding the BVDV E2 protein are potential DNA vaccines against BVDV, but their immunogenicity has been insufficient. Here, we investigated the adjuvant effect of CD40 and CD63 plasmids on the immune responses to a BVDV E2 DNA vaccine in mice. We constructed pUMVC4a-based plasmids encoding the BVDV E2 protein (pE2), mouse CD40 (pCD40), or mouse CD63 (pCD63). Protein expression by each plasmid was confirmed through Western blot analysis and immunofluorescence staining of cultured cell lines. BALB/c mice were immunized intradermally twice with pE2 in combination with, or without, pCD40 or pCD63, with 3 weeks between the two doses. pE2 with pCD40 induced significantly higher neutralizing antibody titers against BVDV than pE2 alone. pE2 with pCD63 induced significantly higher anti-E2 IgG2a antibody titers than pE2 alone. Furthermore, pE2 with pCD40 or pCD63 induced significantly increased lymphocyte proliferation and interferon (IFN)-γ production in response to BVDV, compared with E2 alone. These results suggest that a plasmid encoding CD40 or CD63 can be used as an adjuvant to enhance immune responses to DNA vaccines against BVDV.
Collapse
Affiliation(s)
- Dongze LENG
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Shinji YAMADA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Yusuke CHIBA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Syuji YONEYAMA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Yusuke SAKAI
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | | | - Kenji MURAKAMI
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| |
Collapse
|
2
|
Abstract
Immune principles formulated by Jenner, Pasteur, and early immunologists served as fundamental propositions for vaccine discovery against many dreadful pathogens. However, decisive success in the form of an efficacious vaccine still eludes for diseases such as tuberculosis, leishmaniasis, and trypanosomiasis. Several antileishmanial vaccine trials have been undertaken in past decades incorporating live, attenuated, killed, or subunit vaccination, but the goal remains unmet. In light of the above facts, we have to reassess the principles of vaccination by dissecting factors associated with the hosts' immune response. This chapter discusses the pathogen-associated perturbations at various junctures during the generation of the immune response which inhibits antigenic processing, presentation, or remodels memory T cell repertoire. This can lead to ineffective priming or inappropriate activation of memory T cells during challenge infection. Thus, despite a protective primary response, vaccine failure can occur due to altered immune environments in the presence of pathogens.
Collapse
Affiliation(s)
| | - Sunil Kumar
- National Centre for Cell Science, Pune, Maharashtra, India
| | | | - Bhaskar Saha
- National Centre for Cell Science, Pune, Maharashtra, India.
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India.
| |
Collapse
|
3
|
Immunization of turkeys with a DNA vaccine expressing the haemagglutinin gene of low pathogenic avian influenza virus subtype H9N2. J Virol Methods 2020; 284:113938. [PMID: 32663531 DOI: 10.1016/j.jviromet.2020.113938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/22/2020] [Accepted: 07/09/2020] [Indexed: 01/25/2023]
Abstract
Low pathogenic avian influenza H9N2 is still circulating in the Middle East causing respiratory manifestations and severe economic losses in poultry. In the present study, an H9 plasmid-based DNA vaccine targeting the HA gene of H9N2 A/CK/Egypt/SCU8/2014 was developed and evaluated in turkeys. The full length of HA was cloned into vector plasmids under the control of a cytomegalovirus promoter. The in-vitro expression of the recombinant HA was demonstrated in HeLa cells transfected with the plasmids pVAX1-H9 or pCR-H9 using western blot and Immunofluorescent assay (IFA). The efficacy of pVAX-H9 and pCR- H9, naked or saponin-adjuvanted, was evaluated in turkey poults at 3 weeks and challenged with A/CK/Egypt/SCU8/2014 (106 EID50/bird at 3 weeks post-vaccination. The efficacy was assesses based on virus shedding, oropharyngeal and cloacal, as well as seroconversion using haemagglutination inhibition (HI) test. All immunized birds showed high HI antibody titers (7-8 log2) at 3 weeks post-vaccination. None of the birds vaccinated with naked or saponin-adjuvanted pVAX-H9 or pCR-H9 showed any clinical signs. The pVAX-H9 and pCR-H9 alone did not prevent cloacal and oropharyngeal virus shedding, however, saponin-adjuvanted pVAX1-H9 and pCR-H9 prevented cloacal and oropharyngeal virus shedding at 3 and 5 days post challenge, respectively. In conclusion, DNA vaccination with pVAX1-H9 and pCR-H9 could protect turkey from the H9N2 virus, but vaccination regimes need to be improved.
Collapse
|
4
|
Tran-Nguyen TK, Chandra D, Yuan K, Patibandla PK, Nguyen KT, Sethu P, Zhang Y, Xue J, Mobley JA, Kim YI, Shoushtari A, Leader JK, Bon J, Sciurba FC, Duncan SR. Glucose-Regulated Protein 78 Autoantibodies Are Associated with Carotid Atherosclerosis in Chronic Obstructive Pulmonary Disease Patients. Immunohorizons 2020; 4:108-118. [PMID: 32086320 PMCID: PMC7430561 DOI: 10.4049/immunohorizons.1900098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/01/2020] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis prevalence is increased in chronic obstructive pulmonary disease (COPD) patients, independent of other risk factors. The etiology of the excess vascular disease in COPD is unknown, although it is presumably related to an underlying (if cryptic) systemic immune response. Autoantibodies with specificity for glucose-regulated protein 78 (GRP78), a multifunctional component of the unfolded protein response, are common in COPD patients and linked to comorbidities of this lung disease. We hypothesized anti-GRP78 autoreactivity might also be a risk factor for atherosclerosis in COPD patients. Carotid intima-medial thickness (cIMT) was measured in 144 current and former smokers by ultrasound. Concentrations of circulating IgG autoantibodies against full-length GRP78, determined by ELISA, were greater among subjects with abnormally increased cIMT (p <, 0.01). Plasma levels of autoantibodies against a singular GRP78 peptide segment, amino acids 246–260 (anti-GRP78aa 246–260), were even more highly correlated with cIMT, especially among males with greater than or equal to moderate COPD (rs = 0.62, p = 0.001). Anti-GRP78aa 246–260 concentrations were independent of CRP, IL-6, and TNF-α levels. GRP78 autoantigen expression was upregulated among human aortic endothelial cells (HAECs) stressed by incubation with tunicamycin (an unfolded protein response inducer) or exposure to culture media flow disturbances. Autoantibodies against GRP78aa 246–260, isolated from patient plasma by immunoprecipitation, induced HAEC production of proatherosclerotic mediators, including IL-8. In conclusion, anti-GRP78 autoantibodies are highly associated with carotid atherosclerosis in COPD patients and exert atherogenic effects on HAECs. These data implicate Ag-specific autoimmunity in the pathogenesis of atherosclerosis among COPD patients and raise possibilities that directed autoantibody reduction might ameliorate vascular disease in this high-risk population.
Collapse
Affiliation(s)
- Thi K Tran-Nguyen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Divay Chandra
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - Kaiyu Yuan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Phani K Patibandla
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Khanh T Nguyen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Palaniappan Sethu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - Jianmin Xue
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - James A Mobley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Young-Il Kim
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ali Shoushtari
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - Joseph K Leader
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213; and
| | - Jessica Bon
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213.,Department of Medicine, VA Pittsburgh Healthcare System, Pittsburgh, PA 15213
| | - Frank C Sciurba
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - Steven R Duncan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
5
|
HIV latency can be established in proliferating and nonproliferating resting CD4+ T cells in vitro: implications for latency reversal. AIDS 2019; 33:199-209. [PMID: 30562171 DOI: 10.1097/qad.0000000000002075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To determine whether latency can be established and reversed in both proliferating and nonproliferating CD4+ T cells in the same model in vitro. METHODS Activated CD4+ T cells were infected with either a nonreplication competent, luciferase reporter virus or wild-type full-length enhanced green fluorescent protein (EGFP) reporter virus and cultured for 12 days. The cells were then sorted by flow cytometry to obtain two distinct T-cell populations that did not express the T-cell activation markers, CD69, CD25 and human leukocyte antigen (HLA)-DR: CD69CD25HLA-DR small cells (nonblasts) that had not proliferated in vitro following mitogen stimulation and CD69CD25HLA-DR large cells (which we here call transitional blasts) that had proliferated. The cells were then reactivated with latency-reversing agents and either luciferase or EGFP quantified. RESULTS Inducible luciferase expression, consistent with latent infection, was observed in nonblasts and transitional blasts following stimulation with either phorbol-myristate-acetate/phytohemagglutinin (3.8 ± 1 and 2.9 ± 0.5 fold above dimethyl sulfoxide, respectively) or romidepsin (2.1 ± 0.6 and 1.8 ± 0.2 fold above dimethyl sulfoxide, respectively). Constitutive expression of luciferase was higher in transitional blasts compared with nonblasts. Using wild-type full-length EGFP reporter virus, inducible virus was observed in nonblasts but not in transitional blasts. No significant difference was observed in the response to latency-reversing agents in either nonblasts or transitional blasts. CONCLUSION HIV latency can be established in vitro in resting T cells that have not proliferated (nonblasts) and blasts that have proliferated (transitional blasts). This model could potentially be used to assess new strategies to eliminate latency.
Collapse
|
6
|
Lee LYY, Izzard L, Hurt AC. A Review of DNA Vaccines Against Influenza. Front Immunol 2018; 9:1568. [PMID: 30038621 PMCID: PMC6046547 DOI: 10.3389/fimmu.2018.01568] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/25/2018] [Indexed: 01/07/2023] Open
Abstract
The challenges of effective vaccination against influenza are gaining more mainstream attention, as recent influenza seasons have reported low efficacy in annual vaccination programs worldwide. Combined with the potential emergence of novel influenza viruses resulting in a pandemic, the need for effective alternatives to egg-produced conventional vaccines has been made increasingly clear. DNA vaccines against influenza have been in development since the 1990s, but the initial excitement over success in murine model trials has been tempered by comparatively poor performance in larger animal models. In the intervening years, much progress has been made to refine the DNA vaccine platform-the rational design of antigens and expression vectors, the development of novel vaccine adjuvants, and the employment of innovative gene delivery methods. This review discusses how these advances have been applied in recent efforts to develop an effective influenza DNA vaccine.
Collapse
|
7
|
Chan CC, Lai CW, Wu CJ, Chen LC, Tao MH, Kuo ML. Liver-Specific Allergen Gene Transfer by Adeno-Associated Virus Suppresses Allergic Airway Inflammation in Mice. Hum Gene Ther 2017; 27:631-42. [PMID: 27178525 DOI: 10.1089/hum.2015.161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Allergic airway inflammation driven by T helper 2 (Th2)-type immunity is characterized by airway hyperresponsiveness, eosinophilic infiltration, and elevated IgE production. Various novel strategies for managing asthma have been explored, such as DNA vaccines, T-cell peptides, and allergen-specific immunotherapy. A principal goal of most immunotherapeutic approaches is active and long-term allergen-specific tolerance. Liver-specific gene transfer using adeno-associated virus (AAV) has been shown to favorably induce tolerogenic responses to therapeutic products in various experimental models. AAV8 has strong liver tropism and induces immune tolerance in mice. The present study aimed to determine whether hepatocyte-specific allergen expression by pseudotyped AAV2/8 alleviates asthmatic symptoms in ovalbumin (OVA)-sensitized mice. Mice were intravenously injected with AAV2/8 vector carrying membrane-bound OVA transgene under transcriptional control of a hepatocyte-specific alpha 1 antitrypsin promoter (AAV2/8-OVA) and then sensitized with OVA. AAV2/8-OVA specifically transduced the OVA transgene in the liver. Airway hyperresponsiveness, eosinophilia, mucus hypersecretion, and Th2 cytokines were significantly suppressed in both the lungs and secondary lymphoid organs of asthmatic mice infected with AAV2/8-OVA. Significant reduction of OVA-specific antibodies was detected in the bronchoalveolar lavage fluid from AAV2/8-OVA-treated mice. Moreover, AAV2/8-OVA treatment prominently promoted the expression of Foxp3, IL-10, and TGF-β in the liver. Enhanced Foxp3 expression was also detected in the lungs of asthmatic mice after AAV2/8-OVA treatment. Taken together, these results suggest that the induction of immune tolerance by hepatic AAV gene transfer may be beneficial for modulating allergic asthma.
Collapse
Affiliation(s)
- Cheng-Chi Chan
- 1 Graduate Institute of Biomedical Sciences, Chang Gung University , Taoyuan, Taiwan
| | - Chin-Wen Lai
- 2 Institute of Biomedical Sciences , Academia Sinica, Taipei, Taiwan .,3 Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University , Taoyuan, Taiwan
| | - Chia-Jen Wu
- 2 Institute of Biomedical Sciences , Academia Sinica, Taipei, Taiwan
| | - Li-Chen Chen
- 4 Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital , Taoyuan, Taiwan
| | - Mi-Hua Tao
- 2 Institute of Biomedical Sciences , Academia Sinica, Taipei, Taiwan
| | - Ming-Ling Kuo
- 1 Graduate Institute of Biomedical Sciences, Chang Gung University , Taoyuan, Taiwan .,4 Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital , Taoyuan, Taiwan .,5 Department of Microbiology and Immunology, College of Medicine, Chang Gung University , Taoyuan, Taiwan .,6 Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University , Taoyuan, Taiwan
| |
Collapse
|
8
|
Li FJ, Surolia R, Li H, Wang Z, Kulkarni T, Liu G, de Andrade JA, Kass DJ, Thannickal VJ, Duncan SR, Antony VB. Autoimmunity to Vimentin Is Associated with Outcomes of Patients with Idiopathic Pulmonary Fibrosis. THE JOURNAL OF IMMUNOLOGY 2017; 199:1596-1605. [PMID: 28754682 DOI: 10.4049/jimmunol.1700473] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/06/2017] [Indexed: 02/02/2023]
Abstract
Autoimmunity has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF); however, the repertoire of autoantigens involved in this disease and the clinical relevance of these autoimmune responses are still being explored. Our initial discovery assays demonstrated that circulating and intrapulmonary vimentin levels are increased in IPF patients. Subsequent studies showed native vimentin induced HLA-DR-dependent in vitro proliferation of CD4 T cells from IPF patients and enhanced the production of IL-4, IL-17, and TGF-β1 by these lymphocytes in contrast to normal control specimens. Vimentin supplementation of IPF PBMC cultures also resulted in HLA-DR-dependent production of IgG with anti-vimentin specificities. Circulating anti-vimentin IgG autoantibody levels were much greater in IPF subjects from the University of Alabama at Birmingham (n = 102) and the University of Pittsburgh (U. Pitt., n = 70) than in normal controls. Anti-vimentin autoantibody levels in IPF patients were HLA biased and inversely correlated with physiological measurements of lung function (i.e., forced expiratory volumes and diffusing capacities). Despite considerable intergroup differences in transplant-free survival between these two independent IPF cohorts, serious adverse outcomes were most frequent among the patients within each population that had the highest anti-vimentin autoantibody levels (University of Alabama at Birmingham: hazard ratio 2.5, 95% confidence interval 1.2-5.3, p = 0.012; University of Pittsburgh: hazard ratio 2.7, 95% confidence interval 1.3-5.5, p = 0.006). These data show that anti-vimentin autoreactivity is prevalent in IPF patients and is strongly associated with disease manifestations. These findings have implications with regard to the pathogenesis of this enigmatic disease and raise the possibility that therapies specifically directed at these autoimmune processes could have therapeutic efficacy.
Collapse
Affiliation(s)
- Fu Jun Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ranu Surolia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Huashi Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Zheng Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tejaswini Kulkarni
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Joao A de Andrade
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294.,Birmingham VA Medical Center, Birmingham, AL 35233; and
| | - Daniel J Kass
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294.,Birmingham VA Medical Center, Birmingham, AL 35233; and
| | - Steven R Duncan
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Veena B Antony
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294;
| |
Collapse
|
9
|
Kanuma T, Yamamoto T, Kobiyama K, Moriishi E, Masuta Y, Kusakabe T, Ozasa K, Kuroda E, Jounai N, Ishii KJ. CD63-Mediated Antigen Delivery into Extracellular Vesicles via DNA Vaccination Results in Robust CD8+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2017; 198:4707-4715. [DOI: 10.4049/jimmunol.1600731] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 04/11/2017] [Indexed: 12/16/2022]
|
10
|
Ciabattini A, Pettini E, Medaglini D. CD4(+) T Cell Priming as Biomarker to Study Immune Response to Preventive Vaccines. Front Immunol 2013; 4:421. [PMID: 24363656 PMCID: PMC3850413 DOI: 10.3389/fimmu.2013.00421] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/20/2013] [Indexed: 01/10/2023] Open
Abstract
T cell priming is a critical event in the initiation of the immune response to vaccination since it deeply influences both the magnitude and the quality of the immune response induced. CD4(+) T cell priming, required for the induction of high-affinity antibodies and immune memory, represents a key target for improving and modulating vaccine immunogenicity. A major challenge in the study of in vivo T cell priming is due to the low frequency of antigen-specific T cells. This review discusses the current knowledge on antigen-specific CD4(+) T cell priming in the context of vaccination, as well as the most advanced tools for the characterization of the in vivo T cell priming and the opportunities offered by the application of systems biology.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| |
Collapse
|
11
|
Increasing versatility of the DNA vaccines through modification of the subcellular location of plasmid-encoded antigen expression in the in vivo transfected cells. PLoS One 2013; 8:e77426. [PMID: 24130884 PMCID: PMC3794048 DOI: 10.1371/journal.pone.0077426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022] Open
Abstract
The route of administration of DNA vaccines can play a key role in the magnitude and quality of the immune response triggered after their administration. DNA vaccines containing the gene of the membrane-anchored glycoprotein (gpG) of the fish rhabdoviruses infectious haematopoietic necrosis virus (IHNV) or viral haematopoietic septicaemia virus (VHSV), perhaps the most effective DNA vaccines generated so far, confer maximum protection when injected intramuscularly in contrast to their low efficacy when injected intraperitoneally. In this work, taking as a model the DNA vaccine against VHSV, we focused on developing a more versatile DNA vaccine capable of inducing protective immunity regardless of the administration route used. For that, we designed two alternative constructs to gpG1-507 (the wild type membrane-anchored gpG of VHSV) encoding either a soluble (gpG1-462) or a secreted soluble (gpGLmPle20-462) form of the VHSV-gpG. In vivo immunisation/challenge assays showed that only gpGLmPle20-462 (the secreted soluble form) conferred protective immunity against VHSV lethal challenge via both intramuscular and intraperitoneal injection, being this the first description of a fish viral DNA vaccine that confers protection when administered intraperitoneally. Moreover, this new DNA vaccine construct also conferred protection when administered in the presence of an oil adjuvant suggesting that DNA vaccines against rhabdoviruses could be included in the formulation of current multicomponent-intaperitoneally injectable fish vaccines formulated with an oil adjuvant. On the other hand, a strong recruitment of membrane immunoglobulin expressing B cells, mainly membrane IgT, as well as t-bet expressing T cells, at early times post-immunisation, was specifically observed in the fish immunised with the secreted soluble form of the VHSV-gpG protein; this may indicate that the subcellular location of plasmid-encoded antigen expression in the in vivo transfected cells could be an important factor in determining the ways in which DNA vaccines prime the immune response.
Collapse
|
12
|
Chen J, Lin M, Li N, Lin L, She F. Therapeutic vaccination with Salmonella-delivered codon-optimized outer inflammatory protein DNA vaccine enhances protection in Helicobacter pylori infected mice. Vaccine 2012; 30:5310-5. [PMID: 22749593 DOI: 10.1016/j.vaccine.2012.06.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 05/31/2012] [Accepted: 06/16/2012] [Indexed: 02/07/2023]
Abstract
Vaccination had demonstrated as an alternative way to combat Helicobacter pylori challenge. In the present study, codon-optimized outer inflammatory protein gene (oipA) for Mus species codon usage, the inclusion of optimal Kozak sequence, and modified of GC content was applied to construct a novel DNA construct. The Salmonella-delivered wild type oipA construct (SL7207/poipA) and the Salmonella-delivered codon-optimized oipA construct (SL7207/poipA-opt) were prepared and their therapeutic efficacy was evaluated in H. pylori-infected mice. The codon-optimized oipA construct (poipA-opt) expressed almost six-fold higher protein than that of wild type construct (poipA) as normalized to the β-actin expression in AGS cells. Oral therapeutic immunization with SL7207/poipA-opt significantly eliminated H. pylori colonization in the stomach; and protection was related to a robust Th1/Th2 immune response. Therefore, our results suggested that fine therapeutic efficacy was related to sufficient expression of the antigen. It is supposed that codon-optimized oipA gene improves protein expression and consequently enhances the immunogenicity of DNA vaccine, which resulted in a significant reduction of bacterial loads in H. pylori infected mice. The Salmonella-delivered codon-optimized DNA construct could be a candidate vaccine against H. pylori for the clinical application.
Collapse
Affiliation(s)
- Jiansen Chen
- Department of Hospital Infection Control, Union Hospital, Fujian Medical University, Fuzhou 350001, Fujian, China
| | | | | | | | | |
Collapse
|
13
|
Fissolo N, Montalban X, Comabella M. DNA-based vaccines for multiple sclerosis: Current status and future directions. Clin Immunol 2012; 142:76-83. [DOI: 10.1016/j.clim.2010.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 11/16/2010] [Accepted: 11/17/2010] [Indexed: 01/23/2023]
|
14
|
Moraes MP, Segundo FDS, Dias CC, Pena L, Grubman MJ. Increased efficacy of an adenovirus-vectored foot-and-mouth disease capsid subunit vaccine expressing nonstructural protein 2B is associated with a specific T cell response. Vaccine 2011; 29:9431-40. [DOI: 10.1016/j.vaccine.2011.10.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 10/06/2011] [Accepted: 10/16/2011] [Indexed: 10/15/2022]
|
15
|
The signal peptide sequence impacts the immune response elicited by a DNA epitope vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1776-80. [PMID: 21832097 DOI: 10.1128/cvi.05179-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the effect of two leader sequences, one from a transmembrane molecule (H2-L(d)) and another from a secreted molecule (rat KC chemokine), on the immunogenicity of DNA epitope vaccines. The chemokine leader enhanced vaccine immunogenicity, thus underscoring the importance of the leader sequence in DNA epitope vaccine design.
Collapse
|
16
|
Zeelenberg IS, van Maren WWC, Boissonnas A, Van Hout-Kuijer MA, Den Brok MHMGM, Wagenaars JAL, van der Schaaf A, Jansen EJR, Amigorena S, Théry C, Figdor CG, Adema GJ. Antigen localization controls T cell-mediated tumor immunity. THE JOURNAL OF IMMUNOLOGY 2011; 187:1281-8. [PMID: 21705625 DOI: 10.4049/jimmunol.1003905] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Effective antitumor immunotherapy requires the identification of suitable target Ags. Interestingly, many of the tumor Ags used in clinical trials are present in preparations of secreted tumor vesicles (exosomes). In this study, we compared T cell responses elicited by murine MCA101 fibrosarcoma tumors expressing a model Ag at different localizations within the tumor cell in association with secreted vesicles (exosomes), as a nonsecreted cell-associated protein, or as secreted soluble protein. Remarkably, we demonstrated that only the tumor-secreting vesicle-bound Ag elicited a strong Ag-specific CD8(+) T cell response, CD4(+) T cell help, Ag-specific Abs, and a decrease in the percentage of immunosuppressive regulatory T cells in the tumor. Moreover, in a therapeutic tumor model of cryoablation, only in tumors secreting vesicle-bound Ag could Ag-specific CD8(+) T cells still be detected up to 16 d after therapy. We concluded that the localization of an Ag within the tumor codetermines whether a robust immunostimulatory response is elicited. In vivo, vesicle-bound Ag clearly skews toward a more immunogenic phenotype, whereas soluble or cell-associated Ag expression cannot prevent or even delay outgrowth and results in tumor tolerance. This may explain why particular immunotherapies based on these vesicle-bound tumor Ags are potentially successful. Therefore, we conclude that this study may have significant implications in the discovery of new tumor Ags suitable for immunotherapy and that their location should be taken into account to ensure a strong antitumor immune response.
Collapse
Affiliation(s)
- Ingrid S Zeelenberg
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6525GA Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Doucet JD, Forget MA, Grange C, Rouxel RN, Arbour N, von Messling V, Lapointe R. Endogenously expressed matrix protein M1 and nucleoprotein of influenza A are efficiently presented by class I and class II major histocompatibility complexes. J Gen Virol 2011; 92:1162-1171. [DOI: 10.1099/vir.0.029777-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Current influenza vaccines containing primarily hypervariable haemagglutinin and neuraminidase proteins must be prepared against frequent new antigenic variants. Therefore, there is an ongoing effort to develop influenza vaccines that also elicit strong and sustained cytotoxic responses against highly conserved determinants such as the matrix (M1) protein and nucleoprotein (NP). However, their antigenic presentation properties in humans are less defined. Accordingly, we analysed MHC class I and class II presentation of endogenously processed M1 and NP in human antigen presenting cells and observed expansion of both CD8+- and CD4+-specific effector T lymphocytes secreting gamma interferon and tumour necrosis factor. Further enhancement of basal MHC-II antigenic presentation did not improve CD4+ or CD8+ T-cell quality based on cytokine production upon challenge, suggesting that endogenous M1 and NP MHC-II presentation is sufficient. These new insights about T-lymphocyte expansion following endogenous M1 and NP MHC-I and -II presentation will be important to design complementary heterosubtypic vaccination strategies.
Collapse
Affiliation(s)
- Jean-Daniel Doucet
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM)-Hôpital Notre-Dame, Université de Montréal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Marie-Andrée Forget
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM)-Hôpital Notre-Dame, Université de Montréal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Cécile Grange
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM)-Hôpital Notre-Dame, Université de Montréal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| | | | - Nathalie Arbour
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM)-Hôpital Notre-Dame, Université de Montréal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| | | | - Réjean Lapointe
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM)-Hôpital Notre-Dame, Université de Montréal and Institut du Cancer de Montréal, Montréal, Québec, Canada
| |
Collapse
|
18
|
Rivas EI, Driver JP, Garabatos N, Presa M, Mora C, Rodriguez F, Serreze DV, Stratmann T. Targeting of a T cell agonist peptide to lysosomes by DNA vaccination induces tolerance in the nonobese diabetic mouse. THE JOURNAL OF IMMUNOLOGY 2011; 186:4078-87. [PMID: 21346228 DOI: 10.4049/jimmunol.0902395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD4 T cells are crucial effectors in the pathology of type 1 diabetes (T1D). Successful therapeutic interventions for prevention and cure of T1D in humans are still elusive. Recent research efforts have focused on the manipulation of T cells by treatment with DNA. In this paper, we studied the effects of a DNA treatment strategy designed to target antigenic peptides to the lysosomal compartment on a monospecific T cell population termed 2.5mi(+) T cells that shares reactivity with the diabetogenic T cell clone BDC-2.5 in the NOD mouse. MHC class II tetramer analysis showed that repeated administrations were necessary to expand 2.5mi(+) T cells in vivo. This expansion was independent of Ag presentation by B cells. A single peptide epitope was sufficient to induce protection against T1D, which was not due to Ag-specific T cell anergy. Typical Th2 cytokines such as IL-10 or IL-4 were undetectable in 2.5mi(+) T cells, arguing against a mechanism of immune deviation. Instead, the expanded 2.5mi(+) T cell population produced IFN-γ similar to 2.5mi(+) T cells from naive mice. Protection against T1D by DNA treatment was completely lost in NOD.CD28(-/-) mice which are largely deficient of natural regulatory T cells (Treg). Although Ag-specific Foxp3(+) Treg did not expand in response to DNA treatment, diabetes onset was delayed in Treg-reconstituted and DNA-treated NOD.SCID mice. These observations provide evidence for a Treg-mediated protective mechanism that is independent of the expansion or de novo generation of Ag-specific Treg.
Collapse
Affiliation(s)
- Elisa I Rivas
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Rush CM, Mitchell TJ, Garside P. A detailed characterisation of the distribution and presentation of DNA vaccine encoded antigen. Vaccine 2009; 28:1620-34. [PMID: 20035828 PMCID: PMC2824851 DOI: 10.1016/j.vaccine.2009.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 11/05/2009] [Accepted: 11/06/2009] [Indexed: 11/25/2022]
Abstract
The association between plasmid DNA distribution, the amount of Ag produced, Ag persistence and the identity and localisation of cells presenting DNA-encoded Ag all have important consequences for both quantitative and qualitative aspects of T cell responses induced by DNA vaccines. Using a variety of approaches to detect and quantify the uptake of injected DNA, and the production and presentation of DNA-encoded antigen, we report that injected DNA vaccines rapidly enter the peripheral blood from the injection site and also reach muscle-draining lymph nodes directly as free DNA. 24 h after plasmid injection, MHCII+CD11b+B220−CD11clow/− cells in the draining and distal LNs and spleen contain pDNA. Interestingly, we also observed pDNA+MHCIIlow/−CD11b+ within the bone marrow. Concomitantly, we detected Ag-containing/expressing cells at both the injection site and in draining lymph nodes. Three days after plasmid injection we detected rare pMHC+CD11c+ cells within secondary lymphoid tissue and simultaneously observed Ag-specific CD4+ T cell accumulation and blastogenesis in these tissues. Our results show that the events that determine the induction of DNA vaccine immune responses occur within days of DNA injection and that the response becomes systemic very rapidly, possibly with involvement from resident BM cells.
Collapse
Affiliation(s)
- Catherine M Rush
- Centre for Biophotonics, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 27 Taylor St, Glasgow G4 0NR, UK.
| | | | | |
Collapse
|
20
|
X4 human immunodeficiency virus type 1 gp120 down-modulates expression and immunogenicity of codelivered antigens. J Virol 2009; 83:10941-50. [PMID: 19692474 DOI: 10.1128/jvi.00394-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In order to increase the immune breadth of human immunodeficiency virus (HIV) vaccines, strategies such as immunization with several HIV antigens or centralized immunogens have been examined. HIV-1 gp120 protein is a major immunogen of HIV and has been routinely considered for inclusion in both present and future AIDS vaccines. However, recent studies proposed that gp120 interferes with the generation of immune response to codelivered antigens. Here, we investigate whether coimmunization with plasmid-encoded gp120 alters the immune response to other coadministered plasmid encoded antigens such as luciferase or ovalbumin in a mouse model. We found that the presence of gp120 leads to a significant reduction in the expression level of the codelivered antigen in vivo. Antigen presentation by antigen-presenting cells was also reduced and resulted in the induction of weak antigen-specific cellular and humoral immune responses. Importantly, gp120-mediated immune interference was observed after administration of the plasmids at the same or at distinct locations. To characterize the region in gp120 mediating these effects, we used plasmid constructs encoding gp120 that lacks the V1V2 loops (DeltaV1V2) or the V3 loop (DeltaV3). After immunization, the DeltaV1V2, but not the DeltaV3 construct, was able to reduce antigen expression, antigen presentation, and subsequently the immunogenicity of the codelivered antigen. The V3 loop dependence of this phenomenon seems to be limited to V3 loops known to interact with the CXCR4 molecule but not with CCR5. Our study presents a novel mechanism by which HIV-1 gp120 interferes with the immune response against coadministered antigen in a polyvalent vaccine preparation.
Collapse
|
21
|
Pettini E, Ciabattini A, Pozzi G, Medaglini D. Adoptive transfer of transgenic T cells to study mucosal adjuvants. Methods 2009; 49:340-5. [PMID: 19409994 DOI: 10.1016/j.ymeth.2009.03.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/24/2009] [Accepted: 03/09/2009] [Indexed: 10/20/2022] Open
Abstract
The study of the initiation and regulation of T-cell responses to vaccine antigens is of primary importance in the rational design of mucosal adjuvants. The detection in vivo of T-cell priming following immunization can be performed by using the adoptive transfer model of naïve antigen-specific transgenic T cells into immunocompetent mice. In this work, we discuss the applications of this system for detecting in vivo the primary antigen-specific clonal expansion, the phenotype, and the effector function of transgenic T cells following mucosal immunization. OVA and the mucosal adjuvant CTB were used as a model vaccine formulation and administered by the nasal route to study T-cell priming. T helper and T cytotoxic primary proliferation and expression of activation and migration markers was observed both in draining and distal sites. This method proved to be a powerful tool to study the efficacy of mucosal adjuvants in enhancing T-cell priming.
Collapse
Affiliation(s)
- Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia, Dipartimento di Biologia Molecolare, Università di Siena, 53100 Siena, Italy
| | | | | | | |
Collapse
|
22
|
Isaji K, Kawase A, Matono M, Guan X, Nishikawa M, Takakura Y. Enhanced CTL response by controlled intracellular trafficking of antigen in dendritic cells following DNA vaccination. J Control Release 2009; 135:227-33. [DOI: 10.1016/j.jconrel.2009.01.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Revised: 01/06/2009] [Accepted: 01/30/2009] [Indexed: 11/25/2022]
|
23
|
almani M, Raffaeli S, Vider-Shalit T, Tsaban L, Fishbain V, Louzoun Y. Human self-protein CD8+ T-cell epitopes are both positively and negatively selected. Eur J Immunol 2009; 39:1056-65. [PMID: 19291702 PMCID: PMC2730470 DOI: 10.1002/eji.200838353] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The cellular immune system recognizes self-epitopes in the context of MHC-I molecules. The immunological general view presumes that these self-epitopes are just a background, both positively and negatively selecting T cells. We here estimate the number of epitopes in each human protein for many frequent HLA alleles, and a score representing over or under presentation of epitopes on these proteins. We further show that there is a clear selection for the presentation of specific self-protein types. Proteins presenting many epitopes include, for example, autoimmune regulator (AIRE) upregulated tissue-specific antigens, immune system receptors and proteins with a high expression level. On the other hand, proteins that may be considered less "useful" for the immune system, such as low expression level proteins, are under-presented. We combine our epitope estimate with single nucleotide polymorphism (SNP) measures to show that this selection can be directly observed through the fraction of non-synonymous SNP (replacement fraction), which is significantly higher inside epitopes than outside.
Collapse
Affiliation(s)
- Michal almani
- Math department and Gonda Brain research center, Bar Ilan University, Ramat Gan, Isreal, 52900
| | - Shai Raffaeli
- Math department and Gonda Brain research center, Bar Ilan University, Ramat Gan, Isreal, 52900
| | - Tal Vider-Shalit
- Math department and Gonda Brain research center, Bar Ilan University, Ramat Gan, Isreal, 52900
| | - Lea Tsaban
- Math department and Gonda Brain research center, Bar Ilan University, Ramat Gan, Isreal, 52900
| | - Vered Fishbain
- Department of Molecular Genetics and Biotechnology, Faculty of Medicine, The Hebrew University of Jerusalem
| | - Yoram Louzoun
- Math department and Gonda Brain research center, Bar Ilan University, Ramat Gan, Isreal, 52900
| |
Collapse
|
24
|
Solvason N, Lou YP, Peters W, Evans E, Martinez J, Ramirez U, Ocampo A, Yun R, Ahmad S, Liu E, Yu L, Eisenbarth G, Leviten M, Steinman L, Garren H. Improved Efficacy of a Tolerizing DNA Vaccine for Reversal of Hyperglycemia through Enhancement of Gene Expression and Localization to Intracellular Sites. THE JOURNAL OF IMMUNOLOGY 2008; 181:8298-307. [DOI: 10.4049/jimmunol.181.12.8298] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
25
|
Reduction in RNA levels rather than retardation of translation is responsible for the inhibition of major histocompatibility complex class I antigen presentation by the glutamic acid-rich repeat of herpesvirus saimiri open reading frame 73. J Virol 2008; 83:273-82. [PMID: 18945762 DOI: 10.1128/jvi.01532-08] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Herpesvirus saimiri (HVS) establishes a persistent infection in squirrel monkeys by maintaining its episome within T lymphocytes. The product of open reading frame 73 (ORF73) plays a key role in episomal maintenance and is the functional homologue of Epstein-Barr virus EBNA1 and Kaposi's sarcoma-associated herpesvirus LANA1 proteins. There is little sequence homology among these proteins, although all contain a central domain of repeating amino acids. The repeat domains of EBNA1 and LANA1 enhance the stability of these proteins and cause a retardation in self-protein synthesis, leading to poor recognition by CD8(+) cytotoxic T lymphocytes (CTL). The HVS ORF73 repeat domain is composed of a glutamic acid and glycine repeat linked to a glutamic acid and alanine repeat (EG-EA repeat). Here we show that the EG-EA repeat similarly causes a reduction in the recognition of ORF73 by CD8(+) CTL. However, deletion of the EG-EA repeat from HVS ORF73 had no affect on the stability of the protein or its rate of translation. In contrast, the presence of the EG-EA repeat was found to decrease the steady-state levels of ORF73 mRNA. The inhibitory properties of the EG-EA repeat were maintained when transferred to a heterologous protein, and manipulation of the repeat revealed that the motif EEAEEAEEE was sufficient to cause a reduction in recognition of ORF73 by CD8(+) CTL. Thus, the EG-EA repeat of HVS ORF73 plays a role in immune evasion but utilizes a mechanism distinct from that of the EBNA1 and LANA1 repeats.
Collapse
|
26
|
Sudowe S, Dominitzki S, Montermann E, Bros M, Grabbe S, Reske-Kunz AB. Uptake and presentation of exogenous antigen and presentation of endogenously produced antigen by skin dendritic cells represent equivalent pathways for the priming of cellular immune responses following biolistic DNA immunization. Immunology 2008; 128:e193-205. [PMID: 18800984 DOI: 10.1111/j.1365-2567.2008.02947.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Gene gun-mediated biolistic DNA vaccination with beta-galactosidase (betaGal)-encoding plasmid vectors efficiently modulated antigen-induced immune responses in an animal model of type I allergy, including the inhibition of immunoglobulin E (IgE) production. Here we show that CD4(+) as well as CD8(+) T cells from mice biolistically transfected with a plasmid encoding betaGal under the control of the fascin promoter (pFascin-betaGal) are capable of inhibiting betaGal-specific IgE production after adoptive transfer into naïve recipients. Moreover, suppression of IgE production was dependent on interferon (IFN)-gamma. To analyse the modalities of activation of CD4(+) and CD8(+) T cells regarding the localization of antigen synthesis following gene gun-mediated DNA immunization, we used the fascin promoter and the keratin 5 promoter (pK5-betaGal) to direct betaGal production mainly to dendritic cells (DCs) and to keratinocytes, respectively. Gene gun-mediated DNA immunization with each vector induced considerable activation of betaGal-specific CD8(+) cytotoxic T cells. Cytokine production by re-stimulated CD4(+) T cells in draining lymph nodes and immunoglobulin isotype profiles in sera of immunized mice indicated that immunization with pFascin-betaGal induced a T helper type 1 (Th1)-biased immune response, whereas immunization with pK5-betaGal generated a mixed Th1/Th2 immune response. Nevertheless, DNA vaccination with pFascin-betaGal and pK5-betaGal, respectively, efficiently inhibited specific IgE production in the mouse model of type I allergy. In conclusion, our data show that uptake of exogenous antigen produced by keratinocytes and its presentation by untransfected DCs as well as the presentation of antigen synthesized endogenously in DCs represent equivalent pathways for efficient priming of cellular immune responses.
Collapse
Affiliation(s)
- Stephan Sudowe
- Department of Dermatology, Johannes Gutenberg-University Mainz, Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
27
|
Kadaoui KA, Corthésy B. Secretory IgA mediates bacterial translocation to dendritic cells in mouse Peyer's patches with restriction to mucosal compartment. THE JOURNAL OF IMMUNOLOGY 2008; 179:7751-7. [PMID: 18025221 DOI: 10.4049/jimmunol.179.11.7751] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In addition to fulfilling its function of immune exclusion at mucosal surfaces, secretory IgA (SIgA) Ab exhibits the striking feature to adhere selectively to M cells in the mouse and human intestinal Peyer's patches (PPs). Subsequent uptake drives the SIgA Ab to dendritic cells (DCs), which become partially activated. Using freshly isolated mouse DCs, we found that the interaction with SIgA was tissue and DC subtype dependent. Only DCs isolated from PPs and mesenteric lymph nodes interacted with the Ab. CD11c(+)CD11b(+) DCs internalized SIgA, while CD11c(+)CD19(+) DCs only bound SIgA on their surface, and no interaction occurred with CD11c(+)CD8alpha(+) DCs. We next examined whether SIgA could deliver a sizeable cargo to PP DCs in vivo by administering SIgA-Shigella flexneri immune complexes into a mouse ligated intestinal loop containing a PP. We found that such immune complexes entered the PPs and were internalized by subepithelial dome PP DCs, in contrast to S. flexneri alone that did not penetrate the intestinal epithelium in mice. Dissemination of intraepithelial S. flexneri delivered as immune complexes was limited to PPs and mesenteric lymph nodes. We propose that preexisting SIgA Abs associated with microbes contribute to mucosal defense by eliciting responses that prevent overreaction while maintaining productive immunity.
Collapse
Affiliation(s)
- Khalil A Kadaoui
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon, Switzerland
| | | |
Collapse
|
28
|
Klotz C, Gehre F, Lucius R, Pogonka T. Identification of Eimeria tenella genes encoding for secretory proteins and evaluation of candidates by DNA immunisation studies in chickens. Vaccine 2007; 25:6625-34. [PMID: 17675183 DOI: 10.1016/j.vaccine.2007.06.048] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 06/15/2007] [Accepted: 06/19/2007] [Indexed: 11/21/2022]
Abstract
In order to identify secretory proteins as possible new vaccine candidates, a cDNA-library from E. tenella sporozoites was generated in yeast and was used to select secreted and surface proteins. Herein 191 clones were isolated and analysis of the nucleic acid sequences revealed 162 deduced open reading frames with a prediction for signal peptides. These sequences are characterized by high redundancy, comprising 25 unique protein fragments with a high degree of stage specificity. Only three sequences showed identical homology to already known E. tenella proteins. The majority, 16 fragments, revealed homology to known or hypothetical proteins, and six fragments had no sequence homologues in protein databases. In order to obtain optimised conditions for a DNA vaccination trial in chickens, with which our selected new sequences could be tested, we performed variant DNA immunisations with the well-characterized E. tenella antigen SO7. The cDNA of the SO7 antigen was subcloned into two different eucaryotic expression vectors, i.e. pcDNA3 and pVR1012. In addition, the SO7 sequence was fused to the stabilizing sequence of the enhanced green fluorescence protein (EGFP). All SO7 constructs induced a SO7 specific immune response after intramuscular application and no significant differences were found on using constructs with or without the EGFP fusion or with different vector systems. Full-length open reading frames from six selected Eimeria sequences were introduced into the eucaryotic expression vector pcDNA3. Subsequent immunisation trials revealed a decrease in parasite excretion for three constructs after challenge infection in comparison to the control animals. Our approach represents a rapid screening to identify and test putative new vaccine candidates from E. tenella sporozoites that could also be adopted to other apicomplexan parasites.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Blotting, Western
- COS Cells
- Cell Line
- Chickens
- Chlorocebus aethiops
- Cloning, Molecular
- DNA, Protozoan/genetics
- DNA, Protozoan/immunology
- Eimeria tenella/genetics
- Eimeria tenella/immunology
- Eimeria tenella/metabolism
- Enzyme-Linked Immunosorbent Assay
- Gene Library
- Humans
- Immunization/methods
- Immunoglobulin G/immunology
- Molecular Sequence Data
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- Protozoan Vaccines/genetics
- Protozoan Vaccines/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Christian Klotz
- Department of Molecular Parasitology, Humboldt University Berlin, Philippstrasse 13, 10115 Berlin, Germany
| | | | | | | |
Collapse
|
29
|
Cassataro J, Pasquevich KA, Estein SM, Laplagne DA, Zwerdling A, de la Barrera S, Bowden R, Fossati CA, Giambartolomei GH, Goldbaum FA. A DNA vaccine coding for the chimera BLSOmp31 induced a better degree of protection against B. ovis and a similar degree of protection against B. melitensis than Rev.1 vaccination. Vaccine 2007; 25:5958-67. [PMID: 17600596 DOI: 10.1016/j.vaccine.2007.05.049] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 05/21/2007] [Accepted: 05/23/2007] [Indexed: 01/24/2023]
Abstract
In the present study, we reported an attempt to improve the immunogenicity and protective capacity of the chimera BLSOmp31 using a different antigen delivery: DNA vaccination. Vaccination of BALB/c mice with the DNA vaccine coding for the chimera BLSOmp31 (pCIBLSOmp31) provided the best protection level against Brucella ovis, which was significantly higher than the given by the co-delivery of both plasmids coding for the whole proteins (pcDNABLS+pCIOmp31) and even higher than the control vaccine Rev.1. Moreover, pCIBLSOmp31 induced higher protection against Brucella melitensis than pcDNABLS+pCIOmp31 but similar protection than Rev.1. The chimera induced a strong humoral response against the inserted peptide. It also induced peptide- and BLS-specific cytotoxic T responses. The insertion of this peptide on BLS induced stronger T helper 1 responses specific for the carrier (BLS), thus our results represent a case of synergic strengthening between two Brucella antigens. Hitherto, this is the first indication that a recombinant subunit vaccine elicits greater protection than whole Brucella.
Collapse
Affiliation(s)
- Juliana Cassataro
- Laboratorio de Inmunogenética, Hospital de Clínicas José de San Martín, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Werner-Klein M, Dresch C, Marconi P, Brocker T. Transcriptional targeting of B cells for induction of peripheral CD8 T cell tolerance. THE JOURNAL OF IMMUNOLOGY 2007; 178:7738-46. [PMID: 17548611 DOI: 10.4049/jimmunol.178.12.7738] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Several mechanisms are in place to neutralize autoimmune CD8 T cells by tolerance induction. Developing self-specific CD8 T cells are eliminated in the thymus by Ag-presenting epithelial and dendritic cells (DCs). However, CD8 T cells escaping thymic central tolerance can also be inactivated by tolerance mechanisms in peripheral organs. In contrast to DCs, the role of B cells in generating CD8 T cell tolerance is not well-characterized. To investigate this question in more detail, we transcriptionally targeted Ag to B cells using B cell-specific retroviral vectors in vivo. Although Ag expression could be detected in B cells of thymus, lymph nodes, and spleen, B cells were unable to induce central tolerance of CD8 thymocytes. In contrast, in peripheral organs, we could identify clonal deletion and functional inhibition (anergy) of CD8 T cells as tolerance-inducing mechanisms. Although Ag expressed by B cells was acquired and cross-presented by DCs, B cells were also sufficient to tolerize CD8 T cells directly. These findings suggest exploitation of B cells for Ag-specific immunotherapy of CD8 T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Melanie Werner-Klein
- Institute for Immunology, Ludwig-Maximilians-University, Goethestrasse 31, D-80336 Munich, Germany
| | | | | | | |
Collapse
|
31
|
Loeffler DIM, Schoen CU, Goebel W, Pilgrim S. Comparison of different live vaccine strategies in vivo for delivery of protein antigen or antigen-encoding DNA and mRNA by virulence-attenuated Listeria monocytogenes. Infect Immun 2006; 74:3946-57. [PMID: 16790768 PMCID: PMC1489688 DOI: 10.1128/iai.00112-06] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Listeria monocytogenes can be used to deliver protein antigens or DNA and mRNA encoding such antigens directly into the cytosol of host cells because of its intracellular lifestyle. In this study, we compare the in vivo efficiencies of activation of antigen-specific CD8 and CD4 T cells when the antigen is secreted by L. monocytogenes or when antigen-encoding plasmid DNA or mRNA is released by self-destructing strains of L. monocytogenes. Infection of mice with self-destructing L. monocytogenes carriers delivering mRNA that encodes a nonsecreted form of ovalbumin (OVA) resulted in a significant OVA-specific CD8 T-cell response. In contrast, infection with L. monocytogenes delivering OVA-encoding DNA failed to generate specific T cells. Secretion of OVA by the carrier bacteria yielded the strongest immune response involving OVA-specific CD8 and CD4 T cells. In addition, we investigated the antigen delivery capacity of a self-destructing, virulence-attenuated L. monocytogenes aroA/B mutant. In contrast to the wild-type strain, this mutant exhibited only marginal liver toxicity when high doses (5 x 10(7) CFU per animal administered intravenously) were used, and it was also able to deliver sufficient amounts of secreted OVA into mice. Therefore, the results presented here could lay the groundwork for a rational combination of L. monocytogenes as an attenuated carrier for the delivery of protein and nucleic acid vaccines in novel vaccination strategies.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Cell Line, Tumor
- DNA, Bacterial/administration & dosage
- DNA, Bacterial/immunology
- Drug Delivery Systems
- Female
- Leukemia P388
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/pathogenicity
- Listeriosis/immunology
- Listeriosis/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Ovalbumin/metabolism
- RNA, Bacterial/administration & dosage
- RNA, Bacterial/immunology
- RNA, Messenger/administration & dosage
- RNA, Messenger/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Virulence
Collapse
|
32
|
Lauterbach H, Gruber A, Ried C, Cheminay C, Brocker T. Insufficient APC capacities of dendritic cells in gene gun-mediated DNA vaccination. THE JOURNAL OF IMMUNOLOGY 2006; 176:4600-7. [PMID: 16585550 DOI: 10.4049/jimmunol.176.8.4600] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gene gun-mediated DNA immunization is a powerful mode of vaccination against infectious diseases and tumors. Many studies have identified dendritic cells (DC) as the central players in inducing immunity upon biolistic DNA vaccination; however, none of these studies directly quantify DC-mediated responses in comparison with immunity triggered by all Ag- and MHC-expressing cells. In this study we use two different approaches to decipher the relative role of DC vs other cell types in gene gun-induced immunity. First, we directly compared the immunization efficacy of different DNA constructs, which allow Ag expression ubiquitously (CMV promoter) or specifically in DC (CD11c promoter) and would encode either for soluble or membrane bound forms of Ag. Second, we immunized transgenic mice in which only DC can present MHC-restricted Ag, and directly compared the magnitudes of CTL activation with those obtained in wild-type mice. Surprisingly, our combined data suggest that, although DC-specific Ag expression is sufficient to induce humoral responses, DC alone cannot trigger optimal CD4 and CD8 T cell responses upon gene gun vaccination. Therefore, we conclude that DC alone are insufficient to mediate optimal induction of T cell immunity upon gene gun DNA vaccination and that broad Ag expression rather than DC-restricted approaches are necessary for induction of complete immune responses.
Collapse
Affiliation(s)
- Henning Lauterbach
- Institute for Immunology, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | | | |
Collapse
|
33
|
Stone GW, Barzee S, Snarsky V, Kee K, Spina CA, Yu XF, Kornbluth RS. Multimeric soluble CD40 ligand and GITR ligand as adjuvants for human immunodeficiency virus DNA vaccines. J Virol 2006; 80:1762-72. [PMID: 16439533 PMCID: PMC1367159 DOI: 10.1128/jvi.80.4.1762-1772.2006] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
For use in humans, human immunodeficiency virus (HIV) DNA vaccines may need to include immunostimulatory adjuvant molecules. CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily (TNFSF), is one candidate adjuvant, but it has been difficult to use because it is normally expressed as a trimeric membrane molecule. Soluble trimeric forms of CD40L have been produced, but in vitro data indicate that multimeric, many-trimer forms of soluble CD40L are more active. This multimerization requirement was evaluated in mice using plasmids that encoded either 1-trimer, 2-trimer, or 4-trimer soluble forms of CD40L. Fusion with the body of Acrp30 was used to produce the 2-trimer form, and fusion with the body of surfactant protein D was used to produce the 4-trimer form. Using plasmids for secreted HIV-1 antigens Gag and Env, soluble CD40L was active as an adjuvant in direct proportion to the valence of the trimers (1 < 2 < 4). These CD40L-augmented DNA vaccines elicited strong CD8(+) T-cell responses but did not elicit significant CD4(+) T-cell or antibody responses. To test the applicability of the multimeric fusion protein approach to other TNFSFs, a 4-trimer construct for the ligand of glucocorticoid-induced TNF family-related receptor (GITR) was also prepared. Multimeric soluble GITR ligand (GITRL) augmented the CD8(+) T-cell, CD4(+) T-cell, and antibody responses to DNA vaccination. In summary, multimeric CD40L and GITRL are new adjuvants for DNA vaccines. Plasmids for expressing multimeric TNFSF fusion proteins permit the rapid testing of TNFSF molecules in vivo.
Collapse
Affiliation(s)
- Geoffrey W Stone
- Department of Medicine-0679, Stein Clinical Sciences Bldg., Room 304, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0679, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Rush CM, Mitchell TJ, Burke B, Garside P. Dissecting the components of the humoral immune response elicited by DNA vaccines. Vaccine 2006; 24:776-84. [PMID: 16198029 DOI: 10.1016/j.vaccine.2005.08.080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2005] [Accepted: 08/16/2005] [Indexed: 10/25/2022]
Abstract
Although DNA vaccines appear to be efficient at inducing strong cellular immune responses, a number of questions remain regarding their ability to induce humoral immunity. The essential components for generating an antibody response include B and T cell recognition of antigen, subsequent activation, clonal expansion of each lymphocyte type and migration of T cells into B cell follicles to provide help, all leading to germinal centre formation and antibody production. We have employed a double adoptive transfer system based on ovalbumin (OVA)-specific CD4+ DO11.10 T cells and hen egg lysozyme (HEL)-specific MD4 B cells to assess all of these parameters in the context of DNA vaccination in vivo. We find that vaccination with DNA constructs expressing an OVA-HEL gene fusion (encoding contiguous T and B cell epitopes) can induce T cell activation, clonal expansion and migration into B cell follicles accompanied by B cell activation, blastogenesis, expansion and antibody production. These findings show that DNA vaccination can induce all of the components required for humoral immunity and also provide a system for in depth analysis of factors that influence the development of antibody responses. Such strategies may facilitate the rational design of vaccines capable of inducing effective humoral immunity.
Collapse
Affiliation(s)
- Catherine M Rush
- Division of Immunology, Infection and Inflammation, Western Infirmary, University of Glasgow, Glasgow G116NT, UK.
| | | | | | | |
Collapse
|
35
|
Cassataro J, Estein SM, Pasquevich KA, Velikovsky CA, de la Barrera S, Bowden R, Fossati CA, Giambartolomei GH. Vaccination with the recombinant Brucella outer membrane protein 31 or a derived 27-amino-acid synthetic peptide elicits a CD4+ T helper 1 response that protects against Brucella melitensis infection. Infect Immun 2006; 73:8079-88. [PMID: 16299302 PMCID: PMC1307072 DOI: 10.1128/iai.73.12.8079-8088.2005] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immunogenicity and protective efficacy of the recombinant 31-kDa outer membrane protein from Brucella melitensis (rOmp31), administered with incomplete Freund's adjuvant, were evaluated in mice. Immunization of BALB/c mice with rOmp31 conferred protection against B. ovis and B. melitensis infection. rOmp31 induced a vigorous immunoglobulin G (IgG) response, with higher IgG1 than IgG2 titers. In addition, spleen cells from rOmp31-immunized mice produced interleukin 2 (IL-2) and gamma interferon, but not IL-10 or IL-4, after in vitro stimulation with rOmp31, suggesting the induction of a T helper 1 (Th1) response. Splenocytes from rOmp31-vaccinated animals also induced a specific cytotoxic-T-lymphocyte activity, which led to the in vitro lysis of Brucella-infected macrophages. In vitro T-cell subset depletion indicated that rOmp31 immunization elicited specific CD4+ T cells that secrete IL-2 and gamma interferon, while CD8+ T cells induced cytotoxic-T-lymphocyte activity. In vivo depletion of T-cell subsets showed that the rOmp31-elicited protection against B. melitensis infection is mediated by CD4+ T cells while the contribution of CD8+ T cells may be limited. We then evaluated the immunogenicity and protective efficacy of a known exposed region from Omp31 on the Brucella membrane, a peptide that contains amino acids 48 to 74 of Omp31. Immunization with the synthetic peptide in adjuvant did not elicit a specific humoral response but elicited a Th1 response mediated by CD4+ T cells. The peptide in adjuvant induced levels of protection similar to those induced by rOmp31 against B. melitensis but less protection than was induced by rOmp31 against B. ovis. Our results indicate that rOmp31 could be a useful candidate for the development of subunit vaccines against B. melitensis and B. ovis.
Collapse
Affiliation(s)
- Juliana Cassataro
- Laboratorio de Inmunogenética, Hospital de Clínicas José de San Martín, Facultad de Medicina, UBA, Córdoba 2351 3 Piso Sala 4 (1120), Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Gupta V, Tabiin TM, Sun K, Chandrasekaran A, Anwar A, Yang K, Chikhlikar P, Salmon J, Brusic V, Marques ET, Kellathur SN, August TJ. SARS coronavirus nucleocapsid immunodominant T-cell epitope cluster is common to both exogenous recombinant and endogenous DNA-encoded immunogens. Virology 2006; 347:127-39. [PMID: 16387339 PMCID: PMC7111852 DOI: 10.1016/j.virol.2005.11.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2005] [Revised: 09/22/2005] [Accepted: 11/22/2005] [Indexed: 01/12/2023]
Abstract
Correspondence between the T-cell epitope responses of vaccine immunogens and those of pathogen antigens is critical to vaccine efficacy. In the present study, we analyzed the spectrum of immune responses of mice to three different forms of the SARS coronavirus nucleocapsid (N): (1) exogenous recombinant protein (N-GST) with Freund's adjuvant; (2) DNA encoding unmodified N as an endogenous cytoplasmic protein (pN); and (3) DNA encoding N as a LAMP-1 chimera targeted to the lysosomal MHC II compartment (p-LAMP-N). Lysosomal trafficking of the LAMP/N chimera in transfected cells was documented by both confocal and immunoelectron microscopy. The responses of the immunized mice differed markedly. The strongest T-cell IFN-γ and CTL responses were to the LAMP-N chimera followed by the pN immunogen. In contrast, N-GST elicited strong T cell IL-4 but minimal IFN-γ responses and a much greater antibody response. Despite these differences, however, the immunodominant T-cell ELISpot responses to each of the three immunogens were elicited by the same N peptides, with the greatest responses being generated by a cluster of five overlapping peptides, N76–114, each of which contained nonameric H2d binding domains with high binding scores for both class I and, except for N76–93, class II alleles. These results demonstrate that processing and presentation of N, whether exogenously or endogenously derived, resulted in common immunodominant epitopes, supporting the usefulness of modified antigen delivery and trafficking forms and, in particular, LAMP chimeras as vaccine candidates. Nevertheless, the profiles of T-cell responses were distinctly different. The pronounced Th-2 and humoral response to N protein plus adjuvant are in contrast to the balanced IFN-γ and IL-4 responses and strong memory CTL responses to the LAMP-N chimera.
Collapse
Affiliation(s)
- Vandana Gupta
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
| | - Tani M. Tabiin
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
| | - Kai Sun
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
| | - Ananth Chandrasekaran
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
| | - Azlinda Anwar
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
| | - Kun Yang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Priya Chikhlikar
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Jerome Salmon
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Vladimir Brusic
- Institute for Infocomm Research, 21 Heng Mui Keng Terrace, Singapore 119613, Singapore
- School of Land and Food Sciences and the Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Ernesto T.A. Marques
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
- Department of Medicine, Division of Infectious Diseases, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21218, USA
- Virology and Experimental Therapy Laboratory, Aggeu Magalhaes Research Center, Recife, PE 50670-420, Brazil
| | - Srinivasan N. Kellathur
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Thomas J. August
- Division of Biomedical Sciences, Johns Hopkins in Singapore, 31 Biopolis Way, #02-01 The Nanos, Singapore 138669, Singapore
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
- Corresponding author. Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA. Fax: +1 410 502 3066.
| |
Collapse
|
37
|
Ko HJ, Ko SY, Kim YJ, Lee EG, Cho SN, Kang CY. Optimization of codon usage enhances the immunogenicity of a DNA vaccine encoding mycobacterial antigen Ag85B. Infect Immun 2005; 73:5666-74. [PMID: 16113284 PMCID: PMC1231050 DOI: 10.1128/iai.73.9.5666-5674.2005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In spite of its many other benefits, DNA vaccine is limited in its application by its insufficient immunogenicity. One promising approach for enhancing its immunogenicity is to maximize its expression in the immunized host. In the current study, we investigated whether codon optimization of the mycobacterial antigen Ag85B gene could enhance the expression and immunogenicity of the Ag85B DNA vaccine. We generated a synthetic humanized Ag85B (hAg85B) gene in which codon usage was optimized for expression in human cells. DNA plasmids with codon-optimized hAg85B increased the level of protein expression in vitro and in vivo. DNA vaccine with hAg85B induced stronger Th1-like and cytotoxic T-cell immune responses in BALB/c mice and generated higher protective immunity in a BALB/c mouse model of Mycobacterium tuberculosis aerosol infection than did the DNA vaccine with wild-type Ag85B. Therefore, our results suggest that codon optimization of mycobacterial antigens (e.g., Ag85B) could improve protein expression and thereby enhance the immunogenicity of DNA vaccines against M. tuberculosis.
Collapse
Affiliation(s)
- Hyun-Jeong Ko
- Laboratory of Immunology and Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Shillim-Dong, Kwanak-Gu, Seoul, 151-742, Republic of Korea
| | | | | | | | | | | |
Collapse
|
38
|
Garmory HS, Perkins SD, Phillpotts RJ, Titball RW. DNA vaccines for biodefence. Adv Drug Deliv Rev 2005; 57:1343-61. [PMID: 15935877 DOI: 10.1016/j.addr.2005.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2004] [Accepted: 01/25/2005] [Indexed: 11/30/2022]
Abstract
The advantages associated with DNA vaccines include the speed with which they may be constructed and produced at large-scale, the ability to produce a broad spectrum of immune responses, and the ability for delivery using non-invasive means. In addition, DNA vaccines may be manipulated to express multiple antigens and may be tailored for the induction of appropriate immune responses. These advantages make DNA vaccination a promising approach for the development of vaccines for biodefence. In this review, the potential of DNA vaccines for biodefence is discussed.
Collapse
Affiliation(s)
- Helen S Garmory
- Department of Biomedical Sciences, Defence Science and Technology Laboratory, Porton Down, Salisbury, SP4 0JQ, UK.
| | | | | | | |
Collapse
|
39
|
Bruna-Romero O, Rocha CD, Tsuji M, Gazzinelli RT. Enhanced protective immunity against malaria by vaccination with a recombinant adenovirus encoding the circumsporozoite protein of Plasmodium lacking the GPI-anchoring motif. Vaccine 2004; 22:3575-84. [PMID: 15315836 DOI: 10.1016/j.vaccine.2004.03.050] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 03/18/2004] [Accepted: 03/29/2004] [Indexed: 10/26/2022]
Abstract
A major malaria vaccine candidate, the circumsporozoite (CS) protein of Plasmodium, is a pre-erythrocytic stage antigen that is attached to the surface of the sporozoites through a glycosylphosphatidylinositol (GPI) anchor. However, here we show that the motif that signals for glycosylphosphatidylinositol anchor addition interferes with the immunogenicity of this protein and reduces protection in mice upon immunization with a recombinant adenovirus. The presence of the glycosylphosphatidylinositol-anchoring motif sequentially affected total circumsporozoite protein production, cellular distribution, antigen processing and secretion, leading to less effective antigen presentation. Consistently, vaccination with an adenovirus recombinant carrying the anchoring motif-disrupted circumsporozoite gene, resulted in significant increase of the number of interferon-gamma (IFN-gamma) producing T cells and specific IgG2a isotype antibodies, ensuing more effective vaccination. Given that the anchoring motif is highly conserved among different species of Plasmodium, anti-malaria subunit vaccines encoded by recombinant vectors that aim at the induction of strong cellular immunity could maximize immunogenicity by removing anchoring motifs.
Collapse
Affiliation(s)
- Oscar Bruna-Romero
- Laboratory of Immunopathology, Centro de Pesquisas René Rachou, FIOCRUZ, Av Augusto de Lima 1715, Belo Horizonte 30190-002, MG, Brazil.
| | | | | | | |
Collapse
|
40
|
Tsagozis P, Tseveleki V, Probert L, Dotsika E, Karagouni E. Vaccination with Plasmids Encoding the Leishmania Major GP63 Glycoprotein and CD40L Results in a Partial Suppression of the Inflammatory Reaction after Experimental Infection. EUR J INFLAMM 2004. [DOI: 10.1177/1721727x0400200207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The development of an effective vaccine against leishmaniasis is the aim of an intensive research effort, to bring relief to thousands of people worldwide. DNA vaccination is a promising approach in this direction, since it is able to generate a strong cellular immune response. We tested whether the co-administration of a plasmid encoding a truncated gp63 gene, that allows extracellular secretion of the encoded protein, and a plasmid encoding for CD40L could induce a protective response in genetically susceptible BALB/c mice and reduce the local inflammatory swelling after infection with Leishmania major. We document that vaccination with the combination of plasmids for gp63 and CD40L reduced inflammatory swelling, while vaccination with the truncated gp63-encoding plasmid resulted in an exacerbation of the local inflammatory reaction. These preliminary data indicate that the CD40L expression plasmid is consequently an efficient adjuvant for the induction of protective responses in the context of a DNA vaccination against leishmaniasis.
Collapse
Affiliation(s)
| | - V. Tseveleki
- Laboratory of Molecular Genetics, Institut Pasteur Hellenique, Athens, Greece
| | - L. Probert
- Laboratory of Molecular Genetics, Institut Pasteur Hellenique, Athens, Greece
| | | | | |
Collapse
|
41
|
de Arruda LB, Chikhlikar PR, August JT, Marques ETA. DNA vaccine encoding human immunodeficiency virus-1 Gag, targeted to the major histocompatibility complex II compartment by lysosomal-associated membrane protein, elicits enhanced long-term memory response. Immunology 2004; 112:126-33. [PMID: 15129672 PMCID: PMC1782456 DOI: 10.1111/j.1365-2567.2004.01823.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2003] [Revised: 11/06/2003] [Accepted: 01/06/2004] [Indexed: 12/25/2022] Open
Abstract
Antigen presentation by major histocompatibility complex type II (MHC II) molecules and activation of CD4+ helper T cells are critical for the generation of immunological memory. We previously described a DNA vaccine encoding human immunodeficiency virus-1 p55Gag as a chimera with the lysosome-associated membrane protein (LAMP/gag). The LAMP/gag chimera protein traffics to the MHC II compartment of transfected cells and elicits enhanced immune responses as compared to a DNA vaccine encoding native gag not targeted to the MHC II compartment. We have now investigated the long-term responses of immunized mice and show that the LAMP/gag DNA vaccine promotes long-lasting B cell- and CD4+ and CD8+ T-cell memory responses induced by DNA encoding non-targeted Gag decay rapidly and elicit very low or undetectable levels of gag DNA is sufficient to generate T-cell memory. Following this initial priming immunization with LAMP/gag DNA, booster immunizations with native gag DNA or the LAMP/gag chimera are equally efficient in eliciting B- and T-cell secondary responses, results in accordance with observations that secondary expansion of CD8+ cells in the boost phase does not require additional CD4+ help. These findings underscore the significance of targeting DNA-encoded vaccine antigens to the MHC II processing compartments for induction of long-term immunological memory.
Collapse
|
42
|
Abstract
Distinct populations of both CD4(+) and CD8(+) memory T cells have been identified on the basis of their location (lymphoid versus non-lymphoid tissues) and function. There have been several recent advances in our understanding of the factors that govern the generation and maintenance of T-cell memory in vivo. Utilising this knowledge could contribute to the refinement of DNA vaccine technology to induce more robust and longer-lasting protective T-cell based immunity.
Collapse
Affiliation(s)
- Joanna R Kirman
- Malaghan Institute of Medical Research, PO Box 7060, Wellington South, New Zealand.
| | | |
Collapse
|
43
|
Cunto-Amesty G, Monzavi-Karbassi B, Luo P, Jousheghany F, Kieber-Emmons T. Strategies in cancer vaccines development. Int J Parasitol 2003; 33:597-613. [PMID: 12782058 DOI: 10.1016/s0020-7519(03)00054-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The recent definition of tumour-specific immunity in cancer patients and the identification of tumour-associated antigens have generated renewed enthusiasm for the application of immune-based therapies for the treatment of malignancies. Recent developments in cancer vaccines have also been based on an improved understanding of the cellular interactions required to induce a specific anti-tumour immune response. Consequently, a number of cancer vaccines have entered clinical trials. Targeting broad-spectrum tumour-associated antigens has emerged as a strategy to lower the risk of tumour escape due to the loss of specific nominal antigen. Amongst the most challenging of tumour-associated antigens to which to target in active specific immunotherapy applications are carbohydrate antigens. As carbohydrates are intrinsically T-cell-independent antigens, more novel approaches are perhaps needed to drive specific-T-cell-dependent immune responses to carbohydrate antigens. In this context peptide mimetics of core structures of tumour-associated carbohydrate antigens might be developed to augment immune responses to these broad-spectrum antigens.
Collapse
Affiliation(s)
- Gina Cunto-Amesty
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|