Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Vopr Virusol ; 67(6): 516-526, 2023 02 07.
Article in Russian | MEDLINE | ID: mdl-37264841

ABSTRACT

INTRODUCTION: A vaccine against hepatitis C has not yet been developed. Recombinant proteins and plasmids encoding hepatitis C virus (HCV) proteins, the components of candidate vaccines, induce a weak immune response and require the use of adjuvants. The aim of the work was to study the adjuvant action of an aqueous solution of fullerene C60 during immunization of mice with HCV recombinant protein NS5B (rNS5B) that is an RNA-dependent RNA polymerase, or with NS5B-encoding pcNS5B plasmid. MATERIALS AND METHODS: An aqueous solution of dispersed fullerene (dnC60) was obtained by ultrafiltration. C57BL/6 mice were immunized with rNS5B subcutaneously, pcNS5B intramuscularly mixed with different doses of dnC60 three times, then the humoral and cellular response to HCV was evaluated. RESULTS: Mice immunization with rNS5B in a mixture with dnC60 at doses of 250 g/mouse significantly induced humoral response: a dose-dependent increase in IgG1 antibody titers was 720 times higher than in the absence of fullerene. There was no increase in the cellular response to rNS5B when administered with dnC60. The humoral response to DNA immunization was weak in mice of all groups receiving pcNS5B. The cellular response was suppressed when the plasmid was injected in a mixture with dnC60. CONCLUSIONS: Dispersed fullerene dnC60 is a promising adjuvant for increasing the immunostimulating activity of weakly immunogenic proteins including surface and other HCV proteins, important for a protective response. Further research is needed to enhance the ability of dnC60 to boost the cellular immune response to the components of the candidate vaccine.


Subject(s)
Fullerenes , Hepatitis C , Vaccines, DNA , Viral Hepatitis Vaccines , Mice , Animals , Hepacivirus , Fullerenes/pharmacology , Fullerenes/metabolism , Base Sequence , Amino Acids/genetics , Amino Acids/metabolism , Amino Acids/pharmacology , Mice, Inbred C57BL , Adjuvants, Immunologic/genetics , Immunity, Cellular , Recombinant Proteins/genetics , Mice, Inbred BALB C , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology , Viral Hepatitis Vaccines/genetics , Viral Hepatitis Vaccines/pharmacology
2.
Sci Adv ; 7(45): eabj0611, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34739313

ABSTRACT

This work reports a suction-based cutaneous delivery method for in vivo DNA transfection. Following intradermal Mantoux injection of plasmid DNA in a rat model, a moderate negative pressure is applied to the injection site, a technique similar to Chinese báguàn and Middle Eastern hijama cupping therapies. Strong GFP expression was demonstrated with pEGFP-N1 plasmids where fluorescence was observed as early as 1 hour after dosing. Modeling indicates a strong correlation between focal strain/stress and expression patterns. The absence of visible and/or histological tissue injury contrasts with current in vivo transfection systems such as electroporation. Specific utility was demonstrated with a synthetic SARS-CoV-2 DNA vaccine, which generated host humoral immune response in rats with notable antibody production. This method enables an easy-to-use, cost-effective, and highly scalable platform for both laboratorial transfection needs and clinical applications for nucleic acid­based therapeutics and vaccines.


Subject(s)
COVID-19 Vaccines , COVID-19 , DNA , SARS-CoV-2 , Skin/immunology , Transfection , Vaccines, DNA , Administration, Cutaneous , Animals , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , COVID-19 Vaccines/pharmacology , DNA/genetics , DNA/immunology , DNA/pharmacology , Male , Rats , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Suction , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
3.
Curr Opin Allergy Clin Immunol ; 21(6): 569-575, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34387280

ABSTRACT

PURPOSE OF REVIEW: Molecular forms of allergen-specific immunotherapy (AIT) are continuously emerging to improve the efficacy of the treatment, to shorten the duration of protocols and to prevent any side effects. The present review covers the recent progress in the development of AIT based on nucleic acid encoding allergens or CpG oligodeoxynucleotides (CpG-ODN). RECENT FINDINGS: Therapeutic vaccinations with plasmid deoxyribonucleic acid (DNA) encoding major shrimp Met e 1 or insect For t 2 allergen were effective for the treatment of food or insect bite allergy in respective animal models. DNA expressing hypoallergenic shrimp tropomyosin activated Foxp3+ T regulatory (Treg) cells whereas DNA encoding For t 2 down-regulated the expression of pruritus-inducing IL-31. Co-administrations of major cat allergen Fel d 1 with high doses of CpG-ODN reduced Th2 airway inflammation through tolerance induction mediated by GATA3+ Foxp3hi Treg cells as well as early anti-inflammatory TNF/TNFR2 signaling cascade. Non-canonical CpG-ODN derived from Cryptococcus neoformans as well as methylated CpG sites present in the genomic DNA from Bifidobacterium infantis mediated Th1 or Treg cell differentiation respectively. SUMMARY: Recent studies on plasmid DNA encoding allergens evidenced their therapeutic potential for the treatment of food allergy and atopic dermatitis. Unmethylated or methylated CpG-ODNs were shown to activate dose-dependent Treg/Th1 responses. Large clinical trials need to be conducted to confirm these promising preclinical data. Moreover, tremendous success of messenger ribonucleic acid (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 must encourage as well the re-exploration of mRNA vaccine platform for innovative AIT.


Subject(s)
Desensitization, Immunologic/methods , Hypersensitivity, Immediate/therapy , Oligodeoxyribonucleotides/administration & dosage , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/administration & dosage , Allergens/administration & dosage , Allergens/genetics , Allergens/immunology , Animals , Clinical Trials as Topic , Desensitization, Immunologic/trends , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Hypersensitivity, Immediate/immunology , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Treatment Outcome , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , mRNA Vaccines
4.
Methods Mol Biol ; 2197: 51-85, 2021.
Article in English | MEDLINE | ID: mdl-32827132

ABSTRACT

CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides specifically designed to stimulate Toll-like receptor 9. TLR9 is expressed on human plasmacytoid dendritic cells and B cells and triggers an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. This chapter reviews recent progress in understanding the mechanism of action of CpG ODN and provides an overview of human clinical trial results using CpG ODN to improve vaccines for the prevention/treatment of cancer, allergy, and infectious disease.


Subject(s)
Adjuvants, Immunologic , Oligodeoxyribonucleotides , Vaccines/immunology , Allergens/immunology , Animals , Biomarkers , Clinical Studies as Topic , Drug Evaluation, Preclinical , Host-Pathogen Interactions/immunology , Humans , Immunity , Signal Transduction , Toll-Like Receptor 9/metabolism , Vaccines/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology
5.
Acta Parasitol ; 64(3): 471-478, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31187386

ABSTRACT

BACKGROUND: Toxoplasma gondii has a comprehensive impact on a great range of warm-blood mammals, in which one-third of the population all over the world is involved. Dense granular proteins, regarded as GRA family, mediating substantial interface between host cell cytoplasm and parasite, are widely studied for preventing the infection of T. gondii. PURPOSE: As is handled in our study, the effect of intramuscularly injecting the genetic vaccine pEGFP-C1/GRA41 encoding a novel dense granule protein, GRA41, was evaluated. METHODS: At the beginning, bioinformatics analysis was used to evaluate epitopes of both B cells and T cells on the GRA41 protein of T. gondii. Afterwards, recombinant plasmids (pEGFP-C1/GRA41) were injected into BALB/c mice and the quantity of IgG and its subclass IgG2a remarkably increased. IFN-γ, distinctive from the other cytokines (IL-4, and IL-10), was significant in growth. Afterwards, the intraperitoneal challenge was executed for recording survival time with tachyzoites with high virulence (in RH strain) and counting the number of brain cysts was carried out after the infection of PRU strain (low virulence). RESULTS: In pEGFP-C1/GRA41 group, the survival period was significantly longer (13.3 ± 3.37 days) after tachyzoites attack with the RH strain in high virulence, compared with the other groups (less than 8 days). Additionally, the cyst quantity is remarkably lower and the rate of reduction could reach 59.34%. CONCLUSION: All the results indicated effective protection of DNA vaccine encoding GRA41 against T. gondii.


Subject(s)
Membrane Proteins/immunology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Cytokines/genetics , Cytokines/immunology , Drug Evaluation, Preclinical , Epitopes/genetics , Epitopes/immunology , Female , Humans , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , T-Lymphocytes/immunology , Toxoplasma/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
6.
Proc Natl Acad Sci U S A ; 115(32): 8179-8184, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038013

ABSTRACT

Agents that remodel the tumor microenvironment (TME), prime functional tumor-specific T cells, and block inhibitory signaling pathways are essential components of effective immunotherapy. We are evaluating live-attenuated, double-deleted Listeria monocytogenes expressing tumor antigens (LADD-Ag) in the clinic. Here we show in numerous mouse models that while treatment with nonrecombinant LADD induced some changes in the TME, no antitumor efficacy was observed, even when combined with immune checkpoint blockade. In contrast, LADD-Ag promoted tumor rejection by priming tumor-specific KLRG1+PD1loCD62L- CD8+ T cells. These IFNγ-producing effector CD8+ T cells infiltrated the tumor and converted the tumor from an immunosuppressive to an inflamed microenvironment that was characterized by a decrease in regulatory T cells (Treg) levels, a proinflammatory cytokine milieu, and the shift of M2 macrophages to an inducible nitric oxide synthase (iNOS)+CD206- M1 phenotype. Remarkably, these LADD-Ag-induced tumor-specific T cells persisted for more than 2 months after primary tumor challenge and rapidly controlled secondary tumor challenge. Our results indicate that the striking antitumor efficacy observed in mice with LADD-based immunotherapy stems from TME remodeling which is a direct consequence of eliciting potent, systemic tumor-specific CD8+ T cells.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Listeria monocytogenes/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Listeria monocytogenes/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Treatment Outcome , Vaccination/methods , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Xenograft Model Antitumor Assays
7.
Front Immunol ; 9: 711, 2018.
Article in English | MEDLINE | ID: mdl-29686682

ABSTRACT

The innate immune system provides the first line of defense against pathogen infection though also influences pathways involved in cancer immunosurveillance. The innate immune system relies on a limited set of germ line-encoded sensors termed pattern recognition receptors (PRRs), signaling proteins and immune response factors. Cytosolic receptors mediate recognition of danger damage-associated molecular patterns (DAMPs) signals. Once activated, these sensors trigger multiple signaling cascades, converging on the production of type I interferons and proinflammatory cytokines. Recent studies revealed that PRRs respond to nucleic acids (NA) released by dying, damaged, cancer cells, as danger DAMPs signals, and presence of signaling proteins across cancer types suggests that these signaling mechanisms may be involved in cancer biology. DAMPs play important roles in shaping adaptive immune responses through the activation of innate immune cells and immunological response to danger DAMPs signals is crucial for the host response to cancer and tumor rejection. Furthermore, PRRs mediate the response to NA in several vaccination strategies, including DNA immunization. As route of double-strand DNA intracellular entry, DNA immunization leads to expression of key components of cytosolic NA-sensing pathways. The involvement of NA-sensing mechanisms in the antitumor response makes these pathways attractive drug targets. Natural and synthetic agonists of NA-sensing pathways can trigger cell death in malignant cells, recruit immune cells, such as DCs, CD8+ T cells, and NK cells, into the tumor microenvironment and are being explored as promising adjuvants in cancer immunotherapies. In this minireview, we discuss how cGAS-STING and RIG-I-MAVS pathways have been targeted for cancer treatment in preclinical translational researches. In addition, we present a targeted selection of recent clinical trials employing agonists of cytosolic NA-sensing pathways showing how these pathways are currently being targeted for clinical application in oncology.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/metabolism , Nucleic Acids/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cytosol/immunology , DEAD Box Protein 58/antagonists & inhibitors , Drug Evaluation, Preclinical , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/antagonists & inhibitors , Membrane Proteins/agonists , Membrane Proteins/metabolism , Neoplasms/therapy , Nucleic Acids/metabolism , Signal Transduction/drug effects , Vaccines, DNA/genetics , Vaccines, DNA/immunology
8.
Expert Opin Biol Ther ; 18(2): 159-173, 2018 02.
Article in English | MEDLINE | ID: mdl-29148858

ABSTRACT

INTRODUCTION: The Ebola virus (EBOV) disease epidemic during 2014-16 in West Africa has accelerated the clinical development of several vaccine candidates that have demonstrated efficacy in the gold standard nonhuman primate (NHP) model, namely cynomolgus macaques. AREAS COVERED: This review discusses the pre-clinical research and if available, clinical evaluation of the currently available EBOV vaccine candidates, while emphasizing the translatability of pre-clinical data generated in the NHP model to clinical data in humans. EXPERT OPINION: Despite the existence of many successful EBOV vaccine candidates in the pre-clinical stages, only two platforms became the focus of Phase 2/3 efficacy trials in Liberia, Sierra Leone, and Guinea near the peak of the epidemic: the Vesicular stomatitis virus (VSV)-vectored vaccine and the chimpanzee adenovirus type 3 (ChAd3)-vectored vaccine. The results of three distinct clinical trials involving these candidates may soon pave the way for a licensed, safe and efficacious EBOV vaccine to help combat future epidemics.


Subject(s)
Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animals , Drug Evaluation, Preclinical , Ebolavirus/genetics , Ebolavirus/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Humans , Macaca fascicularis , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Virus-Like Particle/immunology
9.
Arch Virol ; 163(3): 587-597, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29149434

ABSTRACT

To be effective, therapeutic cancer vaccines should stimulate both an effective cell-mediated and a robust cytotoxic CD8+ T-cell response against human papillomavirus (HPV)-infected cells to treat the pre-existing tumors and prevent potential future tumors. In this study, the therapeutic experiments were designed in order to evaluate antitumor effect against the syngeneic TC-1 tumor model. The anti-tumor efficacy of a HPV-16 E7 DNA vaccine adjuvanted with melatonin (MLT) was evaluated in a C57BL/6 mouse tumor model by measuring tumor growth post vaccination and the survival rate of tumor-bearing mice, analyzing the specific lymphocyte proliferation responses in control and vaccinated mice by MTT assay. The E7-specific cytotoxic T cells (CTL) were analyzed by lymphocyte proliferation and lactate dehydrogenates (LDH) release assays. IFN-γ, IL-4 and TNF-α secretion in splenocyte cultures as well as vascular endothelial growth factor (VEGF) and IL-10 in the tumor microenvironment were assayed by ELISA. Our results demonstrated that subcutaneous administration of C57BL/6 mice with a DNA vaccine adjuvanted with MLT dose-dependently and significantly induced strong HPV16 E7-specific CD8+ cytotoxicity and IFN-γ and TNF-α responses capable of reducing HPV-16 E7-expressing tumor volume. A significantly higher level of E7-specific T-cell proliferation was also found in the adjuvanted vaccine group. Furthermore, tumor growth was significantly inhibited when the DNA vaccine was combined with MLT and the survival time of TC-1 tumor bearing mice was also significantly prolonged. In vivo studies further demonstrated that MLT decreased the accumulation of IL-10 and VEGF in the tumor microenvironment of vaccinated mice. These data indicate that melatonin as an adjuvant augmented the cancer vaccine efficiency against HPV-associated tumors in a dose dependent manner.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Human papillomavirus 16/drug effects , Melatonin/administration & dosage , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Vaccines, DNA/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/immunology , Female , Gene Expression Regulation , Human papillomavirus 16/genetics , Human papillomavirus 16/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocyte Activation/drug effects , Melatonin/immunology , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/mortality , Papillomavirus Infections/virology , Papillomavirus Vaccines/genetics , Papillomavirus Vaccines/immunology , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology
10.
Hum Vaccin Immunother ; 13(12): 2996-3009, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28933684

ABSTRACT

Our previous preclinical studies and a Phase I clinical trial DP6-001 have indicated that a polyvalent Env formulation was able to elicit broadly reactive antibody responses including low titer neutralizing antibody responses against viral isolates of subtypes A, B, C and AE. In the current report, a panel of 62 gp120 immunogens were screened in a rabbit model to identify gp120 immunogens that can elicit improved binding and neutralizing antibody responses and some of them can be included in the next polyvalent formulation. Only about 19% of gp120 immunogens in this panel were able to elicit neutralizing antibodies against greater than 50% of the viruses included in a high throughput PhenoSense neutralization assay when these immuongens were tested as a DNA prime followed by a fixed 5-valent gp120 protein vaccine boost. The new polyvalent formulation, using five gp120 immunogens selected from this subgroup, elicited improved quality of antibody responses in rabbits than the previous DP6-001 formulation. More significantly, this new polyvalent formulation elicited higher antibody responses against a panel of gp70V1/V2 antigens expressing V1/V2 sequences from diverse subtypes. Bioinformatics analysis supports the design of a 4-valent or 5-valent formulation using gp120 immunogens from this screening study to achieve a broad coverage against 16 HIV-1 subtypes.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Drug Evaluation, Preclinical , HIV Envelope Protein gp120/genetics , Immunization Schedule , Neutralization Tests , Rabbits , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
11.
Hum Vaccin Immunother ; 13(7): 1586-1597, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28272978

ABSTRACT

The development of safe and potent vaccines for human respiratory syncytial virus (HRSV) is still a challenge for researchers worldwide. DNA-based immunization is currently a promising approach that has been used to generate human vaccines for different age groups. In this study, novel HRSV DNA vaccine candidates were generated and preclinically tested in BALB/c mice. Three different versions of the codon-optimized HRSV fusion (F) gene were individually cloned into the pPOE vector. The new recombinant vectors either express full-length (pPOE-F), secretory (pPOE-TF), or M282-90 linked (pPOE-FM2) forms of the F protein. Distinctive expression of the F protein was identified in HEp-2 cells transfected with the different recombinant vectors using ELISA and immunofluorescence. Mice immunization verified the potential for recombinant vectors to elicit significant levels of neutralizing antibodies and CD8+ T-cell lymphocytes. pPOE-TF showed higher levels of gene expression in cell culture and better induction of the humoral and cellular immune responses. Following virus challenge, mice that had been immunized with the recombinant vectors were able to control virus replication and displayed lower inflammation compared with mice immunized with empty pPOE vector or formalin-inactivated HRSV vaccine. Moreover, pulmonary cytokine profiles of mice immunized with the 3 recombinant vectors were similar to those of the mock infected group. In conclusion, recombinant pPOE vectors are promising HRSV vaccine candidates in terms of their safety, immunogenicity and protective efficiency. These data encourage further evaluation in phase I clinical trials.


Subject(s)
Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Mice, Inbred BALB C , Potexvirus , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/isolation & purification , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/isolation & purification , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
12.
Biomed Res Int ; 2017: 2145386, 2017.
Article in English | MEDLINE | ID: mdl-29359145

ABSTRACT

Growing evidence shows that antioxidant proteins of Leishmania could be used as vaccine candidates. In this study, we report the efficacy of Leishmania donovani iron superoxide dismutase B1 (LdFeSODB1) as a vaccine antigen in BALB/c mice in a DNA-protein prime-boost immunization regimen in the presence or absence of murine granulocyte macrophage colony stimulating factor (mGMCSF) DNA adjuvant. The expression study confirmed that LdFeSODB1 is expressed in mammalian cells and mGMCSF fusion mediates the secretion of the recombinant protein. Heterologous immunization with LdFeSODB1 induced a strong antibody- and cell-mediated immune response in mice. Immunization triggered a mixed Th1/Th2 response as evidenced by the ratio of IgG2a to IgG1. Antigen-stimulated spleen cells from the immunized mice produced high level IFN-γ. Multiparametric flow cytometry data showed that immunization with LdFeSODB1 induced significantly higher expression of TNF-α or IL-2 by antigen-stimulated T cells. Eight weeks after L. major infection, immunization with the antigen shifted the immune response to a more Th1 type than the controls as demonstrated by IgG2a/IgG1 ratio. Moreover, IFN-γ production by antigen-stimulated spleen cells from immunized mice remained high. The footpad swelling experiment showed that immunization with LdFeSODB1 resulted in partial protection of mice from a high dose L. major infection.


Subject(s)
Leishmania donovani/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Recombinant Proteins/immunology , Superoxide Dismutase/immunology , Vaccines, DNA/immunology , Animals , Cytokines/analysis , Cytokines/genetics , Cytokines/metabolism , Female , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/prevention & control , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Vaccines/chemistry , Protozoan Vaccines/genetics , Protozoan Vaccines/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Vaccines, DNA/genetics , Vaccines, DNA/metabolism
13.
Parasitol Res ; 116(2): 609-616, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27909791

ABSTRACT

Many recent studies have been conducted to evaluate protective immunity mediated by DNA vaccines against toxoplasmosis. Cocktail DNA vaccines showed better immune responses compared to single vaccines. The objective of the current study was to evaluate the protective efficacy of rhomboid 4 (ROM4) and cocktail DNA vaccines (ROM4 + GRA14) of the Toxoplasma gondii RH strain with or without coated calcium phosphate nanoparticles (CaPNs) as the adjuvant to improve the immunogenicity against the T. gondii RH strain in BALB/c mice. Cocktail DNA vaccines of pcROM4 + pcGRA14 of the T. gondii RH strain were constructed. CaPNs were synthesized and the cocktail DNA vaccine was coated with the adjuvant of CaPNs. Immunogenicity and the protective effects of cocktail DNA vaccines with or without CaPNs against lethal challenge were evaluated in BALB/c mice. pcROM4 and cocktail DNA vaccine coated with CaPNs significantly enhanced cellular and humoral immune responses against Toxoplasma compared to pcROM4 and cocktail DNA vaccine without CaPNs (p < 0.05). These findings indicate that the survival time of immunized mice after challenge with the RH strain of T. gondii was increased compared to that of controls and the DNA vaccine provided significant protection in mice (p < 0.05). The CaPN-based cocktail DNA vaccine of pcROM4 + pcGRA14 showed the longest survival time compared to the other groups. Co-immunization with CaPN-based cocktail DNA vaccine (pcROM4 + pcGRA14) boosted immune responses and increased the protective efficacy against acute toxoplasmosis in BALB/c mice compared to both single gene and bivalent DNA vaccine without nano-adjuvants.


Subject(s)
Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/genetics , Animals , Antibodies, Protozoan/immunology , Calcium Phosphates/chemistry , DNA , Female , Humans , Immunity, Humoral , Immunization , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Protozoan Proteins/genetics , Protozoan Vaccines/immunology , Toxoplasma/genetics , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/prevention & control , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
14.
Expert Opin Biol Ther ; 16(1): 67-77, 2016.
Article in English | MEDLINE | ID: mdl-26512415

ABSTRACT

INTRODUCTION: Tremendous progress has been achieved during the last decades in electroporation-based technologies for medicine. Understanding the basic underlying mechanisms of gene delivery opens the way for clinical gene therapy and DNA vaccination. AREAS COVERED: This review focuses on the use of gene electrotherapy in cutaneous tissue repair and how it affects healing. EXPERT OPINION: Gene electrotherapy is safe, efficient and promising as shown by the increasing number of publications reporting evidence for its potential in wound healing. Going deeper into the mechanisms of DNA delivery and expression as well as into skin regeneration at the molecular, cellular and tissues levels will help make it an attractive approach for the treatment of skin pathologies in general.


Subject(s)
Electroporation/methods , Gene Transfer Techniques , Genetic Therapy/methods , Skin/injuries , Wound Healing/physiology , Humans , Re-Epithelialization , Vaccines, DNA/genetics , Wound Healing/genetics
15.
Vaccine ; 33(50): 7135-40, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26458805

ABSTRACT

Since the early 1990s, DNA immunization has been used as a platform for developing a tetravalent dengue vaccine in response to the high priority need for protecting military personnel deployed to dengue endemic regions of the world. Several approaches have been explored ranging from naked DNA immunization to the use of live virus vectors to deliver the targeted genes for expression. Pre-clinical animal studies were largely successful in generating anti-dengue cellular and humoral immune responses that were protective either completely or partially against challenge with live dengue virus. However, Phase 1 clinical evaluation of a prototype monovalent dengue 1 DNA vaccine expressing prM and E genes revealed anti-dengue T cell IFNγ responses, but poor neutralizing antibody responses. These less than optimal results are thought to be due to poor uptake and expression of the DNA vaccine plasmids. Because DNA immunization as a vaccine platform has the advantages of ease of manufacture, flexible genetic manipulation and enhanced stability, efforts continue to improve the immunogenicity of these vaccines using a variety of methods.


Subject(s)
Dengue Vaccines/administration & dosage , Dengue Vaccines/immunology , Dengue/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Clinical Trials, Phase I as Topic , Dengue/epidemiology , Dengue Vaccines/genetics , Dengue Vaccines/isolation & purification , Drug Evaluation, Preclinical , Humans , Primates , Vaccines, DNA/genetics , Vaccines, DNA/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
16.
Sci Rep ; 5: 12478, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26212482

ABSTRACT

Immunomodulatory oligodeoxynucleotides (IMODNs) are the short DNA sequences that activate the innate immune system via toll-like receptor 9. These sequences predominantly contain unmethylated CpG motifs. In this work, we describe VaccineDA (Vaccine DNA adjuvants), a web-based resource developed to design IMODN-based vaccine adjuvants. We collected and analyzed 2193 experimentally validated IMODNs obtained from the literature. Certain types of nucleotides (e.g., T, GT, TC, TT, CGT, TCG, TTT) are dominant in IMODNs. Based on these observations, we developed support vector machine-based models to predict IMODNs using various compositions. The developed models achieved the maximum Matthews Correlation Coefficient (MCC) of 0.75 with an accuracy of 87.57% using the pentanucleotide composition. The integration of motif information further improved the performance of our model from the MCC of 0.75 to 0.77. Similarly, models were developed to predict palindromic IMODNs and attained a maximum MCC of 0.84 with the accuracy of 91.94%. These models were evaluated using a five-fold cross-validation technique as well as validated on an independent dataset. The models developed in this study were integrated into VaccineDA to provide a wide range of services that facilitate the design of DNA-based vaccine adjuvants (http://crdd.osdd.net/raghava/vaccineda/).


Subject(s)
Drug Evaluation, Preclinical/methods , Software , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Algorithms , Base Sequence , Chromosome Mapping/methods , Drug Design , Humans , Internet , Molecular Sequence Data , Sequence Analysis, DNA/methods
17.
Hum Vaccin Immunother ; 11(8): 1991-2004, 2015.
Article in English | MEDLINE | ID: mdl-25996997

ABSTRACT

Cynomolgus macaques were vaccinated by intramuscular electroporation with DNA plasmids expressing codon-optimized glycoprotein (GP) genes of Ebola virus (EBOV) or Marburg virus (MARV) or a combination of codon-optimized GP DNA vaccines for EBOV, MARV, Sudan virus and Ravn virus. When measured by ELISA, the individual vaccines elicited slightly higher IgG responses to EBOV or MARV than did the combination vaccines. No significant differences in immune responses of macaques given the individual or combination vaccines were measured by pseudovirion neutralization or IFN-γ ELISpot assays. Both the MARV and mixed vaccines were able to protect macaques from lethal MARV challenge (5/6 vs. 6/6). In contrast, a greater proportion of macaques vaccinated with the EBOV vaccine survived lethal EBOV challenge in comparison to those that received the mixed vaccine (5/6 vs. 1/6). EBOV challenge survivors had significantly higher pre-challenge neutralizing antibody titers than those that succumbed.


Subject(s)
Electroporation , Filoviridae/immunology , Hemorrhagic Fever, Ebola/prevention & control , Marburg Virus Disease/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Codon , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Filoviridae/genetics , Glycoproteins/genetics , Glycoproteins/immunology , Immunoglobulin G/blood , Injections, Intramuscular , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Macaca fascicularis , Male , Neutralization Tests , Plasmids , Survival Analysis , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
18.
Clin Vaccine Immunol ; 21(11): 1474-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165025

ABSTRACT

This study was conducted to evaluate the immunogenicity and protective efficacy of a DNA vaccine encoding Brucella abortus Cu,Zn superoxide dismutase (SOD) using the Toll-like receptor 2/6 agonist S-[2,3-bispalmitoyiloxy-(2R)-propyl]-R-cysteinyl-amido-monomethoxy polyethylene glycol (BPPcysMPEG) as an adjuvant. Intranasal coadministration of BPPcysMPEG with a plasmid carrying the SOD-encoding gene (pcDNA-SOD) into BALB/c mice elicited antigen-specific humoral and cellular immune responses. Humoral responses were characterized by the stimulation of IgG2a and IgG1 and by the presence of SOD-specific secretory IgA in nasal and bronchoalveolar lavage fluids. Furthermore, T-cell proliferative responses and increased production of gamma interferon were also observed upon splenocyte restimulation with recombinant SOD. Cytotoxic responses were also stimulated, as demonstrated by the lysis of RB51-SOD-infected J774.A1 macrophages by cells recovered from immunized mice. The pcDNA-SOD/BPPcysMPEG formulation induced improved protection against challenge with the virulent strain B. abortus 2308 in BALB/c mice over that provided by pcDNA-SOD, suggesting the potential of this vaccination strategy against Brucella infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Brucella Vaccine/immunology , Brucella abortus/enzymology , Brucellosis/prevention & control , Polyethylene Glycols/administration & dosage , Superoxide Dismutase/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bronchoalveolar Lavage Fluid/immunology , Brucella Vaccine/administration & dosage , Brucella Vaccine/genetics , Brucella abortus/genetics , Brucellosis/immunology , Cell Proliferation , Cytotoxicity Tests, Immunologic , Disease Models, Animal , Female , Immunoglobulin A, Secretory/analysis , Immunoglobulin G/blood , Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred BALB C , Nasal Mucosa/immunology , Polyethylene Glycols/pharmacology , Spleen/immunology , Superoxide Dismutase/genetics , T-Lymphocytes/immunology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/agonists , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
19.
Am J Rhinol Allergy ; 28(3): e144-7, 2014.
Article in English | MEDLINE | ID: mdl-24980227

ABSTRACT

BACKGROUND: Allergic rhinitis is one of the most common atopic disorders in children. There is no available method to prevent airway sensitization in newborns except allergen avoidance. Recombinant DNA plasmids encoding allergens have been proven to activate Th1 but attenuate Th2-deviated allergic responses in adult animal studies. However, their preventive effects are not presumptive in neonates because of their immature immune function. The aim of this study was to examine the potential preventive effect of a DNA vaccine encoding grass pollen allergen Cyn d 1 on allergic reaction to grass pollen in neonatal mice. METHODS: Recombinant plasmid Cyn d 1 (pCyn d 1) vaccine was constructed by insertion of Cyn d 1 cDNA into the vector pcDNA3. Neonatal BALB/c mice received the vaccine once on the 3rd day of life or a second dose 2 days later. Control mice received PBS only. Mice were sensitized twice with recombinant Cyn d 1 and alum beginning at 7 weeks of age. Serum antibody responses and cytokine profiles of spleen cells were examined. RESULTS: Neonatal injection with pCyn d 1 vaccine resulted in IgG2a responses and production of interferon gamma in spleen cells. Vaccination with pCyn d 1 also reduced specific IgE responses and spleen cell secretion of IL-4. CONCLUSION: This study shows the prophylactic effects of DNA vaccine encoding Bermuda grass pollen allergen Cyn d 1 on specific IgE responses in neonatal mice.


Subject(s)
Antigens, Plant/immunology , Rhinitis, Allergic, Seasonal/prevention & control , Vaccines, DNA/immunology , Animals , Animals, Newborn , Antigens, Plant/genetics , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Poaceae , Pollen/adverse effects , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Vaccination , Vaccines, DNA/genetics
20.
PLoS One ; 9(1): e86578, 2014.
Article in English | MEDLINE | ID: mdl-24497955

ABSTRACT

BACKGROUND: Schistosomiasis japonica remains a major public-health concern in China. Praziquantel-based chemotherapy effectively reduces both infections and intensity; however, it can not prevent re-infection. Furthermore, there is an increasing concern about praziquantel resistance following long-term repeated use of the drug in endemic areas. Therefore, development of a schistosomiasis vaccine, as a strategy to prevent and control schistosomiasis japonica, has been given high priority. The present study was conducted to develop PAMAM dendrimers as a novel vaccine delivery vector for a schistosomiasis japonica DNA vaccine and evaluate its ability to enhance protective effects against Schistosoma japonicum infection. METHODOLOGY/PRINCIPAL FINDINGS: Lysine was used to modify 4.0G PAMAM, and the modified product PAMAM-Lys was synthesized. PAMAM-Lys showed both high transfection and low cytotocity for gene delivery in vitro. DNA vaccines combined with PAMAM-Lys produced higher level of protection compare with naked DNA vaccines against S. japonicum infection in a mouse model. Futhermore,antibodies from mice immunized with PAMAM-Lys combined DNA vaccines were significantly higher than those of mice immunized with the naked DNA vaccines. The PAMAM-Lys vector elicited a predominantly IgG2a antibody response and a tremendously increase in the production of IL-2 and IFN-γ. CONCLUSION/SIGNIFICANCE: Lysine-modified PAMAM-Lys is an excellent vector. PAMAM-Lys may enhance the immunoreactivity of DNA vaccine and increase the protective effect of the SjC23 DNA vaccine against S. japonicum infection.


Subject(s)
Adjuvants, Immunologic/chemistry , Dendrimers/chemistry , Lysine/analogs & derivatives , Schistosoma japonicum/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Helminth/blood , Dendrimers/pharmacology , Female , HEK293 Cells , Humans , Immunity, Humoral , Liver/parasitology , Lysine/chemistry , Lysine/pharmacology , Mice, Inbred BALB C , Plasmids , Schistosomiasis japonica/immunology , Schistosomiasis japonica/parasitology , Schistosomiasis japonica/prevention & control , Snails/parasitology , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL