Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.694
Filtrer
1.
BMC Biotechnol ; 24(1): 71, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39350162

RÉSUMÉ

BACKGROUND: Human papillomavirus type 16 (HPV-16) infection is strongly associated with considerable parts of cervical, neck, and head cancers. Performed investigations have had moderate clinical success, so research to reach an efficient vaccine has been of great interest. In the present study, the immunization potential of a newly designed HPV-16 construct was evaluated in a mouse model. RESULTS: Initially, a construct containing HPV-16 mutant (m) E6/E7 fusion gene was designed and antigen produced in two platforms (i.e., DNA vaccine and recombinant protein). Subsequently, the immunogenicity of these platforms was investigated in five mice) C57BL/6 (groups based on several administration strategies. Three mice groups were immunized recombinant protein, DNA vaccine, and a combination of them, and two other groups were negative controls. The peripheral blood mononuclear cells (PBMCs) proliferation, Interleukin-5 (IL-5) and interferon-γ (IFN-γ) cytokines, IgG1 and IgG2a antibody levels were measured. After two weeks, TC-1 tumor cells were injected into all mice groups, and subsequently further analysis of tumor growth and metastasis and mice survival were performed according to the schedule. Overall, the results obtained from in vitro immunology and tumor cells challenging assays indicated the potential of the mE6/E7 construct as an HPV16 therapeutic vaccine candidate. The results demonstrated a significant increase in IFN-γ cytokine (P value < 0.05) in the Protein/Protein (D) and DNA/Protein (E) groups. This finding was in agreement with in vivo assays. Control groups show a 10.5-fold increase (P value < 0.001) and (C) DNA/DNA group shows a 2.5-fold increase (P value < 0.01) in tumor growth compared to D and E groups. Also, a significant increase in survival of D and E (P value < 0.001) and C (P value < 0.01) groups were observed. CONCLUSIONS: So, according to the findings, the recombinant protein could induce stronger protection compared to the DNA vaccine form. Protein/Protein and DNA/Protein are promising administration strategies for presenting this construct to develop an HPV-16 therapeutic vaccine candidate.


Sujet(s)
Papillomavirus humain de type 16 , Souris de lignée C57BL , Protéines des oncogènes viraux , Protéines E7 de papillomavirus , Vaccins contre les papillomavirus , Protéines de répression , Vaccins à ADN , Animaux , Protéines des oncogènes viraux/génétique , Protéines des oncogènes viraux/immunologie , Protéines E7 de papillomavirus/génétique , Protéines E7 de papillomavirus/immunologie , Souris , Protéines de répression/génétique , Protéines de répression/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Papillomavirus humain de type 16/génétique , Papillomavirus humain de type 16/immunologie , Vaccins contre les papillomavirus/immunologie , Vaccins contre les papillomavirus/génétique , Vaccins contre les papillomavirus/administration et posologie , Femelle , Infections à papillomavirus/prévention et contrôle , Infections à papillomavirus/virologie , Infections à papillomavirus/immunologie , Modèles animaux de maladie humaine , Humains , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie
2.
J Immunotoxicol ; 21(1): 2400624, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39319829

RÉSUMÉ

Infections caused by the influenza virus lead to both epidemic and pandemic outbreaks in humans and animals. Owing to their rapid production, safety, and stability, DNA vaccines represent a promising avenue for eliciting immunity and thwarting viral infections. While DNA vaccines have demonstrated substantial efficacy in murine models, their effectiveness in larger animals remains subdued. This limitation may be addressed by augmenting the immunogenicity of DNA-based vaccines. In the investigation here, protein expression was enhanced via codon optimization and then mouse cytotoxic T-lymphocyte antigen 4 (CTLA-4) was harnessed as a modulatory adjunct to bind directly to antigen-presenting cells. Further, the study evaluated the immunogenicity of two variants of the hemagglutinin (HA) antigen, i.e. the full-length and the C-terminal deletion versions. The study findings revealed that the codon-optimized HA gene (pcHA) led to increased protein synthesis, as evidenced by elevated mRNA levels. Codon optimization also significantly bolstered both cellular and humoral immune responses. In cytokine assays, all plasmid constructs, particularly pCTLA4-cHA, induced robust interferon (IFN)-γ production, while interleukin (IL)-4 levels remained uniformly non-significant. Mice immunized with pcHA displayed an augmented presence of IFNγ+ T-cells, underscoring the enhanced potency of the codon-optimized HA vaccine. Contrarily, CTLA-4-fused DNA vaccines did not significantly amplify the immune response.


Sujet(s)
Antigène CTLA-4 , Codon , Glycoprotéine hémagglutinine du virus influenza , Vaccins antigrippaux , Infections à Orthomyxoviridae , Vaccins à ADN , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Souris , Antigène CTLA-4/génétique , Antigène CTLA-4/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Codon/génétique , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Humains , Femelle , Souris de lignée BALB C , Modèles animaux de maladie humaine , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Sous-type H1N1 du virus de la grippe A/immunologie
3.
Sci Rep ; 14(1): 21742, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39289418

RÉSUMÉ

Vaccine manufacturing fosters the prevention, control, and eradication of infectious diseases. Recombinant DNA and in vitro (IVT) mRNA vaccine manufacturing technologies were enforced to combat the recent pandemic. Despite the impact of these technologies, there exists no scientific announcement that compares them. Digital Shadows are employed in this study to simulate each technology, investigating root cause deviations, technical merits, and liabilities, evaluating cost scenarios. Under this lens we provide an unbiased, advanced comparative technoeconomic study, one that determines which of these manufacturing platforms are suited for the two types of vaccines considered (monoclonal antibodies or antigens). We find recombinant DNA technology to exhibit higher Profitability Index due to lower capital and starting material requirements, pertaining to lower Minimum Selling Price per Dose values, delivering products of established quality. However, the potency of the mRNA, the streamlined and scalable synthetic processes involved and the raw material availability, facilitate faster market penetration and product flexibility, constituting these vaccines preferable whenever short product development cycles become a necessity.


Sujet(s)
ARN messager , ARN messager/génétique , ARN messager/immunologie , Humains , ADN recombiné/génétique , Vaccins/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Anticorps monoclonaux/immunologie , Développement de vaccin
4.
Antiviral Res ; 230: 105978, 2024 10.
Article de Anglais | MEDLINE | ID: mdl-39117282

RÉSUMÉ

Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.


Sujet(s)
Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Sialidase/immunologie , Sialidase/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/génétique , Animaux , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Souris , Lymphocytes T/immunologie , Souris de lignée BALB C , Femelle , Humains , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Lymphocytes auxiliaires Th1/immunologie , Protéines virales/génétique , Protéines virales/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang
5.
Egypt J Immunol ; 31(3): 95-112, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38995715

RÉSUMÉ

In this study, we aimed to evaluate the immunogenic profile of a chimeric DNA-based hepatitis C virus (HCV) vaccine candidate encoding the full-length viral core-E1-E2 (HCV-CE) fragment. The vaccine candidate was designed to uniformly express the HCV genotype 4 core-E1-E2 protein. The recombinant HCV-CE protein was bacterially expressed in C41 (DE3) cells, and then BALB/c mice were immunized with different combinations of DNA/DNA or DNA/protein prime/boost immunizations. The proper construction of our vaccine candidate was confirmed by specific amplification of the encoded fragments and basic local alignment search tool (BLAST) results of the nucleotide sequence, which revealed a high degree of similarity with several HCV serotypes/genotypes. The platform for bacterial expression was optimized to maximize the yield of the purified recombinant HCV-CE protein. The recombinant protein showed high specific antigenicity against the sera of HCV-infected patients according to the ELISA and western blot results. The predicted B- and T-cell epitopes showed high antigenic and interferon-γ (IFN-γ) induction potential, in addition to cross-genotype conservation and population coverage. The mice antisera further demonstrated a remarkable ability to capture 100% of the native viral antigens circulating in the sera of HCV patients, with no cross-reactivity detected in control sera. In conclusion, the proposed HCV vaccination strategy demonstrated promising potential regarding its safety, immunogenicity, and population coverage.


Sujet(s)
Hepacivirus , Hépatite C , Souris de lignée BALB C , Vaccins à ADN , Vaccins contre les hépatites virales , Animaux , Hepacivirus/immunologie , Hepacivirus/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Souris , Vaccins contre les hépatites virales/immunologie , Hépatite C/prévention et contrôle , Hépatite C/immunologie , Humains , Immunogénicité des vaccins/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines du core viral/immunologie , Protéines du core viral/génétique , Femelle , Anticorps de l'hépatite C/immunologie , Anticorps de l'hépatite C/sang
6.
Acta Trop ; 257: 107302, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38959992

RÉSUMÉ

Toxoplasma gondii is an important protozoan pathogen, which can cause severe diseases in the newborns and immunocompromised individuals. Developing an effective vaccine against Toxoplasma infection is a critically important global health priority. Immunofluorescence staining analysis revealed that TgSAG2 and TgSRS2 are membrane associated and displayed on the surface of the parasite. Immunizations with pBud-SAG2, pBud-SRS2 and pBud-SAG2-SRS2 DNA vaccines significantly increased the production of specific IgG antibodies. Immunization with pBud-SAG2-SRS2 elicited cellular immune response with higher concentrations of IFN-γ and IL-4 compared to the control group. Antigen-specific lymphocyte proliferations in the pBud-SRS2 and pBud-SAG2-SRS2 groups were significantly higher compared to that in the control group. Furthermore, 30 % of mice immunized with pBud-SAG2-SRS2 survived after the challenge infection with virulent T. gondii RH tachyzoites. This study revealed that immunization with pBud-SAG2-SRS2 induced potent immune responses, and has the potential as a promising vaccine candidate for the control of T. gondii infection.


Sujet(s)
Anticorps antiprotozoaires , Antigènes de protozoaire , Immunoglobuline G , Protéines de protozoaire , Vaccins antiprotozoaires , Toxoplasma , Toxoplasmose animale , Vaccins à ADN , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Antigènes de protozoaire/immunologie , Antigènes de protozoaire/génétique , Protéines de protozoaire/immunologie , Protéines de protozoaire/génétique , Toxoplasma/immunologie , Toxoplasma/génétique , Anticorps antiprotozoaires/sang , Vaccins antiprotozoaires/immunologie , Vaccins antiprotozoaires/administration et posologie , Vaccins antiprotozoaires/génétique , Souris , Immunoglobuline G/sang , Femelle , Toxoplasmose animale/prévention et contrôle , Toxoplasmose animale/immunologie , Souris de lignée BALB C , Interféron gamma/immunologie , Modèles animaux de maladie humaine , Prolifération cellulaire , Interleukine-4/immunologie , Analyse de survie
7.
Sci Rep ; 14(1): 13865, 2024 06 15.
Article de Anglais | MEDLINE | ID: mdl-38879684

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 had devastating consequences for human health. Despite the introduction of several vaccines, COVID-19 continues to pose a serious health risk due to emerging variants of concern. DNA vaccines gained importance during the pandemic due to their advantages such as induction of both arms of immune response, rapid development, stability, and safety profiles. Here, we report the immunogenicity and protective efficacy of a DNA vaccine encoding spike protein with D614G mutation (named pcoSpikeD614G) and define a large-scale production process. According to the in vitro studies, pcoSpikeD614G expressed abundant spike protein in HEK293T cells. After the administration of pcoSpikeD614G to BALB/c mice through intramuscular (IM) route and intradermal route using an electroporation device (ID + EP), it induced high level of anti-S1 IgG and neutralizing antibodies (P < 0.0001), strong Th1-biased immune response as shown by IgG2a polarization (P < 0.01), increase in IFN-γ levels (P < 0.01), and increment in the ratio of IFN-γ secreting CD4+ (3.78-10.19%) and CD8+ (5.24-12.51%) T cells. Challenging K18-hACE2 transgenic mice showed that pcoSpikeD614G administered through IM and ID + EP routes conferred 90-100% protection and there was no sign of pneumonia. Subsequently, pcoSpikeD614G was evaluated as a promising DNA vaccine candidate and scale-up studies were performed. Accordingly, a large-scale production process was described, including a 36 h fermentation process of E. coli DH5α cells containing pcoSpikeD614G resulting in a wet cell weight of 242 g/L and a three-step chromatography for purification of the pcoSpikeD614G DNA vaccine.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Souris de lignée BALB C , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Vaccins à ADN , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à ADN/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Animaux , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Souris , COVID-19/prévention et contrôle , COVID-19/immunologie , Cellules HEK293 , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Femelle , Immunogénicité des vaccins , Immunoglobuline G/sang , Immunoglobuline G/immunologie
8.
Curr HIV Res ; 22(2): 109-119, 2024.
Article de Anglais | MEDLINE | ID: mdl-38712371

RÉSUMÉ

BACKGROUND: Heterologous combinations in vaccine design are an effective approach to promote T cell activity and antiviral effects. The goal of this study was to compare the homologous and heterologous regimens targeting the Nef-Tat fusion antigen to develop a human immunodeficiency virus-1 (HIV-1) therapeutic vaccine candidate. METHODS: At first, the DNA and protein constructs harboring HIV-1 Nef and the first exon of Tat as linked form (pcDNA-nef-tat and Nef-Tat protein) were prepared in large scale and high purity. The generation of the Nef-Tat protein was performed in the E. coli expression system using an IPTG inducer. Then, we evaluated and compared immune responses of homologous DNA prime/ DNA boost, homologous protein prime/ protein boost, and heterologous DNA prime/protein boost regimens in BALB/c mice. Finally, the ability of mice splenocytes to secret cytokines after exposure to single-cycle replicable (SCR) HIV-1 was compared between immunized and control groups in vitro. RESULTS: The nef-tat gene was successfully subcloned in eukaryotic pcDNA3.1 (-) and prokaryotic pET-24a (+) expression vectors. The recombinant Nef-Tat protein was generated in the E. coli Rosetta strain under optimized conditions as a clear band of ~ 35 kDa detected on SDS-PAGE. Moreover, transfection of pcDNA-nef-tat into HEK-293T cells was successfully performed using Lipofectamine 2000, as confirmed by western blotting. The immunization studies showed that heterologous DNA prime/protein boost regimen could significantly elicit the highest levels of Ig- G2a, IFN-γ, and Granzyme B in mice as compared to homologous DNA/DNA and protein/protein regimens. Moreover, the secretion of IFN-γ was higher in DNA/protein regimens than in DNA/DNA and protein/protein regimens after exposure of mice splenocytes to SCR HIV-1 in vitro. CONCLUSION: The chimeric HIV-1 Nef-Tat antigen was highly immunogenic, especially when applied in a heterologous prime/ boost regimen. This regimen could direct immune response toward cellular immunity (Th1 and CTL activity) and increase IFN-γ secretion after virus exposure.


Sujet(s)
Vaccins contre le SIDA , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Souris de lignée BALB C , Protéines de fusion recombinantes , Vaccins à ADN , Produits du gène nef du virus de l'immunodéficience humaine , Produits du gène tat du virus de l'immunodéficience humaine , Animaux , Produits du gène nef du virus de l'immunodéficience humaine/immunologie , Produits du gène nef du virus de l'immunodéficience humaine/génétique , Vaccins contre le SIDA/immunologie , Vaccins contre le SIDA/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Produits du gène tat du virus de l'immunodéficience humaine/immunologie , Produits du gène tat du virus de l'immunodéficience humaine/génétique , Souris , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/génétique , Humains , Femelle , Lymphocytes T/immunologie , Infections à VIH/immunologie , Infections à VIH/prévention et contrôle , Rappel de vaccin , Cytokines/métabolisme , Escherichia coli/génétique , Escherichia coli/immunologie
9.
Hum Vaccin Immunother ; 20(1): 2346390, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38691025

RÉSUMÉ

Middle East respiratory coronavirus (MERS-CoV) is a newly emergent, highly pathogenic coronavirus that is associated with 34% mortality rate. MERS-CoV remains listed as priority pathogen by the WHO. Since its discovery in 2012 and despite the efforts to develop coronaviruses vaccines to fight against SARS-CoV-2, there are currently no MERS-CoV vaccine that has been approved. Therefore, there is high demand to continue on the development of prophylactic vaccines against MERS-CoV. Current advancements in vaccine developments can be adapted for the development of improved MERS-CoV vaccines candidates. Nucleic acid-based vaccines, including pDNA and mRNA, are relatively new class of vaccine platforms. In this work, we developed pDNA and mRNA vaccine candidates expressing S.FL gene of MERS-CoV. Further, we synthesized a silane functionalized hierarchical aluminosilicate to encapsulate each vaccine candidates. We tested the nucleic acid vaccine candidates in mice and evaluated humoral antibodies response. Interestingly, we determined that the non-encapsulated, codon optimized S.FL pDNA vaccine candidate elicited the highest level of antibody responses against S.FL and S1 of MERS-CoV. Encapsulation of mRNA with nanoporous aluminosilicate increased the humoral antibody responses, whereas encapsulation of pDNA did not. These findings suggests that MERS-CoV S.FL pDNA vaccine candidate induced the highest level of humoral responses. This study will enhance further optimization of nanosilica as potential carrier for mRNA vaccines. In conclusion, this study suggests MERS-CoV pDNA vaccine candidate as a suitable vaccine platform for further pivotal preclinical testings.


Sujet(s)
Anticorps antiviraux , Infections à coronavirus , Coronavirus du syndrome respiratoire du Moyen-Orient , Nanoparticules , Silice , Vaccins à ADN , Vaccins antiviraux , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Coronavirus du syndrome respiratoire du Moyen-Orient/immunologie , Coronavirus du syndrome respiratoire du Moyen-Orient/génétique , Souris , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vaccins antiviraux/administration et posologie , Anticorps antiviraux/immunologie , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/immunologie , Silice/composition chimique , Souris de lignée BALB C , Femelle , Humains , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Développement de vaccin
10.
Methods Mol Biol ; 2786: 303-319, 2024.
Article de Anglais | MEDLINE | ID: mdl-38814401

RÉSUMÉ

A plasmid production process has been established to manufacture plasmid DNA at a large scale in High-Quality grade. This is used as a starting material to produce mRNA vaccines for clinical trials. Recently, the World Health Organization (WHO) has released regulatory guidelines related to the quality, safety, and efficacy for DNA- as well as for mRNA-based vaccines. Following an extraordinary year of scientific, regulatory, and manufacturing developments, the scientific community today stands considerably better equipped to deal with urgent production requirements in large scale for nucleic acid-based vaccinations and therapies. Going forward, work needs to be done in better coordinating the supply and logistics of essential raw materials for biological manufacturing, especially under emergency conditions.


Sujet(s)
Plasmides , Vaccins à ADN , Plasmides/génétique , Humains , Vaccins à ADN/génétique , Vaccins à ADN/immunologie , Vaccins à ARNm
11.
Mol Biochem Parasitol ; 259: 111630, 2024 09.
Article de Anglais | MEDLINE | ID: mdl-38795969

RÉSUMÉ

Toxoplasma gondii is an intracellular protozoan parasite that infects all nucleated cells except the red blood cells. Currently, nucleic acid vaccines are being widely investigated in Toxoplasma gondii control, and several nucleic acid vaccine candidate antigens have shown good protection in various studies. The aim of this study was to construct a nucleic acid vaccine with Toxoplasma gondii SRS29C as the target gene. We explored the nucleic acid vaccine with Toxoplasma surface protein SRS29C and the combined gene of SRS29C and SAG1 and evaluated its immunoprotective effect against Toxoplasma gondii. To amplify the gene fragment and clone it to the expression vector, the recombinant plasmid pEGFP-SRS29C was constructed by PCR. Eukaryotic cells were transfected with the plasmid, and the expression of the target protein was assessed using the Western blot method. The level of serum IgG was determined via ELISA, and the splenic lymphocyte proliferation ability was detected using the CCK-8 method. The percentages of CD4+ and CD8+ T cells were measured by flow cytometry. Mice were immunised three times with single-gene nucleic acid vaccine and combination vaccine. Splenic lymphocytokine expression was determined using ELISA kits. The mice's survival time was monitored and recorded during an in vivo insect assault experiment, and the vaccine's protective power was assessed. The outcomes showed that PCR-amplification of an SRS29C gene fragment was successful. The 4,733-bp vector fragment and the 1,119-bp target segment were both recognised by double digestion. Additionally, after transfection of the recombinant plasmid pEGFP-SRS29C, Western blot examination of the extracted protein revealed the presence of a target protein strip at 66 kDa. The test results demonstrated that the IgG content in the serum of the pEGFP-SRS29C group and the co-immunization group was significantly higher than that of the PBS group and the empty vector group. The IgG potency induced by the co-immunization group was higher than that of the pEGFP-SRS29C group and the pEGFP-SAG1 group, the number of splenic lymphocyte proliferation number was higher than that of the PBS group and the empty vector group. The CD4+/CD8+ T ratio was higher than that of the PBS group and the empty vector group. The expression of IFN-γ and TNF-α in the splenocytes of the pEGFP-SRS29C group and the combined immunisation group was significantly higher following antigen stimulation. In the worm attack experiments, mice in the PBS and empty vector groups perished within 9 days of the worm attack, whereas mice in the pEGFP-SRS29C group survived for 18 days, mice in the pEGFP-SAG1 group survived for 21 days, and mice in the co-immunization group survived for 24 days. This demonstrates that the constructed Toxoplasma gondii nucleic acid vaccine pEGFP-SRS29C and the combined gene vaccine can induce mice to develop certain humoral and cellular immune responses, and enhance their ability to resist Toxoplasma gondii infection.


Sujet(s)
Anticorps antiprotozoaires , Antigènes de protozoaire , Immunoglobuline G , Protéines de protozoaire , Vaccins antiprotozoaires , Toxoplasma , Vaccins à ADN , Animaux , Toxoplasma/immunologie , Toxoplasma/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Protéines de protozoaire/immunologie , Protéines de protozoaire/génétique , Vaccins antiprotozoaires/immunologie , Vaccins antiprotozoaires/génétique , Souris , Anticorps antiprotozoaires/sang , Anticorps antiprotozoaires/immunologie , Antigènes de protozoaire/immunologie , Antigènes de protozoaire/génétique , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Femelle , Toxoplasmose animale/prévention et contrôle , Toxoplasmose animale/immunologie , Souris de lignée BALB C , Lymphocytes T CD8+/immunologie , Rate/immunologie , Rate/parasitologie , Prolifération cellulaire , Plasmides/génétique , Plasmides/immunologie , Cytokines/métabolisme
12.
Virol Sin ; 39(3): 490-500, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38768713

RÉSUMÉ

As of December 2022, 2603 laboratory-identified Middle East respiratory syndrome coronavirus (MERS-CoV) infections and 935 associated deaths, with a mortality rate of 36%, had been reported to the World Health Organization (WHO). However, there are still no vaccines for MERS-CoV, which makes the prevention and control of MERS-CoV difficult. In this study, we generated two DNA vaccine candidates by integrating MERS-CoV Spike (S) gene into a replicating Vaccinia Tian Tan (VTT) vector. Compared to homologous immunization with either vaccine, mice immunized with DNA vaccine prime and VTT vaccine boost exhibited much stronger and durable humoral and cellular immune responses. The immunized mice produced robust binding antibodies and broad neutralizing antibodies against the EMC2012, England1 and KNIH strains of MERS-CoV. Prime-Boost immunization also induced strong MERS-S specific T cells responses, with high memory and poly-functional (CD107a-IFN-γ-TNF-α) effector CD8+ T cells. In conclusion, the research demonstrated that DNA-Prime/VTT-Boost strategy could elicit robust and balanced humoral and cellular immune responses against MERS-CoV-S. This study not only provides a promising set of MERS-CoV vaccine candidates, but also proposes a heterologous sequential immunization strategy worthy of further development.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Infections à coronavirus , Immunité cellulaire , Immunité humorale , Souris de lignée BALB C , Coronavirus du syndrome respiratoire du Moyen-Orient , Vaccins à ADN , Vaccins antiviraux , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à ADN/génétique , Coronavirus du syndrome respiratoire du Moyen-Orient/immunologie , Coronavirus du syndrome respiratoire du Moyen-Orient/génétique , Anticorps antiviraux/sang , Souris , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/génétique , Femelle , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/immunologie , Lymphocytes T CD8+/immunologie , Virus de la vaccine/génétique , Virus de la vaccine/immunologie , Rappel de vaccin , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique
13.
BMC Infect Dis ; 24(1): 476, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38714948

RÉSUMÉ

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral disease caused by the SFTS virus (Dabie bandavirus), which has become a substantial risk to public health. No specific treatment is available now, that calls for an effective vaccine. Given this, we aimed to develop a multi-epitope DNA vaccine through the help of bioinformatics. The final DNA vaccine was inserted into a special plasmid vector pVAX1, consisting of CD8+ T cell epitopes, CD4+ T cell epitopes and B cell epitopes (six epitopes each) screened from four genome-encoded proteins--nuclear protein (NP), glycoprotein (GP), RNA-dependent RNA polymerase (RdRp), as well as nonstructural protein (NSs). To ascertain if the predicted structure would be stable and successful in preventing infection, an immunological simulation was run on it. In conclusion, we designed a multi-epitope DNA vaccine that is expected to be effective against Dabie bandavirus, but in vivo trials are needed to verify this claim.


Sujet(s)
Déterminants antigéniques des lymphocytes T , Phlebovirus , Syndrome de fièvre sévère avec thrombocytopénie , Vaccins à ADN , Vaccins antiviraux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Phlebovirus/immunologie , Phlebovirus/génétique , Syndrome de fièvre sévère avec thrombocytopénie/prévention et contrôle , Syndrome de fièvre sévère avec thrombocytopénie/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Humains , Conception assistée par ordinateur , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/génétique , Animaux , Biologie informatique
14.
Virulence ; 15(1): 2345019, 2024 12.
Article de Anglais | MEDLINE | ID: mdl-38656137

RÉSUMÉ

Klebsiella pneumoniae is an important gram-negative bacterium that causes severe respiratory and healthcare-associated infections. Although antibiotic therapy is applied to treat severe infections caused by K. pneumoniae, drug-resistant isolates pose a huge challenge to clinical practices owing to adverse reactions and the mismanagement of antibiotics. Several studies have attempted to develop vaccines against K. pneumoniae, but there are no licensed vaccines available for the control of K. pneumoniae infection. In the current study, we constructed a novel DNA vaccine, pVAX1-YidR, which encodes a highly conserved virulence factor YidR and a recombinant expression plasmid pVAX1-IL-17 encoding Interleukin-17 (IL-17) as a molecular adjuvant. Adaptive immune responses were assessed in immunized mice to compare the immunogenicity of the different vaccine schemes. The results showed that the targeted antigen gene was expressed in HEK293T cells using an immunofluorescence assay. Mice immunized with pVAX1-YidR elicited a high level of antibodies, induced strong cellular immune responses, and protected mice from K. pneumoniae challenge. Notably, co-immunization with pVAX1-YidR and pVAX1-IL-17 significantly augmented host adaptive immune responses and provided better protection against K. pneumoniae infections in vaccinated mice. Our study demonstrates that combined DNA vaccines and molecular adjuvants is a promising strategy to develop efficacious antibacterial vaccines against K. pneumoniae infections.


Sujet(s)
Vaccins antibactériens , Interleukine-17 , Infections à Klebsiella , Klebsiella pneumoniae , Vaccins à ADN , Animaux , Femelle , Humains , Souris , Immunité acquise , Adjuvants immunologiques/administration et posologie , Anticorps antibactériens/sang , Anticorps antibactériens/immunologie , Protéines bactériennes/immunologie , Protéines bactériennes/génétique , Vaccins antibactériens/immunologie , Vaccins antibactériens/génétique , Vaccins antibactériens/administration et posologie , Modèles animaux de maladie humaine , Cellules HEK293 , Immunité cellulaire , Immunisation , Interleukine-17/immunologie , Interleukine-17/génétique , Infections à Klebsiella/prévention et contrôle , Infections à Klebsiella/immunologie , Klebsiella pneumoniae/immunologie , Klebsiella pneumoniae/génétique , Souris de lignée BALB C , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Facteurs de virulence/immunologie , Facteurs de virulence/génétique
15.
Acta Biochim Biophys Sin (Shanghai) ; 56(7): 986-996, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38655616

RÉSUMÉ

Vaccines play essential roles in the fight against the COVID-19 pandemic. The development and assessment of COVID-19 vaccines have generally focused on the induction and boosting of neutralizing antibodies targeting the SARS-CoV-2 spike (S) protein. Due to rapid and continuous variation in the S protein, such vaccines need to be regularly updated to match newly emerged dominant variants. T-cell vaccines that target MHC I- or II-restricted epitopes in both structural and non-structural viral proteins have the potential to induce broadly cross-protective and long-lasting responses. In this work, the entire proteome encoded by SARS-CoV-2 (Wuhan-hu-1) is subjected to immunoinformatics-based prediction of HLA-A*02:01-restricted epitopes. The immunogenicity of the predicted epitopes is evaluated using peripheral blood mononuclear cells from convalescent Wuhan-hu-1-infected patients. Furthermore, predicted epitopes that are conserved across major SARS-CoV-2 lineages and variants are used to construct DNA vaccines expressing multi-epitope polypeptides. Most importantly, two DNA vaccine constructs induce epitope-specific CD8 + T-cell responses in a mouse model of HLA-A*02:01 restriction and protect immunized mice from challenge with Wuhan-hu-1 virus after hACE2 transduction. These data provide candidate T-cell epitopes useful for the development of T-cell vaccines against SARS-CoV-2 and demonstrate a strategy for quick T-cell vaccine candidate development applicable to other emerging pathogens.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Biologie informatique , Déterminants antigéniques des lymphocytes T , Antigène HLA-A2 , SARS-CoV-2 , Vaccins à ADN , Déterminants antigéniques des lymphocytes T/immunologie , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Antigène HLA-A2/immunologie , Antigène HLA-A2/génétique , Souris , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Vaccins contre la COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Femelle , Souris de lignée BALB C ,
16.
Curr Microbiol ; 81(5): 125, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38558085

RÉSUMÉ

More than half of the world's population is infected with Helicobacter pylori (H. pylori), which may lead to chronic gastritis, peptic ulcers, and stomach cancer. LeoA, a conserved antigen of H. pylori, aids in preventing this infection by triggering specific CD3+ T-cell responses. In this study, recombinant plasmids containing the LeoA gene of H. pylori are created and conjugated with chitosan nanoparticle (CSNP) to immunize BALB/c mice against the H. pylori infection. We used the online Vaxign tool to analyze the genomes of five distinct strains of H. pylori, and we chose the outer membrane as a prospective vaccine candidate. Afterward, the proteins' immunogenicity was evaluated. The DNA vaccine was constructed and then encapsulated in CSNPs. The effectiveness of the vaccine's immunoprotective effects was evaluated in BALB/c mice. Purified activated splenic CD3+ T cells are used to test the anticancer effects in vitro. Nanovaccines had apparent spherical forms, were small (mean size, 150-250 nm), and positively charged (41.3 ± 3.11 mV). A consistently delayed release pattern and an entrapment efficiency (73.35 ± 3.48%) could be established. Compared to the non-encapsulated DNA vaccine, vaccinated BALB/c mice produced higher amounts of LeoA-specific IgG in plasma and TNF-α in splenocyte lysate. Moreover, BALB/c mice inoculated with nanovaccine demonstrated considerable immunity (87.5%) against the H. pylori challenge and reduced stomach injury and bacterial burdens in the stomach. The immunological state in individuals with GC with chronic infection with H. pylori is mimicked by the H. pylori DNA nanovaccines by inducing a shift from Th1 to Th2 in the response. In vitro human GC cell development is inhibited by activated CD3+ T lymphocytes. According to our findings, the H. pylori vaccine-activated CD3+ has potential immunotherapeutic benefits.


Sujet(s)
Chitosane , Infections à Helicobacter , Helicobacter pylori , Nanoparticules , Vaccins à ADN , Humains , Animaux , Souris , Helicobacter pylori/génétique , Vaccins à ADN/génétique , ADN , Vaccination , Infections à Helicobacter/prévention et contrôle , Infections à Helicobacter/microbiologie , Vaccins antibactériens/génétique , Souris de lignée BALB C , Anticorps antibactériens
17.
Mol Cancer ; 23(1): 46, 2024 03 08.
Article de Anglais | MEDLINE | ID: mdl-38459592

RÉSUMÉ

Nucleic acid vaccines have shown promising potency and efficacy for cancer treatment with robust and specific T-cell responses. Improving the immunogenicity of delivered antigens helps to extend therapeutic efficacy and reduce dose-dependent toxicity. Here, we systematically evaluated chemokine-fused HPV16 E6/E7 antigen to improve the cellular and humoral immune responses induced by nucleotide vaccines in vivo. We found that fusion with different chemokines shifted the nature of the immune response against the antigens. Although a number of chemokines were able to amplify specific CD8 + T-cell or humoral response alone or simultaneously. CCL11 was identified as the most potent chemokine in improving immunogenicity, promoting specific CD8 + T-cell stemness and generating tumor rejection. Fusing CCL11 with E6/E7 antigen as a therapeutic DNA vaccine significantly improved treatment effectiveness and caused eradication of established large tumors in 92% tumor-bearing mice (n = 25). Fusion antigens with CCL11 expanded the TCR diversity of specific T cells and induced the infiltration of activated specific T cells, neutrophils, macrophages and dendritic cells (DCs) into the tumor, which created a comprehensive immune microenvironment lethal to tumor. Combination of the DNA vaccine with anti-CTLA4 treatment further enhanced the therapeutic effect. In addition, CCL11 could also be used for mRNA vaccine design. To summarize, CCL11 might be a potent T cell enhancer against cancer.


Sujet(s)
Vaccins anticancéreux , Tumeurs , Protéines des oncogènes viraux , Vaccins contre les papillomavirus , Vaccins à ADN , Animaux , Souris , Vaccins à base d'acide nucléique , Vaccins à ADN/génétique , Vaccins contre les papillomavirus/génétique , Tumeurs/génétique , Tumeurs/thérapie , Lymphocytes T CD8+ , Protéines E7 de papillomavirus/génétique , Protéines des oncogènes viraux/génétique , Souris de lignée C57BL , Microenvironnement tumoral
18.
Braz J Microbiol ; 55(1): 997-1010, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38311710

RÉSUMÉ

The swine industry across the globe is recently facing a devastating situation imparted by a highly contagious and deadly viral disease, African swine fever. The disease is caused by a DNA virus, the African swine fever virus (ASFV) of the genus Asfivirus. ASFV affects both wild boars and domestic pigs resulting in an acute form of hemorrhagic fever. Since the first report in 1921, the disease remains endemic in some of the African countries. However, the recent occurrence of ASF outbreaks in Asia led to a fresh and formidable challenge to the global swine production industry. Culling of the infected animals along with the implementation of strict sanitary measures remains the only options to control this devastating disease. Efforts to develop an effective and safe vaccine against ASF began as early as in the mid-1960s. Different approaches have been employed for the development of effective ASF vaccines including inactivated vaccines, subunit vaccines, DNA vaccines, virus-vectored vaccines, and live attenuated vaccines (LAVs). Inactivated vaccines are a non-feasible strategy against ASF due to their inability to generate a complete cellular immune response. However genetically engineered vaccines, such as subunit vaccines, DNA vaccines, and virus vector vaccines, represent tailored approaches with minimal adverse effects and enhanced safety profiles. As per the available data, gene deleted LAVs appear to be the most potential vaccine candidates. Currently, a gene deleted LAV (ASFV-G-∆I177L), developed in Vietnam, stands as the sole commercially available vaccine against ASF. The major barrier to the goal of developing an effective vaccine is the critical gaps in the knowledge of ASFV biology and the immune response induced by ASFV infection. The precise contribution of various hosts, vectors, and environmental factors in the virus transmission must also be investigated in depth to unravel the disease epidemiology. In this review, we mainly focus on the recent progress in vaccine development against ASF and the major gaps associated with it.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Vaccins à ADN , Vaccins antiviraux , Suidae , Animaux , Peste porcine africaine/prévention et contrôle , Peste porcine africaine/épidémiologie , Virus de la peste porcine africaine/génétique , Vaccins à ADN/génétique , Sus scrofa , Vaccins antiviraux/génétique , Vaccins atténués/génétique , Développement de vaccin , Vaccins inactivés , Vaccins sous-unitaires
19.
Nat Commun ; 15(1): 590, 2024 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-38238326

RÉSUMÉ

A safe and effective vaccine with long-term protection against SARS-CoV-2 variants of concern (VOCs) is a global health priority. Here, we develop lipid nanoparticles (LNPs) to provide safe and effective delivery of plasmid DNA (pDNA) and show protection against VOCs in female small animal models. Using a library of LNPs encapsulating unique barcoded DNA (b-DNA), we screen for b-DNA delivery after intramuscular administration. The top-performing LNPs are further tested for their capacity of pDNA uptake in antigen-presenting cells in vitro. The lead LNP is used to encapsulate pDNA encoding the HexaPro version of SARS-CoV-2 spike (LNP-HPS) and immunogenicity and protection is tested in vivo. LNP-HPS elicit a robust protective effect against SARS-CoV-2 Gamma (P.1), correlating with reduced lethality, decreased viral load in the lungs and reduced lung damage. LNP-HPS induce potent humoral and T cell responses against P.1, and generate high levels of neutralizing antibodies against P.1 and Omicron (B.1.1.529). Our findings indicate that the protective efficacy and immunogenicity elicited by LNP-HPS are comparable to those achieved by the approved COVID-19 vaccine from Biontech/Pfizer in animal models. Together, these findings suggest that LNP-HPS hold great promise as a vaccine candidate against VOCs.


Sujet(s)
COVID-19 , Forme B de l'ADN , Vaccins à ADN , Femelle , Animaux , Humains , SARS-CoV-2/génétique , Vaccins à ADN/génétique , , Vaccins contre la COVID-19 , COVID-19/prévention et contrôle , ADN , Anticorps neutralisants , Anticorps antiviraux
20.
Adv Biol (Weinh) ; 8(2): e2300402, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37840398

RÉSUMÉ

The most crucial disadvantage of DNA-based vaccines is their low immunogenicity; therefore, finding an effectual adjuvant is essential for their development. Herein, immunostimulatory effects of IFNγ cytokine and a CD40 ligand (CD40L) costimulatory molecule are evaluated as combined with an antigen, and also linked to an antigen in mice. For this purpose, after preparation of the HIV-1 Nef, IFNγ, and CD40L DNA constructs, and also their recombinant protein in an Escherichia coli expression system, nine groups of female BALB/c mice are immunized with different regimens of DNA constructs. About 3 weeks and also 3 months after the last injection, humoral and cellular immune responses are assessed in mice sera and splenocytes. Additionally, mice splenocytes are exposed to single-cycle replicable (SCR) HIV-1 virions for evaluating their potency in the secretion of cytokines in vitro. The data indicate that the linkage of IFNγ and CD40L to Nef antigen can significantly induce the Th-1 pathway and activate cytotoxic T lymphocytes compared to other regimens. Moreover, groups receiving the IFNγ-Nef and CD40L-Nef fusion DNA constructs show higher secretion of IFNγ and TNF-α from virion-infected lymphocytes than other groups. Therefore, the IFNγ-Nef and CD40L-Nef fusion DNA constructs are suggested to be a potential option for development of an efficient HIV-1 vaccine.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Vaccins à ADN , Femelle , Animaux , Souris , Cytokines , Ligand de CD40 , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Vaccins à ADN/pharmacologie , Vaccins à ADN/génétique , ADN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE