Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.550
Filtrar
1.
Scand J Immunol ; 99(5): e13356, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605549

RESUMO

In light of increasing resistance to PD1 antibody therapy among certain patient populations, there is a critical need for in-depth research. Our study assesses the synergistic effects of a MUC1 DNA vaccine and PD1 antibody for surmounting PD1 resistance, employing a murine CT26/MUC1 colon carcinoma model for this purpose. When given as a standalone treatment, PD1 antibodies showed no impact on tumour growth. Additionally, there was no change observed in the intra-tumoural T-cell ratios or in the functionality of T-cells. In contrast, the sole administration of a MUC1 DNA vaccine markedly boosted the cytotoxicity of CD8+ T cells by elevating IFN-γ and granzyme B production. Our compelling evidence highlights that combination therapy more effectively inhibited tumour growth and prolonged survival compared to either monotherapy, thus mitigating the limitations intrinsic to single-agent therapies. This enhanced efficacy was driven by a significant alteration in the tumour microenvironment, skewing it towards pro-immunogenic conditions. This assertion is backed by a raised CD8+/CD4+ T-cell ratio and a decrease in immunosuppressive MDSC and Treg cell populations. On the mechanistic front, the synergistic therapy amplified expression levels of CXCL13 in tumours, subsequently facilitating T-cell ingress into the tumour setting. In summary, our findings advocate for integrated therapy as a potent mechanism for surmounting PD1 antibody resistance, capitalizing on improved T-cell functionality and infiltration. This investigation affords critical perspectives on enhancing anti-tumour immunity through the application of innovative therapeutic strategies.


Assuntos
Neoplasias , Vacinas de DNA , Camundongos , Animais , Humanos , Linfócitos T CD8-Positivos , Receptor de Morte Celular Programada 1/metabolismo , Anticorpos/metabolismo , Neoplasias/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral , Mucina-1/genética
2.
Curr Microbiol ; 81(5): 125, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38558085

RESUMO

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.


Assuntos
Quitosana , Infecções por Helicobacter , Helicobacter pylori , Nanopartículas , Vacinas de DNA , Humanos , Animais , Camundongos , Helicobacter pylori/genética , Vacinas de DNA/genética , DNA , Vacinação , Infecções por Helicobacter/prevenção & controle , Infecções por Helicobacter/microbiologia , Vacinas Bacterianas/genética , Camundongos Endogâmicos BALB C , Anticorpos Antibacterianos
3.
Virulence ; 15(1): 2345019, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38656137

RESUMO

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.


Assuntos
Vacinas Bacterianas , Modelos Animais de Doenças , Interleucina-17 , Infecções por Klebsiella , Klebsiella pneumoniae , Vacinas de DNA , Animais , Klebsiella pneumoniae/imunologia , Klebsiella pneumoniae/genética , Infecções por Klebsiella/prevenção & controle , Infecções por Klebsiella/imunologia , Interleucina-17/imunologia , Interleucina-17/genética , Vacinas de DNA/imunologia , Vacinas de DNA/genética , Vacinas de DNA/administração & dosagem , Camundongos , Humanos , Feminino , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/administração & dosagem , Células HEK293 , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/genética , Imunização , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Fatores de Virulência/imunologia , Fatores de Virulência/genética , Imunidade Adaptativa , Camundongos Endogâmicos BALB C , Adjuvantes Imunológicos/administração & dosagem , Imunidade Celular
4.
Recent Pat Biotechnol ; 18(3): 180-189, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38528666

RESUMO

BACKGROUND: SARS-CoV-2, a highly dynamic beta-coronavirus, can afflict all age groups. Notably, over 16100 mortalities have been recorded among children as yet. In this regard, many vaccine projects are operational to assess immuno-potency among young cohorts. A bulk of reports have evidenced the efficacy of these immunization technologies in the elderly population, though the impact is yet to be determined among children. OBJECTIVES: This review is envisioned to outline the current efficacy of contributing vaccine technologies and examine the dose-dependent impact of immunization regimens in lowering the risks of SARS-CoV-2 infections among children and adolescents. Furthermore, the current review exclusively estimated the vaccine impact at current doses. METHODS: A total of 52 research papers extracted from PubMed, Pubmed Central, Science Direct, Research Gate, Google Scholar and Semantic Scholar were screened along with an emphasis on patents. Inclusion criteria involved all published reports directly or indirectly linked to the contributing vaccine candidates that are operational among the young cohort. Unrelated research papers were excluded from the study. Key search terminologies included information on vaccine identifiers, such as name, type and clinical trial ID, and successively restricted to children and adolscents age groups. RESULTS: Several vaccine designs, such as mRNA-based vaccinations, viral vector vaccines, DNA vaccines, inactivated vaccines, recombinant vaccines, and protein-based immunizations, are being examined at various stages of clinical trials to gauge the effects on children and adolescents. With reference to the published reports, the mRNA 1273 (1610 GMT; 6-10 yrs, 1401 GMT; 12-15 yrs), BNT162b2 (1407 GMT; 6 months- <2 yrs, 1535 GMT; 2-4 yrs, 4583 GMT; 5-11 yrs, 1239.5 GMT; 12-15 yrs) and Ad5 nCoV (1037.5 GMT; 6-17 yrs) offered relatively high neutralization titers with sharp seroconversion rates compared to MVC-COV1901 (648.5 GMT; 12-17 yrs) and ZyCoV-D (133.49 GMT; 12-17 yrs), which produced modest immune responses. CONCLUSION: Currently, the WHO is analyzing emerging evidence to issue an emergency use list of vaccines for vaccinating children and adolescents.


Assuntos
COVID-19 , Vacinas de DNA , Criança , Humanos , Adolescente , Idoso , Vacina BNT162 , Patentes como Assunto , COVID-19/prevenção & controle , Imunização
5.
Viruses ; 16(3)2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38543782

RESUMO

The sudden emergence of SARS-CoV-2 demonstrates the need for new vaccines that rapidly protect in the case of an emergency. In this study, we developed a recombinant MVA vaccine co-expressing SARS-CoV-2 prefusion-stabilized spike protein (ST) and SARS-CoV-2 nucleoprotein (N, MVA-SARS-2-ST/N) as an approach to further improve vaccine-induced immunogenicity and efficacy. Single MVA-SARS-2-ST/N vaccination in K18-hACE2 mice induced robust protection against lethal respiratory SARS-CoV-2 challenge infection 28 days later. The protective outcome of MVA-SARS-2-ST/N vaccination correlated with the activation of SARS-CoV-2-neutralizing antibodies (nABs) and substantial amounts of SARS-CoV-2-specific T cells especially in the lung of MVA-SARS-2-ST/N-vaccinated mice. Emergency vaccination with MVA-SARS-2-ST/N just 2 days before lethal SARS-CoV-2 challenge infection resulted in a delayed onset of clinical disease outcome in these mice and increased titers of nAB or SARS-CoV-2-specific T cells in the spleen and lung. These data highlight the potential of a multivalent COVID-19 vaccine co-expressing S- and N-protein, which further contributes to the development of rapidly protective vaccination strategies against emerging pathogens.


Assuntos
COVID-19 , Melfalan , SARS-CoV-2 , Vacinas de DNA , Vacinas Virais , gama-Globulinas , Animais , Humanos , Camundongos , SARS-CoV-2/genética , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Anticorpos Antivirais , Glicoproteína da Espícula de Coronavírus/genética , Vacinação , Anticorpos Neutralizantes
6.
Viruses ; 16(3)2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38543793

RESUMO

Single-dose, immunogenic DNA (iDNA) vaccines coding for whole live-attenuated viruses are reviewed. This platform, sometimes called immunization DNA, has been used for vaccine development for flavi- and alphaviruses. An iDNA vaccine uses plasmid DNA to launch live-attenuated virus vaccines in vitro or in vivo. When iDNA is injected into mammalian cells in vitro or in vivo, the RNA genome of an attenuated virus is transcribed, which starts replication of a defined, live-attenuated vaccine virus in cell culture or the cells of a vaccine recipient. In the latter case, an immune response to the live virus vaccine is elicited, which protects against the pathogenic virus. Unlike other nucleic acid vaccines, such as mRNA and standard DNA vaccines, iDNA vaccines elicit protection with a single dose, thus providing major improvement to epidemic preparedness. Still, iDNA vaccines retain the advantages of other nucleic acid vaccines. In summary, the iDNA platform combines the advantages of reverse genetics and DNA immunization with the high immunogenicity of live-attenuated vaccines, resulting in enhanced safety and immunogenicity. This vaccine platform has expanded the field of genetic DNA and RNA vaccines with a novel type of immunogenic DNA vaccines that encode entire live-attenuated viruses.


Assuntos
Flavivirus , Vacinas de DNA , Vacinas Virais , Animais , Anticorpos Antivirais , Flavivirus/genética , Vacinas Atenuadas , DNA , Mamíferos
7.
Cell Biochem Funct ; 42(3): e3992, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38551221

RESUMO

Nucleic acid vaccines (NAVs) have the potential to be economical, safe, and efficacious. Furthermore, just the chosen antigen in the pathogen is the target of the immune responses brought on by NAVs. Triple-negative breast cancer (TNBC) treatment shows great promise for nucleic acid-based vaccines, such as DNA (as plasmids) and RNA (as messenger RNA [mRNA]). Moreover, cancer vaccines offer a compelling approach that can elicit targeted and long-lasting immune responses against tumor antigens. Bacterial plasmids that encode antigens and immunostimulatory molecules serve as the foundation for DNA vaccines. In the 1990s, plasmid DNA encoding the influenza A nucleoprotein triggered a protective and targeted cytotoxic T lymphocyte (CTL) response, marking the first instance of DNA vaccine-mediated immunity. Similarly, in vitro transcribed mRNA was first successfully used in animals in 1990. At that point, mice were given an injection of the gene encoding the mRNA sequence, and the researchers saw the production of a protein. We begin this review by summarizing our existing knowledge of NAVs. Next, we addressed NAV delivery, emphasizing the need to increase efficacy in TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Vacinas de DNA , Humanos , Camundongos , Animais , Vacinas Baseadas em Ácido Nucleico , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/patologia , Imunoterapia , DNA , RNA Mensageiro/genética
8.
Fish Shellfish Immunol ; 148: 109502, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38471627

RESUMO

ß-defensin of flounder plays an important role in immunomodulation by recruiting immune cells and has a potential vaccine adjuvant effect in addition to its bactericidal activity. In this study, adjuvant effects of ß-defensin on DNA vaccine OmpC against edwardsiellosis in flounder (Paralichthys olivaceus) were investigated. The bicistronic eukaryotic expression plasmid pBudCE4.1 plasmid vector with two independent coding regions was selected to construct DNA vaccine of p-OmpC which express only the gene for the outer membrane protein of Edwardsiella tarda and the vaccine of p-OmpC-ßdefensin which express both the outer membrane protein of the bacterium and ß-defensin of flounder. In vitro and in vivo studies have shown that the constructed plasmids can be expressed in flounder embryonic cell lines and injection sites of muscles. After vaccination by intramuscular injection, both p-OmpC and p-OmpC-ßdefensin groups showed significant upregulation of immune-response. Compared to the pBbudCE4.1 and the p-OmpC vaccinated groups, the p-OmpC-ßdefensin vaccinated group showed significantly more cell aggregation at the injection site and intense immune response. The proportion of sIgM+ cells, as well as the CD4-1+ and CD4-2+ cells in both spleen and kidney was significantly higher in the p-OmpC-ßdefensin vaccinated group at peak time point than in the control groups. The relative survival rate of the p-OmpC-ßdefensin vaccine was 74.17%, which was significantly higher than that of the p-OmpC vaccinated group 48.33%. The results in this study determined that ß-defensin enhances the responses in cellular and humoral immunity and evokes a high degree of protection against E. tarda, which is a promising candidate for vaccine adjuvant.


Assuntos
Infecções por Enterobacteriaceae , Doenças dos Peixes , Linguado , Vacinas de DNA , beta-Defensinas , Animais , beta-Defensinas/genética , Adjuvantes de Vacinas , Adjuvantes Imunológicos/farmacologia , Edwardsiella tarda , Vacinas Bacterianas , Infecções por Enterobacteriaceae/prevenção & controle , Infecções por Enterobacteriaceae/veterinária
9.
Fish Shellfish Immunol ; 148: 109494, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38499217

RESUMO

Vibrio harveyi poses a significant threat to fish and invertebrates in mariculture, resulting in substantial financial repercussions for the aquaculture sector. Valine-glycine repeat protein G (VgrG) is essential for the type VI secretion system's (T6SS) assembly and secretion. VgrG from V. harveyi QT520 was cloned and analyzed in this study. The localization of VgrG was determined by Western blot, which revealed that it was located in the cytoplasm, secreted extracellularly, and attached to the membrane. The effectiveness of two vaccinations against V. harveyi infection-a subunit vaccine (rVgrG) and a DNA vaccine (pCNVgrG) prepared with VgrG was evaluated. The findings indicated that both vaccines provided a degree of protection against V. harveyi challenge. At 4 weeks post-vaccination (p.v.), the rVgrG and pCNVgrG exhibited relative percent survival rates (RPS) of 71.43% and 76.19%, respectively. At 8 weeks p.v., the RPS for rVgrG and pCNVgrG were 68.21% and 72.71%, respectively. While both rVgrG and pCNVgrG elicited serum antibody production, the subunit vaccinated fish demonstrated significantly higher levels of serum anti-VgrG specific antibodies than the DNA vaccine group. The result of qRT-PCR demonstrated that the expression of major histocompatibility complex (MHC) class Iα, tumor necrosis factor-alpha (TNF-α), interferon γ (IFNγ), and cluster of differentiation 4 (CD4) were up-regulated by both rVgrG and pCNVgrG. Fish vaccinated with rVgrG and pCNVgrG exhibited increased activity of acid phosphatase, alkaline phosphatase, superoxide dismutase, and lysozyme. These findings suggest that VgrG from V. harveyi holds potential for application in vaccination.


Assuntos
Doenças dos Peixes , Vacinas de DNA , Vibrioses , Vibrio , Animais , Vibrioses/prevenção & controle , Vibrioses/veterinária , Valina , Vacinas Bacterianas , Peixes , Doenças dos Peixes/prevenção & controle
10.
Front Immunol ; 15: 1342816, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38515753

RESUMO

Salmonid alphavirus (SAV) causes pancreas disease (PD), which negatively impacts farmed Atlantic salmon. In this study, fish were vaccinated with a DNA-PD vaccine (DNA-PD) and an oil-adjuvanted, inactivated whole virus PD vaccine (Oil-PD). Controls were two non-PD vaccinated groups. Fish were kept in one tank and challenged by cohabitation with SAV genotype 2 in seawater. Protection against infection and mortality was assessed for 84 days (Efficacy study). Nineteen days post challenge (dpc), subgroups of fish from all treatment groups were transferred to separate tanks and cohabited with naïve fish (Transmission study 1) or fish vaccinated with a homologous vaccine (Transmission study 2), to evaluate virus transmission for 26 days (47 dpc). Viremia, heart RT-qPCR and histopathological scoring of key organs affected by PD were used to measure infection levels. RT-droplet digital PCR quantified shedding of SAV into water for transmission studies. The Efficacy study showed that PD associated growth-loss was significantly lower and clearance of SAV2 RNA significantly higher in the PD-DNA group compared to the other groups. The PD-DNA group had milder lesions in the heart and muscle. Cumulative mortality post challenge was low and not different between groups, but the DNA-PD group had delayed time-to-death. In Transmission study 1, the lowest water levels of SAV RNA were measured in the tanks containing the DNA-PD group at 21 and 34 dpc. Despite this, and irrespective of the treatment group, SAV2 was effectively transmitted to the naïve fish during 26-day cohabitation. At 47 dpc, the SAV RNA concentrations in the water were lower in all tanks compared to 34 dpc. In Transmission study 2, none of the DNA-PD immunized cohabitants residing with DNA-PD-vaccinated, pre-challenged fish got infected. In contrast, Oil-PD immunized cohabitants residing with Oil-PD-vaccinated, pre-challenged fish, showed infection levels similar to the naïve cohabitants in Transmission study 1. The results demonstrate that the DNA-PD vaccine may curb the spread of SAV infection as the DNA-PD vaccinated, SAV2 exposed fish, did not spread the infection to cohabiting DNA-PD vaccinated fish. This signifies that herd immunity may be achieved by the DNA-PD vaccine, a valuable tool to control the PD epizootic in farmed Atlantic salmon.


Assuntos
Alphavirus , Doenças dos Peixes , Pancreatopatias , Salmo salar , Vacinas de DNA , Vacinas Virais , Animais , Pancreatopatias/veterinária , Pancreatopatias/patologia , RNA/genética , Água , Pâncreas/patologia , DNA , Genótipo
11.
Int J Biol Macromol ; 264(Pt 2): 130660, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38460634

RESUMO

The emergence of SARS-CoV-2 presents a significant global public health dilemma. Vaccination has long been recognized as the most effective means of preventing the spread of infectious diseases. DNA vaccines have attracted attention due to their safety profile, cost-effectiveness, and ease of production. This study aims to assess the efficacy of plasmid-encoding GM-CSF (pGM-CSF) as an adjuvant to augment the specific humoral and cellular immune response elicited by DNA vaccines based on the receptor-binding domain (RBD) antigen. Compared to the use of plasmid-encoded RBD (pRBD) alone, mice that were immunized with a combination of pRBD and pGM-CSF exhibited significantly elevated levels of RBD-specific antibody titers in serum, BALF, and nasal wash. Furthermore, these mice generated more potent neutralization antibodies against both the wild-type and Omicron pseudovirus, as well as the ancestral virus. In addition, pGM-CSF enhanced pRBD-induced CD4+ and CD8+ T cell responses and promoted central memory T cells storage in the spleen. At the same time, tissue-resident memory T (Trm) cells in the lung also increased significantly, and higher levels of specific responses were maintained 60 days post the final immunization. pGM-CSF may play an adjuvant role by promoting antigen expression, immune cells recruitment and GC B cell responses. In conclusion, pGM-CSF may be an effective adjuvant candidate for the DNA vaccines against SARS-CoV-2.


Assuntos
COVID-19 , Vacinas de DNA , Humanos , Animais , Camundongos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , SARS-CoV-2 , Vacinas contra COVID-19 , COVID-19/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Adjuvantes Farmacêuticos , Vacinação , DNA , Anticorpos Antivirais , Anticorpos Neutralizantes
12.
Front Immunol ; 15: 1362780, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38487527

RESUMO

Dengue, caused by the dengue virus (DENV), affects millions of people worldwide every year. This virus has two distinct life cycles, one in the human and another in the mosquito, and both cycles are crucial to be controlled. To control the vector of DENV, the mosquito Aedes aegypti, scientists employed many techniques, which were later proved ineffective and harmful in many ways. Consequently, the attention shifted to the development of a vaccine; researchers have targeted the E protein, a surface protein of the virus and the NS1 protein, an extracellular protein. There are several types of vaccines developed so far, such as live attenuated vaccines, recombinant subunit vaccines, inactivated virus vaccines, viral vectored vaccines, DNA vaccines, and mRNA vaccines. Along with these, scientists are exploring new strategies of developing improved version of the vaccine by employing recombinant DNA plasmid against NS1 and also aiming to prevent the infection by blocking the DENV life cycle inside the mosquitoes. Here, we discussed the aspects of research in the field of vaccines until now and identified some prospects for future vaccine developments.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Vacinas de DNA , Vacinas Virais , Animais , Humanos , Vírus da Dengue/genética , Mosquitos Vetores , Vacinas Atenuadas , Vacinas de Produtos Inativados
13.
Mol Cancer ; 23(1): 46, 2024 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-38459592

RESUMO

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.


Assuntos
Vacinas Anticâncer , Neoplasias , Proteínas Oncogênicas Virais , Vacinas contra Papillomavirus , Vacinas de DNA , Animais , Camundongos , Vacinas Baseadas em Ácido Nucleico , Vacinas de DNA/genética , Vacinas contra Papillomavirus/genética , Neoplasias/genética , Neoplasias/terapia , Linfócitos T CD8-Positivos , Proteínas E7 de Papillomavirus/genética , Proteínas Oncogênicas Virais/genética , Camundongos Endogâmicos C57BL , Microambiente Tumoral
14.
Vaccine ; 42(9): 2299-2309, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38429153

RESUMO

Toxoplasma gondii is a pervasive protozoan parasite that is responsible for significant zoonoses. A wide array of vaccines using different effector molecules of T. gondii have been studied worldwide to control toxoplasmosis. None of the existing vaccines are sufficiently effective to confer protective immunity. Among the different Toxoplasma-derived effector molecules, T. gondii dense granule protein 15 from the type II strain (GRA15 (II)) was recently characterized as an immunomodulatory molecule that induced host immunity via NF-κB. Therefore, we assessed the immunostimulatory and protective efficacy of recombinant GRA15 (II) (rGRA15) against T. gondii infection in a C57BL/6 mouse model. We observed that rGRA15 treatment increased the production of IL-12p40 from mouse peritoneal macrophages in vitro. Immunization of mice with rGRA15 induced the production of anti-TgGRA15-specific IgG, IgG1 and IgG2c antibodies. The rGRA15-sensitized spleen cells from mice inoculated with the same antigen strongly promoted spleen cell proliferation and IFN-γ production. Immunization with rGRA15 significantly enhanced the survival rate of mice and dramatically decreased parasite burden in mice challenged with the Pru (type II) strain. These results suggested that rGRA15 triggered humoral and cellular immune responses to control infection. However, all of the immunized mice died when challenged with the GRA15-deficient Pru strain or the RH (type I) strain. These results suggest that GRA15 (II)-dependent immunity plays a crucial role in protection against challenge infection with the type II strain of T. gondii. This study is the first report to show GRA15 (II) as a recombinant vaccine antigen against Toxoplasma infection.


Assuntos
Vacinas Protozoárias , Toxoplasma , Toxoplasmose Animal , Toxoplasmose , Vacinas de DNA , Vacinas , Animais , Camundongos , Proteínas de Protozoários , Camundongos Endogâmicos C57BL , Toxoplasmose/prevenção & controle , Proteínas Recombinantes/metabolismo , Anticorpos Antiprotozoários , Toxoplasmose Animal/prevenção & controle , Camundongos Endogâmicos BALB C
15.
AAPS PharmSciTech ; 25(3): 60, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38472523

RESUMO

The protective efficacies of current licensed vaccines against COVID-19 have significantly reduced as a result of SARS-CoV-2 variants of concern (VOCs) which carried multiple mutations in the Spike (S) protein. Considering that these vaccines were developed based on the S protein of the original SARS-CoV-2 Wuhan strain, we designed a recombinant plasmid DNA vaccine based on highly conserved and immunogenic B and T cell epitopes against SARS-CoV-2 Wuhan strain and the Omicron VOC. Literature mining and bioinformatics were used to identify 6 immunogenic peptides from conserved regions of the SARS-CoV-2 S and membrane (M) proteins. Nucleotide sequences encoding these peptides representing highly conserved B and T cell epitopes were cloned into a pVAX1 vector to form the pVAX1/S2-6EHGFP recombinant DNA plasmid vaccine. The DNA vaccine was intranasally or intramuscularly administered to BALB/c mice and evaluations of humoral and cellular immune responses were performed. The intramuscular administration of pVAX1/S2-6EHGFP was associated with a significantly higher percentage of CD8+ T cells expressing IFN-γ when compared with the empty vector and PBS controls. Intramuscular or intranasal administrations of pVAX1/S2-6EHGFP resulted in robust IgG antibody responses. Sera from mice intramuscularly immunized with pVAX1/S2-6EHGFP were found to elicit neutralizing antibodies capable of SARS-CoV-2 Omicron variant with the ACE2 cell surface receptor. This study demonstrated that the DNA vaccine construct encoding highly conserved immunogenic B and T cell epitopes was capable of eliciting potent humoral and cellular immune responses in mice.


Assuntos
COVID-19 , Vacinas de DNA , Animais , Humanos , Camundongos , SARS-CoV-2 , Epitopos de Linfócito T , Camundongos Endogâmicos BALB C , Linfócitos T CD8-Positivos , Vacinas contra COVID-19 , Peptídeos , Anticorpos Antivirais
16.
Int J Pharm ; 654: 123959, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38430949

RESUMO

DNA vaccines can be a potential solution to protect global health, triggering both humoral and cellular immune responses. DNA vaccines are valuable in preventing intracellular pathogen infections, and therefore can be explored against coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2). This work explored different systems based on polyethylenimine (PEI), functionalized for the first time with both cholesterol (CHOL) and mannose (MAN) to deliver parental plasmid (PP) and minicircle DNA (mcDNA) vectors encoding the receptor-binding domain (RBD) of SARS-CoV-2 to antigen-presenting cells (APCs). For comparative purposes, three different systems were evaluated: PEI, PEI-CHOL and PEI-CHOL-MAN. The systems were prepared at various nitrogen-to-phosphate group (N/P) ratios and characterized in terms of encapsulation efficiency, surface charge, size, polydispersity index (PDI), morphology, and stability over time. Moreover, in vitro transfection studies of dendritic cells (JAWS II) and human fibroblast cells were performed. Viability studies assured the biocompatibility of all nanocarriers. Confocal microscopy studies confirmed intracellular localization of systems, resulting in enhanced cellular uptake using PEI-CHOL and PEI-CHOL-MAN systems when compared with the PEI system. Regarding the RBD expression, PEI-CHOL-MAN was the system that led to the highest levels of transcripts and protein expression in JAWS II cells. Furthermore, the nanosystems significantly stimulated pro-inflammatory cytokines production and dendritic cell maturation in vitro. Overall, mannosylated systems can be considered a valuable tool in the delivery of plasmid DNA or mcDNA vaccines to APCs.


Assuntos
COVID-19 , Nanopartículas , Vacinas de DNA , Humanos , Polietilenoimina/química , Vacinas contra COVID-19 , COVID-19/prevenção & controle , SARS-CoV-2/genética , Transfecção , DNA , Células Apresentadoras de Antígenos , Colesterol , Nanopartículas/química
17.
Front Immunol ; 15: 1332939, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38361919

RESUMO

Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.


Assuntos
Vacinas de DNA , Animais , Humanos , Sistemas de Liberação de Medicamentos , Modelos Animais
18.
Discov Med ; 36(181): 355-365, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38409840

RESUMO

BACKGROUND: Arg-gingipain A (rgpA) and Arg-gingipain B (rgpB) are crucial virulence factors associated with Porphyromonas gingivalis (P. gingivalis) and have been recognized as promising targets for antibacterial vaccines. Although vaccines containing rgpA have shown efficacy, the incorporation of rgpB, which lacks the haemagglutinin adhesin (HA) domain, diminishes the vaccine's effectiveness. This study aims to assess the immunogenicity of the functional HA domain of rgpA in mouse periodontitis models. METHODS: A total of 24 mice were randomly divided into four groups, each receiving different immune injections: group A received phosphate-buffered saline (PBS) as an empty control; group B received pVAX1 as a negative control (NC); group C received pVAX1-HA; and group D received pVAX1-rgpA. The mice were subjected to intramuscular injections every two weeks for a total of three administrations. Prior to each immunization, blood samples were collected for antibody detection under isoflurane anesthesia. Following the final immunization, periodontitis was induced two weeks later by using sutures soaked in a P. gingivalis solution. The mice were euthanized after an additional two-week period. To assess the safety of the procedure, major organs were examined through hematoxylin-eosin (HE) staining. Subsequently, the levels of IgG, IgG1, and IgG2a in the serum were quantified via enzyme-linked immunosorbent assay (ELISA). Additionally, the expression of inflammatory factors in the gingiva, including interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and tumor necrosis factor alpha (TNF-α), was determined using quantitative real-time reverse transcript PCR (qRT-PCR). The extent of bone loss in periodontal tissues was evaluated using micro-computed tomography (micro-CT) and HE staining. RESULTS: HE staining of the organs confirmed the absence of vaccine-induced toxicity in vivo. After the second immunization, both the rgpA and HA groups displayed significantly higher specific IgG titers in comparison to the NC and PBS groups (p < 0.05). Furthermore, the rgpA and HA groups exhibited a noteworthy predominance of IgG1 antibodies after three immunization doses, while there was a noticeable reduction in IgG2a levels observed following ligation with P. gingivalis sutures, as opposed to the NC and PBS groups (p < 0.05). Additionally, both the HA and rgpA groups showed a significant decrease in the expression of inflammatory factors such as IL-6, IL-1ß, and TNF-α, as well as a reduction in bone loss around periodontitis-affected teeth, when compared to the NC and PBS groups (p < 0.05). CONCLUSIONS: The results of this study demonstrate that the rgpA-engineered/functionalized HA gene vaccine is capable of eliciting a potent prophylactic immune response against P. gingivalis-induced periodontitis, effectively serving as an immunogenic and protective agent in vivo.


Assuntos
Periodontite , Vacinas de DNA , Camundongos , Animais , Cisteína Endopeptidases Gingipaínas , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Vacinas de DNA/uso terapêutico , Porphyromonas gingivalis/genética , Interleucina-6 , Fator de Necrose Tumoral alfa , Microtomografia por Raio-X , Adesinas Bacterianas , Vacinação , Periodontite/prevenção & controle , Imunoglobulina G
19.
Biomed Pharmacother ; 172: 116264, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38359491

RESUMO

Pseudomonas aeruginosa (PA) is one of the leading pathogens responsible for hospital-acquired infections. With the increasing antibiotic resistance of PA, clinical treatment has become increasingly challenging. DNA vaccines represent a promising approach for combating PA infection. However, the immune response induced by a single antigen is limited, and combination vaccines hold greater therapeutic potential. The highly conserved OprF and PcrV genes are attractive candidate antigens for vaccine development, but the poor delivery of such vaccines has limited their clinical application. In this study, we constructed an OprF/PcrV bivalent DNA vaccine, and a polyaspartamide/polyethylene glycol di-aldehyde (PSIH/PEG DA) hydrogel was formulated to improve DNA delivery. The OprF/PcrV DNA vaccine formulated with the PSIH/PEG DA hydrogel was carefully characterized in vitro and in vivo and showed suitable compatibility. The PSIH/PEG DA hydrogel formulation induced a mixed Th1/Th2/Th17 immune response in mice, leading to a significant increase in antibody titers, lymphocyte proliferation rates, and cytokine levels compared to those in mice treated with single or combined vaccines. The PSIH/PEG DA hydrogel delivery system significantly enhanced the immune protection of the DNA vaccine in a murine pneumonia model, as revealed by the reduced bacterial burden and inflammation in the mouse lungs and increased survival rate. In conclusion, the PSIH/PEG DA hydrogel delivery system can further enhance the immune efficacy of the combination OprF/PcrV DNA vaccine. This research provides a novel optimized strategy for the prevention and treatment of PA infection using DNA vaccines.


Assuntos
Infecções por Pseudomonas , Vacinas de DNA , Animais , Camundongos , Hidrogéis , Pseudomonas aeruginosa , Aldeídos , Materiais Biocompatíveis , Infecções por Pseudomonas/prevenção & controle
20.
Vaccine ; 42(7): 1487-1497, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38350766

RESUMO

H5 highly pathogenic avian influenza (HPAI) viruses of the Asian lineage (A/goose/Guangdong/1/96) belonging to clade 2.3.4.4 have spread worldwide through wild bird migration in two major waves: in 2014/2015 (clade 2.3.4.4c), and since 2016 up to now (clade 2.3.4.4b). Due to the increasing risk of these H5 HPAI viruses to establish and persist in the wild bird population, implementing vaccination in certain sensitive areas could be a complementary measure to the disease control strategies already applied. In this study, the efficacy of a novel DNA vaccine, encoding a H5 gene (A/gyrfalcon/Washington/41088-6/2014 strain) of clade 2.3.4.4c was evaluated in specific pathogen-free (SPF) white leghorn chickens against a homologous and heterologous H5 HPAI viruses. A single vaccination at 2 weeks of age (1 dose), and a vaccination at 2 weeks of age, boosted at 4 weeks (2 doses), with or without adjuvant were characterized. The groups that received 1 dose with or without adjuvant as well as 2 doses with adjuvant demonstrated full clinical protection and a significant or complete reduction of viral shedding against homologous challenge at 6 and 25 weeks of age. The heterologous clade 2.3.4.4b challenge of 6-week-old chickens vaccinated with 2 doses with or without adjuvant showed similar results, indicating good cross-protection induced by the DNA vaccine. Long lasting humoral immunity was observed in vaccinated chickens up to 18 or 25 weeks of age, depending on the vaccination schedule. The analysis of viral transmission after homologous challenge showed that sentinels vaccinated with 2 doses with adjuvant were fully protected against mortality with no excretion detected. This study of H5 DNA vaccine efficacy confirmed the important role that this type of so-called third-generation vaccine could play in the fight against H5 HPAI viruses.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Influenza Aviária , Vacinas de DNA , Animais , Galinhas , Vacinação/veterinária , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...