Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.447
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
BMC Immunol ; 25(1): 27, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38706005

RESUMO

BACKGROUND: Due to antibiotic resistance, the Klebsiella genus is linked to morbidity and death, necessitating the development of a universally protective vaccine against Klebsiella pathogens. METHODS: Core sequence analysis prioritized non-redundant host molecules and expected lipid bilayer peptides from fully sequenced Klebsiella genomes. These proteins were refined to identify epitopes, examining their immunogenicity, toxicity, solubility, and interaction with MHC alleles. Epitopes were linked to CPG ODN C274 via EAAAK, HEYGAEALERAG, and GGGS linkers to enhance immunological responses. The vaccine's tertiary structure was modelled and docked with MHC-I and MHC-II. RESULTS: Fifty-five proteins were recognized in the Vaxign collection as having remarkable features. Twenty-three proteins with potential pathogenicity were then identified. Eight options for vaccines emerged after the immunogenicity of proteins was examined. The best antigens were three proteins: MrkD, Iron-regulated lipid membrane polypeptides, and RmpA. These compounds were selected for their sensitivity. The structural protein sequences of K. pneumoniae were utilized to identify seven CTL epitopes, seven HTL epitopes, and seven LBL epitopes, respectively. The produced immunization displayed a stable contact with the receptors, based on molecular dynamic simulations lasting 250 nanoseconds. Intermolecular binding free energies also indicated the dominance of the van der Waals and electrostatic energies. CONCLUSION: In summary, the results of this study might help scientists develop a novel vaccine to prevent K. pneumoniae infections.


Assuntos
Vacinas Bacterianas , Infecções por Klebsiella , Klebsiella pneumoniae , Klebsiella pneumoniae/imunologia , Vacinas Bacterianas/imunologia , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/prevenção & controle , Animais , Epitopos de Linfócito T/imunologia , Camundongos , Humanos , Simulação de Dinâmica Molecular , Antígenos de Bactérias/imunologia , Oligodesoxirribonucleotídeos/imunologia , Epitopos/imunologia , Simulação de Acoplamento Molecular
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 441-446, 2024 Mar 20.
Artigo em Chinês | MEDLINE | ID: mdl-38645870

RESUMO

Objective: To study the immunoadjuvant effects of chitosan oligosaccharide (COS), including the immune activation and the triggering of lysosomal escape, and to explore whether COS can be used as an adjuvant for attenuated live bacteria vector vaccines. Methods: 1) Mouse macrophages RAW264.7 cells were cultured with COS at 0 mg/mL (the control group) and 0.1-4 mg/mL for 24 h and the effect on cell viability was measured by CCK8 assay. Mouse macrophages RAW264.7 were treated with COS at 0 (the control group), 1, 2, and 4 mg/mL for 24 h. Then, the mRNA expression levels of the cytokines, including IFN-γ, IL-10, TGF-ß, and TLR4, were determined by RT-qPCR assay. 2) RAW264.7 cells were treated with 1 mL of PBS containing different components, including calcein at 50 µg/mL, COS at 2 mg/mL, and bafilomycin A1, an inhibitor, at 1 µmol/mL, for culturing. The cells were divided into the Calcein group, Calcein+COS group, and Calcein+COS+Bafilomycin A1 group accordingly. Laser scanning confocal microscopy was used to observe the phagocytosis and the intracellular fluorescence distribution of calcein, a fluorescent dye, in RAW264.7 cells in the presence or absence of COS intervention to determine whether COS was able to trigger lysosomal escape. 3) LM∆E6E7 and LI∆E6E7, the attenuated Listeria vector candidate therapeutic vaccines for cervical cancer, were encapsulated with COS at the mass concentrations of 0.5 mg/mL, 1 mg/mL, 2 mg/mL , 4 mg/mL, and 8 mg/mL. Then, the changes in zeta potential were measured to select the concentration of COS that successfully encapsulated the bacteria. Phagocytosis of the vaccine strains by RAW264.7 cells was measured before and after LM∆E6E7 and LI∆E6E7 were coated with COS at 2 mg/mL. Results: 1) CCK8 assays showed that, compared with the findings for the control group, the intervention of RAW264.7 cells with COS at different concentrations for 24 h was not toxic to the cells and promoted cell proliferation, with the difference being statistically significant (P<0.05). According to the RT-qPCR results, compared with those of the control group, the COS intervention up-regulated the mRNA levels of TLR4 and IFN-γ in RAW264.7 cells, while it inhibited the mRNA expression levels of TGF-ß and IL-10, with the most prominent effect being observed in the 4 mg/mL COS group (P<0.05). 2) Laser scanning confocal microscopy revealed that the amount of fluorescent dye released from lysosomes into the cells was greater in the Calcein+COS group than that in the Calcein group. In other words, a greater amount of fluorescent dye was released from lysosomes into the cells under COS intervention. Furthermore, this process could be blocked by bafilomycin A1. 3) The zeta potential results showed that COS could successfully encapsulate the surface of bacteria when its mass concentration reached 2 mg/mL. Before and after the vaccine strain was encapsulated by COS, the phagocytosis of LM∆E6E7 by RAW264.7 cells was 5.70% and 22.00%, respectively, showing statistically significant differences (P<0.05); the phagocytosis of LI∆E6E7 by RAW264.7 cells was 1.55% and 6.12%, respectively, showing statistically significant differences (P<0.05). Conclusion: COS has the effect of activating the immune response of macrophages and triggering lysosomal escape. The candidates strains of coated live attenuated bacterial vector vaccines can promote the phagocytosis of bacteria by macrophages. Further research is warranted to develop COS into an adjuvant for bacterial vector vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas Bacterianas , Quitosana , Oligossacarídeos , Animais , Camundongos , Células RAW 264.7 , Oligossacarídeos/farmacologia , Adjuvantes Imunológicos/farmacologia , Vacinas Bacterianas/imunologia , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Vacinas Atenuadas/imunologia , Citocinas/metabolismo , Sobrevivência Celular/efeitos dos fármacos
3.
Microb Pathog ; 172: 105782, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36150556

RESUMO

Listeria monocytogenes is the causative agent of listeriosis, which is dangerous for pregnant women, the elderly or individuals with a weakened immune system. Individuals with leukaemia, cancer, HIV/AIDS, kidney transplant and steroid therapy suffer from immunological damage are menaced. World Health Organization (WHO) reports that human listeriosis has a high mortality rate of 20-30% every year. To date, no vaccine is available to treat listeriosis. Thereby, it is high time to design novel vaccines against L. monocytogenes. Here, we present computational approaches to design an antigenic, stable and safe vaccine against the L. monocytogenes that could help to control the infections associated with the pathogen. Three vital pathogenic proteins of L. monocytogenes, such as Listeriolysin O (LLO), Phosphatidylinositol-specific phospholipase C (PI-PLC), and Actin polymerization protein (ActA), were selected using a subtractive proteomics approach to design the multi-epitope vaccine (MEV). A total of 5 Cytotoxic T-lymphocyte (CTL) and 9 Helper T-lymphocyte (HTL) epitopes were predicted from these selected proteins. To design the multi-epitope vaccine (MEV) from the selected proteins, CTL epitopes were joined with the AAY linker, and HTL epitopes were joined with the GPGPG linker. Additionally, a human ß-defensin-3 (hBD-3) adjuvant was added to the N-terminal side of the final MEV construct to increase the immune response to the vaccine. The final MEV was predicted to be antigenic, non-allergen and non-toxic in nature. Physicochemical property analysis suggested that the MEV construct is stable and could be easily purified through the E. coli expression system. This in-silico study showed that MEV has a robust binding interaction with Toll-like receptor 2 (TLR2), a key player in the innate immune system. Current subtractive proteomics and immunoinformatics study provides a background for designing a suitable, safe and effective vaccine against pathogenic L. monocytogenes.


Assuntos
Vacinas Bacterianas , Listeriose , Humanos , Actinas , beta-Defensinas , Biologia Computacional , Epitopos de Linfócito B , Epitopos de Linfócito T , Escherichia coli , Listeriose/prevenção & controle , Simulação de Acoplamento Molecular , Fosfoinositídeo Fosfolipase C , Proteômica , Esteroides , Receptor 2 Toll-Like , Vacinas de Subunidades Antigênicas , Vacinas Bacterianas/imunologia , Desenvolvimento de Vacinas
4.
Helicobacter ; 27(2): e12875, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35092634

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM: This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS: We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of ßdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION: Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.


Assuntos
Vacinas Bacterianas , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Infecções por Helicobacter , Helicobacter pylori , Animais , Vacinas Bacterianas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/prevenção & controle , Camundongos , Células Th17 , Vacinação
5.
J Clin Pharm Ther ; 47(2): 139-145, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34328230

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Live-attenuated bacterial veterinary vaccines can constitute an infection risk for individuals with any defect in their phagocytic function, including chronic granulomatous disease, leukocyte adhesion deficiency, myeloperoxidase deficiency, as well as Chediak-Higashi syndrome, from accidental acquisition of licenced attenuated live bacterial vaccine, at vaccination or from their vaccinated pet. Ownership of small companion animals, including cats and dogs, is popular within the cystic fibrosis (CF) community. These animals require vaccines as part of their routine care, which may involve live viral and bacterial vaccines, with potential for infection in the CF owner. This report examines the scope of current canine and feline vaccines, with particular emphasis on veterinary vaccination strategies against the Gram-negative pathogen, Bordetella bronchiseptica and describes new vaccine innovations offering protection to both pet and CF owner. COMMENT: The Gram-negative bacterium, Bordetella bronchoseptica, may cause respiratory disease in small companion animals, as well as in certain human vulnerable groups, including those with CF. Live licenced veterinary bacterial vaccines for Bordetella bronchiseptica (Kennel Cough) are available for cats and dogs, which are an infection concern for humans with CF who may come into contact with vaccinated animals. Live licenced veterinary bacterial vaccines for Bordetella bronchiseptica (Kennel Cough) are available for intranasal administration to cats and dogs. These vaccines require a withdrawal period of vaccinated animal from vulnerable owner, ranging from 35 days - 11 weeks. Recently, a new dead IM vaccine is now available not requiring exclusion of the vaccinated pet from CF owner. WHAT IS NEW & CONCLUSION: CF pharmacists, hospital pharmacists and community pharmacists are important custodians of vaccine-related advice to people with CF, who are frequently consulted for such advice. Pharmacists should be aware of the recent innovations in veterinary medicines, so that they can give appropriate advice to people with CF when asked. Immunocompromised patients, that is those with CF or those with any defect in their phagocytic function (chronic granulomatous disease, leukocyte adhesion deficiency, myeloperoxidase deficiency, Chediak-Higashi syndrome) should avoid exposure to live veterinary bacterial vaccines and seek animal vaccination utilising non-live vaccines. Most importantly, this manuscript highlights the development of a new veterinary vaccine for dogs, which we want to make the CF healthcare community aware of, which is an acellular dead vaccine, so that those patients with dogs needing annual vaccination can select this vaccine pathway, thereby minimising risk of infection from the vaccine strains and avoiding the social exclusion between CF patient and their pet. CF patients should understand the potential infection implications of live-attenuated viral and bacterial strains as vaccines, whether these are small companion animals, exotic animals or large farm animals. Patients should make their veterinarian aware of their CF status, so that a safe and efficacious vaccine strategy is used, both mitigating the potential infection risks from live vaccine components with the CF patient, but simultaneously offering maximum immunological protection to the animal.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Bordetella/prevenção & controle , Doenças do Gato/prevenção & controle , Fibrose Cística/epidemiologia , Doenças do Cão/prevenção & controle , Zoonoses/prevenção & controle , Animais , Vacinas Bacterianas/imunologia , Bordetella bronchiseptica , Doenças do Gato/microbiologia , Gatos , Doenças do Cão/microbiologia , Cães , Humanos , Animais de Estimação , Medicina Veterinária
6.
Fish Shellfish Immunol ; 120: 133-138, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34780977

RESUMO

Pseudomonas plecoglossicida is the causative agent of visceral granulomas disease (VGD) in large yellow croaker (LYC, Larimichthys crocea) farming. However, multi-antibiotic resistant of P. plecoglossicida creates an urgent need of an efficient vaccine to combat this pathogen. In this study, an inactivated vaccine added polyactin (PA), CpG-riched plasmid (pCpG) and aluminum adjuvant (Al) was developed. As a result, its relative percentage survival (RPS) against P. plecoglossicida were up to 64%. Comparatively, RPS of groups that vaccinated with vaccines adjuvanted with PA and Al or CpG and Al were 49% and 39%. However, an interesting result that the vaccine combined with PA, CpG and Al did not show the strongest activation of total serum protein and antibody levels in serum among three vaccinated groups. According to expressions of some cellular immune related genes, we found that the inactivated vaccine combined with PA, CpG and Al was more likely to induce a cellular immune response rather than humoral immune response. Totally, our study demonstrated that the mixture of PA, CpG and aluminum adjuvant is a potential adjuvant system for LYC vaccine development.


Assuntos
Vacinas Bacterianas/imunologia , Ilhas de CpG , Doenças dos Peixes , Glicopeptídeos/farmacologia , Perciformes , Infecções por Pseudomonas , Potência de Vacina , Adjuvantes Imunológicos , Alumínio , Animais , Doenças dos Peixes/prevenção & controle , Perciformes/imunologia , Perciformes/microbiologia , Pseudomonas , Infecções por Pseudomonas/prevenção & controle , Infecções por Pseudomonas/veterinária , Desenvolvimento de Vacinas , Vacinas de Produtos Inativados
7.
Front Immunol ; 12: 768820, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34917086

RESUMO

Fowl cholera (FC) caused by Pasteurella multocida is among the serious infectious diseases of poultry. Currently, formalin inactivated FC (FI-FC) vaccine is widely used in Ethiopia. However, reports of the disease complaint remain higher despite the use of the vaccine. The aim of this study was to develop and evaluate gamma-irradiated mucosal FC vaccines that can be used nationally. In a vaccination-challenge experiment, the performance of gamma-irradiated P. multocida (at 1 kGy) formulated with Montanide gel/01 PR adjuvant was evaluated at different dose rates (0.5 and 0.3 ml) and routes (intranasal, intraocular, and oral), in comparison with FI-FC vaccine in chicken. Chickens received three doses of the candidate vaccine at 3-week intervals. Sera, and trachea and crop lavage were collected to assess the antibody levels using indirect and sandwich ELISAs, respectively. Challenge exposure was conducted by inoculation at 3.5×109 CFU/ml of P. multocida biotype A intranasally 2 weeks after the last immunization. Repeated measures ANOVA test and Kaplan Meier curve analysis were used to examine for statistical significance of antibody titers and survival analysis, respectively. Sera IgG and secretory IgA titers were significantly raised after second immunization (p=0.0001). Chicken survival analysis showed that intranasal and intraocular administration of the candidate vaccine at the dose of 0.3 ml resulted in 100% protection as compared to intramuscular injection of FI-FC vaccine, which conferred 85% protection (p=0.002). In conclusion, the results of this study showed that gamma-irradiated FC mucosal vaccine is safe and protective, indicating its potential use for immunization of chicken against FC.


Assuntos
Vacinas Bacterianas/imunologia , Infecções por Pasteurella/veterinária , Pasteurella multocida/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinação/veterinária , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/efeitos adversos , Galinhas , Raios gama , Infecções por Pasteurella/prevenção & controle , Pasteurella multocida/efeitos da radiação
8.
Infect Genet Evol ; 96: 105136, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34775078

RESUMO

Sexually transmitted diseases (STDs) have a profound effect on reproductivity and sexual health worldwide. According to world health organization (WHO) 375 million new case of STD, including chlamydia trachomatis (chlamydia), Neisseria gonorrhoeae, HSV, HPV has been reported in 2016. More than 30 diverse pathogenesis have identified to be transmitted through sexual intercourse. Of these, viral infections (hepatitis B, herpes simplex virus (HSV or herpes), HIV, and human papillomavirus (HPV) are incurable. However, symptoms caused by the incurable viral infections can be alleviated through treatment. Antimicrobial resistance (AMR) of sexually transmitted infections (STIs) to antibiotics has increased recent years, in this regard, vaccination is proposed as an important strategy for prevention or treatment of STDs. Vaccine against HPV 16 and 18 suggests a new approach for controlling STDs but until now, there is no prophylactic or therapeutic vaccine have been approved for HSV-2 and Chlamydia trachomatis (CT); in this reason, developing an efficient vaccine is inevitable. Recently, different combinatorial forms of subunit vaccines against two or three type of bacteria have been designed. In this study, to design a combinatorial vaccine against HSV, CT, and HPV, the E7 and L2 from HPV, glycoprotein D from HSV-2 and ompA from CT were selected as final antigens. Afterward, the immunodominant helper T lymphocytes (HTLs) and cytolytic T lymphocytes (CTLs) epitopes were chosen from aforesaid antigens. P30 (tetanus toxoid epitope) as universal T-helper were also added to the vaccine. Moreover, flagellin D1/D0 as TLR5 agonist and the RS09 as a TLR4 ligand were incorporated to N and C-terminals of peptide vaccine, respectively. Finally, all selected parts were fused together by appropriate linkers to enhance vaccine efficiency. The physicochemical, structural, and immunological properties of the designed vaccine protein were assessed. To achieve the best 3D model of the protein vaccine, modeling, refinement, and validation of modeled structures were also done. Docking evaluation demonstrated suitable interaction between the vaccine and TLR5. Moreover, molecular dynamics (MD) studies showed an appropriate and stable structure of protein and TLR5. Based on immunoinformatic analysis, our vaccine candidate could potentially incite humoral and cellular immunities, which are critical for protection against HPV, HSV-2, and chlamydia trachomatis. It should be noted that, experimental studies are needed to confirm the efficacy of the designed vaccine.


Assuntos
Vacinas Bacterianas/imunologia , Chlamydia trachomatis/imunologia , Herpesvirus Humano 2/imunologia , Papillomaviridae/imunologia , Infecções Sexualmente Transmissíveis/prevenção & controle , Vacinas Virais/imunologia , Infecções por Chlamydia/prevenção & controle , Epitopos de Linfócito B/imunologia , Herpes Simples/prevenção & controle , Humanos , Infecções por Papillomavirus/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia
9.
Sci Rep ; 11(1): 22377, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789792

RESUMO

Mycoplasma (M.) hyopneumoniae is the main pathogen of porcine enzootic pneumonia (PEP). Its controlling is challenging, and requires alternative strategies. This study aimed to develop an oral vaccine against M. hyopneumoniae using a nanostructured mesoporous silica (SBA-15) as an adjuvant, and compare its effect with an intramuscular (IM) commercial vaccine (CV). Fifty 24 day-old M. hyopneumoniae-free piglets composed five equal groups for different immunization protocols, consisting of a CV and/or oral immunization (OI). Control piglets did not receive any form of immunization. All piglets were challenged with M. hyopneumoniae strain 232 on D49 by tracheal route. IgA antibody response in the respiratory tract, bacterial shedding and serum IgG were evaluated. The piglets were euthanized on 28 (D77) and 56 (D105) days post-infection. Lung lesions were macroscopically evaluated; lung fragments and bronchoalveolar fluid (BALF) were collected for estimation of bacterial loads by qPCR and/or histopathology examination. All immunization protocols induced reduction on Mycoplasma-like macroscopic lung lesions. IgA Ab responses anti-M. hyopneumoniae, the expression of IL-4 cytokine and a lower expression of IL-8 were induced by CV and OI vaccines, while IgG was induced only by CV. Oral immunization using silica as a carrier-adjuvant can be viable in controlling M. hyopneumoniae infection.


Assuntos
Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Mycoplasma hyopneumoniae/imunologia , Pneumonia Suína Micoplasmática/prevenção & controle , Adjuvantes Imunológicos , Administração Oral , Animais , Biópsia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Imuno-Histoquímica , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Mycoplasma hyopneumoniae/classificação , Mycoplasma hyopneumoniae/genética , Pneumonia Suína Micoplasmática/microbiologia , Pneumonia Suína Micoplasmática/patologia , Reação em Cadeia da Polimerase em Tempo Real , Dióxido de Silício , Suínos , Resultado do Tratamento , Vacinação/métodos
10.
Front Immunol ; 12: 716676, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659206

RESUMO

Peptide-based subunit vaccines are coming to the forefront of current vaccine approaches, with safety and cost-effective production among their top advantages. Peptide vaccine formulations consist of multiple synthetic linear epitopes that together trigger desired immune responses that can result in robust immune memory. The advantages of linear compared to conformational epitopes are their simple structure, ease of synthesis, and ability to stimulate immune responses by means that do not require complex 3D conformation. Prediction of linear epitopes through use of computational tools is fast and cost-effective, but typically of low accuracy, necessitating extensive experimentation to verify results. On the other hand, identification of linear epitopes through experimental screening has been an inefficient process that requires thorough characterization of previously identified full-length protein antigens, or laborious techniques involving genetic manipulation of organisms. In this study, we apply a newly developed generalizable screening method that enables efficient identification of B-cell epitopes in the proteomes of pathogenic bacteria. As a test case, we used this method to identify epitopes in the proteome of Francisella tularensis (Ft), a Select Agent with a well-characterized immunoproteome. Our screen identified many peptides that map to known antigens, including verified and predicted outer membrane proteins and extracellular proteins, validating the utility of this approach. We then used the method to identify seroreactive peptides in the less characterized immunoproteome of Select Agent Burkholderia pseudomallei (Bp). This screen revealed known Bp antigens as well as proteins that have not been previously identified as antigens. Although B-cell epitope prediction tools Bepipred 2.0 and iBCE-EL classified many of our seroreactive peptides as epitopes, they did not score them significantly higher than the non-reactive tryptic peptides in our study, nor did they assign higher scores to seroreactive peptides from known Ft or Bp antigens, highlighting the need for experimental data instead of relying on computational epitope predictions alone. The present workflow is easily adaptable to detecting peptide targets relevant to the immune systems of other mammalian species, including humans (depending upon the availability of convalescent sera from patients), and could aid in accelerating the discovery of B-cell epitopes and development of vaccines to counter emerging biological threats.


Assuntos
Mapeamento de Epitopos/métodos , Epitopos de Linfócito B/imunologia , Proteoma , Proteômica , Animais , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Biologia Computacional/métodos , Francisella tularensis/imunologia , Humanos , Imunização , Camundongos , Peptídeos/imunologia , Proteômica/métodos , Vacinas de Subunidades Antigênicas/imunologia
11.
Mol Immunol ; 140: 22-34, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34649027

RESUMO

Acinetobacter baumannii is an opportunistic multidrug-resistant pathogen that causes a significant mortality rate. The proteins containing Tetratrico Peptide Repeats (TPRs) are involved in the pathogenicity and virulence of bacteria and have different roles such as transfer of bacterial virulence factors to host cells, binding to the host cells and inhibition of phagolysosomal maturation. So, in this study, physicochemical properties of a new protein containing TPRs in A. baumannii which was named PcTPRs1 by this study were characterized and its 3D structure was predicted by in-silico tools. The protein B and T cell epitopes were mapped and its vaccine potential was in-silico and in-vivo investigated. Domain analysis indicated that the protein contains the Flp pilus assembly protein TadD domain which has three TPRs. The helix is dominant in the protein structure, and this protein is an outer membrane antigen which, is extremely conserved among A. baumannii strains; thus, has good properties to be applied as a recombinant vaccine. The best-predicted and refined model was applied in ligand-binding sites and conformational epitopes prediction. Based on epitope mapping results, several epitopes were characterized which could stimulate both immune systems. BLAST results showed the introduced epitopes are completely conserved among A. baumannii strains. The in-vivo analysis indicates that a 101 amino acid fragment of the protein which contains the best selected epitope, can produce a good protectivity against A. baumannii as well as the whole TPR protein and thus could be investigated as an effective subunit and potential vaccines.


Assuntos
Acinetobacter baumannii/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Fenômenos Químicos , Simulação por Computador , Mapeamento de Epitopos , Repetições de Tetratricopeptídeos , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Camundongos Endogâmicos BALB C , Modelos Moleculares , Peptídeos/química , Peptídeos/imunologia , Domínios Proteicos , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Frações Subcelulares/metabolismo , Análise de Sobrevida , Virulência
12.
mSphere ; 6(5): e0069921, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34612675

RESUMO

Along with surging threats and antibiotic resistance of Pseudomonas aeruginosa in health care settings, it is imperative to develop effective vaccines against P. aeruginosa infection. In this study, we used an Asd (aspartate-semialdehyde dehydrogenase)-based balanced-lethal host-vector system of a recombinant Yersinia pseudotuberculosis mutant to produce self-adjuvanting outer membrane vesicles (OMVs). The OMVs were used as a carrier to deliver the heterologous PcrV-HitAT (PH) fusion antigen of P. aeruginosa for vaccine evaluation. Intramuscular vaccination with OMVs carrying the PH antigen (referred to rOMV-PH) afforded 73% protection against intranasal challenge with 5 × 106 (25 50% lethal doses) of the cytotoxic PA103 strain and complete protection against a noncytotoxic PAO1 strain. In contrast, vaccination with the PH-deficient OMVs or PH antigen alone failed to offer effective protection against the same challenge. Immune analysis showed that the rOMV-PH vaccination induced potent humoral and Th1/Th17 responses compared to the PH vaccination. The rOMV-PH vaccination rapidly cleared P. aeruginosa burdens with coordinated production of proinflammatory cytokines in mice. Moreover, antigen-specific CD4+ and CD8+ T cells and their producing cytokines (tumor necrosis factor alpha and interleukin-17A), rather than antibodies, were essential for protection against pneumonic P. aeruginosa infection. Our studies demonstrated that the recombinant Y. pseudotuberculosis OMVs delivering heterologous P. aeruginosa antigens could be a new promising vaccine candidate for preventing the spread of drug-resistant P. aeruginosa. IMPORTANCE Hospital- and community-acquired infections with Pseudomonas aeruginosa cause a high rate of morbidity and mortality in patients who have underlying medical conditions. The spread of multidrug-resistant P. aeruginosa strains is becoming a great challenge for treatment using antibiotics. Thus, a vaccine as one of the alternative strategies is urgently required to prevent P. aeruginosa infection.


Assuntos
Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Proteínas Citotóxicas Formadoras de Poros/imunologia , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/imunologia , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Antibacterianos/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/sangue , Feminino , Imunização , Pneumopatias/imunologia , Pneumopatias/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Pseudomonas/imunologia
13.
Front Immunol ; 12: 698737, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34249004

RESUMO

Efficacious vaccines are needed to control genital chlamydial diseases in humans and the veterinary industry. We previously reported a C. abortus (Cab) vaccine comprising recombinant Vibrio cholerae ghosts (rVCG) expressing the conserved and immunogenic N-terminal region of the Cab polymorphic membrane protein D (rVCG-Pmp18.1) protein that protected mice against intravaginal challenge. In this study, we investigated the immunomodulatory effect of the hematopoietic progenitor activator cytokine, Fms-like tyrosine kinase 3-ligand (FL) when co-administered with the rVCG-Pmp18.1 vaccine as a strategy to enhance the protective efficacy and the potential mechanism of immunomodulation. Groups of female C57BL/6J mice were immunized and boosted twice intranasally (IN) with rVCG-PmpD18.1 with and without FL or purified rPmp18.1 or rVCG-gD2 (antigen control) or PBS (medium) per mouse. The results revealed that co-administration of the vaccine with FL enhanced antigen-specific cellular and humoral immune responses and protected against live Cab genital infection. Comparative analysis of immune cell phenotypes infiltrating mucosal and systemic immune inductive tissue sites following immunization revealed that co-administration of rVCG-Pmp18.1 with FL significantly enhanced the number of macrophages, dendritic and NK cells, γδ and NK T cells in the spleen (systemic) and iliac lymph nodes (ILN) draining the genital tract (mucosal) tissues compared to rVCG-Pmp18.1 alone. Furthermore, FL enhanced monocyte infiltration in the ILN, while CD19+ B cells and CD4+ T cells were enhanced in the spleen. These results indicate that the immunomodulatory effect of FL is associated with its ability to mobilize innate immune cells and subsequent activation of robust antigen-specific immune effectors in mucosal and systemic lymphoid tissues.


Assuntos
Adjuvantes de Vacinas/farmacocinética , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/farmacologia , Infecções por Chlamydia , Proteínas de Membrana/imunologia , Animais , Chlamydia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Vibrio cholerae
14.
Glycoconj J ; 38(4): 397-398, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34169357

RESUMO

Glycoconjugate vaccines, obtained by carbohydrates covalently bound to protein carriers, have contributed to fight diseases such as meningitidis, pneumonia and typhoid fever. Despite new technologies such as RNA and adenovirus based vaccine have now reached the market, these approaches are unable to target carbohydrates which are key virulence factors.This issue intends to provide an overview on relevant directions where the field is evolving and serve as starting point to increase interest in this exciting and fundamental part of vaccinology.


Assuntos
Vacinas Bacterianas/imunologia , Glicoconjugados/química , Vacinas Conjugadas/imunologia , Vacinas Virais/imunologia , Vacinas Bacterianas/classificação , Humanos , Vacinas Virais/classificação
15.
Sci Rep ; 11(1): 9940, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976336

RESUMO

Clostridioides difficile (C. difficile) is an opportunistic anaerobic bacterium that causes severe diseases of the digestive tract of humans and animals. One of the possible methods of preventing C. difficile infection is to develop a vaccine. The most promising candidates for vaccine antigens are the proteins involved in the adhesion phenomena. Among them, the FliC and FliD are considered to be suitable candidates. In this paper, the FliC and FliD protein polypeptide epitopes were mapped in silico and by using PEPSCAN procedure. We identified four promising epitopes: 117QRMRTLS123, 205MSKAG209 of FliC and 226NKVAS230, 306TTKKPKD312 of FliD protein. We showed that 117QRMRTLS123 sequence is not only located in TLR5-binding and activating region, as previously shown, but forms an epitope recognized by C. difficile-infected patients' antibodies. 205MSKAG209 is a C. difficile-unique, immunogenic sequence that forms an exposed epitope on the polymerized flagella structure which makes it a suitable vaccine antigen. 226NKVAS230 and 306TTKKPKD312 are well exposed and possess potential protective properties according to VaxiJen analysis. Our results open the possibility to use these epitopes as suitable anti-C. difficile vaccine antigens.


Assuntos
Clostridioides difficile/imunologia , Infecções por Clostridium/diagnóstico , Flagelos/imunologia , Sequência de Aminoácidos/genética , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Clostridioides/genética , Clostridioides/imunologia , Clostridioides difficile/patogenicidade , Infecções por Clostridium/imunologia , Epitopos/imunologia , Flagelina/imunologia , Humanos , Coelhos , Alinhamento de Sequência/métodos
16.
Vet Immunol Immunopathol ; 237: 110256, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33971523

RESUMO

Lawsonia intracellularis is an economically important bacterium that causes ileitis in pigs. Current vaccines for L. intracellularis do not allow for differentiation between infected and vaccinated animals (DIVA), which is beneficial for disease tracking and surveillance. Previously, we identified five putative surface L. intracellularis proteins that were targeted by antibodies from pigs infected with L. intracellularis which could serve as antigens in a subunit vaccine. We conducted two trials to determine whether these antigens were immunogenic and provided protection against infectious challenge and whether truncated glycoprotein D could be used as a DIVA antigen. For Trial 1, 5 week-old piglets were administered intramuscular monovalent vaccines comprised of a recombinant (r) flagella subunit protein (rFliC,) and DIVA antigen (truncated glycoprotein D (TgD), a herpes virus antigen) both formulated with a combination adjuvant consisting of polyinosinic:polycytidylic acid(poly I:C), host defense peptide 1002 and polyphosphazene, referred to as Triple Adjuvant (TriAdj). Relative to control animals, animals vaccinated with rFliC and rTgD had significantly elevated antigen-specific humoral immunity in sera suggesting that rFliC and TgD are immunogenic. Control animals had negligible anti-TgD titres suggesting that TgD may be a suitable DIVA antigen for pigs. For Trial 2, piglets were immunized with a trivalent vaccine (FOG vaccine consisting of rFLiC, rOppA protein (a ABC Type dipeptide transport system) and rGroEL (a stress response protein)) and a divalent vaccine (CM vaccine consisting of rClpP (an ATP-dependent Clp protease proteolytic subunit) and rMetK (a S-adenosyl methionine synthase)) formulated with Emulsigen®. Relative to the control pigs, pigs immunized with the FOG vaccine produced robust and significantly higher serum IgG antibodies against rFliC and rGroEL, and significantly higher anti-FliC and anti-GroEL IgA antibodies in jejunal (GroEL only) and ileal intestinal mucosa. Pigs immunized with CM vaccine produced significantly higher serum antibodies against rClpP and rMetK and significantly higher anti-rClpP IgA antibodies in the ileum relative to the control pigs. Quantitative polymerase chain reaction (qPCR) analysis showed that 18 days after challenge with infectious L. intracellularis, challenged/control pigs and pigs that received the CM vaccine, but not the pigs vaccinated with the FOG vaccine, shed significantly more bacteria in feces than the unchallenged controls pigs. These data suggest that the FOG vaccinated pigs showed limited protection. While promising, more work is needed to enhance the efficiency of the intramuscular vaccine to show significant disease protection.


Assuntos
Vacinas Bacterianas/imunologia , Infecções por Desulfovibrionaceae/prevenção & controle , Imunogenicidade da Vacina , Lawsonia (Bactéria)/imunologia , Doenças dos Suínos/prevenção & controle , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/imunologia , Infecções por Desulfovibrionaceae/imunologia , Feminino , Gravidez , Suínos , Doenças dos Suínos/microbiologia , Vacinas Combinadas/imunologia , Vacinas de Subunidades Antigênicas/imunologia
17.
J Bacteriol ; 203(15): e0008221, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-33972353

RESUMO

Treponema pallidum, an obligate human pathogen, has an outer membrane (OM) whose physical properties, ultrastructure, and composition differ markedly from those of phylogenetically distant Gram-negative bacteria. We developed structural models for the outer membrane protein (OMP) repertoire (OMPeome) of T. pallidum Nichols using solved Gram-negative structures, computational tools, and small-angle X-ray scattering (SAXS) of selected recombinant periplasmic domains. The T. pallidum "OMPeome" harbors two "stand-alone" proteins (BamA and LptD) involved in OM biogenesis and four paralogous families involved in the influx/efflux of small molecules: 8-stranded ß-barrels, long-chain-fatty-acid transporters (FadLs), OM factors (OMFs) for efflux pumps, and T. pallidum repeat proteins (Tprs). BamA (TP0326), the central component of a ß-barrel assembly machine (BAM)/translocation and assembly module (TAM) hybrid, possesses a highly flexible polypeptide-transport-associated (POTRA) 1-5 arm predicted to interact with TamB (TP0325). TP0515, an LptD ortholog, contains a novel, unstructured C-terminal domain that models inside the ß-barrel. T. pallidum has four 8-stranded ß-barrels, each containing positively charged extracellular loops that could contribute to pathogenesis. Three of five FadL-like orthologs have a novel α-helical, presumptively periplasmic C-terminal extension. SAXS and structural modeling further supported the bipartite membrane topology and tridomain architecture of full-length members of the Tpr family. T. pallidum's two efflux pumps presumably extrude noxious small molecules via four coexpressed OMFs with variably charged tunnels. For BamA, LptD, and OMFs, we modeled the molecular machines that deliver their substrates into the OM or external milieu. The spirochete's extended families of OM transporters collectively confer a broad capacity for nutrient uptake. The models also furnish a structural road map for vaccine development. IMPORTANCE The unusual outer membrane (OM) of T. pallidum, the syphilis spirochete, is the ultrastructural basis for its well-recognized capacity for invasiveness, immune evasion, and persistence. In recent years, we have made considerable progress in identifying T. pallidum's repertoire of OMPs. Here, we developed three-dimensional (3D) models for the T. pallidum Nichols OMPeome using structural modeling, bioinformatics, and solution scattering. The OM contains three families of OMP transporters, an OMP family involved in the extrusion of noxious molecules, and two "stand-alone" proteins involved in OM biogenesis. This work represents a major advance toward elucidating host-pathogen interactions during syphilis; understanding how T. pallidum, an extreme auxotroph, obtains a wide array of biomolecules from its obligate human host; and developing a vaccine with global efficacy.


Assuntos
Membrana Externa Bacteriana/química , Vacinas Bacterianas/química , Sífilis/prevenção & controle , Treponema pallidum/imunologia , Membrana Externa Bacteriana/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Humanos , Modelos Estruturais , Conformação Proteica , Sífilis/microbiologia , Treponema pallidum/química , Treponema pallidum/genética , Difração de Raios X
18.
Vet Immunol Immunopathol ; 237: 110272, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34029878

RESUMO

The bacterium Clostridium chauvoei is the causative agent of blackleg in livestock, and vaccination is the most effective means of prevention. The aim of this study was to assess the effect of short-term supplementation with Bacillus toyonensis and Saccharomyces boulardii on the immune response to a C. chauvoei vaccine in sheep. Sheep were vaccinated subcutaneously on day 0 and received a booster dose on day 21, with 2 mL of a commercial vaccine formulated with inactivated C. chauvoei bacterin adsorbed on aluminum hydroxide. Probiotics were orally administered B. toyonensis (3 × 108 cfu) and S. boulardii (3 × 108 cfu) over five days prior to the first and second doses of the vaccine. Sheep supplemented with B. toyonensis and S. boulardii showed significantly higher specific IgG, IgG1, and IgG2 titers (P<0.05), with approximately 24- and 14-fold increases in total IgG levels, respectively, than the nonsupplemented group. Peripheral blood mononuclear cells from the supplemented group had increased mRNA transcription levels of the IFN-γ, IL2, and Bcl6 genes. These results demonstrate an adjuvant effect of short-term supplementation with B. toyonensis and S. boulardii on the immune response against the C. chauvoei vaccine in sheep.


Assuntos
Bacillus/imunologia , Vacinas Bacterianas/imunologia , Infecções por Clostridium/veterinária , Clostridium chauvoei/imunologia , Saccharomyces boulardii/imunologia , Doenças dos Ovinos/prevenção & controle , Animais , Anticorpos Antibacterianos/imunologia , Infecções por Clostridium/imunologia , Infecções por Clostridium/prevenção & controle , Feminino , Imunoglobulina G/imunologia , Imunomodulação , Interferon gama/genética , Interleucina-2/genética , Probióticos/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-6/genética , Ovinos , Doenças dos Ovinos/imunologia , Transcrição Gênica
19.
Vet Immunol Immunopathol ; 236: 110253, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33940537

RESUMO

Coxiella burnetii causes Q fever in individuals exposed to infected ruminants. Vaccination in 3-4-month-old goats, has been reported to result in significantly greater reduction in C. burnetii shedding compared to goats vaccinated one month before breeding, the most commonly used strategy of controlling Q fever on infected intensively-managed herds. It is possible that an even greater reduction in the number of animals shedding C. burnetii could be achieved if vaccination were administered shortly after protection from colostrum antibodies wanes and animals become susceptible to infection with C. burnetii. This study aimed to evaluate the immunogenicity and safety of a formaldehyde-inactivated phase 1 C. burnetii vaccine in 8-week-old goats. Two injections, four weeks apart, elicited specific IgM and IgG responses in all vaccinated goats (n = 6), while no antibodies were detected in two control groups (n = 12). Swelling at the site of inoculation was observed in all the vaccinated and in 10/11 of the placebo-treated goats but receded after 3 weeks. Weight change and rectal temperatures were also comparable between vaccinated and control goats. The data indicated that this vaccine could be suitable for immunising 8-week-old goats, although further trials to determine level of protection against challenge are required.


Assuntos
Vacinas Bacterianas/imunologia , Formaldeído/química , Doenças das Cabras/prevenção & controle , Imunogenicidade da Vacina , Vacinação/veterinária , Fatores Etários , Animais , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/administração & dosagem , Fezes/microbiologia , Feminino , Doenças das Cabras/imunologia , Doenças das Cabras/microbiologia , Cabras , Imuno-Histoquímica/métodos , Masculino , Gravidez , Distribuição Aleatória , Vacinas de Produtos Inativados/imunologia
20.
Infect Immun ; 89(7): e0013421, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33875472

RESUMO

Francisella tularensis is a facultative, intracellular, Gram-negative bacterium that causes a fatal disease known as tularemia. Due to its extremely high virulence, ease of spread by aerosolization, and potential to be used as a bioterror agent, F. tularensis is classified by the CDC as a tier 1 category A select agent. Previous studies have demonstrated the roles of the inflammasome sensors absent in melanoma 2 (AIM2) and NLRP3 in the generation of innate immune responses to F. tularensis infection. However, contributions of both the AIM2 and NLRP3 to the development of vaccine-induced adaptive immune responses against F. tularensis are not known. This study determined the contributions of Aim2 and Nlrp3 inflammasome sensors to vaccine-induced immune responses in a mouse model of respiratory tularemia. We developed a model to vaccinate Aim2- and Nlrp3-deficient (Aim2-/- and Nlrp3-/-) mice using the emrA1 mutant of the F. tularensis live vaccine strain (LVS). The results demonstrate that the innate immune responses in Aim2-/- and Nlrp3-/- mice vaccinated with the emrA1 mutant differ from those of their wild-type counterparts. However, despite these differences in the innate immune responses, both Aim2-/- and Nlrp3-/- mice are fully protected against an intranasal lethal challenge dose of F. tularensis LVS. Moreover, the lack of both Aim2 and Nlrp3 inflammasome sensors does not affect the production of vaccination-induced antibody and cell-mediated responses. Overall, this study reports a novel finding that both Aim2 and Nlrp3 are dispensable for vaccination-induced immunity against respiratory tularemia caused by F. tularensis.


Assuntos
Vacinas Bacterianas/imunologia , Proteínas de Ligação a DNA/genética , Francisella tularensis/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Tularemia/genética , Tularemia/imunologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Humoral , Imunização , Camundongos , Camundongos Knockout , Mutação , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Atenuadas , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA