Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Microbiol ; 73(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38963177

RESUMO

Introduction. Administered nasally, spores of the Gram-positive bacterium Bacillus subtilis have been shown to be able to induce innate immunity sufficient to confer protection to influenza and respiratory syncytial virus.Hypothesis. Although members of the aerobiome, intranasal delivery of high numbers of live spores carries potential safety issues.Aim. To address the potential safety risk of using live spores, we assessed the safety of spores that had been completely inactivated using heat sterilization.Methodology. Using autoclaved, and therefore killed, spores of a generally recognized as safe-notified B. subtilis strain (DSM 32444), safety was assessed in vitro (biotype, genome and cell based cytoxicity) and in vivo, using intranasal administration in rodent models and lastly in human volunteers.Results. Using a 15-day, repeat-dose, regimen in a rodent model, no indication of toxicity was observed. In a registered human study (NCT05984004), a formulated preparation of inactivated DSM 32444 spores referred to as SPEROVID was developed, and tolerance in human volunteers was assessed following 7 days of nasal dosing (2-4 times/day).Conclusion. Our study demonstrated that in humans an intranasal dose of up to 3×108 killed spores was safe and well tolerated.


Assuntos
Administração Intranasal , Bacillus subtilis , Esporos Bacterianos , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Ratos , Adulto Jovem
2.
ACS Infect Dis ; 10(3): 928-937, 2024 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-38334357

RESUMO

Clostridioides difficile causes life-threatening diarrhea and is one of the leading causes of nosocomial infections. During infection, C. difficile releases two gut-damaging toxins, TcdA and TcdB, which are the primary determinants of disease pathogenesis and are important therapeutic targets. Once in the cytosol of mammalian cells, TcdA and TcdB use UDP-glucose to glucosylate host Rho GTPases, which leads to cytoskeletal changes that result in a loss of intestinal integrity. Isofagomine inhibits TcdA and TcdB as a mimic of the glucocation transition state of the glucosyltransferase reaction. However, sequence variants of TcdA and TcdB across the clades of infective C. difficile continue to be identified, and therefore, evaluation of isofagomine inhibition against multiple toxin variants is required. Here, we show that isofagomine inhibits the glucosyltransferase domain of multiple TcdB variants and protects TcdB-induced cell rounding of the most common full-length toxin variants. Furthermore, we demonstrate that isofagomine protects against C. difficile-induced mortality in two murine models of C. difficile infection. Isofagomine treatment of mouse C. difficile infection also permitted the recovery of the gastrointestinal microbiota, an important barrier to preventing recurring C. difficile infection. The broad specificity of isofagomine supports its potential as a prophylactic to protect against C. difficile-induced morbidity and mortality.


Assuntos
Toxinas Bacterianas , Compostos de Boro , Clostridioides difficile , Imino Piranoses , Animais , Camundongos , Toxinas Bacterianas/genética , Enterotoxinas , Clostridioides difficile/genética , Proteínas de Bactérias/genética , Glucosiltransferases/genética , Mamíferos
3.
bioRxiv ; 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37781587

RESUMO

Clostridioides difficile causes life-threatening diarrhea and is the leading cause of healthcare associated bacterial infections in the United States. During infection, C. difficile releases the gut-damaging toxins, TcdA and TcdB, the primary determinants of disease pathogenesis and are therefore therapeutic targets. TcdA and TcdB contain a glycosyltransferase domain that uses UDP-glucose to glycosylate host Rho GTPases, causing cytoskeletal changes that result in a loss of intestinal integrity. Isofagomine inhibits TcdA and TcdB as a mimic of the oxocarbenium ion transition state of the glycosyltransferase reaction. However, sequence variants of TcdA and TcdB across the clades of infective C. difficile continue to be identified and therefore, evaluation of isofagomine inhibition against multiple toxin variants are required. Here we show that Isofagomine inhibits the glycosyltransferase activity of multiple TcdB variants and also protects TcdB toxin-induced cell rounding of the most common full-length toxin variants. Further, isofagomine protects against C. difficile induced mortality in two murine models of C. difficile infection. Isofagomine treatment of mouse C. difficile infection permitted recovery of the gastrointestinal microbiota, an important barrier to prevent recurring C. difficile infection. The broad specificity of isofagomine supports its potential as a prophylactic to protect against C. difficile induced morbidity and mortality.

4.
Vaccines (Basel) ; 10(11)2022 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36366408

RESUMO

Background: Current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are administered systemically and typically result in poor immunogenicity at the mucosa. As a result, vaccination is unable to reduce viral shedding and transmission, ultimately failing to prevent infection. One possible solution is that of boosting a systemic vaccine via the nasal route resulting in mucosal immunity. Here, we have evaluated the potential of bacterial spores as an intranasal boost. Method: Spores engineered to express SARS-CoV-2 antigens were administered as an intranasal boost following a prime with either recombinant Spike protein or the Oxford AZD1222 vaccine. Results: In mice, intranasal boosting following a prime of either Spike or vaccine produced antigen-specific sIgA at the mucosa together with the increased production of Th1 and Th2 cytokines. In a hamster model of infection, the clinical and virological outcomes resulting from a SARS-CoV-2 challenge were ameliorated. Wuhan-specific sIgA were shown to cross-react with Omicron antigens, suggesting that this strategy might offer protection against SARS-CoV-2 variants of concern. Conclusions: Despite being a genetically modified organism, the spore vaccine platform is attractive since it offers biological containment, the rapid and cost-efficient production of vaccines together with heat stability. As such, employed in a heterologous systemic prime-mucosal boost regimen, spore vaccines might have utility for current and future emerging diseases.

5.
Vaccines (Basel) ; 10(9)2022 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-36146637

RESUMO

Background: Influenza is a respiratory infection that continues to present a major threat to human health, with ~500,000 deaths/year. Continued circulation of epidemic subtypes in humans and animals potentially increases the risk of future pandemics. Vaccination has failed to halt the evolution of this virus and next-generation prophylactic approaches are under development. Naked, "heat inactivated", or inert bacterial spores have been shown to protect against influenza in murine models. Methods: Ferrets were administered intranasal doses of inert bacterial spores (DSM 32444K) every 7 days for 4 weeks. Seven days after the last dose, the animals were challenged with avian H7N9 influenza A virus. Clinical signs of infection and viral shedding were monitored. Results: Clinical symptoms of infection were significantly reduced in animals dosed with DSM 32444K. The temporal kinetics of viral shedding was reduced but not prevented. Conclusion: Taken together, nasal dosing using heat-stable spores could provide a useful approach for influenza prophylaxis in both humans and animals.

6.
Biomedicines ; 10(5)2022 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-35625667

RESUMO

Clostridioides difficile is an environmentally acquired, anaerobic, spore-forming bacterium which ordinarily causes disease following antibiotic-mediated dysbiosis of the intestinal microbiota. Although much is understood regarding the life cycle of C. difficile, the fate of C. difficile spores upon ingestion remains unclear, and the underlying factors that predispose an individual to colonization and subsequent development of C. difficile infection (CDI) are not fully understood. Here, we show that Bacillus, a ubiquitous and environmentally acquired, spore-forming bacterium is associated with colonization resistance to C. difficile. Using animal models, we first provide evidence that animals housed under conditions that mimic reduced environmental exposure have an increased susceptibility to CDI, correlating with a loss in Bacillus. Lipopeptide micelles (~10 nm) produced by some Bacilli isolated from the gastro-intestinal (GI)-tract and shown to have potent inhibitory activity to C. difficile have recently been reported. We show here that these micelles, that we refer to as heterogenous lipopeptide lytic micelles (HELMs), act synergistically with components present in the small intestine to augment inhibitory activity against C. difficile. Finally, we show that provision of HELM-producing Bacillus to microbiota-depleted animals suppresses C. difficile colonization thereby demonstrating the significant role played by Bacillus in colonization resistance. In the wider context, our study further demonstrates the importance of environmental microbes on susceptibility to pathogen colonization.

7.
Pharmaceutics ; 13(8)2021 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-34452257

RESUMO

Members of the Bacillus genus, particularly the "Bacillus subtilis group", are known to produce amphipathic lipopeptides with biosurfactant activity. This includes the surfactins, fengycins and iturins that have been associated with antibacterial, antifungal, and anti-viral properties. We have screened a large collection of Bacillus, isolated from human, animal, estuarine water and soil samples and found that the most potent lipopeptide producers are members of the species Bacillus velezensis. B. velezensis lipopeptides exhibited anti-bacterial activity which was localised on the surface of both vegetative cells and spores. Interestingly, lipopeptide micelles (6-10 nm diameter) were detectable in strains exhibiting the highest levels of activity. Micelles were stable (heat and gastric stable) and shown to entrap other antimicrobials produced by the host bacterium (exampled here was the dipeptide antibiotic chlorotetaine). Commercially acquired lipopeptides did not exhibit similar levels of inhibitory activity and we suspect that micelle formation may relate to the particular isomeric forms produced by individual bacteria. Using naturally produced micelle formulations we demonstrated that they could entrap antimicrobial compounds (e.g., clindamycin, vancomycin and resveratrol). Micellar incorporation of antibiotics increased activity. Bacillus is a prolific producer of antimicrobials, and this phenomenon could be exploited naturally to augment antimicrobial activity. From an applied perspective, the ability to readily produce Bacillus micelles and formulate with drugs enables a possible strategy for enhanced drug delivery.

8.
PLoS Pathog ; 14(9): e1007191, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30208103

RESUMO

Clostridium difficile is a Gram-positive spore-forming anaerobe and a major cause of antibiotic-associated diarrhoea. Disruption of the commensal microbiota, such as through treatment with broad-spectrum antibiotics, is a critical precursor for colonisation by C. difficile and subsequent disease. Furthermore, failure of the gut microbiota to recover colonisation resistance can result in recurrence of infection. An unusual characteristic of C. difficile among gut bacteria is its ability to produce the bacteriostatic compound para-cresol (p-cresol) through fermentation of tyrosine. Here, we demonstrate that the ability of C. difficile to produce p-cresol in vitro provides a competitive advantage over gut bacteria including Escherichia coli, Klebsiella oxytoca and Bacteroides thetaiotaomicron. Metabolic profiling of competitive co-cultures revealed that acetate, alanine, butyrate, isobutyrate, p-cresol and p-hydroxyphenylacetate were the main metabolites responsible for differentiating the parent strain C. difficile (630Δerm) from a defined mutant deficient in p-cresol production. Moreover, we show that the p-cresol mutant displays a fitness defect in a mouse relapse model of C. difficile infection (CDI). Analysis of the microbiome from this mouse model of CDI demonstrates that colonisation by the p-cresol mutant results in a distinctly altered intestinal microbiota, and metabolic profile, with a greater representation of Gammaproteobacteria, including the Pseudomonales and Enterobacteriales. We demonstrate that Gammaproteobacteria are susceptible to exogenous p-cresol in vitro and that there is a clear divide between bacterial Phyla and their susceptibility to p-cresol. In general, Gram-negative species were relatively sensitive to p-cresol, whereas Gram-positive species were more tolerant. This study demonstrates that production of p-cresol by C. difficile has an effect on the viability of intestinal bacteria as well as the major metabolites produced in vitro. These observations are upheld in a mouse model of CDI, in which p-cresol production affects the biodiversity of gut microbiota and faecal metabolite profiles, suggesting that p-cresol production contributes to C. difficile survival and pathogenesis.


Assuntos
Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Cresóis/metabolismo , Microbioma Gastrointestinal/fisiologia , Bactérias Gram-Negativas/fisiologia , Animais , Antibacterianos/efeitos adversos , Biodiversidade , Membrana Celular/efeitos dos fármacos , Clostridioides difficile/genética , Clostridioides difficile/patogenicidade , Cresóis/farmacologia , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Humanos , Metaboloma , Camundongos , Camundongos Endogâmicos C57BL , Mutação
9.
J Infect Dis ; 216(11): 1452-1459, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-28968845

RESUMO

Clostridium difficile infection (CDI) is an important hospital-acquired infection resulting from the germination of spores in the intestine as a consequence of antibiotic-mediated dysbiosis of the gut microbiota. Key to this is CotE, a protein displayed on the spore surface and carrying 2 functional elements, an N-terminal peroxiredoxin and a C-terminal chitinase domain. Using isogenic mutants, we show in vitro and ex vivo that CotE enables binding of spores to mucus by direct interaction with mucin and contributes to its degradation. In animal models of CDI, we show that when CotE is absent, both colonization and virulence were markedly reduced. We demonstrate here that the attachment of spores to the intestine is essential in the development of CDI. Spores are usually regarded as biochemically dormant, but our findings demonstrate that rather than being simply agents of transmission and dissemination, spores directly contribute to the establishment and promotion of disease.


Assuntos
Adesinas Bacterianas/fisiologia , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Esporos Bacterianos/química , Animais , Proteínas de Bactérias/genética , Quitinases/metabolismo , Clostridioides difficile/genética , Clostridioides difficile/metabolismo , Contagem de Colônia Microbiana , Cricetinae , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Parasita/fisiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Mesocricetus , Camundongos , Mucinas/metabolismo , Mutação , Peroxirredoxinas/metabolismo , Esporos Bacterianos/genética , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/patogenicidade , Virulência
10.
Front Microbiol ; 8: 1793, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28983286

RESUMO

Clostridium difficile remains a leading nosocomial pathogen, putting considerable strain on the healthcare system. The ability to form endospores, highly resistant to environmental insults, is key to its persistence and transmission. However, important differences exist between the sporulation pathways of C. difficile and the model Gram-positive organism Bacillus subtilis. Amongst the challenges in studying sporulation in C. difficile is the relatively poor levels of sporulation and high heterogeneity in the sporulation process. To overcome these limitations we placed Ptet regulatory elements upstream of the master regulator of sporulation, spo0A, generating a new strain that can be artificially induced to sporulate by addition of anhydrotetracycline (ATc). We demonstrate that this strain is asporogenous in the absence of ATc, and that ATc can be used to drive faster and more efficient sporulation. Induction of Spo0A is titratable and this can be used in the study of the spo0A regulon both in vitro and in vivo, as demonstrated using a mouse model of C. difficile infection (CDI). Insights into differences between the sporulation pathways in B. subtilis and C. difficile gained by study of the inducible strain are discussed, further highlighting the universal interest of this tool. The Ptet-spo0A strain provides a useful background in which to generate mutations in genes involved in sporulation, therefore providing an exciting new tool to unravel key aspects of sporulation in C. difficile.

12.
Infect Immun ; 85(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28167669

RESUMO

Mucosal immunity is considered important for protection against Clostridium difficile infection (CDI). We show that in hamsters immunized with Bacillus subtilis spores expressing a carboxy-terminal segment (TcdA26-39) of C. difficile toxin A, no colonization occurs in protected animals when challenged with C. difficile strain 630. In contrast, animals immunized with toxoids showed no protection and remained fully colonized. Along with neutralizing toxins, antibodies to TcdA26-39 (but not to toxoids), whether raised to the recombinant protein or to TcdA26-39 expressed on the B. subtilis spore surface, cross-react with a number of seemingly unrelated proteins expressed on the vegetative cell surface or spore coat of C. difficile These include two dehydrogenases, AdhE1 and LdhA, as well as the CdeC protein that is present on the spore. Anti-TcdA26-39 mucosal antibodies obtained following immunization with recombinant B. subtilis spores were able to reduce the adhesion of C. difficile to mucus-producing intestinal cells. This cross-reaction is intriguing yet important since it illustrates the importance of mucosal immunity for complete protection against CDI.


Assuntos
Toxinas Bacterianas/imunologia , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Enterotoxinas/imunologia , Imunoglobulina A Secretora/imunologia , Mucosa/imunologia , Mucosa/microbiologia , Domínios e Motivos de Interação entre Proteínas/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Toxinas Bacterianas/química , Linhagem Celular , Infecções por Clostridium/prevenção & controle , Cricetinae , Reações Cruzadas , Enterotoxinas/química , Humanos , Imunidade nas Mucosas , Imunização , Camundongos , Fragmentos de Peptídeos/imunologia , Esporos Bacterianos/imunologia
13.
FEMS Microbiol Lett ; 358(2): 170-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24990572

RESUMO

Recombinant Bacillus subtilis spores expressing a TB antigen, MPT64, were tested for their ability to protect mice against tuberculosis challenge. A chimeric gene consisting of the spore coat gene cotB fused to mpt64 was constructed, and expression of a stable CotB-MPT64 hybrid protein of the spore coat verified. Spores were evaluated as a live vaccine and also formaldehyde inactivated. Mice were given three doses of spores or alternatively used in a prime-boost regimen with BCG. The results showed that inactivated recombinant spores were able to reduce the bacterial burden in the lungs of mice to comparable levels to that of BCG. In the prime-boost regimen, both live and inactivated spores showed a reduction in bacterial load in comparison with BCG. ELISPOT and polyfunctional T-cell analysis were performed to examine cellular responses and showed that antigen-specific secretion of Th1 cytokines was stimulated after immunisation with inactive recombinant spores and BCG. In summary, recombinant spores can elicit Th1 responses, which are important for protection against TB disease.


Assuntos
Antígenos de Bactérias/imunologia , Bacillus subtilis/genética , Portadores de Fármacos , Esporos Bacterianos/genética , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Animais , Antígenos de Bactérias/genética , Carga Bacteriana , Proteínas de Bactérias/genética , Técnicas de Visualização da Superfície Celular , Citocinas/metabolismo , Modelos Animais de Doenças , ELISPOT , Feminino , Vetores Genéticos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Tuberculose/imunologia , Vacinas contra a Tuberculose/genética , Vacinação/métodos , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
14.
Mol Microbiol ; 92(5): 1025-38, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24720767

RESUMO

The BclA protein is a major component of the outermost layer of spores of a number of bacterial species and Clostridium difficile carries three bclA genes. Using insertional mutagenesis each gene was characterized and spores devoid of these proteins had surface aberrations, reduced hydrophobicity and germinated faster than wild-type spores. Therefore the BclA proteins were likely major components of the spore surface and when absent impaired the protective shield effect of this outermost layer. Analysis of infection and colonization in mice and hamsters revealed that the 50% infectious dose (ID50 ) of spores was significantly higher (2-logs) in the bclA1(-) mutant compared to the isogenic wild-type control, but that levels of toxins (A and B) were indistinguishable from animals dosed with wild-type spores. bclA1(-) spores germinated faster than wild-type spores yet mice were less susceptible to infection suggesting that BclA1 must play a key role in the initial (i.e. pre-spore germination) stages of infection. We also show that the ID50 was higher in mice infected with R20291, a 'hypervirulent' 027 strain, that carries a truncated BclA1 protein.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridioides difficile/patogenicidade , Enterocolite Pseudomembranosa/metabolismo , Esporos Bacterianos/patogenicidade , Animais , Clostridioides difficile/metabolismo , Cricetinae , Regulação Bacteriana da Expressão Gênica , Camundongos , Esporos Bacterianos/metabolismo
15.
Infect Immun ; 81(11): 4071-80, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23959722

RESUMO

Needle-free, mucosal immunization is a highly desirable strategy for vaccination against many pathogens, especially those entering through the respiratory mucosa, such as Mycobacterium tuberculosis. Unfortunately, mucosal vaccination against tuberculosis (TB) is impeded by a lack of suitable adjuvants and/or delivery platforms that could induce a protective immune response in humans. Here, we report on a novel biotechnological approach for mucosal vaccination against TB that overcomes some of the current limitations. This is achieved by coating protective TB antigens onto the surface of inert bacterial spores, which are then delivered to the respiratory tract. Our data showed that mice immunized nasally with coated spores developed humoral and cellular immune responses and multifunctional T cells and, most importantly, presented significantly reduced bacterial loads in their lungs and spleens following pathogenic challenge. We conclude that this new vaccine delivery platform merits further development as a mucosal vaccine for TB and possibly also other respiratory pathogens.


Assuntos
Antígenos de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Vacinação/métodos , Administração Intranasal , Administração através da Mucosa , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/administração & dosagem , Carga Bacteriana , Técnicas de Visualização da Superfície Celular , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Feminino , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/microbiologia , Esporos Bacterianos/imunologia , Linfócitos T/imunologia , Tuberculose/imunologia , Vacinas contra a Tuberculose/administração & dosagem
16.
J Bacteriol ; 195(7): 1492-503, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23335421

RESUMO

Spores of Clostridium difficile play a key role in the dissemination of this important human pathogen, and until recently little has been known of their functional characteristics. Genes encoding six spore coat proteins (cotA, cotB, cotCB, cotD, cotE, and sodA) were disrupted by ClosTron insertional mutagenesis. Mutation of one gene, cotA, presented a major structural defect in spore assembly, with a clear misassembly of the outermost layers of the spore coat. The CotA protein is most probably subject to posttranslational modification and could play a key role in stabilizing the spore coat. Surprisingly, mutation of the other spore coat genes did not affect the integrity of the spore, although for the cotD, cotE, and sodA mutants, enzyme activity was reduced or abolished. This could imply that these enzymatic proteins are located in the exosporium or alternatively that they are structurally redundant. Of the spore coat proteins predicted to carry enzymatic activity, three were confirmed to be enzymes using both in vivo and in vitro methods, the latter using recombinant expressed proteins. These were a manganese catalase, encoded by cotD, a superoxide dismutase (SOD), encoded by sodA, and a bifunctional enzyme with peroxiredoxin and chitinase activity, encoded by cotE. These enzymes being exposed on the spore surface would play a role in coat polymerization and detoxification of H2O2. Two additional proteins, CotF (a tyrosine-rich protein and potential substrate for SodA) and CotG (a putative manganese catalase) were shown to be located at the spore surface.


Assuntos
Proteínas de Bactérias/metabolismo , Clostridioides difficile/crescimento & desenvolvimento , Clostridioides difficile/metabolismo , Esporos Bacterianos/crescimento & desenvolvimento , Esporos Bacterianos/metabolismo , Proteínas de Bactérias/genética , Clostridioides difficile/genética , Enzimas/genética , Enzimas/metabolismo , Técnicas de Inativação de Genes , Mutagênese Insercional , Esporos Bacterianos/genética
17.
Vaccine ; 30(22): 3266-77, 2012 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22446640

RESUMO

Heat killed spores of the Gram-positive bacterium Bacillus subtilis have been evaluated as a vaccine delivery system with mucosal adjuvant properties for influenza. Killed spores were able to bind H5N1 virions (NIBRG-14; clade 1) and, when intra-nasally administered to mice, resulting immune responses, both humoral and cell mediated, were enhanced compared to immunization with the virion alone. Levels of both systemic IgG and mucosal sIgA specific to the virion were elevated. Levels of IgG2a (a Th(1) antibody type) were strongly enhanced when the virion was co-administered with killed spores. Cytokine induction in stimulated splenocytes was also apparent indicating balanced T(h)1 and T(h)2 responses. Evidence of cross-neutralization of clade 2.2 viruses was shown. In a challenge experiment mice dosed two times with spores adsorbed with just 20 ng HA (hemagglutinin) of inactivated NIBRG-14 were fully protected against challenge with 20 LD(50) of H5N2 virus. Interestingly, partial protection (60%) was observed in animals dosed only with killed spores. Mice dosed only with killed spores were shown to be fully protected against H5N2 (5 LD(50)) infection indicating that innate immunity and its stimulation by spores may play an important role in protection. Supporting this killed spores were (i) shown to stimulate TLR-mediated expression of NF-κB, and (ii) able to recruit NK cells into lungs and induce maturation of DCs. This work demonstrates the potential and underlying mechanism for the use of bacterial spores as an adjuvant for H5N1 vaccination.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Bacillus subtilis/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Esporos Bacterianos/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Reações Cruzadas , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina G/sangue , Vírus da Influenza A Subtipo H5N2/imunologia , Vírus da Influenza A Subtipo H5N2/patogenicidade , Vacinas contra Influenza/administração & dosagem , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/prevenção & controle , Baço/imunologia , Análise de Sobrevida
18.
Infect Immun ; 79(6): 2295-302, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21482682

RESUMO

Clostridium difficile is a leading cause of nosocomial infection in the developed world. Two toxins, A and B, produced by most strains of C. difficile are implicated as virulence factors, yet only recently has the requirement of these for infection been investigated by genetic manipulation. Current vaccine strategies are focused mostly on parenteral delivery of toxoids. In this work, we have used bacterial spores (Bacillus subtilis) as a delivery vehicle to evaluate the carboxy-terminal repeat domains of toxins A and B as protective antigens. Our findings are important and show that oral immunization of the repeat domain of toxin A is sufficient to confer protection in a hamster model of infection designed to closely mimic the human course of infection. Importantly, neutralizing antibodies to the toxin A repeat domain were shown to be cross-reactive with the analogous domain of toxin B and, being of high avidity, provided protection against challenge with a C. difficile strain producing toxins A and B (A(+)B(+)). Thus, although many strains produce both toxins, antibodies to only toxin A can mediate protection. Animals vaccinated with recombinant spores were fully able to survive reinfection, a property that is particularly important for a disease with which patients are prone to relapse. We show that mucosal immunization, not parenteral delivery, is required to generate secretory IgA and that production of these neutralizing polymeric antibodies correlates with protection. This work demonstrates that an effective vaccine against C. difficile can be designed around two attributes, mucosal delivery and the repeat domain of toxin A.


Assuntos
Bacillus subtilis/imunologia , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Clostridioides difficile/imunologia , Enterocolite Pseudomembranosa/prevenção & controle , Enterotoxinas/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Cricetinae , Proteção Cruzada/imunologia , Enterocolite Pseudomembranosa/imunologia , Enterocolite Pseudomembranosa/microbiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Esporos Bacterianos/imunologia , Vacinas Sintéticas/imunologia
19.
Vaccine ; 28(4): 1021-30, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19914191

RESUMO

The development of new-generation vaccines has followed a number of strategic avenues including the use of live recombinant bacteria. Of these, the use of genetically engineered bacterial spores has been shown to offer promise as both a mucosal as well as a heat-stable vaccine delivery system. Spores of the genus Bacillus are currently in widespread use as probiotics enabling a case to be made for their safety. In this work we have discovered that the negatively charged and hydrophobic surface layer of spores provides a suitable platform for adsorption of protein antigens. Binding can be promoted under conditions of low pH and requires a potent combination of electrostatic and hydrophobic interactions between spore and immunogen. Using appropriately adsorbed spores we have shown that mice immunised mucosally can be protected against challenge with tetanus toxin, Clostridium perfringens alpha toxin and could survive challenge with anthrax toxin. In some cases protection is actually greater than using a recombinant vaccine. Remarkably, killed or inactivated spores appear equally effective as live spores. The spore appears to present a bound antigen in its native conformation promoting a cellular (T(h)1-biased) response coupled with a strong antibody response. Spores then, should be considered as mucosal adjuvants, most similar to particulate adjuvants, by enhancing responses against soluble antigens. The broad spectrum of immune responses elicited coupled with the attendant benefits of safety suggest that spore adsorption could be appropriate for improving the immunogenicity of some vaccines as well as the delivery of biotherapeutic molecules.


Assuntos
Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Bacillus subtilis/química , Bacillus subtilis/imunologia , Esporos Bacterianos/química , Esporos Bacterianos/imunologia , Adsorção , Animais , Antraz/prevenção & controle , Anticorpos Antibacterianos/sangue , Infecções por Clostridium/prevenção & controle , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Linfócitos T/imunologia , Tétano/prevenção & controle
20.
Int Rev Immunol ; 28(6): 487-505, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19954360

RESUMO

Over the past few decades, advancements in molecular and cell biology have allowed scientists to identify a large number of new antigens from a variety of viral and bacterial pathogens. However, successful development of these antigens into effective vaccines strongly relies on delivery systems able to avoid the rapid loss of biological activity that often impairs antigen efficacy. Various delivery systems have been proposed as alternative vaccine vehicles, from live microorganisms to nanoparticles, and all of them have shown advantages but also drawbacks. The bacterial spore is a quiescent cell form that, as a vaccine vehicle, may conjugate some advantages of live microorganisms with those of synthetic nanoparticles and that has recently been proposed as a potentially powerful tool to deliver antigens to mucosal surfaces. Here we review the use of bacterial spores as a delivery system for mucosal immunizations. We will first analyze the nature of the interaction between wild type spores and the gut-associated lymphoid tissue and then address the immune responses that are induced by oral immunizations with recombinant spores displaying heterologous antigens.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Probióticos/administração & dosagem , Esporos Bacterianos/crescimento & desenvolvimento , Vacinas/administração & dosagem , Administração Oral , Animais , Bacillus subtilis/fisiologia , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/microbiologia , Intestinos/citologia , Intestinos/imunologia , Intestinos/microbiologia , Esporos Bacterianos/genética , Vacinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...