Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38968329

RESUMO

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Assuntos
Sistemas CRISPR-Cas , Influenza Humana , Estabilidade de RNA , RNA Mensageiro , RNA Viral , Animais , RNA Viral/genética , RNA Viral/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Humanos , Influenza Humana/virologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N2/genética , Infecções por Orthomyxoviridae/virologia , Antivirais/farmacologia , Cães , Cricetinae , Proteínas Virais/genética , Proteínas Virais/metabolismo , Mesocricetus , Células Madin Darby de Rim Canino
2.
Nat Mater ; 22(3): 369-379, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36443576

RESUMO

Messenger RNA has now been used to vaccinate millions of people. However, the diversity of pulmonary pathologies, including infections, genetic disorders, asthma and others, reveals the lung as an important organ to directly target for future RNA therapeutics and preventatives. Here we report the screening of 166 polymeric nanoparticle formulations for functional delivery to the lungs, obtained from a combinatorial synthesis approach combined with a low-dead-volume nose-only inhalation system for mice. We identify P76, a poly-ß-amino-thio-ester polymer, that exhibits increased expression over formulations lacking the thiol component, delivery to different animal species with varying RNA cargos and low toxicity. P76 allows for dose sparing when delivering an mRNA-expressed Cas13a-mediated treatment in a SARS-CoV-2 challenge model, resulting in similar efficacy to a 20-fold higher dose of a neutralizing antibody. Overall, the combinatorial synthesis approach allowed for the discovery of promising polymeric formulations for future RNA pharmaceutical development for the lungs.


Assuntos
COVID-19 , Animais , Camundongos , RNA Mensageiro/genética , SARS-CoV-2/genética , Polímeros/metabolismo , Pulmão , RNA/metabolismo
3.
J Infect Dis ; 219(9): 1448-1455, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30496439

RESUMO

BACKGROUND: Persistence of bacterial pathogens in the airways has profound consequences on the course and pathogenesis of chronic obstructive pulmonary disease (COPD). Patients with COPD continuously acquire and clear strains of Moraxella catarrhalis, a major pathogen in COPD. Some strains are cleared quickly and some persist for months to years. The mechanism of the variability in duration of persistence is unknown. METHODS: Guided by genome sequences of selected strains, we studied the expression of Hag/MID, hag/mid gene sequences, adherence to human cells, and autoaggregation in longitudinally collected strains of M. catarrhalis from adults with COPD. RESULTS: Twenty-eight of 30 cleared strains of M. catarrhalis expressed Hag/MID whereas 17 of 30 persistent strains expressed Hag/MID upon acquisition by patients. All persistent strains ceased expression of Hag/MID during persistence. Expression of Hag/MID in human airways was regulated by slipped-strand mispairing. Virulence-associated phenotypes (adherence to human respiratory epithelial cells and autoaggregation) paralleled Hag/MID expression in airway isolates. CONCLUSIONS: Most strains of M. catarrhalis express Hag/MID upon acquisition by adults with COPD and all persistent strains shut off expression during persistence. These observations suggest that Hag/MID is important for initial colonization by M. catarrhalis and that cessation of expression facilitates persistence in COPD airways.


Assuntos
Adesinas Bacterianas/genética , Moraxella catarrhalis/genética , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/microbiologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Sistema Respiratório/microbiologia , Adulto , Aderência Bacteriana , Expressão Gênica , Humanos , Moraxella catarrhalis/fisiologia , Fenótipo , Fatores de Virulência/genética
4.
Vet Pathol ; 55(2): 258-267, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29145795

RESUMO

Burkholderia mallei causes the highly contagious and debilitating zoonosis glanders, which infects via inhalation or percutaneous inoculation and often culminates in life-threatening pneumonia and sepsis. In humans, glanders is difficult to diagnose and requires prolonged antibiotic therapy with low success rates. No vaccine exists to protect against B. mallei, and there is concern regarding its use as a bioweapon. The authors previously identified the protein BpaB as a potential target for devising therapies due to its role in adherence to host cells and the formation of biofilms in vitro and its contribution to pathogenicity in a mouse model of glanders. In the present study, the authors developed an immunostaining approach to probe tissues of experimentally infected animals and demonstrated that BpaB is produced exclusively in vivo by wild-type B. mallei in target organs from mice and marmosets. They detected the expression of BpaB by B. mallei both extracellularly and within macrophages, neutrophils, and epithelial cells in respiratory tissues (7/10 marmoset; 2/2 mouse). The authors also noted the intracellular expression of BpaB by B. mallei in macrophages in the regional lymph nodes of mice (2/2 tissues) and MALT of marmosets (4/5 tissues). It is interesting that B. mallei bacteria infecting distal organs did not express BpaB (2/2 mice; 3/3 marmosets), suggesting that the protein is not necessary for bacterial fitness in these anatomic locations. These findings underscore the value of BpaB as a target for developing medical countermeasures and provide insight into its role in pathogenesis.


Assuntos
Burkholderia mallei/patogenicidade , Mormo/microbiologia , Fatores de Virulência/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Burkholderia mallei/imunologia , Burkholderia mallei/metabolismo , Callithrix/microbiologia , Mormo/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Virulência/imunologia
5.
Infect Immun ; 85(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28507073

RESUMO

Burkholderia mallei, a facultative intracellular bacterium and tier 1 biothreat, causes the fatal zoonotic disease glanders. The organism possesses multiple genes encoding autotransporter proteins, which represent important virulence factors and targets for developing countermeasures in pathogenic Gram-negative bacteria. In the present study, we investigated one of these autotransporters, BatA, and demonstrate that it displays lipolytic activity, aids in intracellular survival, is expressed in vivo, elicits production of antibodies during infection, and contributes to pathogenicity in a mouse aerosol challenge model. A mutation in the batA gene of wild-type strain ATCC 23344 was found to be particularly attenuating, as BALB/c mice infected with the equivalent of 80 median lethal doses cleared the organism. This finding prompted us to test the hypothesis that vaccination with the batA mutant strain elicits protective immunity against subsequent infection with wild-type bacteria. We discovered that not only does vaccination provide high levels of protection against lethal aerosol challenge with B. mallei ATCC 23344, it also protects against infection with multiple isolates of the closely related organism and causative agent of melioidosis, Burkholderia pseudomallei Passive-transfer experiments also revealed that the protective immunity afforded by vaccination with the batA mutant strain is predominantly mediated by IgG antibodies binding to antigens expressed exclusively in vivo Collectively, our data demonstrate that BatA is a target for developing medical countermeasures and that vaccination with a mutant lacking expression of the protein provides a platform to gain insights regarding mechanisms of protective immunity against B. mallei and B. pseudomallei, including antigen discovery.


Assuntos
Anticorpos Antibacterianos/imunologia , Burkholderia mallei/imunologia , Burkholderia pseudomallei/imunologia , Melioidose/prevenção & controle , Animais , Proteínas de Bactérias/genética , Burkholderia mallei/genética , Burkholderia mallei/crescimento & desenvolvimento , Burkholderia mallei/patogenicidade , Burkholderia pseudomallei/patogenicidade , Modelos Animais de Doenças , Mormo/imunologia , Mormo/microbiologia , Mormo/prevenção & controle , Imunoglobulina G/imunologia , Melioidose/imunologia , Melioidose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Vacinação , Fatores de Virulência/genética
6.
Inflamm Res ; 65(1): 71-80, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26608498

RESUMO

OBJECTIVE AND DESIGN: Our aim was to study whether an extracellular, oxidative antimicrobial mechanism inherent to tracheal epithelial cells is capable of inactivating influenza H1N2 virus. MATERIAL OR SUBJECTS: Epithelial cells were isolated from tracheas of male Sprague-Dawley rats. Both primary human and rat tracheobronchial epithelial cells were differentiated in air-liquid interface cultures. TREATMENT: A/swine/Illinois/02860/09 (swH1N2) influenza A virions were added to the apical side of airway cells for 1 h in the presence or absence of lactoperoxidase or thiocyanate. METHODS: Characterization of rat epithelial cells (morphology, Duox expression) occurred via western blotting, PCR, hydrogen peroxide production measurement and histology. The number of viable virions was determined by plaque assays. Statistical difference of the results was analyzed by ANOVA and Tukey's test. RESULTS: Our data show that rat tracheobronchial epithelial cells develop a differentiated, polarized monolayer with high transepithelial electrical resistance, mucin production and expression of dual oxidases. Influenza A virions are inactivated by human and rat epithelial cells via a dual oxidase-, lactoperoxidase- and thiocyanate-dependent mechanism. CONCLUSIONS: Differentiated air-liquid interface cultures of rat tracheal epithelial cells provide a novel model to study airway epithelium-influenza interactions. The dual oxidase/lactoperoxidase/thiocyanate extracellular oxidative system producing hypothiocyanite is a fast and potent anti-influenza mechanism inactivating H1N2 viruses prior to infection of the epithelium.


Assuntos
Células Epiteliais/metabolismo , Vírus da Influenza A Subtipo H1N2/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Tiocianatos/metabolismo , Animais , Cães , Humanos , Peróxido de Hidrogênio/metabolismo , Lactoperoxidase/metabolismo , Células Madin Darby de Rim Canino , Masculino , Mucinas/biossíntese , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Mucosa Respiratória/citologia
7.
J Bacteriol ; 196(1): 107-20, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24142255

RESUMO

The major phospholipid constituents of Moraxella catarrhalis membranes are phosphatidylglycerol, phosphatidylethanolamine, and cardiolipin (CL). However, very little is known regarding the synthesis and function of these phospholipids in M. catarrhalis. In this study, we discovered that M. catarrhalis expresses a cardiolipin synthase (CLS), termed MclS, that is responsible for the synthesis of CL within the bacterium. The nucleotide sequence of mclS is highly conserved among M. catarrhalis isolates and is predicted to encode a protein with significant amino acid similarity to the recently characterized YmdC/ClsC protein of Escherichia coli. Isogenic mclS mutant strains were generated in M. catarrhalis isolates O35E, O12E, and McGHS1 and contained no observable levels of CL. Site-directed mutagenesis of a highly conserved HKD motif of MclS also resulted in a CL-deficient strain. Moraxella catarrhalis, which depends on adherence to epithelial cells for colonization of the human host, displays significantly reduced levels of adherence to HEp-2 and A549 cell lines in the mclS mutant strains compared to wild-type bacteria. The reduction in adherence appears to be attributed to the absence of CL. These findings mark the first instance in which a CLS has been related to a virulence-associated trait.


Assuntos
Aderência Bacteriana , Cardiolipinas/biossíntese , Células Epiteliais/microbiologia , Proteínas de Membrana/biossíntese , Moraxella catarrhalis/enzimologia , Moraxella catarrhalis/fisiologia , Transferases (Outros Grupos de Fosfato Substituídos)/biossíntese , Linhagem Celular , Análise Mutacional de DNA , DNA Bacteriano/química , DNA Bacteriano/genética , Proteínas de Escherichia coli/genética , Deleção de Genes , Hepatócitos/microbiologia , Humanos , Proteínas de Membrana/genética , Dados de Sequência Molecular , Moraxella catarrhalis/genética , Mutagênese Sítio-Dirigida , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transferases (Outros Grupos de Fosfato Substituídos)/genética
8.
BMC Microbiol ; 14: 92, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24731253

RESUMO

BACKGROUND: Autotransporters form a large family of outer membrane proteins specifying diverse biological traits of Gram-negative bacteria. In this study, we report the identification and characterization of a novel autotransporter gene product of Burkholderia mallei (locus tag BMA1027 in strain ATCC 23344). RESULTS: Database searches identified the gene in at least seven B. mallei isolates and the encoded proteins were found to be 84% identical. Inactivation of the gene encoding the autotransporter in the genome of strain ATCC 23344 substantially reduces adherence to monolayers of HEp-2 laryngeal cells and A549 type II pneumocytes, as well as to cultures of normal human bronchial epithelium (NHBE). Consistent with these findings, expression of the autotransporter on the surface of recombinant E. coli bacteria increases adherence to these cell types by 5-7 fold. The gene specifying the autotransporter was identified in the genome of 29 B. pseudomallei isolates and disruption of the gene in strain DD503 reduced adherence to NHBE cultures by 61%. Unlike B. mallei, the mutation did not impair binding of B. pseudomallei to A549 or HEp-2 cells. Analysis of sera from mice infected via the aerosol route with B. mallei and B. pseudomallei revealed that animals inoculated with as few as 10 organisms produce antibodies against the autotransporter, therefore indicating expression in vivo. CONCLUSIONS: Our data demonstrate that we have identified an autotransporter protein common to the pathogenic species B. mallei and B. pseudomallei which mediates adherence to respiratory epithelial cells and is expressed in vivo during the course of aerosol infection.


Assuntos
Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Burkholderia mallei/fisiologia , Burkholderia pseudomallei/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Adesinas Bacterianas/genética , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Burkholderia mallei/genética , Burkholderia pseudomallei/genética , Células Epiteliais/microbiologia , Escherichia coli/genética , Escherichia coli/fisiologia , Feminino , Deleção de Genes , Expressão Gênica , Humanos , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
BMC Microbiol ; 13: 140, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23782650

RESUMO

BACKGROUND: Moraxella catarrhalis is a human-specific gram-negative bacterium readily isolated from the respiratory tract of healthy individuals. The organism also causes significant health problems, including 15-20% of otitis media cases in children and ~10% of respiratory infections in adults with chronic obstructive pulmonary disease. The lack of an efficacious vaccine, the rapid emergence of antibiotic resistance in clinical isolates, and high carriage rates reported in children are cause for concern. Virtually all Moraxella catarrhalis isolates are resistant to ß-lactam antibiotics, which are generally the first antibiotics prescribed to treat otitis media in children. The enzymes responsible for this resistance, BRO-1 and BRO-2, are lipoproteins and the mechanism by which they are secreted to the periplasm of M. catarrhalis cells has not been described. RESULTS: Comparative genomic analyses identified M. catarrhalis gene products resembling the TatA, TatB, and TatC proteins of the well-characterized Twin Arginine Translocation (TAT) secretory apparatus. Mutations in the M. catarrhalis tatA, tatB and tatC genes revealed that the proteins are necessary for optimal growth and resistance to ß-lactams. Site-directed mutagenesis was used to replace highly-conserved twin arginine residues in the predicted signal sequence of M. catarrhalis strain O35E BRO-2, which abolished resistance to the ß-lactam antibiotic carbanecillin. CONCLUSIONS: Moraxella catarrhalis possesses a TAT secretory apparatus, which plays a key role in growth of the organism and is necessary for secretion of BRO-2 into the periplasm where the enzyme can protect the peptidoglycan cell wall from the antimicrobial activity of ß-lactam antibiotics.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Moraxella catarrhalis/metabolismo , beta-Lactamases/metabolismo , Adulto , Criança , Pré-Escolar , Biologia Computacional , Análise Mutacional de DNA , Técnicas de Inativação de Genes , Genoma Bacteriano , Humanos , Proteínas de Membrana Transportadoras/genética , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/genética , Moraxella catarrhalis/crescimento & desenvolvimento , Moraxella catarrhalis/isolamento & purificação , Infecções por Moraxellaceae/microbiologia , Mutagênese Sítio-Dirigida , Resistência beta-Lactâmica
10.
bioRxiv ; 2022 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-35702147

RESUMO

Immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has greatly reduced coronavirus disease 2019 (COVID-19)-related deaths and hospitalizations, but waning immunity and the emergence of variants capable of immune escape indicate the need for novel SARS-CoV-2 vaccines. An intranasal parainfluenza virus 5 (PIV5)-vectored COVID-19 vaccine CVXGA1 has been proven efficacious in animal models and blocks contact transmission of SARS-CoV-2 in ferrets. CVXGA1 vaccine is currently in human clinical trials in the United States. This work investigates the immunogenicity and efficacy of CVXGA1 and other PIV5-vectored vaccines expressing additional antigen SARS-CoV-2 nucleoprotein (N) or SARS-CoV-2 variant spike (S) proteins of beta, delta, gamma, and omicron variants against homologous and heterologous challenges in hamsters. A single intranasal dose of CVXGA1 induces neutralizing antibodies against SARS-CoV-2 WA1 (ancestral), delta variant, and omicron variant and protects against both homologous and heterologous virus challenges. Compared to mRNA COVID-19 vaccine, neutralizing antibody titers induced by CVXGA1 were well-maintained over time. When administered as a boost following two doses of a mRNA COVID-19 vaccine, PIV5-vectored vaccines expressing the S protein from WA1 (CVXGA1), delta, or omicron variants generate higher levels of cross-reactive neutralizing antibodies compared to three doses of a mRNA vaccine. In addition to the S protein, the N protein provides added protection as assessed by the highest body weight gain post-challenge infection. Our data indicates that PIV5-vectored COVID-19 vaccines, such as CVXGA1, can serve as booster vaccines against emerging variants.

11.
Adv Sci (Weinh) ; 9(34): e2202771, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36316224

RESUMO

Despite the success of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, there remains a clear need for new classes of preventatives for respiratory viral infections due to vaccine hesitancy, lack of sterilizing immunity, and for at-risk patient populations, including the immunocompromised. While many neutralizing antibodies have been identified, and several approved, to treat COVID-19, systemic delivery, large doses, and high costs have the potential to limit their widespread use, especially in low- and middle-income countries. To use these antibodies more efficiently, an inhalable formulation is developed that allows for the expression of mRNA-encoded, membrane-anchored neutralizing antibodies in the lung to mitigate SARS-CoV-2 infections. First, the ability of mRNA-encoded, membrane-anchored, anti-SARS-CoV-2 antibodies to prevent infections in vitro is demonstrated. Next, it is demonstrated that nebulizer-based delivery of these mRNA-expressed neutralizing antibodies potently abrogates disease in the hamster model. Overall, these results support the use of nebulizer-based mRNA expression of neutralizing antibodies as a new paradigm for mitigating respiratory virus infections.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , RNA Mensageiro/genética , Anticorpos Neutralizantes/uso terapêutico
12.
Nat Biotechnol ; 39(6): 717-726, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33536629

RESUMO

Cas13a has been used to target RNA viruses in cell culture, but efficacy has not been demonstrated in animal models. In this study, we used messenger RNA (mRNA)-encoded Cas13a for mitigating influenza virus A and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in mice and hamsters, respectively. We designed CRISPR RNAs (crRNAs) specific for PB1 and highly conserved regions of PB2 of influenza virus, and against the replicase and nucleocapsid genes of SARS-CoV-2, and selected the crRNAs that reduced viral RNA levels most efficiently in cell culture. We delivered polymer-formulated Cas13a mRNA and the validated guides to the respiratory tract using a nebulizer. In mice, Cas13a degraded influenza RNA in lung tissue efficiently when delivered after infection, whereas in hamsters, Cas13a delivery reduced SARS-CoV-2 replication and reduced symptoms. Our findings suggest that Cas13a-mediated targeting of pathogenic viruses can mitigate respiratory infections.


Assuntos
COVID-19/terapia , Influenza Humana/terapia , RNA Mensageiro/farmacologia , SARS-CoV-2/genética , Animais , COVID-19/genética , COVID-19/virologia , Sistemas CRISPR-Cas/genética , Cricetinae , Modelos Animais de Doenças , Humanos , Influenza Humana/genética , Influenza Humana/virologia , Camundongos , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Orthomyxoviridae/patogenicidade , RNA Mensageiro/genética , RNA Viral/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , SARS-CoV-2/patogenicidade
13.
BMC Microbiol ; 10: 250, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20920184

RESUMO

BACKGROUND: Burkholderia pseudomallei and Burkholderia mallei cause the diseases melioidosis and glanders, respectively. A well-studied aspect of pathogenesis by these closely-related bacteria is their ability to invade and multiply within eukaryotic cells. In contrast, the means by which B. pseudomallei and B. mallei adhere to cells are poorly defined. The purpose of this study was to identify adherence factors expressed by these organisms. RESULTS: Comparative sequence analyses identified a gene product in the published genome of B. mallei strain ATCC23344 (locus # BMAA0649) that resembles the well-characterized Yersinia enterocolitica autotransporter adhesin YadA. The gene encoding this B. mallei protein, designated boaA, was expressed in Escherichia coli and shown to significantly increase adherence to human epithelial cell lines, specifically HEp2 (laryngeal cells) and A549 (type II pneumocytes), as well as to cultures of normal human bronchial epithelium (NHBE). Consistent with these findings, disruption of the boaA gene in B. mallei ATCC23344 reduced adherence to all three cell types by ~50%. The genomes of the B. pseudomallei strains K96243 and DD503 were also found to contain boaA and inactivation of the gene in DD503 considerably decreased binding to monolayers of HEp2 and A549 cells and to NHBE cultures.A second YadA-like gene product highly similar to BoaA (65% identity) was identified in the published genomic sequence of B. pseudomallei strain K96243 (locus # BPSL1705). The gene specifying this protein, termed boaB, appears to be B. pseudomallei-specific. Quantitative attachment assays demonstrated that recombinant E. coli expressing BoaB displayed greater binding to A549 pneumocytes, HEp2 cells and NHBE cultures. Moreover, a boaB mutant of B. pseudomallei DD503 showed decreased adherence to these respiratory cells. Additionally, a B. pseudomallei strain lacking expression of both boaA and boaB was impaired in its ability to thrive inside J774A.1 murine macrophages, suggesting a possible role for these proteins in survival within professional phagocytic cells. CONCLUSIONS: The boaA and boaB genes specify adhesins that mediate adherence to epithelial cells of the human respiratory tract. The boaA gene product is shared by B. pseudomallei and B. mallei whereas BoaB appears to be a B. pseudomallei-specific adherence factor.


Assuntos
Adesinas Bacterianas/genética , Burkholderia mallei/genética , Burkholderia pseudomallei/genética , Células Epiteliais/microbiologia , Sequência de Aminoácidos , Animais , Burkholderia mallei/classificação , Burkholderia mallei/patogenicidade , Burkholderia pseudomallei/classificação , Burkholderia pseudomallei/patogenicidade , Linhagem Celular , Feminino , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sequência de DNA , Especificidade da Espécie
14.
Hum Vaccin Immunother ; 16(12): 3194-3200, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401688

RESUMO

Conserved Moraxella catarrhalis (Mcat) proteins, oligopeptide permease (Opp)A, hemagglutinin (Hag), outer membrane protein (OMP) CD, Pilin A clade 2 (PilA2), and Moraxella surface protein (Msp) 22 have been studied as vaccine candidates. Children who experience frequent acute otitis media (AOM) confirmed with pathogen identification by tympanocentesis are referred to as stringently-defined otitis prone (sOP). Synchrony of serum antibody responses against 5 Mcat proteins, OppA, Hag, OMP CD, PilA2, and Msp22 resulting from nasopharyngeal colonization and AOM was studied for 85 non-otitis prone (NOP) children and 34 sOP children. Changes in serum IgG were quantitated with ELISA. Serum IgG antibody levels against OppA, Hag, OMP CD, and Msp22 rose in synchrony in NOP and sOP children; that is, the proteins appeared equally and highly immunogenic in children at age 6 to 22-25 months old and then leveled off in their rise at 22-25 to 30 months old. In contrast, rises of PilA2 were slow from 6 months old and kept constant and did not level off significantly before 30 months old. OppA, Hag, OMP CD, and Msp22 elicited a synchronous acquisition of naturally-induced serum antibody in young children. A multi-valent Mcat protein vaccine combining OppA, Hag, OMP CD, and Msp22 may exhibit less antigen competition when administered as a combination vaccine in young children.


Assuntos
Formação de Anticorpos , Moraxella catarrhalis , Otite Média , Anticorpos Antibacterianos , Proteínas da Membrana Bacteriana Externa , Criança , Pré-Escolar , Humanos , Lactente , Moraxella catarrhalis/imunologia , Nasofaringe
15.
Virulence ; 11(1): 1024-1040, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32799724

RESUMO

BURKHOLDERIA MALLEI: is a highly pathogenic bacterium that causes the fatal zoonosis glanders. The organism specifies multiple membrane proteins, which represent prime targets for the development of countermeasures given their location at the host-pathogen interface. We investigated one of these proteins, Pal, and discovered that it is involved in the ability of B. mallei to resist complement-mediated killing and replicate inside host cells in vitro, is expressed in vivo and induces antibodies during the course of infection, and contributes to virulence in a mouse model of aerosol infection. A mutant in the pal gene of the B. mallei wild-type strain ATCC 23344 was found to be especially attenuated, as BALB/c mice challenged with the equivalent of 5,350 LD50 completely cleared infection. Based on these findings, we tested the hypothesis that a vaccine containing the Pal protein elicits protective immunity against aerosol challenge. To achieve this, the pal gene was cloned in the vaccine vector Parainfluenza Virus 5 (PIV5) and mice immunized with the virus were infected with a lethal dose of B. mallei. These experiments revealed that a single dose of PIV5 expressing Pal provided 80% survival over a period of 40 days post-challenge. In contrast, only 10% of mice vaccinated with a PIV5 control virus construct survived infection. Taken together, our data establish that the Peptidoglycan-associated lipoprotein Pal is a critical virulence determinant of B. mallei and effective target for developing a glanders vaccine.


Assuntos
Vacinas Bacterianas/imunologia , Burkholderia mallei/química , Burkholderia mallei/patogenicidade , Lipoproteínas/imunologia , Melioidose/prevenção & controle , Peptidoglicano/química , Aerossóis , Animais , Vacinas Bacterianas/administração & dosagem , Burkholderia mallei/imunologia , Linhagem Celular , Feminino , Vetores Genéticos , Imunização , Lipoproteínas/administração & dosagem , Macrófagos/microbiologia , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 5/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Virulência
16.
Infect Immun ; 77(10): 4597-608, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19667048

RESUMO

Moraxella catarrhalis is a human pathogen causing otitis media in infants and respiratory infections in adults, particularly patients with chronic obstructive pulmonary disease. The surface protein Hag (also designated MID) has previously been shown to be a key adherence factor for several epithelial cell lines relevant to pathogenesis by M. catarrhalis, including NCIH292 lung cells, middle ear cells, and A549 type II pneumocytes. In this study, we demonstrate that Hag mediates adherence to air-liquid interface cultures of normal human bronchial epithelium (NHBE) exhibiting mucociliary activity. Immunofluorescent staining and laser scanning confocal microscopy experiments demonstrated that the M. catarrhalis wild-type isolates O35E, O12E, TTA37, V1171, and McGHS1 bind principally to ciliated NHBE cells and that their corresponding hag mutant strains no longer associate with cilia. The hag gene product of M. catarrhalis isolate O35E was expressed in the heterologous genetic background of a nonadherent Haemophilus influenzae strain, and quantitative assays revealed that the adherence of these recombinant bacteria to NHBE cultures was increased 27-fold. These experiments conclusively demonstrate that the hag gene product is responsible for the previously unidentified tropism of M. catarrhalis for ciliated NHBE cells.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Células Epiteliais/microbiologia , Moraxella catarrhalis/patogenicidade , Mucosa Respiratória/microbiologia , Fatores de Virulência/fisiologia , Adesinas Bacterianas/genética , Adulto , Proteínas de Bactérias/genética , Linhagem Celular , Imunofluorescência , Deleção de Genes , Teste de Complementação Genética , Haemophilus influenzae/genética , Haemophilus influenzae/patogenicidade , Humanos , Lactente , Microscopia Confocal , Fatores de Virulência/genética
17.
mSphere ; 4(1)2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30602525

RESUMO

Burkholderia pseudomallei and Burkholderia mallei are the causative agents of melioidosis and glanders, respectively. There is no vaccine to protect against these highly pathogenic bacteria, and there is concern regarding their emergence as global public health (B. pseudomallei) and biosecurity (B. mallei) threats. In this issue of mSphere, an article by Khakhum and colleagues (N. Khakhum, P. Bharaj, J. N. Myers, D. Tapia, et al., mSphere 4:e00570-18, 2019, https://doi.org/10.1128/mSphere.00570-18) describes a novel vaccination platform with excellent potential for cross-protection against both Burkholderia species. The report also highlights the importance of antibodies in immunity against these facultative intracellular organisms.


Assuntos
Burkholderia pseudomallei , Mormo , Aerossóis , Animais , Burkholderia , Burkholderia mallei , Cavalos
18.
BMC Med Genomics ; 12(1): 127, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31492148

RESUMO

BACKGROUND: Burkholderia mallei (Bm) is a facultative intracellular bacterial pathogen causing highly-fatal glanders in solipeds and humans. The ability of Bm to thrive intracellularly is thought to be related to exploitation of host immune response-related genes and pathways. Relatively little is known of the molecular strategies employed by this pathogen to modulate these pathways and evade intracellular killing. This manuscript seeks to fill gaps in the understanding of the interface between Bm and innate immunity by examining gene expression changes during infection of host monocytes. METHODS: The transcriptome of Bm-infected human Mono Mac-6 (MM6) monocytes was profiled on Affymetrix Human Transcriptome GeneChips 2.0. Gene expression changes in Bm-infected monocytes were compared to those of Burkholderia thailandensis (Bt)-infected monocytes and to uninfected monocytes. The resulting dataset was normalized using Robust Multichip Average and subjected to statistical analyses employing a univariate F test with a random variance model. Differentially expressed genes significant at p < 0.001 were subjected to leave-one-out cross-validation studies and 1st and 3rd nearest neighbor prediction model. Significant probe sets were used to populate human pathways in Ingenuity Pathway Analysis, with statistical significance determined by Fisher's exact test or z-score. RESULTS: The Pattern Recognition Receptor (PRR) pathway was represented among significantly enriched immune response-related human canonical pathways, with evidence of upregulation across both infections. Among members of this pathway, pentraxin-3 was significantly upregulated by Bm- or Bt-infected monocytes. Pentraxin-3 (PTX3) was demonstrated to bind to both Bt and Burkholderia pseudomallei (Bp), but not Bm. Subsequent assays did not identify a role for PTX3 in potentiating complement-mediated lysis of Bt or in enhancing phagocytosis or replication of Bt in human monocytes. CONCLUSION: We report on the novel binding of PTX3 to Bt and Bp, with lack of interaction with Bm, suggesting that a possible evasive mechanism by Bm warrants further exploration. We determined that (1) PTX3 may not play a role in activating the lytic pathway of complement in different bacterial species and that (2) the opsonophagocytic properties of PTX3 should be investigated in different primary or immortalized cell lines representing host phagocytes, given lack of binding of PTX3 to MM6 monocytes.


Assuntos
Burkholderia/imunologia , Proteína C-Reativa/metabolismo , Perfilação da Expressão Gênica , Imunidade Inata , Monócitos/imunologia , Monócitos/microbiologia , Componente Amiloide P Sérico/metabolismo , Anticorpos/metabolismo , Burkholderia/crescimento & desenvolvimento , Linhagem Celular , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunidade Inata/genética , Viabilidade Microbiana , Proteínas Opsonizantes/metabolismo , Fagocitose , Ligação Proteica , Regulação para Cima/genética
19.
Vaccine ; 37(32): 4637-4645, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-28755833

RESUMO

BACKGROUND: Moraxella catarrhalis (Mcat) is a frequent pathogen of acute otitis media (AOM) in young children. Here we prospectively assessed naturally-induced serum antibodies to four Mcat vaccine candidate proteins in stringently defined otitis prone (sOP) and non-otitis prone (NOP) children age 6-36months old following nasopharyngeal (NP) colonization, at onset of AOM and convalescence from AOM. METHODS: Serum IgG and IgM antibody against recombinant Mcat proteins, oligopeptide permease A (OppA), outer membrane protein (OMP) CD, hemagglutinin (Hag), and PilA clade 2 (PilA2), were quantitated by ELISA. RESULTS: During NP colonization by Mcat all four antigens were immunogenic in both sOP and NOP children. However, sOP children had lower antibody responses than NOP children across age 6-36months, similar to our findings for protein vaccine candidates of Streptococcus pneumoniae (Spn) and Nontypeable Haemophilus influenzae (NTHi). sOP children displayed a later and lower peak of antibody rise than NOP children for all four antigens during NP colonization of Mcat. The age-dependent increase of antibody ranked as OppA>Hag5-9>OMP CD>PilA2 in both sOP and NOP children. Lower serum antibody levels to the Mcat antigens were measured in sOP compared to NOP children at the onset of AOM. We did not find a consistent significant increase of antibody at the convalescence phase after an AOM event. CONCLUSIONS: sOP children is a highly vulnerable population that mount lower serum antibody responses to Mcat candidate vaccine proteins compared to NOP children during asymptomatic NP carriage and at onset of AOM.


Assuntos
Anticorpos Antibacterianos/sangue , Formação de Anticorpos/imunologia , Proteínas de Bactérias/imunologia , Moraxella catarrhalis/imunologia , Otite/imunologia , Soro/imunologia , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Pré-Escolar , Feminino , Infecções por Haemophilus/sangue , Infecções por Haemophilus/imunologia , Haemophilus influenzae/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Lactente , Masculino , Proteínas de Membrana Transportadoras/imunologia , Nasofaringe/imunologia , Otite/sangue , Otite Média/imunologia , Infecções Pneumocócicas/sangue , Infecções Pneumocócicas/imunologia , Estudos Prospectivos , Streptococcus pneumoniae/imunologia
20.
Vaccine X ; 1: 100002, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-33826684

RESUMO

BACKGROUND: Burkholderia mallei and Burkholderia pseudomallei are the causative agents of glanders and melioidosis, respectively. There is no vaccine to protect against these highly-pathogenic and intrinsically antibiotic-resistant bacteria, and there is concern regarding their use as biological warfare agents. For these reasons, B. mallei and B. pseudomallei are classified as Tier 1 organisms by the U.S. Federal Select Agent Program and the availability of effective countermeasures represents a critical unmet need. METHODS: Vaccines (subunit and vectored) containing the surface-exposed passenger domain of the conserved Burkholderia autotransporter protein BatA were administered to BALB/c mice and the vaccinated animals were challenged with lethal doses of wild-type B. mallei and B. pseudomallei strains via the aerosol route. Mice were monitored for signs of illness for a period of up to 40 days post-challenge and tissues from surviving animals were analyzed for bacterial burden at study end-points. RESULTS: A single dose of recombinant Parainfluenza Virus 5 (PIV5) expressing BatA provided 74% and 60% survival in mice infected with B. mallei and B. pseudomallei, respectively. Vaccination with PIV5-BatA also resulted in complete bacterial clearance from the lungs and spleen of 78% and 44% of animals surviving lethal challenge with B. pseudomallei, respectively. In contrast, all control animals vaccinated with a PIV5 construct expressing an irrelevant antigen and infected with B. pseudomallei were colonized in those tissues. CONCLUSION: Our study indicates that the autotransporter BatA is a valuable target for developing countermeasures against B. mallei and B. pseudomallei and demonstrates the utility of the PIV5 viral vaccine delivery platform to elicit cross-protective immunity against the organisms.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA