Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Infect Immun ; 91(12): e0041623, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37909751

RESUMO

Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long infections of the respiratory mucosa caused by a diverse array of opportunists, which are leading causes of morbidity and mortality. In recent years, there has been increased appreciation for the range and diversity of microbes causing CF-related respiratory infections. The introduction of new therapeutics and improved detection methodology has revealed CF-related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species which is widely distributed in environmental sources and has been increasingly observed in sputa and other samples from pwCF, typically in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors including flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized cultures of CFBE41o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization of cell layers. Ax colonized and persisted in mouse lungs for up to 72 h post infection, with inflammatory consequences that include increased neutrophil influx in the lung, lung damage, cytokine production, and mortality. We also identified genes that are differentially expressed in synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.


Assuntos
Achromobacter denitrificans , Fibrose Cística , Infecções por Bactérias Gram-Negativas , Infecções Respiratórias , Animais , Camundongos , Humanos , Achromobacter denitrificans/genética , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Escarro/microbiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Perfilação da Expressão Gênica
2.
Microbiology (Reading) ; 169(1)2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36748431

RESUMO

Cystic fibrosis (CF) is a genetic disorder affecting epithelial ion transport, which among other impacts results in defective mucociliary clearance and innate defenses in the respiratory tract. Consequently, people with CF experience lifelong infections of the respiratory mucosa that are chronic and polymicrobial in nature. Young children with CF are initially colonized by opportunists like nontypeable Haemophilus influenzae (NTHi), which normally resides within the microbiome of the nasopharynx and upper airways and can also cause infections of the respiratory mucosa that include bronchitis and otitis media. NTHi is typically supplanted by other microbes as patients age; for example, people with CF are often chronically infected with mucoid strains of Pseudomonas aeruginosa, which prior work in our laboratory has shown to promote colonization and persistence by other opportunists that include Stenotrophomonas maltophilia. Our previous work has shown that polymicrobial infection impacts host colonization and persistence of incoming microbes via diverse mechanisms that include priming of host immunity that can promote microbial clearance, and cooperativity within polymicrobial biofilms, which can promote persistence. In infection studies with BALB/c Cftrtm1UNC mice, results showed, as previously observed for WT BALB/c mice, preceding infection with NTHi decreased colonization and persistence by P. aeruginosa. Likewise, polymicrobial infection of BALB/c Cftrtm1UNC and C57BL/6 Cftrtm1UncTg(FABPhCFTR)1Jaw/J mice showed correlation between S. maltophilia and P. aeruginosa, with increased bacterial colonization and lung pathology. Based on these results, we conclude that our previous observations regarding polymicrobial infections with CF opportunists in WT mice are also validated using CF mice.


Assuntos
Coinfecção , Fibrose Cística , Infecções por Pseudomonas , Camundongos , Animais , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Coinfecção/microbiologia , Camundongos Endogâmicos C57BL , Sistema Respiratório , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética
3.
Microbiology (Reading) ; 169(11)2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37942787

RESUMO

Stenotrophomonas maltophilia is a Gram-negative emerging opportunistic pathogen often present in people with respiratory diseases such as cystic fibrosis (CF). People with CF (pwCF) experience lifelong polymicrobial infections of the respiratory mucosa. Our prior work showed that Pseudomonas aeruginosa promotes persistence of S. maltophilia in mouse respiratory infections. As is typical for environmental opportunistic pathogens, S. maltophilia has a large genome and a high degree of genetic diversity. In this study, we evaluated the genomic content of S. maltophilia, combining short and long read sequencing to construct nearly complete genomes of 10 clinical isolates. The genomes of these isolates were then compared with all publicly available S. maltophilia genome assemblies, and each isolate was then evaluated for colonization/persistence in vivo, both alone and in coinfection with P. aeruginosa. We found that while the overall genome size and GC content were fairly consistent between strains, there was considerable variability in both genome structure and gene content. Similarly, there was significant variability in S. maltophilia colonization and persistence in experimental mouse respiratory infections in the presence or absence of P. aeruginosa. Ultimately, this study gives us a greater understanding of the genomic diversity of clinical S. maltophilia isolates, and how this genomic diversity relates to both interactions with other pulmonary pathogens and to host disease progression. Identifying the molecular determinants of infection with S. maltophilia can facilitate development of novel antimicrobial strategies for a highly drug-resistant pathogen.


Assuntos
Coinfecção , Fibrose Cística , Infecções por Bactérias Gram-Negativas , Infecções Respiratórias , Stenotrophomonas maltophilia , Humanos , Camundongos , Animais , Stenotrophomonas maltophilia/genética , Genômica , Fibrose Cística/complicações , Pseudomonas aeruginosa/genética , Variação Genética
4.
Infect Immun ; 90(2): e0056821, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34780275

RESUMO

Patients with cystic fibrosis (CF) experience lifelong respiratory infections, which are a significant cause of morbidity and death. These infections are polymicrobial in nature, and the predominant bacterial species undergo a predictable series of changes as patients age. Young patients have populations dominated by opportunists that are typically found within the microbiome of the human nasopharynx, such as nontypeable Haemophilus influenzae (NTHi); these are eventually supplanted, and the population within the CF lung is later dominated by pathogens such as Pseudomonas aeruginosa. In this study, we investigated how initial colonization with NTHi impacts colonization and persistence of P. aeruginosa in the respiratory tract. Analysis of polymicrobial biofilms in vitro by confocal microscopy revealed that NTHi promoted greater P. aeruginosa biofilm volume and diffusion. However, sequential respiratory infection of mice with NTHi followed by P. aeruginosa resulted in significantly lower levels of P. aeruginosa, compared to infection with P. aeruginosa alone. Coinfected mice also had reduced airway tissue damage and lower levels of inflammatory cytokines, compared with P. aeruginosa-infected mice. Similar results were observed after instillation of heat-inactivated NTHi bacteria or purified NTHi lipooligosaccharide endotoxin prior to P. aeruginosa introduction. Based on these results, we conclude that NTHi significantly reduces susceptibility to subsequent P. aeruginosa infection, most likely due to priming of host innate immunity rather than a direct competitive interaction between species. These findings have potential significance with regard to therapeutic management of early-life infections in patients with CF.


Assuntos
Fibrose Cística , Infecções por Haemophilus , Infecções Respiratórias , Animais , Biofilmes , Infecções por Haemophilus/microbiologia , Haemophilus influenzae , Humanos , Camundongos , Pseudomonas aeruginosa , Sistema Respiratório
5.
Microbiology (Reading) ; 168(1)2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35077346

RESUMO

Pseudomonas aeruginosa is a common opportunistic pathogen that can cause chronic infections in multiple disease states, including respiratory infections in patients with cystic fibrosis (CF) and non-CF bronchiectasis. Like many opportunists, P. aeruginosa forms multicellular biofilm communities that are widely thought to be an important determinant of bacterial persistence and resistance to antimicrobials and host immune effectors during chronic/recurrent infections. Poly (acetyl, arginyl) glucosamine (PAAG) is a glycopolymer that has antimicrobial activity against a broad range of bacterial species, and also has mucolytic activity, which can normalize the rheological properties of cystic fibrosis mucus. In this study, we sought to evaluate the effect of PAAG on P. aeruginosa bacteria within biofilms in vitro, and in the context of experimental pulmonary infection in a rodent infection model. PAAG treatment caused significant bactericidal activity against P. aeruginosa biofilms, and a reduction in the total biomass of preformed P. aeruginosa biofilms on abiotic surfaces, as well as on the surface of immortalized cystic fibrosis human bronchial epithelial cells. Studies of membrane integrity indicated that PAAG causes changes to P. aeruginosa cell morphology and dysregulates membrane polarity. PAAG treatment reduced infection and consequent tissue inflammation in experimental P. aeruginosa rat infections. Based on these findings we conclude that PAAG represents a novel means to combat P. aeruginosa infection, and may warrant further evaluation as a therapeutic.


Assuntos
Fibrose Cística , Infecções por Pseudomonas , Animais , Antibacterianos/farmacologia , Biofilmes , Fibrose Cística/microbiologia , Glucosamina/farmacologia , Glucosamina/uso terapêutico , Humanos , Pulmão/microbiologia , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Ratos
6.
Infect Immun ; 88(4)2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-31932329

RESUMO

Stenotrophomonas maltophilia is a Gram-negative bacterium found ubiquitously in the environment that has historically been regarded as nonpathogenic. S. maltophilia is increasingly observed in patient sputa in cystic fibrosis (CF), and while existing epidemiology indicates that patients with S. maltophilia have poorer diagnoses, its clinical significance remains unclear. Moreover, as multidrug resistance is common among S. maltophilia isolates, treatment options for these infections may be limited. Here, we investigated the pathogenicity of S. maltophilia alone and during polymicrobial infection with Pseudomonas aeruginosa Colonization, persistence, and virulence of S. maltophilia were assessed in experimental respiratory infections of mice. The results of this study indicate that S. maltophilia transiently colonizes the lung accompanied by significant weight loss and immune cell infiltration and the expression of early inflammatory markers, including interleukin 6 (IL-6), IL-1α, and tumor necrosis factor alpha (TNF-α). Importantly, polymicrobial infection with P. aeruginosa elicited significantly higher S. maltophilia counts in bronchoalveolar lavages and lung tissue homogenates. This increase in bacterial load was directly correlated with the density of the P. aeruginosa population and required viable P. aeruginosa bacteria. Microscopic analysis of biofilms formed in vitro revealed that S. maltophilia formed well-integrated biofilms with P. aeruginosa, and these organisms colocalize in the lung during dual-species infection. Based on these results, we conclude that active cellular processes by P. aeruginosa afford a significant benefit to S. maltophilia during polymicrobial infections. Furthermore, these results indicate that S. maltophilia may have clinical significance in respiratory infections.


Assuntos
Coinfecção/microbiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Interações Microbianas , Pneumonia Bacteriana/microbiologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Stenotrophomonas maltophilia/crescimento & desenvolvimento , Animais , Carga Bacteriana , Peso Corporal , Líquido da Lavagem Broncoalveolar/microbiologia , Coinfecção/patologia , Modelos Animais de Doenças , Infecções por Bactérias Gram-Negativas/patologia , Imunidade Inata , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Pneumonia Bacteriana/patologia
7.
J Infect Dis ; 220(9): 1399-1405, 2019 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-31369673

RESUMO

Bacterial vaginosis (BV) is the most common cause of vaginal discharge. It is associated with an increased risk of preterm delivery, pelvic inflammatory disease, and an increased risk of acquisition of sexually transmitted infections including human immunodeficiency virus (HIV). The epidemiology of BV supports sexual transmission. However, its etiology remains unknown. At the center of the debate is whether BV is caused by a primary pathogen or a polymicrobial consortium of microorganisms that are sexually transmitted. We previously published a conceptual model hypothesizing that BV is initiated by sexual transmission of Gardnerella vaginalis. Critics of this model have iterated that G. vaginalis is found in virginal women and in sexually active women with a normal vaginal microbiota. In addition, colonization does not always lead to BV. However, recent advances in BV pathogenesis research have determined the existence of 13 different species within the genus Gardnerella. It may be that healthy women are colonized by nonpathogenic Gardnerella species, whereas virulent strains are involved in BV development. Based on our results from a recent prospective study, in addition to an extensive literature review, we present an updated conceptual model for the pathogenesis of BV that centers on the roles of virulent strains of G. vaginalis, as well as Prevotella bivia and Atopobium vaginae.


Assuntos
Actinobacteria/crescimento & desenvolvimento , Gardnerella vaginalis/crescimento & desenvolvimento , Prevotella/crescimento & desenvolvimento , Vagina/microbiologia , Vaginose Bacteriana/fisiopatologia , Actinobacteria/patogenicidade , Feminino , Gardnerella vaginalis/patogenicidade , Humanos , Modelos Biológicos , Prevotella/patogenicidade , Virulência
8.
J Immunol ; 199(6): 2020-2029, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28768724

RESUMO

B-1 cells produce natural Abs which provide an integral first line of defense against pathogens while also performing important homeostatic housekeeping functions. In this study, we demonstrate that programmed cell death 1 ligand 2 (PD-L2) regulates the production of natural Abs against phosphorylcholine (PC). Naive PD-L2-deficient (PD-L2-/-) mice produced significantly more PC-reactive IgM and IgA. This afforded PD-L2-/- mice with selectively enhanced protection against PC-expressing nontypeable Haemophilus influenzae, but not PC-negative nontypeable Haemophilus influenzae, relative to wild-type mice. PD-L2-/- mice had significantly increased PC-specific CD138+ splenic plasmablasts bearing a B-1a phenotype, and produced PC-reactive Abs largely of the T15 Id. Importantly, PC-reactive B-1 cells expressed PD-L2 and irradiated chimeras demonstrated that B cell-intrinsic PD-L2 expression regulated PC-specific Ab production. In addition to increased PC-specific IgM, naive PD-L2-/- mice and irradiated chimeras reconstituted with PD-L2-/- B cells had significantly higher levels of IL-5, a potent stimulator of B-1 cell Ab production. PD-L2 mAb blockade of wild-type B-1 cells in culture significantly increased CD138 and Blimp1 expression and PC-specific IgM, but did not affect proliferation. PD-L2 mAb blockade significantly increased IL-5+ T cells in culture. Both IL-5 neutralization and STAT5 inhibition blunted the effects of PD-L2 mAb blockade on B-1 cells. Thus, B-1 cell-intrinsic PD-L2 expression inhibits IL-5 production by T cells and thereby limits natural Ab production by B-1 cells. These findings have broad implications for the development of therapeutic strategies aimed at altering natural Ab levels critical for protection against infectious disease, autoimmunity, allergy, cancer, and atherosclerosis.


Assuntos
Formação de Anticorpos , Linfócitos B/imunologia , Imunoglobulina M/metabolismo , Fosforilcolina/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Linfócitos T/imunologia , Animais , Anticorpos Bloqueadores/farmacologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Homeostase , Imunidade Inata , Interleucina-5/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteína 2 Ligante de Morte Celular Programada 1/imunologia , Sindecana-1/genética , Sindecana-1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Infect Immun ; 86(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30249749

RESUMO

Nontypeable Haemophilus influenzae (NTHi) is an extremely common human pathobiont that persists on the airway mucosal surface within biofilm communities, and our previous work has shown that NTHi biofilm maturation is coordinated by the production and uptake of autoinducer 2 (AI-2) quorum signals. To directly test roles for AI-2 in maturation and maintenance of NTHi biofilms, we generated an NTHi 86-028NP mutant in which luxS transcription was under the control of the xylA promoter (NTHi 86-028NP luxS xylA::luxS), rendering AI-2 production inducible by xylose. Comparison of biofilms under inducing and noninducing conditions revealed a biofilm defect in the absence of xylose, whereas biofilm maturation increased following xylose induction. The removal of xylose resulted in the interruption of luxS expression and biofilm dispersal. Measurement of luxS transcript levels by real-time reverse transcription-PCR (RT-PCR) showed that luxS expression peaked as biofilms matured and waned before dispersal. Transcript profiling revealed significant changes following the induction of luxS, including increased transcript levels for a predicted family 8 glycosyltransferase (NTHI1750; designated gstA); this result was confirmed by real-time RT-PCR. An isogenic NTHi 86-028NP gstA mutant had a biofilm defect, including decreased levels of sialylated matrix and significantly altered biofilm structure. In experimental chinchilla infections, we observed a significant decrease in the number of bacteria in the biofilm population (but not in effusions) for NTHi 86-028NP gstA compared to the parental strain. Therefore, we conclude that AI-2 promotes NTHi biofilm maturation and the maintenance of biofilm integrity, due at least in part to the expression of a probable glycosyltransferase that is potentially involved in the synthesis of the biofilm matrix.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Proteínas de Transporte/metabolismo , Glicosiltransferases/metabolismo , Haemophilus influenzae/metabolismo , Homosserina/análogos & derivados , Lactonas/metabolismo , Animais , Proteínas de Bactérias/genética , Liases de Carbono-Enxofre/genética , Proteínas de Transporte/genética , Chinchila/microbiologia , Perfilação da Expressão Gênica , Glicosiltransferases/genética , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/genética , Homosserina/genética , Homosserina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Mutação , Otite Média/microbiologia , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica , Xilose/metabolismo
10.
Infect Immun ; 85(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28674033

RESUMO

Haemophilus parainfluenzae is a nutritionally fastidious, Gram-negative bacterium with an oropharyngeal/nasopharyngeal carriage niche that is associated with a range of opportunistic infections, including infectious endocarditis and otitis media (OM). These infections are often chronic/recurrent in nature and typically involve bacterial persistence within biofilm communities that are highly resistant to host clearance. This study addresses the primary hypothesis that H. parainfluenzae forms biofilm communities that are important determinants of persistence in vivo The results from in vitro biofilm studies confirmed that H. parainfluenzae formed biofilm communities within which the polymeric matrix was mainly composed of extracellular DNA and proteins. Using a chinchilla OM infection model, we demonstrated that H. parainfluenzae formed surface-associated biofilm communities containing bacterial and host components that included neutrophil extracellular trap (NET) structures and that the bacteria mainly persisted in these biofilm communities. We also used this model to examine the possible interaction between H. parainfluenzae and its close relative Haemophilus influenzae, which is also commonly carried within the same host environments and can cause OM. The results showed that coinfection with H. influenzae promoted clearance of H. parainfluenzae from biofilm communities during OM infection. The underlying mechanisms for bacterial persistence and biofilm formation by H. parainfluenzae and knowledge about the survival defects of H. parainfluenzae during coinfection with H. influenzae are topics for future work.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções por Haemophilus/microbiologia , Haemophilus parainfluenzae/fisiologia , Otite Média/microbiologia , Animais , Antibiose , Chinchila , Modelos Animais de Doenças , Infecções por Haemophilus/patologia , Haemophilus influenzae/crescimento & desenvolvimento , Haemophilus influenzae/fisiologia , Haemophilus parainfluenzae/crescimento & desenvolvimento , Otite Média/patologia
11.
Infect Immun ; 85(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28096183

RESUMO

Even in the vaccine era, Streptococcus pneumoniae (the pneumococcus) remains a leading cause of otitis media, a significant public health burden, in large part because of the high prevalence of nasal colonization with the pneumococcus in children. The primary pneumococcal neuraminidase, NanA, which is a sialidase that catalyzes the cleavage of terminal sialic acids from host glycoconjugates, is involved in both of these processes. Coinfection with influenza A virus, which also expresses a neuraminidase, exacerbates nasal colonization and disease by S. pneumoniae, in part via the synergistic contributions of the viral neuraminidase. The specific role of its pneumococcal counterpart, NanA, in this interaction, however, is less well understood. We demonstrate in a mouse model that NanA-deficient pneumococci are impaired in their ability to cause both nasal colonization and middle ear infection. Coinfection with neuraminidase-expressing influenza virus and S. pneumoniae potentiates both colonization and infection but not to wild-type levels, suggesting an intrinsic role of NanA. Using in vitro models, we show that while NanA contributes to both epithelial adherence and biofilm viability, its effect on the latter is actually independent of its sialidase activity. These data indicate that NanA contributes both enzymatically and nonenzymatically to pneumococcal pathogenesis and, as such, suggest that it is not a redundant bystander during coinfection with influenza A virus. Rather, its expression is required for the full synergism between these two pathogens.


Assuntos
Biofilmes , Vírus da Influenza A/fisiologia , Neuraminidase/metabolismo , Otite Média/microbiologia , Otite Média/virologia , Streptococcus pneumoniae/fisiologia , Simbiose , Animais , Aderência Bacteriana , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Camundongos , Mucosa Nasal/microbiologia , Neuraminidase/genética
12.
J Immunol ; 193(10): 5076-87, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25311807

RESUMO

Infection with influenza A virus can lead to increased susceptibility to subsequent bacterial infection, often with Streptococcus pneumoniae. Given the substantial modification of the lung environment that occurs following pathogen infection, there is significant potential for modulation of immune responses. In this study, we show that infection of mice with influenza virus, followed by the noninvasive EF3030 strain of Streptococcus pneumoniae, leads to a significant decrease in the virus-specific CD8(+) T cell response in the lung. Adoptive-transfer studies suggest that this reduction contributes to disease in coinfected animals. The reduced number of lung effector cells in coinfected animals was associated with increased death, as well as a reduction in cytokine production in surviving cells. Further, cells that retained the ability to produce IFN-γ exhibited a decreased potential for coproduction of TNF-α. Reduced cytokine production was directly correlated with a decrease in the level of mRNA. Negative regulation of cells in the mediastinal lymph node was minimal compared with that present in the lung, supporting a model of selective regulation in the tissue harboring high pathogen burden. These results show that entry of a coinfecting pathogen can have profound immunoregulatory effects on an ongoing immune response. Together, these findings reveal a novel dynamic interplay between concurrently infecting pathogens and the adaptive immune system.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia Pneumocócica/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Carga Bacteriana , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Coinfecção , Feminino , Imunomodulação , Vírus da Influenza A Subtipo H1N1/imunologia , Interferon gama/biossíntese , Interferon gama/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia , Índice de Gravidade de Doença , Streptococcus pneumoniae/imunologia , Análise de Sobrevida , Subpopulações de Linfócitos T/microbiologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Carga Viral
13.
Infect Immun ; 83(1): 239-46, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25348637

RESUMO

Nontypeable Haemophilus influenzae (NTHI) is a common commensal and opportunistic pathogen of the human airways. For example, NTHI is a leading cause of otitis media and is the most common cause of airway infections associated with chronic obstructive pulmonary disease (COPD). These infections are often chronic/recurrent in nature and involve bacterial persistence within biofilm communities that are highly resistant to host clearance. Our previous work has shown that NTHI within biofilms has increased expression of factors associated with oxidative stress responses. The goal of this study was to define the roles of catalase (encoded by hktE) and a bifunctional peroxiredoxin-glutaredoxin (encoded by pdgX) in resistance of NTHI to oxidants and persistence in vivo. Isogenic NTHI strain 86-028NP mutants lacking hktE and pdgX had increased susceptibility to peroxide. Moreover, these strains had persistence defects in the chinchilla infection model for otitis media, as well as in a murine model for COPD. Additional work showed that pdgX and hktE were important determinants of NTHI survival within neutrophil extracellular traps (NETs), which we have shown to be an integral part of NTHI biofilms in vivo. Based on these data, we conclude that catalase and peroxiredoxin-glutaredoxin are determinants of bacterial persistence during chronic/recurrent NTHI infections that promote bacterial survival within NETs.


Assuntos
Catalase/metabolismo , Tolerância a Medicamentos , Glutarredoxinas/metabolismo , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/enzimologia , Oxidantes/toxicidade , Peroxirredoxinas/metabolismo , Animais , Catalase/genética , Chinchila , Modelos Animais de Doenças , Deleção de Genes , Glutarredoxinas/genética , Haemophilus influenzae/genética , Viabilidade Microbiana/efeitos dos fármacos , Otite Média/microbiologia , Oxidantes/metabolismo , Peroxirredoxinas/genética
14.
PLoS Pathog ; 9(5): e1003377, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696740

RESUMO

Pili of pathogenic Neisseria are major virulence factors associated with adhesion, twitching motility, auto-aggregation, and DNA transformation. Pili of N. meningitidis are subject to several different post-translational modifications. Among these pilin modifications, the presence of phosphorylcholine (ChoP) and a glycan on the pilin protein are phase-variable (subject to high frequency, reversible on/off switching of expression). In this study we report the location of two ChoP modifications on the C-terminus of N. meningitidis pilin. We show that the surface accessibility of ChoP on pili is affected by phase variable changes to the structure of the pilin-linked glycan. We identify for the first time that the platelet activating factor receptor (PAFr) is a key, early event receptor for meningococcal adherence to human bronchial epithelial cells and tissue, and that synergy between the pilin-linked glycan and ChoP post-translational modifications is required for pili to optimally engage PAFr to mediate adherence to human airway cells.


Assuntos
Aderência Bacteriana , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Neisseria meningitidis/metabolismo , Processamento de Proteína Pós-Traducional , Mucosa Respiratória/metabolismo , Linhagem da Célula , Membrana Celular/microbiologia , Células Epiteliais/microbiologia , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Humanos , Neisseria meningitidis/genética , Neisseria meningitidis/patogenicidade , Fosforilcolina/metabolismo , Mucosa Respiratória/microbiologia
15.
J Infect Dis ; 209(1): 87-97, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23964109

RESUMO

The efficacy of different vaccines in protecting elderly individuals against Streptococcus pneumoniae infections is not clear. In the current study, aged mice (22-25 months old) exhibited significantly increased susceptibility to respiratory infection with serotype 3 S. pneumoniae relative to younger adult mice, regardless of whether mice were naive or immunized with native pneumococcal polysaccharide (PPS; Pneumovax23) or protein-PPS conjugate (Prevnar-13) vaccines. Nonetheless, Pneumovax-immunized aged mice developed limited bacteremia following respiratory challenge and exhibited significantly increased survival following systemic challenge relative to Prevnar-immune aged mice and young mice that had received either vaccine. This was explained by >10-fold increases in PPS-specific immunoglobulin G (IgG) levels in Pneumovax-immunized aged mice relative to other groups. Remarkably, PPS3-specific B-cell expansion, IgG switching, plasmablast differentiation, and spleen and bone marrow antibody-secreting cell frequencies were 10-fold higher in aged mice following Pneumovax immunization relative to young mice, due to significantly increased B-1b cell participation. In summary, this study highlights (1) the need to devise strategies to enhance respiratory immunity in aged populations, (2) the diverse responses young and aged populations generate to Pneumovax vs Prevnar vaccines, and (3) the potential value of exploiting B-1b cell responses in aged individuals for increased vaccine efficacy.


Assuntos
Subpopulações de Linfócitos B/imunologia , Vacinas Pneumocócicas/imunologia , Pneumonia Pneumocócica/imunologia , Fatores Etários , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Suscetibilidade a Doenças/imunologia , Imunidade Humoral/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Pneumocócicas/farmacologia , Polissacarídeos Bacterianos/imunologia
16.
Infect Immun ; 82(11): 4802-12, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25156728

RESUMO

Streptococcus pneumoniae (pneumococcus) is both a widespread nasal colonizer and a leading cause of otitis media, one of the most common diseases of childhood. Pneumococcal phase variation influences both colonization and disease and thus has been linked to the bacteria's transition from colonizer to otopathogen. Further contributing to this transition, coinfection with influenza A virus has been strongly associated epidemiologically with the dissemination of pneumococci from the nasopharynx to the middle ear. Using a mouse infection model, we demonstrated that coinfection with influenza virus and pneumococci enhanced both colonization and inflammatory responses within the nasopharynx and middle ear chamber. Coinfection studies were also performed using pneumococcal populations enriched for opaque or transparent phase variants. As shown previously, opaque variants were less able to colonize the nasopharynx. In vitro, this phase also demonstrated diminished biofilm viability and epithelial adherence. However, coinfection with influenza virus ameliorated this colonization defect in vivo. Further, viral coinfection ultimately induced a similar magnitude of middle ear infection by both phase variants. These data indicate that despite inherent differences in colonization, the influenza A virus exacerbation of experimental middle ear infection is independent of the pneumococcal phase. These findings provide new insights into the synergistic link between pneumococcus and influenza virus in the context of otitis media.


Assuntos
Vírus da Influenza A , Nariz/microbiologia , Infecções por Orthomyxoviridae/complicações , Otite Média/microbiologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/fisiologia , Animais , Portador Sadio , Coinfecção , Camundongos , Otite Média/complicações , Infecções Pneumocócicas/complicações
17.
Phys Biol ; 12(1): 016003, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25473880

RESUMO

Host-to-host variability with respect to interactions between microorganisms and multicellular hosts are commonly observed in infection and in homeostasis. However, the majority of mechanistic models used to analyze host-microorganism relationships, as well as most of the ecological theories proposed to explain coevolution of hosts and microbes, are based on averages across a host population. By assuming that observed variations are random and independent, these models overlook the role of differences between hosts. Here, we analyze mechanisms underlying host-to-host variations of bacterial infection kinetics, using the well characterized experimental infection model of polymicrobial otitis media (OM) in chinchillas, in combination with population dynamic models and a maximum entropy (MaxEnt) based inference scheme. We find that the nature of the interactions between bacterial species critically regulates host-to-host variations in these interactions. Surprisingly, seemingly unrelated phenomena, such as the efficiency of individual bacterial species in utilizing nutrients for growth, and the microbe-specific host immune response, can become interdependent in a host population. The latter finding suggests a potential mechanism that could lead to selection of specific strains of bacterial species during the coevolution of the host immune response and the bacterial species.


Assuntos
Infecções Bacterianas/veterinária , Chinchila/microbiologia , Coinfecção/veterinária , Otite Média/veterinária , Animais , Infecções Bacterianas/epidemiologia , Coinfecção/epidemiologia , Fenômenos Ecológicos e Ambientais , Modelos Biológicos , Otite Média/epidemiologia , Dinâmica Populacional
18.
bioRxiv ; 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-37066231

RESUMO

Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long respiratory mucosal infections caused by a diverse array of opportunists, and these infections are a leading cause of morbidity and mortality for pwCF. In recent years, there has been increased appreciation for the range and diversity of microbes in CF-related respiratory infections. Introduction of new therapeutics and improved detection methodology has revealed CF related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species that is widely distributed in the environment and has been increasingly observed in sputa and other samples from pwCF; typically Ax infections occur in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors involved in flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized CFBE14o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization. Ax colonized and persisted in mouse lung for up to 72 hours post infection, with inflammatory consequences that include increased neutrophilia, lung damage, cytokine production, and mortality. Transcript profiling reveled differential expression of Ax genes during growth in SCFM2 synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.

19.
bioRxiv ; 2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37503051

RESUMO

Stenotrophomonas maltophilia is a Gram-negative emerging opportunistic pathogen often found in respiratory diseases such as cystic fibrosis (CF). Patients with CF experience lifelong polymicrobial infections of the respiratory mucosa. Our prior work showed that P. aeruginosa promotes persistence of S. maltophilia mouse respiratory infections. As is typical for environmental opportunistic pathogens, S. maltophilia has a large genome and a high degree of genetic diversity. In this study, we evaluated the genomic content of S. maltophilia, combining short and long read sequencing to construct complete genomes of 10 clinical isolates which were then compared with the larger phylogeny of S. maltophilia genomic sequence data, and compared colonization/persistence in vivo, alone and in coinfection with P. aeruginosa. We found that while the overall genome size and GC content were fairly consistent, there was considerable variability in arrangement and gene content. Similarly, there was significant variability in S. maltophilia colonization and persistence in vivo in experimental mouse respiratory infection. Ultimately, this study gives us a greater understanding of the genomic diversity of S. maltophilia isolated from patients, and how this genomic diversity relates to interactions with other pulmonary pathogens, and to host disease progression. Identifying the molecular determinants of infection with S. maltophilia can facilitate development of novel antimicrobial strategies for a highly drug-resistant pathogen.

20.
Microbiol Spectr ; 11(1): e0384622, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36472421

RESUMO

Stenotrophomonas maltophilia is an emerging opportunistic respiratory pathogen in people with cystic fibrosis (CF). S. maltophilia is frequently observed in polymicrobial infections, and we have previously shown that Pseudomonas aeruginosa promotes colonization and persistence of S. maltophilia in mouse respiratory infections. In this study, we used host and bacterial RNA sequencing to further understand the molecular underpinnings of this interaction. To evaluate S. maltophilia transcript profiles, we used a recently described method for selective capture of bacterial mRNA transcripts with strain-specific RNA probes. We found that factors associated with the type IV pilus, including the histidine kinase subunit of a chemotactic two-component signaling system (chpA), had increased transcript levels during dual-species infection. Using immortalized CF respiratory epithelial cells, we found that infection with P. aeruginosa increases adherence of S. maltophilia, at least in part due to disruption of epithelial tight junctions. In contrast, an isogenic S. maltophilia chpA mutant strain lacked cooperative adherence to CF epithelia and decreased bacterial burden in vivo in dual-species infections with P. aeruginosa. Similarly, P. aeruginosa lacking elastase (lasB) failed to promote S. maltophilia adherence or bacterial colonization and persistence in vivo. Based on these results, we propose that disruption of lung tissue integrity by P. aeruginosa facilitates adherence of S. maltophilia to the lung epithelia, likely in a type IV pilus-dependent manner. These data lend insight into S. maltophilia colonization and persistence in people in later stages of CF disease and may have implications for interactions with other bacterial opportunists. IMPORTANCE Despite advances in treatment options for people with CF, complications of bacterial infections remain the greatest driver of morbidity and mortality in this patient population. These infections often involve more than one bacterial pathogen, and our understanding of how interspecies interactions impact disease progression is lacking. Previous work in our lab found that two CF pathogens, Stenotrophomonas maltophilia and Pseudomonas aeruginosa, can work together in the lung to cause more severe infection. In the present study, we found that infection with P. aeruginosa promotes persistence of S. maltophilia by interfering with epithelial barrier integrity. Depolarization of the epithelial cell layer by P. aeruginosa-secreted elastase increased S. maltophilia adherence, likely in a type IV pilus-dependent manner. Ultimately, this work sheds light on the molecular mechanisms governing an important multispecies interaction seen in pulmonary diseases such as CF.


Assuntos
Fibrose Cística , Infecções por Bactérias Gram-Negativas , Stenotrophomonas maltophilia , Humanos , Animais , Camundongos , Pseudomonas aeruginosa/genética , Stenotrophomonas maltophilia/genética , Stenotrophomonas maltophilia/metabolismo , Células Epiteliais/microbiologia , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Mucosa Respiratória , Infecções por Bactérias Gram-Negativas/microbiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa