Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 771
Filtrar
1.
Infect Immun ; 92(5): e0045323, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38602405

RESUMO

Nontypeable Haemophilus influenzae (NTHi) is a major otitis media (OM) pathogen, with colonization a prerequisite for disease development. Most acute OM is in children <5 years old, with recurrent and chronic OM impacting hearing and learning. Therapies to prevent NTHi colonization and/or disease are needed, especially for young children. Respiratory viruses are implicated in driving the development of bacterial OM in children. We have developed an infant mouse model of influenza-driven NTHi OM, as a preclinical tool for the evaluation of safety and efficacy of clinical therapies to prevent NTHi colonization and the development of OM. In this model, 100% of infant BALB/cARC mice were colonized with NTHi, and all developed NTHi OM. Influenza A virus (IAV) facilitated the establishment of dense (1 × 105 CFU/mL) and long-lasting (6 days) NTHi colonization. IAV was essential for the development of NTHi OM, with 100% of mice in the IAV/NTHi group developing NTHi OM compared with 8% of mice in the NTHi only group. Histological analysis and cytokine measurements revealed that the inflammation observed in the middle ear of the infant mice with OM reflected inflammation observed in children with OM. We have developed the first infant mouse model of NTHi colonization and OM. This ascension model uses influenza-driven establishment of OM and reflects the clinical pathology of bacterial OM developing after a respiratory virus infection. This model provides a valuable tool for testing therapies to prevent or treat NTHi colonization and disease in young children.


Assuntos
Modelos Animais de Doenças , Infecções por Haemophilus , Haemophilus influenzae , Vírus da Influenza A , Otite Média , Animais , Otite Média/microbiologia , Haemophilus influenzae/crescimento & desenvolvimento , Haemophilus influenzae/patogenicidade , Haemophilus influenzae/fisiologia , Infecções por Haemophilus/microbiologia , Camundongos , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/complicações , Humanos , Animais Recém-Nascidos
2.
J Med Microbiol ; 71(8)2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36006824

RESUMO

Introduction. Haemophilus influenzae is a commensal of the respiratory tract that is frequently present in cystic fibrosis (CF) patients and may cause infection. Antibiotic resistance is well described for CF strains, and virulence factors have been proposed.Hypothesis/Gap. The genetic diversity of H. influenzae strains present in the lungs of persons with CF is largely unknown despite the fact that this organism is considered to be a pathogen in this condition. The aim was to establish the genetic diversity and susceptibility of H. influenzae strains from persons with CF, and to screen the whole genomes of these strains for the presence of antibiotic resistance determinants and proposed virulence factors.Methods. A total of 67 strains, recovered from respiratory samples from persons with CF from the UK (n=1), Poland (n=2), Spain (n=24) and the Netherlands (n=40), were subjected to whole-genome sequencing using Illumina technology and tested for antibiotic susceptibility. Forty-nine of these strains (one per different sequence type) were analysed for encoded virulence factors and resistance determinants.Results. The 67 strains represented 49 different sequence types. Susceptibility testing showed that all strains were susceptible to aztreonam, ciprofloxacin, imipenem and tetracycline. Susceptibility to ampicillin, ampicillin/sulbactam, amoxicillin/clavulanic acid, cefuroxime, cefixime, ceftriaxone, cefepime, meropenem, clarithromycin, co-trimoxazole and levofloxacin ranged from 70.2-98.5%. Only 6/49 strains (12.2%) harboured acquired resistance genes. Mutations associated with a ß-lactamase-negative ampicillin-resistant phenotype were present in four strains (8.2 %). The potential virulence factors, urease, haemoglobin- and haptoglobin-binding protein/carbamate kinase, and OmpP5 (OmpA), were encoded in more than half of the strains. The genes for HMW1, HMW2, H. influenzae adhesin, a IgA-specific serine endopeptidase autotransporter precursor, a TonB-dependent siderophore, an ABC-transporter ATP-binding protein, a methyltransferase, a BolA-family transcriptional regulator, glycosyltransferase Lic2B, a helix-turn-helix protein, an aspartate semialdehyde dehydrogenase and another glycosyltransferase were present in less than half of the strains.Conclusion. The H. influenzae strains showed limited levels of resistance, with the highest being against co-trimoxazole. Sequences encoding a carbamate kinase and a haemoglobin- and haemoglobin-haptoglobin-binding-like protein, a glycosyl transferase and an urease may aid the colonization of the CF lung. The adhesins and other identified putative virulence factors did not seem to be necessary for colonization.


Assuntos
Fibrose Cística , Infecções por Haemophilus , Haemophilus influenzae/classificação , Haemophilus influenzae/isolamento & purificação , Fibrose Cística/complicações , Farmacorresistência Bacteriana , Genoma Bacteriano , Infecções por Haemophilus/tratamento farmacológico , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/patogenicidade , Humanos , Testes de Sensibilidade Microbiana , Fatores de Virulência , Sequenciamento Completo do Genoma
3.
Ann Vasc Surg ; 79: 442.e1-442.e4, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34655753

RESUMO

Mycotic aneurysms arise from infection of an arterial wall secondary to septic emboli from endocarditis. Although rare, most mycotic aneurysms involve the abdominal aorta, with Staphylococcus aureus and Salmonella spp being the most common causative organisms. We report a case of an 81-year-old woman with a ruptured mycotic popliteal aneurysm from Haemophilus influenzae infection.


Assuntos
Aneurisma Infectado/microbiologia , Aneurisma Roto/microbiologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/patogenicidade , Artéria Poplítea/microbiologia , Idoso de 80 Anos ou mais , Aneurisma Infectado/diagnóstico por imagem , Aneurisma Infectado/cirurgia , Aneurisma Roto/diagnóstico por imagem , Aneurisma Roto/cirurgia , Antibacterianos/uso terapêutico , Desbridamento , Feminino , Infecções por Haemophilus/complicações , Infecções por Haemophilus/diagnóstico , Infecções por Haemophilus/cirurgia , Humanos , Artéria Poplítea/diagnóstico por imagem , Artéria Poplítea/cirurgia , Técnicas de Sutura , Resultado do Tratamento , Procedimentos Cirúrgicos Vasculares
4.
J Immunol Res ; 2021: 2185568, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34765679

RESUMO

We recently showed that both nontypeable Haemophilus influenzae (NTHi) and its surface plasminogen- (Plg-) binding proteins interact with lipoprotein(a) (Lp(a)) in a lysine-dependent manner. Because Lp(a) can be taken up by macrophages, we postulated that it serves as an opsonin to enhance phagocytosis of NTHi by macrophages. Based on colony-forming unit (CFU) counts, Lp(a) was found to increase U937 macrophage-mediated phagocytosis of NTHi49247 and NTHi49766 by 34% and 43%, respectively, after 120 min. In contrast, Lp(a) did not enhance phagocytosis of Escherichia coli BL21 or E. coli JM109, which were unable to bind to Lp(a). As with U937 macrophages, Lp(a) was capable of increasing phagocytosis of NTHi49247 by peripheral blood mononuclear cell-derived macrophages. Opsonic phagocytosis by Lp(a) was inhibited by the addition of recombinant kringle IV type 10 (rKIV10), a lysine-binding competitor; moreover, Lp(a) did not increase phagocytosis of NTHi by U937 macrophages that were pretreated with a monoclonal antibody against the scavenger receptor CD36. Taken together, our observation suggests that Lp(a) might serve as a lysine-binding opsonin to assist macrophages in rapid recognition and phagocytosis of NTHi.


Assuntos
Infecções por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidade , Lipoproteína(a)/metabolismo , Macrófagos/metabolismo , Proteínas Opsonizantes/metabolismo , Fagocitose/fisiologia , Antígenos CD36/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Escherichia coli/patogenicidade , Infecções por Haemophilus/microbiologia , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Macrófagos/microbiologia , Células U937
6.
Clin Sci (Lond) ; 135(17): 2067-2083, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34405230

RESUMO

Dipeptidyl peptidase 4 (DPP4) expression is increased in the lungs of chronic obstructive pulmonary disease (COPD). DPP4 is known to be associated with inflammation in various organs, including LPS-induced acute lung inflammation. Since non-typeable Haemophilus influenzae (NTHi) causes acute exacerbations in COPD patients, we examined the contribution of DPP4 in NTHi-induced lung inflammation in COPD. Pulmonary macrophages isolated from COPD patients showed higher expression of DPP4 than the macrophages isolated from normal subjects. In response to NTHi infection, COPD, but not normal macrophages show a further increase in the expression of DPP4. COPD macrophages also showed higher expression of IL-1ß, and CCL3 responses to NTHi than normal, and treatment with DPP4 inhibitor, diprotin A attenuated this response. To examine the contribution of DPP4 in NTHi-induced lung inflammation, COPD mice were infected with NTHi, treated with diprotin A or PBS intraperitoneally, and examined for DPP4 expression, lung inflammation, and cytokine expression. Mice with COPD phenotype showed increased expression of DPP4, which increased further following NTHi infection. DPP4 expression was primarily observed in the infiltrated inflammatory cells. NTHi-infected COPD mice also showed sustained neutrophilic lung inflammation and expression of CCL3, and this was inhibited by DPP4 inhibitor. These observations indicate that enhanced expression of DPP4 in pulmonary macrophages may contribute to sustained lung inflammation in COPD following NTHi infection. Therefore, inhibition of DPP4 may reduce the severity of NTHi-induced lung inflammation in COPD.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Infecções por Haemophilus/enzimologia , Haemophilus influenzae/patogenicidade , Macrófagos Alveolares/enzimologia , Pneumonia Bacteriana/enzimologia , Doença Pulmonar Obstrutiva Crônica/enzimologia , Idoso , Animais , Estudos de Casos e Controles , Quimiocina CCL20/metabolismo , Quimiocina CCL3/metabolismo , Modelos Animais de Doenças , Feminino , Infecções por Haemophilus/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Interleucina-1beta/metabolismo , Macrófagos Alveolares/microbiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Pneumonia Bacteriana/microbiologia , Doença Pulmonar Obstrutiva Crônica/microbiologia
7.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34344825

RESUMO

Nontypeable Haemophilus influenzae (NTHi) is a common cause of localized respiratory tract disease and results in significant morbidity. The pathogenesis of NTHi disease begins with nasopharyngeal colonization, and therefore, the prevention of colonization represents a strategy to prevent disease. The NTHi HMW1 and HMW2 proteins are a family of conserved adhesins that are present in 75 to 80% of strains and have been demonstrated to play a critical role in colonization of the upper respiratory tract in rhesus macaques. In this study, we examined the vaccine potential of HMW1 and HMW2 using a mouse model of nasopharyngeal colonization. Immunization with HMW1 and HMW2 by either the subcutaneous or the intranasal route resulted in a strain-specific antibody response associated with agglutination of bacteria and restriction of bacterial adherence. Despite the specificity of the antibody response, immunization resulted in protection against colonization by both the parent NTHi strain and heterologous strains expressing distinct HMW1 and HMW2 proteins. Pretreatment with antibody against IL-17A eliminated protection against heterologous strains, indicating that heterologous protection is IL-17A dependent. This work demonstrates the vaccine potential of the HMW1 and HMW2 proteins and highlights the importance of IL-17A in protection against diverse NTHi strains.


Assuntos
Adesinas Bacterianas/imunologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/patogenicidade , Adesinas Bacterianas/genética , Testes de Aglutinação , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Aderência Bacteriana , Feminino , Infecções por Haemophilus/imunologia , Infecções por Haemophilus/prevenção & controle , Haemophilus influenzae/genética , Haemophilus influenzae/imunologia , Imunização , Interleucina-17/sangue , Camundongos Endogâmicos BALB C , Nasofaringe/microbiologia
8.
mBio ; 12(3): e0078921, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34154422

RESUMO

Genetic variants arising from within-patient evolution shed light on bacterial adaptation during chronic infection. Contingency loci generate high levels of genetic variation in bacterial genomes, enabling adaptation to the stringent selective pressures exerted by the host. A significant gap in our understanding of phase-variable contingency loci is the extent of their contribution to natural infections. The human-adapted pathogen nontypeable Haemophilus influenzae (NTHi) causes persistent infections, which contribute to underlying disease progression. The phase-variable high-molecular-weight (HMW) adhesins located on the NTHi surface mediate adherence to respiratory epithelial cells and, depending on the allelic variant, can also confer high epithelial invasiveness or hyperinvasion. In this study, we characterize the dynamics of HMW-mediated hyperinvasion in living cells and identify a specific HMW binding domain shared by hyperinvasive NTHi isolates of distinct pathological origins. Moreover, we observed that HMW expression decreased over time by using a longitudinal set of persistent NTHi strains collected from chronic obstructive pulmonary disease (COPD) patients, resulting from increased numbers of simple-sequence repeats (SSRs) downstream of the functional P2hmw1A promoter, which is the one primarily driving HMW expression. Notably, the increased SSR numbers at the hmw1 promoter region also control a phenotypic switch toward lower bacterial intracellular invasion and higher biofilm formation, likely conferring adaptive advantages during chronic airway infection by NTHi. Overall, we reveal novel molecular mechanisms of NTHi pathoadaptation based on within-patient lifestyle switching controlled by phase variation. IMPORTANCE Human-adapted bacterial pathogens have evolved specific mechanisms to colonize their host niche. Phase variation is a contingency strategy to allow adaptation to changing conditions, as phase-variable bacterial loci rapidly and reversibly switch their expression. Several NTHi adhesins are phase variable. These adhesins are required for colonization but also immunogenic, in such a way that bacteria with lower adhesin levels are better equipped to survive an immune response, making their contribution to natural infections unclear. We show here that the major NTHi adhesin HMW1A displays allelic variation, which can drive a phase-variable epithelial hyperinvasion phenotype. Over time, hmw1A phase variation lowers adhesin expression, which controls an NTHi lifestyle switch from high epithelial invasiveness to lower invasion and higher biofilm formation. This reversible loss of function aligns with the previously stated notion that epithelial infection is essential for NTHi infection establishment, but once established, persistence favors gene inactivation, in this case facilitating biofilm growth.


Assuntos
Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Variação Genética , Genoma Bacteriano , Haemophilus influenzae/genética , Haemophilus influenzae/metabolismo , Adaptação Fisiológica/genética , Adesinas Bacterianas/classificação , Aderência Bacteriana/genética , Aderência Bacteriana/fisiologia , Biofilmes , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/patogenicidade , Humanos , Regiões Promotoras Genéticas
9.
Respir Res ; 22(1): 113, 2021 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-33879129

RESUMO

BACKGROUND: Lower airway bacterial colonisation (LABC) in COPD patients is associated with increased exacerbation frequency and faster lung function decline. Defective macrophage phagocytosis in COPD drives inflammation, but how defective macrophage function contributes to exacerbations is not clear. This study investigated the association between macrophage phagocytosis and exacerbation frequency, LABC and clinical parameters. METHODS: Monocyte-derived macrophages (MDM) were generated from 92 stable COPD patients, and at the onset of exacerbation in 39 patients. Macrophages were exposed to fluorescently labelled Haemophilus influenzae or Streptococcus pneumoniae for 4 h, then phagocytosis measured by fluorimetry and cytokine release by ELISA. Sputum bacterial colonisation was measured by PCR. RESULTS: Phagocytosis of H. influenzae was negatively correlated with exacerbation frequency (r = 0.440, p < 0.01), and was significantly reduced in frequent vs. infrequent exacerbators (1.9 × 103 RFU vs. 2.5 × 103 RFU, p < 0.01). There was no correlation for S. pneumoniae. There was no association between phagocytosis of either bacteria with age, lung function, smoking history or treatment with inhaled corticosteroids, or long-acting bronchodilators. Phagocytosis was not altered during an exacerbation, or in the 2 weeks post-exacerbation. In response to phagocytosis, MDM from exacerbating patients showed increased release of CXCL-8 (p < 0.001) and TNFα (p < 0.01) compared to stable state. CONCLUSION: Impaired COPD macrophage phagocytosis of H. influenzae, but not S. pneumoniae is associated with exacerbation frequency, resulting in pro-inflammatory macrophages that may contribute to disease progression. Targeting these frequent exacerbators with drugs that improve macrophage phagocytosis may prove beneficial.


Assuntos
Haemophilus influenzae/imunologia , Pulmão/microbiologia , Macrófagos/microbiologia , Fagocitose , Doença Pulmonar Obstrutiva Crônica/microbiologia , Idoso , Estudos de Casos e Controles , Células Cultivadas , Progressão da Doença , Feminino , Haemophilus influenzae/patogenicidade , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-8/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Fenótipo , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Fator de Necrose Tumoral alfa/metabolismo
10.
Sci Rep ; 11(1): 11, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420151

RESUMO

Haemophilus influenzae is a predominant pathogen for conjunctivitis, acute otitis media and acute bacterial paranasal sinusitis in children. We undertook this study to investigate the possible association among these diseases. Children younger than 18-year-old with a diagnosis of bacterial conjunctivitis plus acute otitis media and/or acute bacterial paranasal sinusitis during 2009-2018 were included. Sampling for bacterial cultures was obtained from the lower palpebral conjunctiva and/or ear discharge with cotton-tipped swabs. A total of 67 children were recruited and the age was 29.5 (± 22.4) months in average. Fifty-seven children had conjunctivitis-otitis media syndrome and eight of them had a concurrent diagnosis of acute paranasal sinusitis. Ten children had conjunctivitis and acute paranasal sinusitis simultaneously. Clusters in household were observed in 50.7% children. Most common isolates were Haemophilus influenzae (70%), Moraxella catarrhalis (18%), and Staphylococcus aureus (8%). Antibiotic resistance rate of H. influenzae was 80% for ampicillin, 18% for amoxicillin-clavulanate, and 11% for the second or third-generation cephalosporins. Apart from well-known conjunctivitis-otitis media syndrome, acute paranasal sinusitis may also be linked to conjunctivitis with a similar pathogenic process. Simultaneous presence of these infections may guide the choice of empiric antibiotics toward H. influenzae.


Assuntos
Conjuntivite Bacteriana/microbiologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/patogenicidade , Otite Média/microbiologia , Sinusite/microbiologia , Doença Aguda , Criança , Pré-Escolar , Resistência Microbiana a Medicamentos , Feminino , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/isolamento & purificação , Humanos , Lactente , Masculino , Testes de Sensibilidade Microbiana
11.
PLoS One ; 15(12): e0243942, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33306743

RESUMO

Some children are more susceptible to viral and bacterial respiratory infections in the first few years of life than others. However, the factors contributing to this susceptibility are incompletely understood. In a retrospective analysis of clinical samples collected from a prospectively-enrolled cohort of 358 children we sought associations between physician-attended illness visits and bacterial colonization in the first five years of life. A subset of children was identified by unsupervised clustering analysis as infection and allergy prone (IAP). Several respiratory infection- and allergy-mediated illnesses co-occurred at higher rates in IAP children, while the rates of other illnesses were not significantly different between the groups. Analyses of nasopharyngeal (NP) pathobionts and microbiota commensals showed that early age of first colonization with pathobionts Streptococcus pneumonia, non-typeable Haemophilus influenzae, and Moraxella catarrhalis was associated with IAP children, and particularly Moraxella abundance was negatively associated with NP microbiome diversity. We conclude that mucosal pathobiont exposures in early life can influence susceptibility to respiratory illnesses in children.


Assuntos
Portador Sadio/epidemiologia , Doenças Nasofaríngeas/epidemiologia , Pneumonia Pneumocócica/epidemiologia , Infecções Respiratórias/epidemiologia , Portador Sadio/microbiologia , Criança , Pré-Escolar , Feminino , Haemophilus influenzae/isolamento & purificação , Haemophilus influenzae/patogenicidade , Humanos , Lactente , Masculino , Microbiota , Moraxella catarrhalis/isolamento & purificação , Moraxella catarrhalis/patogenicidade , Doenças Nasofaríngeas/microbiologia , Nasofaringe/microbiologia , Nasofaringe/patologia , Pneumonia Pneumocócica/microbiologia , Infecções Respiratórias/microbiologia , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/patogenicidade , Streptococcus pneumoniae/isolamento & purificação , Streptococcus pneumoniae/patogenicidade
12.
Genes (Basel) ; 11(9)2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32967147

RESUMO

Bacterial meningitis (BM) is a severe disease caused by various bacterial pathogens. Toll-like receptors (TLRs) protect humans from invading pathogens. In this study, we determined whether single nucleotide polymorphisms (SNPs) of TLR4 and TLR9 are associated with susceptibility to and outcome of BM in Angolan children. Samples were taken from 241 patients and 265 age-matched ethnic controls. The SNPs TLR4 rs4986790 (896A > G) and TLR9 rs187084 (-1486T > C) were determined by high-resolution melting analysis (HRMA). The frequency of variant genotypes in TLR4 was significantly higher in patients with Haemophilus influenzae meningitis than controls (odds ratio (OR), 2.5; 95% confidence interval (CI), 1.2-5.4; p = 0.021), whereas the frequency of variant genotypes in TLR9 was significantly lower in patients with H. influenzae meningitis than controls (OR, 0.4; 95% CI, 0.2-0.9; p = 0.036). No such differences were found with other causative pathogens, such as Streptococcus pneumoniae and Neisseria meningitidis. At the time of discharge, patients with meningitis caused by Gram-negative bacteria who were carriers of variant TLR4 genotypes had a higher risk of ataxia (OR, 12.91; 95% CI, 1.52-109.80; p = 0.019) and other neurological sequelae (OR, 11.85; 95% CI, 1.07-131.49; p = 0.044) than those with the wild-type TLR4 genotype. Our study suggests an association between H. influenzae meningitis and genetic variation between TLR4 and TLR9 in Angolan children.


Assuntos
Predisposição Genética para Doença/genética , Infecções por Haemophilus/genética , Haemophilus influenzae/patogenicidade , Meningite por Haemophilus/genética , Polimorfismo de Nucleotídeo Único/genética , Receptor 4 Toll-Like/genética , Receptor Toll-Like 9/genética , Adolescente , Criança , Pré-Escolar , Feminino , Frequência do Gene/genética , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Neisseria meningitidis/patogenicidade , Streptococcus pneumoniae/patogenicidade
14.
PLoS One ; 15(8): e0235634, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32760078

RESUMO

Otitis media, the most common disease of childhood, is characterized by extensive changes in the morphology of the middle ear cavity. This includes hyperplasia of the mucosa that lines the tympanic cavity, from a simple monolayer of squamous epithelium into a greatly thickened, respiratory-type mucosa. The processes that control this response, which is critical to otitis media pathogenesis and recovery, are incompletely understood. Given the central role of protein phosphorylation in most intracellular processes, including cell proliferation and differentiation, we screened a library of kinase inhibitors targeting members of all the major families in the kinome for their ability to influence the growth of middle ear mucosal explants in vitro. Of the 160 inhibitors, 30 were found to inhibit mucosal growth, while two inhibitors enhanced tissue proliferation. The results suggest that the regulation of infection-mediated tissue growth in the ME mucosa involves multiple cellular processes that span the kinome. While some of the pathways and processes identified have been previously implicated in mucosa hyperplasia others are novel. The results were used to generate a global model of growth regulation by kinase pathways. The potential for therapeutic applications of the results are discussed.


Assuntos
Proliferação de Células/efeitos dos fármacos , Otite Média/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Avaliação Pré-Clínica de Medicamentos , Haemophilus influenzae/patogenicidade , Ensaios de Triagem em Larga Escala , Humanos , Hiperplasia/tratamento farmacológico , Hiperplasia/microbiologia , Hiperplasia/patologia , Camundongos , Mucosa/efeitos dos fármacos , Mucosa/microbiologia , Mucosa/patologia , Otite Média/microbiologia , Otite Média/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Técnicas de Cultura de Tecidos
15.
IUBMB Life ; 72(10): 2097-2111, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32770825

RESUMO

The pandemic coronavirus disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has affected millions of people worldwide. To date, there are no proven effective therapies for this virus. Efforts made to develop antiviral strategies for the treatment of COVID-19 are underway. Respiratory viral infections, such as influenza, predispose patients to co-infections and these lead to increased disease severity and mortality. Numerous types of antibiotics such as azithromycin have been employed for the prevention and treatment of bacterial co-infection and secondary bacterial infections in patients with a viral respiratory infection (e.g., SARS-CoV-2). Although antibiotics do not directly affect SARS-CoV-2, viral respiratory infections often result in bacterial pneumonia. It is possible that some patients die from bacterial co-infection rather than virus itself. To date, a considerable number of bacterial strains have been resistant to various antibiotics such as azithromycin, and the overuse could render those or other antibiotics even less effective. Therefore, bacterial co-infection and secondary bacterial infection are considered critical risk factors for the severity and mortality rates of COVID-19. Also, the antibiotic-resistant as a result of overusing must be considered. In this review, we will summarize the bacterial co-infection and secondary bacterial infection in some featured respiratory viral infections, especially COVID-19.


Assuntos
Antibacterianos/uso terapêutico , Antivirais/uso terapêutico , Infecções Bacterianas/epidemiologia , COVID-19/epidemiologia , Pandemias , Pneumonia Bacteriana/epidemiologia , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/patogenicidade , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/microbiologia , Infecções Bacterianas/virologia , COVID-19/microbiologia , COVID-19/virologia , Coinfecção , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/patogenicidade , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/patogenicidade , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/virologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/microbiologia , Sistema Respiratório/patologia , Sistema Respiratório/virologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/patogenicidade , Tratamento Farmacológico da COVID-19
16.
Int J Mol Sci ; 21(16)2020 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-32785145

RESUMO

Non-typeable Haemophilus influenzae (NTHI) is a pathogen of the human respiratory tract causing the majority of invasive H. influenzae infections. Severe invasive infections such as septicemia and meningitis occur rarely, but the lack of a protecting vaccine and the increasing antibiotic resistance of NTHI impede treatment and emphasize its relevance as a potential meningitis causing pathogen. Meningitis results from pathogens crossing blood-brain barriers and invading the immune privileged central nervous system (CNS). In this study, we addressed the potential of NTHI to enter the brain by invading cells of the choroid plexus (CP) prior to meningeal inflammation to enlighten NTHI pathophysiological mechanisms. A cell culture model of human CP epithelial cells, which form the blood-cerebrospinal fluid barrier (BCSFB) in vivo, was used to analyze adhesion and invasion by immunofluorescence and electron microscopy. NTHI invade CP cells in vitro in a polar fashion from the blood-facing side. Furthermore, NTHI invasion rates are increased compared to encapsulated HiB and HiF strains. Fimbriae occurrence attenuated adhesion and invasion. Thus, our findings underline the role of the BCSFB as a potential entry port for NTHI into the brain and provide strong evidence for a function of the CP during NTHI invasion into the CNS during the course of meningitis.


Assuntos
Plexo Corióideo/citologia , Plexo Corióideo/microbiologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Infecções por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidade , Interações Hospedeiro-Patógeno , Aderência Bacteriana , Barreira Hematoencefálica , Linhagem Celular Tumoral , Polaridade Celular , Sobrevivência Celular , DNA Bacteriano/genética , Fímbrias Bacterianas , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/genética , Haemophilus influenzae/isolamento & purificação , Humanos , Meningite/líquido cefalorraquidiano , Meningite/microbiologia , Virulência , Fatores de Virulência
17.
mSphere ; 5(4)2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669474

RESUMO

Moraxella catarrhalis, Streptococcus pneumoniae, and nontypeable Haemophilus influenzae (NTHi) are ubiquitous upper respiratory opportunistic pathogens. Together, these three microbes are the most common causative bacterial agents of pediatric otitis media (OM) and have therefore been characterized as the primary human otopathogens. OM is the most prevalent bacterial infection in children and the primary reason for antibiotic administration in this population. Moreover, biofilm formation has been confirmed as a primary mechanism of chronic and recurrent OM disease. As bacterial biofilms are inherently metabolically recalcitrant to most antibiotics and these complex structures also present a significant challenge to the immune system, there is a clear need to identify novel antimicrobial approaches to treat OM infections. In this study, we evaluated the potential efficacy of antibacterial photodynamic therapy (aPDT) with the photosensitizer chlorin e6 (Ce6) against planktonic as well as biofilm-associated M. catarrhalis, S. pneumoniae, and NTHi. Our data indicate aPDT with Ce6 elicits significant bactericidal activity against both planktonic cultures and established biofilms formed by the three major otopathogens (with an efficacy of ≥99.9% loss of viability). Notably, the implementation of a novel, dual-treatment aPDT protocol resulted in this disinfectant effect on biofilm-associated bacteria and, importantly, inhibited bacterial regrowth 24 h posttreatment. Taken together, these data suggest this novel Ce6-aPDT treatment may be a powerful and innovative therapeutic strategy to effectively treat and eradicate bacterial OM infections and, significantly, prevent the development of recurrent disease.IMPORTANCE Otitis media (OM), or middle ear disease, is the most prevalent bacterial infection in children and the primary reason for antibiotic use and surgical intervention in the pediatric population. Biofilm formation by the major bacterial otopathogens, Moraxella catarrhalis, Streptococcus pneumoniae, and nontypeable Haemophilus influenzae, has been shown to occur within the middle ears of OM patients and is a key factor in the development of recurrent disease, which may result in hearing impairment and developmental delays. Bacterial biofilms are inherently impervious to most antibiotics and present a significant challenge to the immune system. In this study, we demonstrate that antimicrobial photodynamic therapy (aPDT) using the photosensitizer chlorin e6 elicits significant bactericidal activity versus planktonic and biofilm-associated otopathogens and supports further analyses of this novel, efficacious, and promising technology as an adjunctive treatment for acute and recurrent OM.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Otite Média/microbiologia , Fotoquimioterapia , Porfirinas/farmacologia , Bactérias/classificação , Bactérias/patogenicidade , Clorofilídeos , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/patogenicidade , Humanos , Viabilidade Microbiana/efeitos dos fármacos , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/patogenicidade , Otite Média/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade
18.
Microb Genom ; 6(8)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32706329

RESUMO

Nontypeable Haemophilus influenzae (NTHi) colonizes human upper respiratory airways and plays a key role in the course and pathogenesis of acute exacerbations of chronic obstructive pulmonary disease (COPD). Currently, it is not possible to distinguish COPD isolates of NTHi from other clinical isolates of NTHi using conventional genotyping methods. Here, we analysed the core and accessory genome of 568 NTHi isolates, including 40 newly sequenced isolates, to look for genetic distinctions between NTHi isolates from COPD with respect to other illnesses, including otitis media, meningitis and pneumonia. Phylogenies based on polymorphic sites in the core-genome did not show discrimination between NTHi strains collected from different clinical phenotypes. However, pan-genome-wide association studies identified 79 unique NTHi accessory genes that were significantly associated with COPD. Furthermore, many of the COPD-related NTHi genes have known or predicted roles in virulence, transmembrane transport of metal ions and nutrients, cellular respiration and maintenance of redox homeostasis. This indicates that specific genes may be required by NTHi for its survival or virulence in the COPD lung. These results advance our understanding of the pathogenesis of NTHi infection in COPD lungs.


Assuntos
Infecções por Haemophilus/microbiologia , Haemophilus influenzae , Doença Pulmonar Obstrutiva Crônica/microbiologia , Virulência/genética , Genoma Bacteriano , Estudo de Associação Genômica Ampla , Haemophilus influenzae/genética , Haemophilus influenzae/patogenicidade , Humanos , Meningite/microbiologia , Otite/microbiologia , Fenótipo , Pneumonia/microbiologia
19.
Infect Immun ; 88(10)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32719154

RESUMO

Haemophilus influenzae is a Gram-negative bacterium that can be classified into typeable (types a through f) and nontypeable (NTHi) groups. This opportunistic pathogen asymptomatically colonizes the mucosal epithelium of the upper respiratory tract, from where it spreads to other neighboring regions, potentially leading to disease. Infection with NTHi can cause otitis media, sinusitis, conjunctivitis, exacerbations of chronic obstructive pulmonary disease, and pneumonia, but it is increasingly causing invasive disease, including bacteremia and meningitis. Invasive NTHi strains are more resistant to complement-mediated killing. However, the mechanisms of complement resistance have never been studied in large numbers of invasive NTHi strains. In this study, we determined the relationship between binding of IgG or IgM and the bacterial survival in normal human serum for 267 invasive H. influenzae strains from Spain, Portugal, and the Netherlands, of which the majority (200 [75%]) were NTHi. NTHi bacteria opsonized with high levels of IgM had the lowest survival in human serum. IgM binding to the bacterial surface, but not IgG binding, was shown to be associated with complement-mediated killing of NTHi strains. We conclude that evasion of IgM binding by NTHi strains increases survival in blood, thereby potentially contributing to their ability to cause severe invasive diseases.


Assuntos
Proteínas do Sistema Complemento/imunologia , Infecções por Haemophilus/imunologia , Haemophilus influenzae/imunologia , Imunoglobulina M/imunologia , Adulto , Idoso , Ativação do Complemento , Europa (Continente)/epidemiologia , Feminino , Infecções por Haemophilus/epidemiologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/isolamento & purificação , Haemophilus influenzae/patogenicidade , Humanos , Evasão da Resposta Imune , Imunoglobulina G/imunologia , Masculino , Viabilidade Microbiana , Pessoa de Meia-Idade , Soro/microbiologia
20.
Genes (Basel) ; 11(7)2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32650475

RESUMO

We aimed to explore the role of TLR4 (rs4986790) polymorphism in the nasopharyngeal (NP) bacterial colonization and its consequent impact on the development of childhood asthma. A semi-quantitative culture of NP swabs was performed on 473 children at 2 months of age and on 213 children at 13 months of age. TLR4 polymorphism was analyzed for 396 children. Children were followed from birth to the age of 7.5 years and the final outcome was physician-diagnosed asthma. The associations between TLR4 genotype, bacterial colonization, and asthma were analyzed. Children with TLR4 AG or GG genotype were more often colonized with Moraxella catarrhalis at 2 months of age (p = 0.009) and Haemophilus influenzae at 13 months of age (p = 0.018). Children who were colonized with H. influenzae at 13 months of age had a significantly higher risk of later development of asthma (p = 0.004). M. catarrhalis or H. Influenzae colonization at 2 months of age or TLR4 genotype Asp299Gly were not associated with the development of childhood asthma. TLR4 Asp299Gly polymorphism was associated with an increased risk of colonization of M. catarrhalis and H. influenzae in children. The colonization with H. influenzae at 13 months of age was associated with a higher risk of later development of childhood asthma.


Assuntos
Asma/genética , Infecções por Haemophilus/genética , Infecções por Moraxellaceae/genética , Polimorfismo de Nucleotídeo Único , Receptor 4 Toll-Like/genética , Asma/epidemiologia , Asma/patologia , Criança , Pré-Escolar , Feminino , Infecções por Haemophilus/epidemiologia , Infecções por Haemophilus/patologia , Haemophilus influenzae/patogenicidade , Humanos , Lactente , Masculino , Microbiota , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/epidemiologia , Infecções por Moraxellaceae/patologia , Cavidade Nasal/microbiologia , Faringe/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...