Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 754
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(28): e2408092121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968106

RESUMEN

The multinuclear nonheme iron-dependent oxidases (MNIOs) are a rapidly growing family of enzymes involved in the biosynthesis of ribosomally synthesized, posttranslationally modified peptide natural products (RiPPs). Recently, a secreted virulence factor from nontypeable Haemophilus influenzae (NTHi) was found to be expressed from an operon, which we designate the hvf operon, that also encodes an MNIO. Here, we show by Mössbauer spectroscopy that the MNIO HvfB contains a triiron cofactor. We demonstrate that HvfB works together with HvfC [a RiPP recognition element (RRE)-containing partner protein] to perform six posttranslational modifications of cysteine residues on the virulence factor precursor peptide HvfA. Structural characterization by tandem mass spectrometry and NMR shows that these six cysteine residues are converted to oxazolone and thioamide pairs, similar to those found in the RiPP methanobactin. Like methanobactin, the mature virulence factor, which we name oxazolin, uses these modified residues to coordinate Cu(I) ions. Considering the necessity of oxazolin for host cell invasion by NTHi, these findings point to a key role for copper during NTHi infection. Furthermore, oxazolin and its biosynthetic pathway represent a potential therapeutic target for NTHi.


Asunto(s)
Proteínas Bacterianas , Cobre , Haemophilus influenzae , Oxazolona , Factores de Virulencia , Haemophilus influenzae/metabolismo , Haemophilus influenzae/enzimología , Haemophilus influenzae/genética , Haemophilus influenzae/patogenicidad , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Cobre/metabolismo , Cobre/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Oxazolona/metabolismo , Tioamidas/metabolismo , Tioamidas/química , Hierro/metabolismo , Procesamiento Proteico-Postraduccional , Oxidorreductasas/metabolismo , Oxidorreductasas/genética , Operón , Cisteína/metabolismo
2.
Front Cell Infect Microbiol ; 14: 1397940, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38751999

RESUMEN

Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) are two common respiratory tract pathogens often associated with acute exacerbations in Chronic Obstructive Pulmonary Disease (COPD) as well as with otitis media (OM) in children. Although there is evidence that these pathogens can adopt persistence mechanisms such as biofilm formation, the precise means through which they contribute to disease severity and chronicity remains incompletely understood, posing challenges for their effective eradication. The identification of potential vaccine candidates frequently entails the characterization of the host-pathogen interplay in vitro even though this approach is limited by the fact that conventional models do not permit long term bacterial infections. In the present work, by using air-liquid-interface (ALI) human airway in vitro models, we aimed to recreate COPD-related persistent bacterial infections. In particular, we explored an alternative use of the ALI system consisting in the assembly of an inverted epithelium grown on the basal part of a transwell membrane with the aim to enable the functionality of natural defense mechanisms such as mucociliary clearance and cellular extrusion that are usually hampered during conventional ALI infection experiments. The inversion of the epithelium did not affect tissue differentiation and considerably delayed NTHi or Mcat infection progression, allowing one to monitor host-pathogen interactions for up to three weeks. Notably, the use of these models, coupled with confocal and transmission electron microscopy, revealed unique features associated with NTHi and Mcat infection, highlighting persistence strategies including the formation of intracellular bacterial communities (IBCs) and surface-associated biofilm-like structures. Overall, this study demonstrates the possibility to perform long term host-pathogen investigations in vitro with the aim to define persistence mechanisms adopted by respiratory pathogens and individuate potential new vaccine targets.


Asunto(s)
Biopelículas , Haemophilus influenzae , Moraxella catarrhalis , Infecciones por Moraxellaceae , Moraxella catarrhalis/fisiología , Humanos , Haemophilus influenzae/fisiología , Haemophilus influenzae/patogenicidad , Biopelículas/crecimiento & desarrollo , Infecciones por Moraxellaceae/microbiología , Infección Persistente/microbiología , Interacciones Huésped-Patógeno , Infecciones por Haemophilus/microbiología , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Modelos Biológicos , Infecciones del Sistema Respiratorio/microbiología , Células Epiteliales/microbiología
3.
Infect Immun ; 92(5): e0045323, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38602405

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Haemophilus , Haemophilus influenzae , Virus de la Influenza A , Otitis Media , Animales , Otitis Media/microbiología , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/patogenicidad , Haemophilus influenzae/fisiología , Infecciones por Haemophilus/microbiología , Ratones , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/crecimiento & desarrollo , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/complicaciones , Humanos , Animales Recién Nacidos
4.
J Infect Dis ; 229(6): 1674-1678, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38114092

RESUMEN

Biofilm formation has been suggested to be associated with phenotype changes compared with the planktonic form. We screened 1092 Haemophilus influenzae isolates for their genetic relationships and then selected 29 isolates from different genotypes and phenotypes and tested their ability to form biofilm. Our data showed a higher capacity of nontypable isolates, particularly isolates from respiratory and genital infections to form biofilm, compared with typable isolates. This ability to form biofilm was also correlated with reduced deposition of the complement component C3b on biofilm-involved bacteria. These data suggest that the biofilm formation contributes to the virulence of nontypable H. influenzae.


Asunto(s)
Biopelículas , Infecciones por Haemophilus , Haemophilus influenzae , Haemophilus influenzae/genética , Haemophilus influenzae/fisiología , Haemophilus influenzae/patogenicidad , Biopelículas/crecimiento & desarrollo , Humanos , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/inmunología , Proteínas del Sistema Complemento/inmunología , Genotipo , Virulencia , Fenotipo
5.
J Med Microbiol ; 71(8)2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36006824

RESUMEN

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.


Asunto(s)
Fibrosis Quística , Infecciones por Haemophilus , Haemophilus influenzae/clasificación , Haemophilus influenzae/aislamiento & purificación , Fibrosis Quística/complicaciones , Farmacorresistencia Bacteriana , Genoma Bacteriano , Infecciones por Haemophilus/tratamiento farmacológico , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Humanos , Pruebas de Sensibilidad Microbiana , Factores de Virulencia , Secuenciación Completa del Genoma
6.
Ann Vasc Surg ; 79: 442.e1-442.e4, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34655753

RESUMEN

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.


Asunto(s)
Aneurisma Infectado/microbiología , Aneurisma Roto/microbiología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Arteria Poplítea/microbiología , Anciano de 80 o más Años , Aneurisma Infectado/diagnóstico por imagen , Aneurisma Infectado/cirugía , Aneurisma Roto/diagnóstico por imagen , Aneurisma Roto/cirugía , Antibacterianos/uso terapéutico , Desbridamiento , Femenino , Infecciones por Haemophilus/complicaciones , Infecciones por Haemophilus/diagnóstico , Infecciones por Haemophilus/cirugía , Humanos , Arteria Poplítea/diagnóstico por imagen , Arteria Poplítea/cirugía , Técnicas de Sutura , Resultado del Tratamiento , Procedimientos Quirúrgicos Vasculares
7.
J Immunol Res ; 2021: 2185568, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34765679

RESUMEN

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.


Asunto(s)
Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidad , Lipoproteína(a)/metabolismo , Macrófagos/metabolismo , Proteínas Opsoninas/metabolismo , Fagocitosis/fisiología , Antígenos CD36/metabolismo , Línea Celular , Línea Celular Tumoral , Escherichia coli/patogenicidad , Infecciones por Haemophilus/microbiología , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Macrófagos/microbiología , Células U937
9.
Clin Sci (Lond) ; 135(17): 2067-2083, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34405230

RESUMEN

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.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Infecciones por Haemophilus/enzimología , Haemophilus influenzae/patogenicidad , Macrófagos Alveolares/enzimología , Neumonía Bacteriana/enzimología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Anciano , Animales , Estudios de Casos y Controles , Quimiocina CCL20/metabolismo , Quimiocina CCL3/metabolismo , Modelos Animales de Enfermedad , Femenino , Infecciones por Haemophilus/microbiología , Interacciones Huésped-Patógeno , Humanos , Interleucina-1beta/metabolismo , Macrófagos Alveolares/microbiología , Masculino , Ratones , Persona de Mediana Edad , Neumonía Bacteriana/microbiología , Enfermedad Pulmonar Obstructiva Crónica/microbiología
10.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34344825

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/inmunología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Adhesinas Bacterianas/genética , Pruebas de Aglutinación , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Adhesión Bacteriana , Femenino , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/prevención & control , Haemophilus influenzae/genética , Haemophilus influenzae/inmunología , Inmunización , Interleucina-17/sangre , Ratones Endogámicos BALB C , Nasofaringe/microbiología
11.
mBio ; 12(3): e0078921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34154422

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Variación Genética , Genoma Bacteriano , Haemophilus influenzae/genética , Haemophilus influenzae/metabolismo , Adaptación Fisiológica/genética , Adhesinas Bacterianas/clasificación , Adhesión Bacteriana/genética , Adhesión Bacteriana/fisiología , Biopelículas , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Humanos , Regiones Promotoras Genéticas
12.
Respir Res ; 22(1): 113, 2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879129

RESUMEN

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.


Asunto(s)
Haemophilus influenzae/inmunología , Pulmón/microbiología , Macrófagos/microbiología , Fagocitosis , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Anciano , Estudios de Casos y Controles , Células Cultivadas , Progresión de la Enfermedad , Femenino , Haemophilus influenzae/patogenicidad , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-8/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Fenotipo , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo
13.
Sci Rep ; 11(1): 11, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420151

RESUMEN

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.


Asunto(s)
Conjuntivitis Bacteriana/microbiología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/patogenicidad , Otitis Media/microbiología , Sinusitis/microbiología , Enfermedad Aguda , Niño , Preescolar , Farmacorresistencia Microbiana , Femenino , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/aislamiento & purificación , Humanos , Lactante , Masculino , Pruebas de Sensibilidad Microbiana
14.
PLoS One ; 15(12): e0243942, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33306743

RESUMEN

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.


Asunto(s)
Portador Sano/epidemiología , Enfermedades Nasofaríngeas/epidemiología , Neumonía Neumocócica/epidemiología , Infecciones del Sistema Respiratorio/epidemiología , Portador Sano/microbiología , Niño , Preescolar , Femenino , Haemophilus influenzae/aislamiento & purificación , Haemophilus influenzae/patogenicidad , Humanos , Lactante , Masculino , Microbiota , Moraxella catarrhalis/aislamiento & purificación , Moraxella catarrhalis/patogenicidad , Enfermedades Nasofaríngeas/microbiología , Nasofaringe/microbiología , Nasofaringe/patología , Neumonía Neumocócica/microbiología , Infecciones del Sistema Respiratorio/microbiología , Staphylococcus aureus/aislamiento & purificación , Staphylococcus aureus/patogenicidad , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pneumoniae/patogenicidad
15.
Genes (Basel) ; 11(9)2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32967147

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Infecciones por Haemophilus/genética , Haemophilus influenzae/patogenicidad , Meningitis por Haemophilus/genética , Polimorfismo de Nucleótido Simple/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 9/genética , Adolescente , Niño , Preescolar , Femenino , Frecuencia de los Genes/genética , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Neisseria meningitidis/patogenicidad , Streptococcus pneumoniae/patogenicidad
16.
Int J Mol Sci ; 21(16)2020 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-32785145

RESUMEN

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.


Asunto(s)
Plexo Coroideo/citología , Plexo Coroideo/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/patogenicidad , Interacciones Huésped-Patógeno , Adhesión Bacteriana , Barrera Hematoencefálica , Línea Celular Tumoral , Polaridad Celular , Supervivencia Celular , ADN Bacteriano/genética , Fimbrias Bacterianas , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/genética , Haemophilus influenzae/aislamiento & purificación , Humanos , Meningitis/líquido cefalorraquídeo , Meningitis/microbiología , Virulencia , Factores de Virulencia
18.
IUBMB Life ; 72(10): 2097-2111, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32770825

RESUMEN

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.


Asunto(s)
Antibacterianos/uso terapéutico , Antivirales/uso terapéutico , Infecciones Bacterianas/epidemiología , COVID-19/epidemiología , Pandemias , Neumonía Bacteriana/epidemiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/patogenicidad , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/virología , COVID-19/microbiología , COVID-19/virología , Coinfección , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/patogenicidad , Legionella pneumophila/efectos de los fármacos , Legionella pneumophila/patogenicidad , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/virología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/microbiología , Sistema Respiratorio/patología , Sistema Respiratorio/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad , Streptococcus pyogenes/efectos de los fármacos , Streptococcus pyogenes/patogenicidad , Tratamiento Farmacológico de COVID-19
19.
PLoS One ; 15(8): e0235634, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32760078

RESUMEN

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.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Otitis Media/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Evaluación Preclínica de Medicamentos , Haemophilus influenzae/patogenicidad , Ensayos Analíticos de Alto Rendimiento , Humanos , Hiperplasia/tratamiento farmacológico , Hiperplasia/microbiología , Hiperplasia/patología , Ratones , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/microbiología , Membrana Mucosa/patología , Otitis Media/microbiología , Otitis Media/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Técnicas de Cultivo de Tejidos
20.
Genes (Basel) ; 11(7)2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32650475

RESUMEN

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.


Asunto(s)
Asma/genética , Infecciones por Haemophilus/genética , Infecciones por Moraxellaceae/genética , Polimorfismo de Nucleótido Simple , Receptor Toll-Like 4/genética , Asma/epidemiología , Asma/patología , Niño , Preescolar , Femenino , Infecciones por Haemophilus/epidemiología , Infecciones por Haemophilus/patología , Haemophilus influenzae/patogenicidad , Humanos , Lactante , Masculino , Microbiota , Moraxella catarrhalis/patogenicidad , Infecciones por Moraxellaceae/epidemiología , Infecciones por Moraxellaceae/patología , Cavidad Nasal/microbiología , Faringe/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA