Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Bull Exp Biol Med ; 176(3): 342-346, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38342813

RESUMO

Dormant forms of causative agents of healthcare-acquired infections Moraxella catarrhalis and Kocuria rhizophila have been obtained. Dormant forms cells retained viability during long-term storage (≈107 CFU/ml after 2 months) under provocative conditions (lack of nutrient sources; temperature 20°C, oxygen access) were characterized by heat resistance, and acquired special ultrastructural organization typical of dormant forms (compacted nucleoid, thickened cell wall). They were also capable of forming alternative phenotypes (dominant and small colony variants) in a new cycle of germination in a fresh medium. These results demonstrate that the dormant forms can be responsible both for survival in the environment and persistence in the host organism.


Assuntos
Micrococcaceae , Moraxella catarrhalis , Moraxella catarrhalis/genética , Moraxella catarrhalis/metabolismo , Fenótipo
2.
Int J Mol Sci ; 24(7)2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37047578

RESUMO

Neutrophil elastase (NE) contributes to innate antibacterial defense at both the intracellular (phagocytosis) and extracellular (degranulation, NETosis) levels. Moraxella catarrhalis, a human respiratory pathogen, can exist in an inflammatory milieu which contains NE. No data are available on the action of NE against M. catarrhalis or on the counteraction of NE-dependent host defenses by this pathogen. Using time-kill assays we found that bacteria are able to survive and replicate in the presence of NE. Transmission electron microscopy and flow cytometry studies with NE-treated bacteria revealed that while NE admittedly destabilizes the outer membrane leaflet, it does not cause cytoplasmic membrane rupture, suggesting that the enzyme does not target components that are essential for cell integrity. Using LC-MS/MS spectroscopy we determined that NE cleaved at least three virulent surface proteins in outer membrane vesicles (OMVs) of M. catarrhalis, including OMP CD, McaP, and TbpA. The cleavage of OMP CD contributes to the significant decrease in resistance to serum complement in the complement-resistant strain Mc6. The cleavage of McaP did not cause any sensitization to erythromycin nor did NE disturb its drug action. Identifying NE as a novel but subtle anti-virulence agent together with its extracellularly not-efficient bactericidal activity against M. catarrhalis may facilitate the pathogen's existence in the airways under inflammation.


Assuntos
Elastase de Leucócito , Moraxella catarrhalis , Humanos , Moraxella catarrhalis/metabolismo , Elastase de Leucócito/metabolismo , Cromatografia Líquida , Proteínas da Membrana Bacteriana Externa/metabolismo , Espectrometria de Massas em Tandem , Bactérias/metabolismo
3.
EMBO J ; 42(6): e112558, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36762431

RESUMO

Moraxella catarrhalis is an important human respiratory pathogen and a major causative agent of otitis media and chronic obstructive pulmonary disease. Toll-like receptors contribute to, but cannot fully account for, the complexity of the immune response seen in M. catarrhalis infection. Using primary mouse bone marrow-derived macrophages to examine the host response to M. catarrhalis infection, our global transcriptomic and targeted cytokine analyses revealed activation of immune signalling pathways by both membrane-bound and cytosolic pattern-recognition receptors. We show that M. catarrhalis and its outer membrane vesicles or lipooligosaccharide (LOS) can activate the cytosolic innate immune sensor caspase-4/11, gasdermin-D-dependent pyroptosis, and the NLRP3 inflammasome in human and mouse macrophages. This pathway is initiated by type I interferon signalling and guanylate-binding proteins (GBPs). We also show that inflammasomes and GBPs, particularly GBP2, are required for the host defence against M. catarrhalis in mice. Overall, our results reveal an essential role for the interferon-inflammasome axis in cytosolic recognition and immunity against M. catarrhalis, providing new molecular targets that may be used to mitigate pathological inflammation triggered by this pathogen.


Assuntos
Caspases , Inflamassomos , Camundongos , Humanos , Animais , Caspases/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Moraxella catarrhalis/metabolismo , Proteínas de Transporte , Imunidade Inata
4.
Am J Respir Cell Mol Biol ; 65(6): 593-602, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34192508

RESUMO

Chronic obstructive pulmonary disease (COPD) is associated with colonization by bacterial pathogens and repeated airway infections, leading to exacerbations and impaired lung function. The highly glycosylated mucins in the mucus lining the airways are an important part of the host defense against pathogens. However, mucus accumulation can contribute to COPD pathology. Here, we examined whether inflammation is associated with glycosylation changes that affect interactions between airway mucins and pathogens. We isolated mucins from lower airway samples (n = 4-9) from long-term smokers with and without COPD and from never-smokers. The most abundant terminal glycan moiety was N-acetylneuraminic acid (Neu5Ac) among smokers with and without COPD and N-acetyl-hexoseamine among never-smokers. Moraxella catarrhalis bound to MUC5 mucins from smokers with and without COPD. M. catarrhalis binding correlated with inflammatory parameters and Neu5Ac content. M. catarrhalis binding was abolished by enzymatic removal of Neu5Ac. Furthermore, M. catarrhalis bound to α2,6 sialyl-lactose, suggesting that α2,6 sialic acid contributes to M. catarrhalis binding to mucins. Furthermore, we detected more M. catarrhalis binding to mucins from patients with pneumonia than to those from control subjects (n = 8-13), and this binding correlated with C-reactive protein and Neu5Ac levels. These results suggest a key role of inflammation-induced Neu5Ac in the adhesion of M. catarrhalis to airway mucins. The inflammation-induced ability of MUC5 mucins to bind M. catarrhalis is likely a host defense mechanism in the healthy lung, although it cannot be excluded that impaired mucociliary clearance limits the effectiveness of this defense in patients with COPD.


Assuntos
Pulmão/metabolismo , Moraxella catarrhalis/metabolismo , Mucina-5B/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Mucosa Respiratória/metabolismo , Humanos , Inflamação , Pulmão/microbiologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Mucosa Respiratória/microbiologia , Ácidos Siálicos/metabolismo
5.
Front Immunol ; 11: 2122, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32983170

RESUMO

Moraxella catarrhalis is a human-specific commensal of the respiratory tract and an opportunistic pathogen. It is one of the leading cause of otitis media in children and of acute exacerbations in patients with chronic obstructive pulmonary disease, resulting in significant morbidity and economic burden. Vaccines and new immunotherapeutic strategies to treat this emerging pathogen are needed. Complement is a key component of innate immunity that mediates the detection, response, and subsequent elimination of invading pathogens. Many pathogens including M. catarrhalis have evolved complement evasion mechanisms, which include the binding of human complement inhibitors such as C4b-binding protein (C4BP) and Factor H (FH). Inhibiting C4BP and FH acquisition by M. catarrhalis may provide a novel therapeutic avenue to treat infections. To achieve this, we created two chimeric proteins that combined the Moraxella-binding domains of C4BP and FH fused to human immunoglobulin Fcs: C4BP domains 1 and 2 and FH domains 6 and 7 fused to IgM and IgG Fc, respectively. As expected, FH6-7/IgG displaced FH from the bacterial surface while simultaneously activating complement via Fc-C1q interactions, together increasing pathogen elimination. C4BP1-2/IgM also increased serum killing of the bacteria through enhanced complement deposition, but did not displace C4BP from the surface of M. catarrhalis. These Fc fusion proteins could act as anti-infective immunotherapies. Many microbes bind the complement inhibitors C4BP and FH through the same domains as M. catarrhalis, therefore these Fc fusion proteins may be promising candidates as adjunctive therapy against many different drug-resistant pathogens.


Assuntos
Proteína de Ligação ao Complemento C4b/farmacologia , Fator H do Complemento/farmacologia , Fragmentos Fc das Imunoglobulinas/farmacologia , Moraxella catarrhalis/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Ligação Competitiva , Atividade Bactericida do Sangue , Células CHO , Complemento C3b/análise , Complemento C3d/análise , Proteína de Ligação ao Complemento C4b/genética , Proteína de Ligação ao Complemento C4b/metabolismo , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Cricetinae , Cricetulus , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Imunoglobulina M/genética , Imunoglobulina M/metabolismo , Imunoglobulina M/farmacologia , Moraxella catarrhalis/metabolismo , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
6.
Mol Cell Proteomics ; 19(3): 518-528, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31941798

RESUMO

Mass spectrometry (MS) and proteomics offer comprehensive characterization and identification of microorganisms and discovery of protein biomarkers that are applicable for diagnostics of infectious diseases. The use of biomarkers for diagnostics is widely applied in the clinic and the use of peptide biomarkers is increasingly being investigated for applications in the clinical laboratory. Respiratory-tract infections are a predominant cause for medical treatment, although, clinical assessments and standard clinical laboratory protocols are time-consuming and often inadequate for reliable diagnoses. Novel methods, preferably applied directly to clinical samples, excluding cultivation steps, are needed to improve diagnostics of infectious diseases, provide adequate treatment and reduce the use of antibiotics and associated development of antibiotic resistance. This study applied nano-liquid chromatography (LC) coupled with tandem MS, with a bioinformatics pipeline and an in-house database of curated high-quality reference genome sequences to identify species-unique peptides as potential biomarkers for four bacterial pathogens commonly found in respiratory tract infections (RTIs): Staphylococcus aureus; Moraxella catarrhalis; Haemophilus influenzae and Streptococcus pneumoniae The species-unique peptides were initially identified in pure cultures of bacterial reference strains, reflecting the genomic variation in the four species and, furthermore, in clinical respiratory tract samples, without prior cultivation, elucidating proteins expressed in clinical conditions of infection. For each of the four bacterial pathogens, the peptide biomarker candidates most predominantly found in clinical samples, are presented. Data are available via ProteomeXchange with identifier PXD014522. As proof-of-principle, the most promising species-unique peptides were applied in targeted tandem MS-analyses of clinical samples and their relevance for identifications of the pathogens, i.e. proteotyping, was validated, thus demonstrating their potential as peptide biomarker candidates for diagnostics of infectious diseases.


Assuntos
Proteínas de Bactérias/metabolismo , Haemophilus influenzae/metabolismo , Moraxella catarrhalis/metabolismo , Peptídeos/metabolismo , Staphylococcus aureus/metabolismo , Streptococcus pneumoniae/metabolismo , Biomarcadores/metabolismo , Haemophilus influenzae/isolamento & purificação , Humanos , Moraxella catarrhalis/isolamento & purificação , Sistema Respiratório/microbiologia , Infecções Respiratórias/microbiologia , Especificidade da Espécie , Staphylococcus aureus/isolamento & purificação , Streptococcus pneumoniae/isolamento & purificação , Espectrometria de Massas em Tandem
7.
Glycobiology ; 30(2): 86-94, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31616921

RESUMO

Moraxella catarrhalis (M. catarrhalis) is a pathogenic gram-negative bacterium that causes otitis media and sinusitis in children. Three major serotypes A, B and C are identified to account for approximately 95% of the clinical isolates. Understanding the conformational properties of different serotypes of M. catarrhalis provides insights into antigenic determinants. In this work, all-atom molecular dynamics simulations were conducted for M. catarrhalis lipooligosaccharide (LOS) bilayer systems and oligosaccharides (OS) in water solution to investigate the conformational similarities and differences of three serotypes. For up to 10 neutral monosaccharides in the core part, the conformational ensembles described by the pair-wise root mean square deviation distributions are similar among the three serotypes of either the LOS or OS. At the central ß-($1\to4$)-linkage, anti-$\psi$ conformation in conjunction with the gauche-gauche (g-) conformation of the central trisubstituted glucosyl residue is observed as the dominant conformation to sustain the structural characteristics of M. catarrhalis three types, which is further supported by calculated transglycosidic ${}^3{J}_{C,H}\Big({\psi}_H\Big)$ of serotype A in comparison to experimental data. Interestingly, the conformational variability of three serotypes is more restricted for the OS in water solution than that in the LOS bilayer systems. The LOS-LOS interactions in the bilayer systems are responsible for the increased conformational diversity despite of tight packing. Solvent-accessible surface area analysis suggests that a trisaccharide attached to the ß-($1\to 6$)-linked sugar in all three serotypes of LOS could be the common epitope and have the possibility to interact with antibodies.


Assuntos
Lipopolissacarídeos/química , Moraxella catarrhalis/química , Oligossacarídeos/química , Sorogrupo , Configuração de Carboidratos , Lipopolissacarídeos/metabolismo , Moraxella catarrhalis/metabolismo , Oligossacarídeos/metabolismo
8.
J Struct Biol ; 208(2): 77-85, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31400508

RESUMO

The gram-negative bacterium Moraxella catarrhalis infects humans exclusively, causing various respiratory tract diseases, including acute otitis media in children, septicaemia or meningitis in adults, and pneumonia in the elderly. To do so, M. catarrhalis expresses virulence factors facilitating its entry and survival in the host. Among them are the ubiquitous surface proteins (Usps): A1, A2, and A2H, which all belong to the trimeric autotransporter adhesin family. They bind extracellular matrix molecules and inhibit the classical and alternative pathways of the complement cascade by recruiting complement regulators C3d and C4b binding protein. Here, we report the 2.5 Šresolution X-ray structure of UspA1299-452, which previous work had suggested contained the canonical C3d binding site found in both UspA1 and UspA2. We show that this fragment of the passenger domain contains part of the long neck domain (residues 299-336) and a fragment of the stalk (residues 337-452). The coiled-coil stalk is left-handed, with 7 polar residues from each chain facing the core and coordinating chloride ions or water molecules. Despite the previous reports of tight binding in serum-based assays, we were not able to demonstrate binding between C3d and UspA1299-452 using ELISA or biolayer interferometry, and the two proteins run separately on size-exclusion chromatography. Microscale thermophoresis suggested that the dissociation constant was 140.5 ±â€¯8.4 µM. We therefore suggest that full-length proteins or other additional factors are important in UspA1-C3d interactions. Other molecules on the bacterial surface or present in serum may enhance binding of those two molecules.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Complemento C3d/química , Complemento C3d/metabolismo , Moraxella catarrhalis/metabolismo , Anisotropia , Sítios de Ligação , Cromatografia em Gel , Cristalografia por Raios X , Ligação Proteica , Estrutura Secundária de Proteína
9.
Eur Respir Rev ; 27(148)2018 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-29950304

RESUMO

Haemophilus influenzae, Moraxella catarrhalis and Pseudomonas aeruginosa are common Gram-negative pathogens associated with an array of pulmonary diseases. All three species have multiple adhesins in their outer membrane, i.e. surface structures that confer the ability to bind to surrounding cells, proteins or tissues. This mini-review focuses on proteins with high affinity for the components of the extracellular matrix such as collagen, laminin, fibronectin and vitronectin. Adhesins are not structurally related and may be lipoproteins, transmembrane porins or large protruding trimeric auto-transporters. They enable bacteria to avoid being cleared together with mucus by attaching to patches of exposed extracellular matrix, or indirectly adhering to epithelial cells using matrix proteins as bridging molecules. As more adhesins are being unravelled, it is apparent that bacterial adhesion is a highly conserved mechanism, and that most adhesins target the same regions on the proteins of the extracellular matrix. The surface exposed adhesins are prime targets for new vaccines and the interactions between proteins are often possible to inhibit with interfering molecules, e.g heparin. In conclusion, this highly interesting research field of microbiology has unravelled host-pathogen interactions with high therapeutic potential.


Assuntos
Adesinas Bacterianas/imunologia , Aderência Bacteriana , Proteínas da Matriz Extracelular/imunologia , Bactérias Aeróbias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Evasão da Resposta Imune , Pulmão/imunologia , Infecções Respiratórias/imunologia , Adesinas Bacterianas/metabolismo , Animais , Proteínas da Matriz Extracelular/metabolismo , Bactérias Aeróbias Gram-Negativas/metabolismo , Bactérias Aeróbias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Haemophilus influenzae/imunologia , Haemophilus influenzae/metabolismo , Haemophilus influenzae/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Moraxella catarrhalis/imunologia , Moraxella catarrhalis/metabolismo , Moraxella catarrhalis/patogenicidade , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Infecções Respiratórias/metabolismo , Infecções Respiratórias/microbiologia
10.
Sci Rep ; 8(1): 4955, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29563531

RESUMO

Moraxella catarrhalis is a common human respiratory tract pathogen. Its virulence factors associated with whole bacteria or outer membrane vesicles (OMVs) aid infection, colonization and may induce specific antibodies. To investigate pathogen-host interactions, we applied integrated bioinformatic and immunoproteomic (2D-electrophoresis, immunoblotting, LC-MS/MS) approaches. We showed that OMV proteins engaged exclusively in complement evasion and colonization strategies, but not those involved in iron transport and metabolism, are major targets for cross-reacting antibodies produced against phylogenetically divergent M. catarrhalis strains. The analysis of 31 complete genomes of M. catarrhalis and other Moraxella revealed that OMV protein-coding genes belong to 64 orthologous groups, five of which are restricted to M. catarrhalis. This species showed a two-fold increase in the number of OMV protein-coding genes relative to its ancestors and animal-pathogenic Moraxella. The appearance of specific OMV factors and the increase in OMV-associated virulence proteins during M. catarrhalis evolution is an interesting example of pathogen adaptation to optimize colonization. This precisely targeted cross-reactive immunity against M. catarrhalis may be an important strategy of host defences to counteract this phenomenon. We demonstrate that cross-reactivity is closely associated with the anti-virulent antibody repertoire which we have linked with adaptation of this pathogen to the host.


Assuntos
Anticorpos Antibacterianos/imunologia , Vesículas Extracelulares/imunologia , Moraxella catarrhalis/imunologia , Infecções por Moraxellaceae/imunologia , Fatores de Virulência/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Parede Celular/imunologia , Parede Celular/metabolismo , Biologia Computacional , Reações Cruzadas/imunologia , Modelos Animais de Doenças , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Genoma Bacteriano/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Moraxella catarrhalis/genética , Moraxella catarrhalis/metabolismo , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/sangue , Infecções por Moraxellaceae/microbiologia , Proteômica , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
Mol Microbiol ; 107(1): 47-56, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29114953

RESUMO

Asymmetry in the outer membrane has long defined the cell envelope of Gram-negative bacteria. This asymmetry, with lipopolysaccharide (LPS) or lipooligosaccharide (LOS) exclusively in the outer leaflet of the membrane, establishes an impermeable barrier that protects the cell from a number of stressors in the environment. Work done over the past 5 years has shown that Acinetobacter baumannii has the remarkable capability to survive with inactivated production of lipid A biosynthesis and the absence of LOS in its outer membrane. The implications of LOS-deficient A. baumannii are far-reaching - from impacts on cell envelope biogenesis and maintenance, bacterial physiology, antibiotic resistance and virulence. This review examines recent work that has contributed to our understanding of LOS-deficiency and compares it to studies done on Neisseria meningitidis and Moraxella catarrhalis; the two other organisms with this capability.


Assuntos
Acinetobacter baumannii/metabolismo , Lipopolissacarídeos/metabolismo , Acinetobacter baumannii/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Endotoxinas/metabolismo , Bactérias Gram-Negativas/metabolismo , Lipídeo A/metabolismo , Lipopolissacarídeos/genética , Moraxella catarrhalis/metabolismo , Neisseria meningitidis/metabolismo , Virulência
12.
Vaccine ; 35(33): 4105-4111, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28668567

RESUMO

BACKGROUND: The effect of pneumococcal vaccination is widely variable when measured by nasopharyngeal carriage of vaccine and non-vaccine targets. The aim of this study was to compare the carriage rates and metabolic activity of Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae and Moraxella catarrhalis among children who were or were not vaccinated with PCV10. METHODS: We included children with acute respiratory infection aged 6-23months from a cross-sectional study (CHIADO-IVAS). Nasopharyngeal aspirates were collected and respiratory pathogens were quantified by nCounter digital transcriptomics (Nanostring) and metagenomic sequencing of 16S ribosomal RNA (Illumina). The metabolic rate was calculated by the ratio between RNA transcripts and 16S DNA reads. RESULTS: Out of the 80 patients in this study, 53 were vaccinated with PCV10 and 27 were unvaccinated. There was no difference in nasopharyngeal carriage rates of S. pneumoniae, S. aureus, H. influenzae or M. catarrhalis by either transcriptomic analysis or 16S metagenomics. However, unvaccinated children presented a higher metabolic rate for S. pneumoniae compared to PCV10-vaccinated children (Median [25-75th percentiles]: 126 [22.75-218.41] vs. 0[0-47.83], p=0.004). Furthermore, unvaccinated children presented a positive correlation between mRNA counts and 16S DNA reads for S. pneumoniae (r=0.707; p<0.001) and H. influenzae (r=0.525; p=0.005), in contrast to vaccinated children. No such effect was observed for S. aureus and M. catarrhalis. CONCLUSIONS: Vaccination by PCV10 exerts a pathogen-specific effect on pneumococcal metabolic rate. Pathogen RNA/DNA ratio might represent a more sensitive readout for vaccine follow-up, as compared to nasopharyngeal carriage.


Assuntos
Portador Sadio/epidemiologia , Haemophilus influenzae/isolamento & purificação , Nasofaringe/microbiologia , Infecções Pneumocócicas/epidemiologia , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/imunologia , Streptococcus pneumoniae/isolamento & purificação , Portador Sadio/microbiologia , Estudos Transversais , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Feminino , Haemophilus influenzae/metabolismo , Humanos , Lactente , Masculino , Moraxella catarrhalis/isolamento & purificação , Moraxella catarrhalis/metabolismo , Vacinas Pneumocócicas/administração & dosagem , Prevalência , Estudos Prospectivos , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/metabolismo , Streptococcus pneumoniae/metabolismo
13.
Microbiol Res ; 202: 71-79, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28647125

RESUMO

Moraxella catarrhalis is becoming an important human respiratory tract pathogen affecting significant proportions from the population. However, still little is known about its physiology and molecular regulation. To this end, the CydDC, which is a heterodimeric ATP binding cassette transporter that has been shown to contribute to the maintenance of the redox homeostasis across the periplasm in other Gram-negative bacteria, is studied here. Amino acids multiple sequence alignments indicated that M. catarrhalis CydC is different from the CydC proteins of the bacterial species in which this system has been previously studied. These findings prompted further interest in studying this system in M. catarrhalis. Isogenic mutant in the CydDC system showed suppression in growth rate, hypersensitivity to oxidative and reductive stress and increased accumulation of intracellular cysteine levels. In addition, the growth of cydC- mutant exhibited hypersensitivity to exogenous cysteine; however, it did not display a significant difference from its wild-type counterpart in the murine pulmonary clearance model. Moreover, a palindrome was detected 94bp upstream of the cydD ORF suggesting it might act as a potential regulatory element. Real-time reverse transcription-PCR analysis showed that deletion/change in the palindrome resulted into alterations in the transcription levels of cydC. A better understanding of such system and its regulation helps in developing better ways to combat M. catarrhalis infections.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Regulação Bacteriana da Expressão Gênica , Sequências Repetidas Invertidas/fisiologia , Moraxella catarrhalis/genética , Fenótipo , Deleção de Sequência , Transportadores de Cassetes de Ligação de ATP/genética , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Cisteína/metabolismo , DNA Recombinante , Escherichia coli/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Sequências Repetidas Invertidas/genética , Camundongos , Moraxella catarrhalis/metabolismo , Oxirredução , Estresse Oxidativo , Periplasma/metabolismo , Alinhamento de Sequência
14.
PLoS One ; 11(7): e0158689, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391026

RESUMO

Moraxella catarrhalis is a human respiratory tract pathogen that causes otitis media (middle ear infections) in children and respiratory tract infections in adults with chronic obstructive pulmonary disease. In view of the huge global burden of disease caused by M. catarrhalis, the development of vaccines to prevent these infections and better approaches to treatment have become priorities. In previous work, we used a genome mining approach that identified three substrate binding proteins (SBPs) of ATP-binding cassette (ABC) transporters as promising candidate vaccine antigens. In the present study, we performed a comprehensive assessment of 19 SBPs of 15 ABC transporter systems in the M. catarrhalis genome by engineering knockout mutants and studying their role in assays that assess mechanisms of infection. The capacity of M. catarrhalis to survive and grow in the nutrient-limited and hostile environment of the human respiratory tract, including intracellular growth, account in part for its virulence. The results show that ABC transporters that mediate uptake of peptides, amino acids, cations and anions play important roles in pathogenesis by enabling M. catarrhalis to 1) grow in nutrient-limited conditions, 2) invade and survive in human respiratory epithelial cells and 3) persist in the lungs in a murine pulmonary clearance model. The knockout mutants of SBPs and ABC transporters showed different patterns of activity in the assay systems, supporting the conclusion that different SBPs and ABC transporters function at different stages in the pathogenesis of infection. These results indicate that ABC transporters are nutritional virulence factors, functioning to enable the survival of M catarrhalis in the diverse microenvironments of the respiratory tract. Based on the role of ABC transporters as virulence factors of M. catarrhalis, these molecules represent potential drug targets to eradicate the organism from the human respiratory tract.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/metabolismo , Moraxella catarrhalis/metabolismo , Moraxella catarrhalis/patogenicidade , Infecções Respiratórias/metabolismo , Células A549 , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Bactérias/genética , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Genoma Bacteriano/genética , Humanos , Camundongos Endogâmicos BALB C , Sistema Respiratório/metabolismo , Sistema Respiratório/microbiologia , Infecções Respiratórias/genética , Virulência
15.
Infect Immun ; 84(2): 432-8, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26597985

RESUMO

Moraxella catarrhalis is an exclusively human pathogen that is an important cause of otitis media in children and lower respiratory tract infections in adults with chronic obstructive pulmonary disease. A vaccine to prevent M. catarrhalis infections would have an enormous global impact in reducing morbidity resulting from these infections. Substrate binding protein 2 (SBP2) of an ABC transporter system has recently been identified as a promising vaccine candidate antigen on the bacterial surface of M. catarrhalis. In this study, we showed that SBP1, -2, and -3 individually bind different basic amino acids with exquisite specificity. We engineered mutants that each expressed a single SBP from this gene cluster and showed in growth experiments that SBP1, -2, and -3 serve a nutritional function through acquisition of amino acids for the bacterium. SBP2 mediates uptake of arginine, a strict growth requirement of M. catarrhalis. Adherence and invasion assays demonstrated that SBP1 and SBP3 play a role in invasion of human respiratory epithelial cells, consistent with a nutritional role in intracellular survival in the human respiratory tract. This work demonstrates that the SBPs of an ABC transporter system function in the uptake of basic amino acids to support growth of M. catarrhalis. The critical role of SBP2 in arginine uptake may contribute to its potential as a vaccine antigen.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Arginina/metabolismo , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Moraxella catarrhalis/crescimento & desenvolvimento , Moraxella catarrhalis/metabolismo , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Humanos , Moraxella catarrhalis/genética , Mutação , Proteínas Recombinantes/metabolismo
16.
J Immunol ; 196(3): 1249-58, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26712944

RESUMO

Moraxella catarrhalis is a respiratory tract pathogen commonly causing otitis media in children and acute exacerbations in patients suffering from chronic obstructive pulmonary disease. Cartilage oligomeric matrix protein (COMP) functions as a structural component in cartilage, as well as a regulator of complement activity. Importantly, COMP is detected in resident macrophages and monocytes, alveolar fluid, and the endothelium of blood vessels in lung tissue. We show that the majority of clinical isolates of M. catarrhalis (n = 49), but not other tested bacterial pathogens, bind large amounts of COMP. COMP interacts directly with the ubiquitous surface protein A2 of M. catarrhalis. Binding of COMP correlates with survival of M. catarrhalis in human serum by inhibiting bactericidal activity of the complement membrane attack complex. Moreover, COMP inhibits phagocytic killing of M. catarrhalis by human neutrophils. We further observed that COMP reduces bacterial adhesion and uptake by human lung epithelial cells, thus protecting M. catarrhalis from intracellular killing by epithelial cells. Taken together, our findings uncover a novel mechanism that M. catarrhalis uses to evade host innate immunity.


Assuntos
Proteína de Matriz Oligomérica de Cartilagem/imunologia , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Moraxella catarrhalis/imunologia , Infecções por Moraxellaceae/imunologia , Aderência Bacteriana/imunologia , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Linhagem Celular , Citometria de Fluxo , Humanos , Moraxella catarrhalis/metabolismo , Infecções por Moraxellaceae/metabolismo
17.
Infect Immun ; 83(9): 3458-69, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26099590

RESUMO

Several bacterial species recruit the complement regulators C4b-binding protein, factor H, and vitronectin, resulting in resistance against the bactericidal activity of human serum. It was recently demonstrated that bacteria also bind plasminogen, which is converted to plasmin that degrades C3b and C5. In this study, we found that a series of clinical isolates (n = 58) of the respiratory pathogen Moraxella catarrhalis, which is commonly isolated from preschool children and adults with chronic obstructive pulmonary disease (COPD), significantly binds human plasminogen. Ubiquitous surface protein A2 (UspA2) and hybrid UspA2 (UspA2H) were identified as the plasminogen-binding factors in the outer membrane proteome of Moraxella. Furthermore, expression of a series of truncated recombinant UspA2 and UspA2H proteins followed by a detailed analysis of protein-protein interactions suggested that the N-terminal head domains bound to the kringle domains of plasminogen. The binding affinity constant (KD) values of full-length UspA2(30-539) (amino acids 30 to 539 of UspA2) and full-length UspA2H(50-720) for immobilized plasminogen were 4.8 × 10(-8) M and 3.13 × 10(-8) M, respectively, as measured by biolayer interferometry. Plasminogen bound to intact M. catarrhalis or to recombinant UspA2/UspA2H was readily accessible for a urokinase plasminogen activator that converted the zymogen into active plasmin, as verified by the specific substrate S-2251 and a degradation assay with fibrinogen. Importantly, plasmin bound at the bacterial surface also degraded C3b and C5, which consequently may contribute to reduced bacterial killing. Our findings suggest that binding of plasminogen to M. catarrhalis may lead to increased virulence and, hence, more efficient colonization of the host.


Assuntos
Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/imunologia , Plasminogênio/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Moraxella catarrhalis/imunologia , Moraxella catarrhalis/metabolismo , Infecções por Moraxellaceae/metabolismo
18.
Microb Drug Resist ; 21(5): 507-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25923017

RESUMO

Previous studies indicate that macrolide resistance in Moraxella catarrhalis isolates is less common in adults than in children. However, few studies have investigated M. catarrhalis macrolide resistance mechanisms in adult patients. In this study, 124 M. catarrhalis isolates were collected from adult patients in a Chinese tertiary hospital, between 2010 and 2013, and investigated for antimicrobial resistance. We found that only seven isolates were macrolide resistant and all exhibited high-level macrolide resistance (minimum inhibitory concentrations >256 µg/ml). Multilocus sequence typing (MLST) suggested that M. catarrhalis has a diverse population; in particular, both pulsed-field gel electrophoresis and MLST revealed that all the seven high-level macrolide-resistant M. catarrhalis belonged to different clones. A 934-bp 23S rRNA gene sequencing showed that only nine isolates (including all the seven macrolide-resistant isolates) had mutations within the studied region, and only the seven macrolide-resistant isolates had mutation of A2330T. No other known macrolide-resistance determinant genes (ermA, ermB, mefA, or mefE) were detected. These findings support previous studies in children on M. catarrhalis macrolide-resistant isolates and suggest that the 23S rRNA gene A2330T mutation is responsible for the high M. catarrhalis macrolide resistance. The findings prompted us to successfully develop a simple allele-specific polymerase chain reaction assay for high-level macrolide-resistant 23S rRNA gene A2330T mutation for future clinical and further surveillance use.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana/genética , Genes Bacterianos , Macrolídeos/farmacologia , Moraxella catarrhalis/genética , Mutação , Reação em Cadeia da Polimerase/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , China , Feminino , Genótipo , Humanos , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/isolamento & purificação , Moraxella catarrhalis/metabolismo , Infecções por Moraxellaceae/tratamento farmacológico , Infecções por Moraxellaceae/microbiologia , Tipagem de Sequências Multilocus , RNA Ribossômico 23S/genética , Estudos Retrospectivos , Centros de Atenção Terciária
19.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 2): 239-55, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25664734

RESUMO

KdsC, the third enzyme of the 3-deoxy-D-manno-octulosonic acid (KDO) biosynthetic pathway, catalyzes a substrate-specific reaction to hydrolyze 3-deoxy-D-manno-octulosonate 8-phosphate to generate a molecule of KDO and phosphate. KdsC is a phosphatase that belongs to the C0 subfamily of the HAD superfamily. To understand the molecular basis for the substrate specificity of this tetrameric enzyme, the crystal structures of KdsC from Moraxella catarrhalis (Mc-KdsC) with several combinations of ligands, namely metal ion, citrate and products, were determined. Various transition states of the enzyme have been captured in these crystal forms. The ligand-free and ligand-bound crystal forms reveal that the binding of ligands does not cause any specific conformational changes in the active site. However, the electron-density maps clearly showed that the conformation of KDO as a substrate is different from the conformation adopted by KDO when it binds as a cleaved product. Furthermore, structural evidence for the existence of an intersubunit tunnel has been reported for the first time in the C0 subfamily of enzymes. A role for this tunnel in transferring water molecules from the interior of the tetrameric structure to the active-site cleft has been proposed. At the active site, water molecules are required for the formation of a water bridge that participates as a proton shuttle during the second step of the two-step phosphoryl-transfer reaction. In addition, as the KDO biosynthesis pathway is a potential antibacterial target, pharmacophore-based virtual screening was employed to identify inhibitor molecules for the Mc-KdsC enzyme.


Assuntos
Aquaporinas/química , Proteínas de Bactérias/química , Moraxella catarrhalis/enzimologia , Infecções por Moraxellaceae/microbiologia , Monoéster Fosfórico Hidrolases/química , Sequência de Aminoácidos , Aquaporinas/metabolismo , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Ácido Cítrico/metabolismo , Cristalografia por Raios X , Humanos , Ligantes , Dados de Sequência Molecular , Moraxella catarrhalis/química , Moraxella catarrhalis/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Ligação Proteica , Conformação Proteica , Alinhamento de Sequência , Especificidade por Substrato
20.
Antimicrob Agents Chemother ; 59(4): 1886-94, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25583725

RESUMO

Moraxella catarrhalis is a common pathogen of the human respiratory tract. Multidrug efflux pumps play a major role in antibiotic resistance and virulence in many Gram-negative organisms. In the present study, the role of the AcrAB-OprM efflux pump in antibiotic resistance was investigated by constructing mutants that lack the acrA, acrB, and oprM genes in M. catarrhalis strain O35E. We observed a moderate (1.5-fold) decrease in the MICs of amoxicillin and cefotaxime and a marked (4.7-fold) decrease in the MICs of clarithromycin for acrA, acrB, and oprM mutants in comparison with the wild-type O35E strain. Exposure of the M. catarrhalis strains O35E and 300 to amoxicillin triggered an increased transcription of all AcrAB-OprM pump genes, and exposure of strains O35E, 300, and 415 to clarithromycin enhanced the expression of acrA and oprM mRNA. Inactivation of the AcrAB-OprM efflux pump genes demonstrated a decreased ability to invade epithelial cells compared to the parental strain, suggesting that acrA, acrB, and oprM are required for efficient invasion of human pharyngeal epithelial cells. Cold shock increases the expression of AcrAB-OprM efflux pump genes in all three M. catarrhalis strains tested. Increased expression of AcrAB-OprM pump genes after cold shock leads to a lower accumulation of Hoechst 33342 (H33342), a substrate of AcrAB-OprM efflux pumps, indicating that cold shock results in increased efflux activity. In conclusion, the AcrAB-OprM efflux pump appears to play a role in the antibiotic resistance and virulence of M. catarrhalis and is involved in the cold shock response.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Resposta ao Choque Frio , Farmacorresistência Bacteriana/genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Moraxella catarrhalis/genética , Moraxella catarrhalis/metabolismo , Amoxicilina/farmacologia , Antibacterianos/farmacologia , Cefotaxima/farmacologia , Linhagem Celular , Claritromicina/farmacologia , DNA Bacteriano/genética , Células Epiteliais/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Mutação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...