Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Viruses ; 15(3)2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36992312

RESUMO

Bacteriophages have been identified as a potential treatment option to treat lung infection in the context of antibiotic resistance. We performed a preclinical study to predict the efficacy of delivery of bacteriophages against Pseudomonas aeruginosa (PA) when administered via nebulization during mechanical ventilation (MV). We selected a mix of four anti-PA phages containing two Podoviridae and two Myoviridae, with a coverage of 87.8% (36/41) on an international PA reference panel. When administered via nebulization, a loss of 0.30-0.65 log of infective phage titers was measured. No difference between jet, ultrasonic and mesh nebulizers was observed in terms of loss of phage viability, but a higher output was measured with the mesh nebulizer. Interestingly, Myoviridae are significantly more sensitive to nebulization than Podoviridae since their long tail is much more prone to damage. Phage nebulization has been measured as compatible with humidified ventilation. Based on in vitro measurement, the lung deposition prediction of viable phage particles ranges from 6% to 26% of the phages loaded in the nebulizer. Further, 8% to 15% of lung deposition was measured by scintigraphy in three macaques. A phage dose of 1 × 109 PFU/mL nebulized by the mesh nebulizer during MV predicts an efficient dose in the lung against PA, comparable with the dose chosen to define the susceptibility of the strain.


Assuntos
Bacteriófagos , Podoviridae , Animais , Respiração Artificial , Macaca , Nebulizadores e Vaporizadores , Myoviridae , Pulmão , Aerossóis
2.
Microbiol Spectr ; 10(5): e0063522, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-35980045

RESUMO

Regulatory small RNAs (sRNAs) are involved in the adaptation of bacteria to their environment. CiaR-dependent sRNAs (csRNAs) are controlled by the regulatory two-component system (TCS) CiaRH, which is widely conserved in streptococci. Except for Streptococcus pneumoniae and Streptococcus sanguinis, the targets of these csRNAs have not yet been investigated. Streptococcus agalactiae, the leading cause of neonatal infections, has four conserved csRNA genes, namely, srn015, srn024, srn070, and srn085. Here, we demonstrate the importance of the direct repeat TTTAAG-N5-TTTAAG in the regulation of these csRNAs by CiaRH. A 24-nucleotide Srn024-sap RNA base-pairing region is predicted in silico. The sap gene encodes a LPXTG-cell wall-anchored pullulanase. This protein cleaves α-glucan polysaccharides such as pullulan and glycogen present in the environment to release glucose and is involved in adhesion to human cervical epithelial cells. Inactivation of S. agalactiae pullulanase (SAP) leads to no bacterial growth in a medium with only pullulan as a carbon source and reduced biofilm formation, while deletion of ciaRH and srn024 genes significantly increases bacterial growth and biofilm formation. Using a new translational fusion vector, we demonstrated that Srn024 is involved in the posttranscriptional regulation of sap expression. Complementary base pair exchanges in S. agalactiae suggest that Srn024 interacts directly with sap mRNA and that disruption of this RNA pairing is sufficient to yield the biofilm phenotype of Srn024 deletion. These results suggest the involvement of Srn024 in the adaptation of S. agalactiae to environmental changes and biofilm formation, likely through the regulation of the sap gene. IMPORTANCE Although Streptococcus agalactiae is a commensal bacterium of the human digestive and genitourinary tracts, it is also an opportunistic pathogen for humans and other animals. As the main cause of neonatal infections, it is responsible for pneumonia, bacteremia, and meningitis. However, its adaptation to these different ecological niches is not fully understood. Bacterial regulatory networks are involved in this adaptation, and the regulatory TCSs (e.g., CiaRH), as well as the regulatory sRNAs, are part of it. This study is the first step to understand the role of csRNAs in the adaptation of S. agalactiae. This bacterium does not currently exhibit extensive antibiotic resistance. However, it is crucial to find alternatives before multidrug resistance emerges. Therefore, we propose that drugs targeting regulatory RNAs with Srn024-like activities would affect pathogens by reducing their abilities to form biofilm and to adapt to host niches.


Assuntos
Regulação Bacteriana da Expressão Gênica , Streptococcus agalactiae , Animais , Recém-Nascido , Humanos , Streptococcus agalactiae/genética , RNA , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes , Glucanos , Nucleotídeos , RNA Mensageiro , Glicogênio/metabolismo , Glucose , Carbono/metabolismo
3.
Front Immunol ; 11: 573040, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33101296

RESUMO

Granulomatosis with polyangiitis (GPA) is a severe autoimmune vasculitis associated with the presence of anti-neutrophil cytoplasmic antibodies (ANCA) mainly targeting proteinase 3 (PR3), a neutrophilic serine proteinase. PR3-ANCA binding to membrane-bound PR3 on neutrophils induce their auto-immune activation responsible for vascular lesions. However, the correlation between PR3-ANCA level and disease activity remains inconsistent, suggesting the existence of non-pathogenic PR3-ANCA. In order to prove their existence, we immortalized B lymphocytes from blood samples of GPA patients in remission having persistent PR3-ANCA to isolate non-activating PR3-ANCA. We obtained for the first time a non-activating human IgG1κ anti-PR3 monoclonal antibody (mAb) named 4C3. This new mAb binds soluble PR3 with a high affinity and membrane-bound PR3 on an epitope close to the PR3 hydrophobic patch and in the vicinity of the active site. 4C3 is able to bind FcγRIIA and FcγRIIIB and has a G2F glycosylation profile on asparagine 297. 4C3 did not induce activation of neutrophils and could inhibit human polyclonal PR3-ANCA-induced activation suggesting that 4C3 is non-pathogenic. This characteristic relies on the recognized epitope on PR3 rather than to the Fc portion properties. The existence of non-pathogenic PR3-ANCA, which do not activate neutrophils, could explain the persistence of high PR3-ANCA levels in some GPA patients in remission and why PR3-ANCA would not predict relapse. Finally, these results offer promising perspectives particularly regarding the understanding of PR3-ANCA pathogenicity and the development of new diagnostic and therapeutic strategies in GPA.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/imunologia , Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Granulomatose com Poliangiite/imunologia , Mieloblastina/imunologia , Idoso , Anticorpos Anticitoplasma de Neutrófilos/metabolismo , Anticorpos Monoclonais/metabolismo , Afinidade de Anticorpos , Especificidade de Anticorpos , Linfócitos B/enzimologia , Sítios de Ligação de Anticorpos , Biomarcadores/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Mapeamento de Epitopos , Epitopos , Feminino , Glicosilação , Granulomatose com Poliangiite/diagnóstico , Granulomatose com Poliangiite/enzimologia , Humanos , Masculino , Pessoa de Meia-Idade , Ativação de Neutrófilo , Estudo de Prova de Conceito
4.
Front Microbiol ; 10: 1826, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31474948

RESUMO

Pseudomonas aeruginosa is an opportunistic bacteria and a major cause of nosocomial pneumonia. P. aeruginosa has many virulence factors contributing to its ability to colonize the host. LoxA is a lipoxygenase enzyme secreted by P. aeruginosa that oxidizes polyunsaturated fatty acids. Based on previous in vitro biochemical studies, several biological roles of LoxA have been hypothesized, including interference of the host lipid signaling, and modulation of bacterial invasion properties. However, the contribution of LoxA to P. aeruginosa lung pathogenesis per se remained unclear. In this study, we used complementary in vitro and in vivo approaches, clinical strains of P. aeruginosa as well as lipidomics technology to investigate the role of LoxA in lung infection. We found that several P. aeruginosa clinical isolates express LoxA. When secreted in the lungs, LoxA processes a wide range of host polyunsaturated fatty acids, which further results in the production of bioactive lipid mediators (including lipoxin A4). LoxA also inhibits the expression of major chemokines (e.g., MIPs and KC) and the recruitment of key leukocytes. Remarkably, LoxA promotes P. aeruginosa persistence in lungs tissues. Hence, our study suggests that LoxA-dependent interference of the host lipid pathways may contribute to P. aeruginosa lung pathogenesis.

5.
Virulence ; 9(1): 1163-1175, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30070169

RESUMO

Pseudomonas aeruginosa, an opportunistic pathogen involved in skin and lung diseases, possesses numerous virulence factors, including type 2 and 3 secretion systems (T2SS and T3SS) and its flagellum, whose functions remain poorly known during cutaneous infection. Using isogenic mutants deleted from genes encoding each or all of these three virulence factors, we investigated their role in induction of inflammatory response and in tissue invasiveness in human primary keratinocytes and reconstructed epidermis. Our results showed that flagellum, but not T2SS and T3SS, is involved in induction of a large panel of cytokine, chemokine, and antimicrobial peptide (AMP) mRNA in the infected keratinocytes. Chemokine secretion and AMP tissular production were also dependent on the presence of the bacterial flagellum. This pro-inflammatory effect was significantly reduced in keratinocytes infected in presence of anti-toll-like receptor 5 (TLR5) neutralizing antibody. Bacterial invasion of human epidermis and persistence in a mouse model of sub-cutaneous infection were dependent on the P. aeruginosa flagellum. We demonstrated that flagellum constitutes the main virulence factor of P. aeruginosa involved not only in early induction of the epidermis inflammatory response but also in bacterial invasion and cutaneous persistence. P. aeruginosa is mainly sensed by TLR5 during the early innate immune response of human primary keratinocytes.


Assuntos
Epiderme/microbiologia , Flagelos/fisiologia , Inflamação/microbiologia , Queratinócitos/imunologia , Pseudomonas aeruginosa/patogenicidade , Animais , Anticorpos Neutralizantes/farmacologia , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/imunologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Inata , Queratinócitos/efeitos dos fármacos , Queratinócitos/microbiologia , Masculino , Camundongos , Mutação , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/ultraestrutura , Receptor 5 Toll-Like/imunologia , Fatores de Virulência/deficiência , Fatores de Virulência/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-30104272

RESUMO

The endotracheal tube (ETT) is an essential interface between the patient and ventilator in mechanically ventilated patients. However, a microbial biofilm is formed gradually on this tube and is associated with the development of ventilator-associated pneumonia. The bacteria present in the biofilm are more resistant to antibiotics, and current medical practices do not make it possible to eliminate. Pseudomonas aeruginosa is one of the leading pathogens that cause biofilm infections and ventilator-associated pneumonia. Poly-l-lysine (pLK) is a cationic polypeptide possessing antibacterial properties and mucolytic activity by compacting DNA. Here, we explored the antibiofilm activity of pLK to treat P. aeruginosa biofilms on ETTs while taking into consideration the necessary constraints for clinical translation in our experimental designs. First, we showed that pLK eradicates a P. aeruginosa biofilm formed in vitro on 96-well microplates. We further demonstrated that pLK alters bacterial membrane integrity, as revealed by scanning electron microscopy, and eventually eradicates biofilm formed either by reference or clinical strains of P. aeruginosa biofilms generated in vitro on ETTs. Second, we collected the ETT from patients with P. aeruginosa ventilator-associated pneumonia. We observed that a single dose of pLK is able to immediately disrupt the biofilm structure and kills more than 90% of bacteria present in the biofilm. Additionally, we did not observe any lung tolerance issue when the pLK solution was instilled into the ETT of ventilated pigs, an animal model particularly relevant to mimic invasive mechanical ventilation in humans. In conclusion, pLK appears as an innovative antibiofilm molecule, which could be applied in the ETT of mechanically ventilated patients.


Assuntos
Biofilmes/efeitos dos fármacos , Intubação Intratraqueal/efeitos adversos , Polilisina/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Respiração Artificial/efeitos adversos , Animais , Antibacterianos/farmacologia , Contaminação de Equipamentos , Humanos , Microscopia Eletrônica de Varredura/métodos , Pneumonia Associada à Ventilação Mecânica/tratamento farmacológico , Suínos
7.
Cell Host Microbe ; 22(1): 38-47.e4, 2017 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-28704651

RESUMO

The rise of multi-drug-resistant (MDR) bacteria has spurred renewed interest in the use of bacteriophages in therapy. However, mechanisms contributing to phage-mediated bacterial clearance in an animal host remain unclear. We investigated the effects of host immunity on the efficacy of phage therapy for acute pneumonia caused by MDR Pseudomonas aeruginosa in a mouse model. Comparing efficacies of phage-curative and prophylactic treatments in healthy immunocompetent, MyD88-deficient, lymphocyte-deficient, and neutrophil-depleted murine hosts revealed that neutrophil-phage synergy is essential for the resolution of pneumonia. Population modeling of in vivo results further showed that neutrophils are required to control both phage-sensitive and emergent phage-resistant variants to clear infection. This "immunophage synergy" contrasts with the paradigm that phage therapy success is largely due to bacterial permissiveness to phage killing. Lastly, therapeutic phages were not cleared by pulmonary immune effector cells and were immunologically well tolerated by lung tissues.


Assuntos
Bacteriófagos/imunologia , Sistema Imunitário/imunologia , Terapia por Fagos/métodos , Infecções por Pseudomonas/terapia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/virologia , Animais , Bacteriófagos/patogenicidade , Citocinas/metabolismo , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla , Feminino , Pulmão/microbiologia , Pulmão/patologia , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Fator 88 de Diferenciação Mieloide/genética , Neutrófilos/imunologia , Infecções por Pseudomonas/microbiologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/terapia
8.
Front Immunol ; 8: 123, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28243234

RESUMO

Recent in silico studies suggested that the transcription cofactor LIM-only protein FHL2 is a major transcriptional regulator of mouse natural killer (NK) cells. However, the expression and role of FHL2 in NK cell biology are unknown. Here, we confirm that FHL2 is expressed in both mouse and human NK cells. Using FHL2-/- mice, we found that FHL2 controls NK cell development in the bone marrow and maturation in peripheral organs. To evaluate the importance of FHL2 in NK cell activation, FHL2-/- mice were infected with Streptococcus pneumoniae. FHL2-/- mice are highly susceptible to this infection. The activation of lung NK cells is altered in FHL2-/- mice, leading to decreased IFNγ production and a loss of control of bacterial burden. Collectively, our data reveal that FHL2 is a new transcription cofactor implicated in NK cell development and activation during pulmonary bacterial infection.

9.
Virulence ; 8(6): 810-820, 2017 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27792459

RESUMO

The IL-22 signaling pathway is critical for regulating mucosal defense and limiting bacterial dissemination. IL-22 is unusual among interleukins because it does not directly regulate the function of conventional immune cells, but instead targets cells at outer body barriers, such as respiratory epithelial cells. Consequently, IL-22 signaling participates in the maintenance of the lung mucosal barrier by controlling cell proliferation and tissue repair, and enhancing the production of specific chemokines and anti-microbial peptides. Pseudomonas aeruginosa is a major pathogen of ventilator-associated pneumonia and causes considerable lung tissue damage. A feature underlying the pathogenicity of this bacterium is its capacity to persist and develop in the host, particularly in the clinical context of nosocomial lung infections. We aimed to investigate the ability of P. auruginosa to disrupt immune-epithelial cells cross-talk. We found that P. aeruginosa escapes the host mucosal defenses by degrading IL-22, leading to severe inhibition of IL-22-mediated immune responses. We demonstrated in vitro that, protease IV, a type 2 secretion system-dependent serine protease, is responsible for the degradation of IL-22 by P. aeruginosa. Moreover, the major anti-proteases molecules present in the lungs were unable to inhibit protease IV enzymatic activity. In addition, tracheal aspirates of patients infected by P. aeruginosa contain protease IV activity which further results in IL-22 degradation. This so far undescribed cleavage of IL-22 by a bacterial protease is likely to be an immune-evasion strategy that contributes to P. aeruginosa-triggered respiratory infections.


Assuntos
Interleucinas/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/metabolismo , Animais , Infecção Hospitalar , Humanos , Evasão da Resposta Imune , Interleucinas/deficiência , Interleucinas/genética , Interleucinas/imunologia , Pulmão/fisiopatologia , Camundongos , Camundongos Knockout , Peptídeo Hidrolases/biossíntese , Peptídeo Hidrolases/metabolismo , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Proteólise , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/patogenicidade , Transdução de Sinais , Interleucina 22
10.
Expert Opin Drug Deliv ; 14(8): 959-972, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27776446

RESUMO

INTRODUCTION: Bacterial respiratory tract infections (RTIs) are increasingly difficult to treat due to evolving antibiotic resistance. In this context, bacteriophages (or phages) are part of the foreseen alternatives or combination therapies. Delivering phages through the airways seems more relevant to accumulate these natural antibacterial viruses in proximity to their bacterial host, within the infectious site. Areas covered: This review addresses the potential of phage therapy to treat RTIs and discusses preclinical and clinical results of phages administration in this context. Recent phage formulation and aerosolization attempts are also reviewed, raising technical challenges to achieve efficient pulmonary deposition via inhalation. Expert opinion: Overall, the inhalation of phages as antibacterial treatment seems both clinically relevant and technically feasible. Several crucial points still need to be investigated, such as phage product pharmacokinetics and immunogenicity. Furthermore, given phage-specific features, appropriate regulatory and manufacturing guidelines will need to be defined. Finally, randomized controlled clinical trials should be carried out to establish phage therapy's clinical positioning in the antimicrobial arsenal against RTIs.


Assuntos
Infecções Bacterianas/terapia , Terapia por Fagos , Infecções Respiratórias/terapia , Administração por Inalação , Animais , Bacteriófagos , Humanos
11.
Am J Physiol Lung Cell Mol Physiol ; 311(3): L664-75, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27496898

RESUMO

Cystic fibrosis (CF) is an inherited disease associated with chronic severe lung inflammation, leading to premature death. To develop innovative anti-inflammatory treatments, we need to characterize new cellular and molecular components contributing to the mechanisms of lung inflammation. Here, we focused on the potential role of "transient receptor potential vanilloid-4" (TRPV4), a nonselective calcium channel. We used both in vitro and in vivo approaches to demonstrate that TRPV4 expressed in airway epithelial cells triggers the secretion of major proinflammatory mediators such as chemokines and biologically active lipids, as well as a neutrophil recruitment in lung tissues. We characterized the contribution of cytosolic phospholipase A2, MAPKs, and NF-κB in TRPV4-dependent signaling. We also showed that 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids, i.e., four natural lipid-based TRPV4 agonists, are present in expectorations of CF patients. Also, TRPV4-induced calcium mobilization and inflammatory responses were enhanced in cystic fibrosis transmembrane conductance regulator-deficient cellular and animal models, suggesting that TRPV4 is a promising target for the development of new anti-inflammatory treatments for diseases such as CF.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fibrose Cística/metabolismo , Canais de Cátion TRPV/fisiologia , Células A549 , Animais , Sinalização do Cálcio , Fibrose Cística/imunologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Feminino , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos Sprague-Dawley
12.
PLoS One ; 10(9): e0138103, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26407005

RESUMO

Streptococcus agalactiae (or Group B Streptococcus, GBS) is a commensal bacterium present in the intestinal and urinary tracts of approximately 30% of humans. We and others previously showed that the PI-2a pilus polymers, made of the backbone pilin PilB, the tip adhesin PilA and the cell wall anchor protein PilC, promote adhesion to host epithelia and biofilm formation. Affinity-purified PI-2a pili from GBS strain NEM316 were recognized by N-acetylneuraminic acid (NeuNAc, also known as sialic acid) specific lectins such as Elderberry Bark Lectin (EBL) suggesting that pili are sialylated. Glycan profiling with twenty different lectins combined with monosaccharide composition by HPLC suggested that affinity-purified PI-2a pili are modified by N-glycosylation and decorated with sialic acid attached to terminal galactose. Analysis of various relevant mutants in the PI-2a pilus operon by flow-cytometry and electron microscopy analyses pointed to PilA as the pilus subunit modified by glycosylation. Double labeling using PilB antibody and EBL lectin, which specifically recognizes N-acetylneuraminic acid attached to galactose in α-2, 6, revealed a characteristic binding of EBL at the tip of the pilus structures, highly reminiscent of PilA localization. Expression of a secreted form of PilA using an inducible promoter showed that this recombinant PilA binds specifically to EBL lectin when produced in the native GBS context. In silico search for potentially glycosylated asparagine residues in PilA sequence pointed to N427 and N597, which appear conserved and exposed in the close homolog RrgA from S. pneumoniae, as likely candidates. Conversion of these two asparagyl residues to glutamyl resulted in a higher instability of PilA. Our results provide the first evidence that the tip PilA adhesin can be glycosylated, and suggest that this modification is critical for PilA stability and may potentially influence interactions with the host.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas de Fímbrias/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Processamento de Proteína Pós-Traducional , Streptococcus agalactiae , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Aderência Bacteriana/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/ultraestrutura , Glucosiltransferases/metabolismo , Modelos Moleculares , Organismos Geneticamente Modificados , Lectinas de Plantas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Inativadoras de Ribossomos/metabolismo , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo
13.
Eur Respir J ; 46(3): 771-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26250498

RESUMO

Chronic obstructive pulmonary disease (COPD) is punctuated by episodes of infection-driven acute exacerbations. Despite the life-threatening nature of these exacerbations, the underlying mechanisms remain unclear, although a high number of neutrophils in the lungs of COPD patients is known to correlate with poor prognosis. Interleukin (IL)-22 is a cytokine that plays a pivotal role in lung antimicrobial defence and tissue protection. We hypothesised that neutrophils secrete proteases that may have adverse effects in COPD, by altering the IL-22 receptor (IL-22R)-dependent signalling.Using in vitro and in vivo approaches as well as reverse transcriptase quantitative PCR, flow cytometry and/or Western blotting techniques, we first showed that pathogens such as the influenza virus promote IL-22R expression in human bronchial epithelial cells, whereas Pseudomonas aeruginosa, bacterial lipopolysaccharide or cigarette smoke do not. Most importantly, neutrophil proteases cleave IL-22R and impair IL-22-dependent immune signalling and expression of antimicrobial effectors such as ß-defensin-2. This proteolysis resulted in the release of a soluble fragment of IL-22R, which was detectable both in cellular and animal models as well as in sputa from COPD patients with acute exacerbations.Hence, our study reveals an unsuspected regulation by the proteolytic action of neutrophil enzymes of IL-22-dependent lung host response. This process probably enhances pathogen replication, and ultimately COPD exacerbations.


Assuntos
Células Epiteliais/enzimologia , Imunidade Inata/efeitos dos fármacos , Neutrófilos/metabolismo , Doença Pulmonar Obstrutiva Crônica/microbiologia , Receptores de Interleucina/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Humanos , Imunidade Inata/fisiologia , Camundongos , Neutrófilos/efeitos dos fármacos , Peptídeo Hidrolases/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina/imunologia , Estudos de Amostragem , Sensibilidade e Especificidade , Fumar/efeitos adversos , Estatísticas não Paramétricas , beta-Defensinas/farmacologia
14.
Microbes Infect ; 14(12): 1044-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22841805

RESUMO

Group B Streptococcus (GBS) is the leading cause of neonatal septicemia and meningitis. Pili appendages were shown to play a critical role in bacterial adhesion and colonization of human tissues. Recently it was claimed that binding of the pilus-associated adhesin PilA to collagen is a critical, initial step in promoting interactions with the α2ß1 integrin expressed on brain endothelial cells. Here we show that strain NCTC10/84 used in this study is not representative for GBS isolates and question the importance of collagen as a critical extracellular matrix component for GBS infections of the central nervous system.


Assuntos
Aderência Bacteriana , Fibrinogênio/metabolismo , Streptococcus agalactiae/patogenicidade , Colágeno/metabolismo , Humanos , Ligação Proteica , Streptococcus agalactiae/fisiologia
15.
PLoS One ; 7(3): e33516, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22442694

RESUMO

BACKGROUND: Protein folding in the envelope is a crucial limiting step of protein export and secretion. In order to better understand this process in Lactococcus lactis, a lactic acid bacterium, genes encoding putative exported folding factors like Peptidyl Prolyl Isomerases (PPIases) were searched for in lactococcal genomes. RESULTS: In L. lactis, a new putative membrane PPIase of the cyclophilin subfamily, PpiA, was identified and characterized. ppiA gene was found to be constitutively expressed under normal and stress (heat shock, H(2)O(2)) conditions. Under normal conditions, PpiA protein was synthesized and released from intact cells by an exogenously added protease, showing that it was exposed at the cell surface. No obvious phenotype could be associated to a ppiA mutant strain under several laboratory conditions including stress conditions, except a very low sensitivity to H(2)O(2). Induction of a ppiA copy provided in trans had no effect i) on the thermosensitivity of an mutant strain deficient for the lactococcal surface protease HtrA and ii) on the secretion and stability on four exported proteins (a highly degraded hybrid protein and three heterologous secreted proteins) in an otherwise wild-type strain background. However, a recombinant soluble form of PpiA that had been produced and secreted in L. lactis and purified from a culture supernatant displayed both PPIase and chaperone activities. CONCLUSIONS: Although L. lactis PpiA, a protein produced and exposed at the cell surface under normal conditions, displayed a very moderate role in vivo, it was found, as a recombinant soluble form, to be endowed with folding activities in vitro.


Assuntos
Proteínas de Bactérias/metabolismo , Membrana Celular/enzimologia , Lactococcus lactis/enzimologia , Proteínas de Membrana/metabolismo , Peptidilprolil Isomerase/metabolismo , Dobramento de Proteína , Proteínas de Bactérias/genética , Membrana Celular/genética , Ciclofilinas/genética , Ciclofilinas/metabolismo , Peróxido de Hidrogênio/farmacologia , Lactococcus lactis/genética , Proteínas de Membrana/genética , Oxidantes/farmacologia , Peptidilprolil Isomerase/genética , Estresse Fisiológico/efeitos dos fármacos
16.
PLoS One ; 7(1): e29963, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22291899

RESUMO

Glyceraldehyde 3-phosphate dehydrogenases (GAPDH) are cytoplasmic glycolytic enzymes that, despite lacking identifiable secretion signals, have been detected at the surface of several prokaryotic and eukaryotic organisms where they exhibit non-glycolytic functions including adhesion to host components. Group B Streptococcus (GBS) is a human commensal bacterium that has the capacity to cause life-threatening meningitis and septicemia in newborns. Electron microscopy and fluorescence-activated cell sorter (FACS) analysis demonstrated the surface localization of GAPDH in GBS. By addressing the question of GAPDH export to the cell surface of GBS strain NEM316 and isogenic mutant derivatives of our collection, we found that impaired GAPDH presence in the surface and supernatant of GBS was associated with a lower level of bacterial lysis. We also found that following GBS lysis, GAPDH can associate to the surface of many living bacteria. Finally, we provide evidence for a novel function of the secreted GAPDH as an inducer of apoptosis of murine macrophages.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/farmacologia , Macrófagos/efeitos dos fármacos , Streptococcaceae/enzimologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo , Antígenos de Superfície/fisiologia , Apoptose/fisiologia , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/análise , Proteínas de Bactérias/fisiologia , Extratos Celulares/química , Extratos Celulares/metabolismo , Células Cultivadas , Feminino , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/fisiologia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/análise , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/fisiologia , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados , Ligação Proteica , Streptococcaceae/classificação , Streptococcaceae/crescimento & desenvolvimento , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/patologia
17.
Environ Microbiol ; 14(8): 1844-54, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22118225

RESUMO

Bacteriophages have been known to be present in the gut for many years, but studies of relationships between these viruses and their hosts in the intestine are still in their infancy. We isolated three bacteriophages specific for an enteroaggregative O104:H4 Escherichia coli (EAEC) strain responsible for diarrhoeal diseases in humans. We studied the replication of these bacteriophages in vitro and in vivo in a mouse model of gut colonization. Each bacteriophage was able to replicate in vitro in both aerobic and anaerobic conditions. Each bacteriophage individually reduced biofilms formed on plastic pegs and a cocktail of the three bacteriophages was found to be more efficient. The cocktail was also able to infect bacterial aggregates formed on the surface of epithelial cells. In the mouse intestine, bacteriophages replicated for at least 3 weeks, provided the host was present, with no change in host levels in the faeces. This model of stable and continuous viral replication provides opportunities for studying the long-term coevolution of virulent bacteriophages with their hosts within a mammalian polymicrobial ecosystem.


Assuntos
Bacteriófagos/fisiologia , Escherichia coli/virologia , Animais , Bacteriófagos/classificação , Bacteriófagos/isolamento & purificação , Bacteriófagos/ultraestrutura , Biofilmes , Caudovirales/classificação , Caudovirales/isolamento & purificação , Caudovirales/fisiologia , Caudovirales/ultraestrutura , Fezes/microbiologia , Fezes/virologia , Especificidade de Hospedeiro , Intestinos/microbiologia , Intestinos/virologia , Camundongos , Replicação Viral
18.
PLoS One ; 6(2): e16963, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21347240

RESUMO

Multidrug-resistant bacteria are the cause of an increasing number of deadly pulmonary infections. Because there is currently a paucity of novel antibiotics, phage therapy--the use of specific viruses that infect bacteria--is now more frequently being considered as a potential treatment for bacterial infections. Using a mouse lung-infection model caused by a multidrug resistant Pseudomonas aeruginosa mucoid strain isolated from a cystic fibrosis patient, we evaluated bacteriophage treatments. New bacteriophages were isolated from environmental samples and characterized. Bacteria and bacteriophages were applied intranasally to the immunocompetent mice. Survival was monitored and bronchoalveolar fluids were analysed. Quantification of bacteria, bacteriophages, pro-inflammatory and cytotoxicity markers, as well as histology and immunohistochemistry analyses were performed. A curative treatment (one single dose) administrated 2 h after the onset of the infection allowed over 95% survival. A four-day preventive treatment (one single dose) resulted in a 100% survival. All of the parameters measured correlated with the efficacy of both curative and preventive bacteriophage treatments. We also showed that in vitro optimization of a bacteriophage towards a clinical strain improved both its efficacy on in vivo treatments and its host range on a panel of 20 P. aeruginosa cystic fibrosis strains. This work provides an incentive to develop clinical studies on pulmonary bacteriophage therapy to combat multidrug-resistant lung infections.


Assuntos
Bacteriófagos/fisiologia , Fibrose Cística/prevenção & controle , Fibrose Cística/terapia , Pulmão/microbiologia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/virologia , Sequência de Aminoácidos , Animais , Bacteriófagos/patogenicidade , Fibrose Cística/microbiologia , Farmacorresistência Bacteriana Múltipla , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Especificidade da Espécie , Proteínas Virais/química , Proteínas Virais/metabolismo
19.
J Bacteriol ; 193(8): 1919-29, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21317326

RESUMO

In the family Streptococcaceae, the genes encoding zinc ABC uptake systems (called zit or adc) are regulated by a coencoded MarR family member (i.e., ZitR or AdcR), whereas in the great majority of bacteria, these genes are regulated by Zur, the Fur-like zinc-responsive repressor. We studied the zit operon from Lactococcus lactis and its regulation in response to Zn(II) in vivo. zit transcription is repressed by Zn(II) in a wide concentration range starting from nontoxic micromolar levels and is derepressed at nanomolar concentrations. The level of zit promoter downregulation by environmental Zn(II) is correlated with the intracellular zinc content. The helix-turn-helix domain of ZitR is required for downregulation. In vitro, the purified protein is a dimer that complexes up to two zinc ligands per monomer and specifically binds two intact palindromic operator sites overlapping the -35 and -10 boxes of the zit promoter. DNA binding is abolished by the chelator EDTA or TPEN and fully restored by Zn(II) addition, indicating that the active repressor complexes Zn(II) with high affinity. These results suggest that derepression under starvation conditions could be an essential emergency mechanism for preserving Zn(II) homeostasis by uptake; under Zn(II)-replete conditions, the function of ZitR repression could be to help save energy rather than to avoid Zn(II) toxicity. The characterization of a MarR family zinc-responsive repressor in this report gives insight into the way Streptococcaceae efficiently adapt to Zn(II) fluctuations in their diverse ecological niches.


Assuntos
Regulação Bacteriana da Expressão Gênica , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Proteínas Repressoras/metabolismo , Zinco/metabolismo , Sequência de Aminoácidos , DNA Bacteriano/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Perfilação da Expressão Gênica , Sequências Hélice-Alça-Hélice , Dados de Sequência Molecular , Óperon , Regiões Promotoras Genéticas , Mapeamento de Interação de Proteínas , Multimerização Proteica , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/isolamento & purificação , Alinhamento de Sequência
20.
J Infect Dis ; 201(7): 1096-104, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20196657

RESUMO

Antibiotic-resistant bacteria threaten life worldwide. Although new antibiotics are scarce, the use of bacteriophages, viruses that infect bacteria, is rarely proposed as a means of offsetting this shortage. Doubt also remains widespread about the efficacy of phage therapy despite recent encouraging results. Using a bioluminescent Pseudomonas aeruginosa strain, we monitored and quantified the efficacy of a bacteriophage treatment in mice during acute lung infection. Bacteriophage treatment not only was effective in saving animals from lethal infection, but also was able to prevent lung infection when given 24 h before bacterial infection, thereby extending the potential use of bacteriophages as therapeutic agents to combat bacterial lung infection.


Assuntos
Pneumopatias/microbiologia , Pneumopatias/prevenção & controle , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/prevenção & controle , Fagos de Pseudomonas/fisiologia , Pseudomonas aeruginosa/virologia , Sequência de Aminoácidos , Animais , Citocinas/metabolismo , Inflamação/metabolismo , Medições Luminescentes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fagos de Pseudomonas/química , Fagos de Pseudomonas/genética , Análise de Sobrevida , Imagem Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA