Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
PLoS Pathog ; 18(8): e1010771, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35960766

RESUMO

ESX type VII secretion systems are complex secretion machineries spanning across the mycobacterial membrane and play an important role in pathogenicity, nutrient uptake and conjugation. We previously reported the role of ESX-4 in modulating Mycobacterium abscessus intracellular survival. The loss of EccB4 was associated with limited secretion of two effector proteins belonging to the WXG-100 family, EsxU and EsxT, and encoded by the esx-4 locus. This prompted us to investigate the function of M. abscessus EsxU and EsxT in vitro and in vivo. Herein, we show that EsxU and EsxT are substrates of ESX-4 and form a stable 1:1 heterodimer that permeabilizes artificial membranes. While expression of esxU and esxT was up-regulated in M. abscessus-infected macrophages, their absence in an esxUT deletion mutant prevented phagosomal membrane disruption while maintaining M. abscessus in an unacidified phagosome. Unexpectedly, the esxUT deletion was associated with a hyper-virulent phenotype, characterised by increased bacterial loads and mortality in mouse and zebrafish infection models. Collectively, these results demonstrate that the presence of EsxU and EsxT dampens survival and persistence of M. abscessus during infection.


Assuntos
Mycobacterium abscessus , Mycobacterium marinum , Mycobacterium tuberculosis , Mycobacterium , Sistemas de Secreção Tipo VII , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Camundongos , Mycobacterium/genética , Mycobacterium abscessus/genética , Mycobacterium marinum/metabolismo , Mycobacterium tuberculosis/genética , Sistemas de Secreção Tipo VII/genética , Sistemas de Secreção Tipo VII/metabolismo , Peixe-Zebra/metabolismo
2.
Am J Respir Cell Mol Biol ; 66(4): 439-451, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35081328

RESUMO

Persistent neutrophilic inflammation associated with chronic pulmonary infection causes progressive lung injury and, eventually, death in individuals with cystic fibrosis (CF), a genetic disease caused by biallelic mutations in the CF transmembrane conductance regulator (CFTR) gene. Therefore, we examined whether roscovitine, a cyclin-dependent kinase inhibitor that (in other conditions) reduces inflammation while promoting host defense, might provide a beneficial effect in the context of CF. Herein, using CFTR-depleted zebrafish larvae as an innovative vertebrate model of CF immunopathophysiology, combined with murine and human approaches, we sought to determine the effects of roscovitine on innate immune responses to tissue injury and pathogens in the CF condition. We show that roscovitine exerts antiinflammatory and proresolution effects in neutrophilic inflammation induced by infection or tail amputation in zebrafish. Roscovitine reduces overactive epithelial reactive oxygen species (ROS)-mediated neutrophil trafficking by reducing DUOX2/NADPH-oxidase activity and accelerates inflammation resolution by inducing neutrophil apoptosis and reverse migration. It is important to note that, although roscovitine efficiently enhances intracellular bacterial killing of Mycobacterium abscessus in human CF macrophages ex vivo, we found that treatment with roscovitine results in worse infection in mouse and zebrafish models. By interfering with DUOX2/NADPH oxidase-dependent ROS production, roscovitine reduces the number of neutrophils at infection sites and, consequently, compromises granuloma formation and maintenance, favoring extracellular multiplication of M. abscessus and more severe infection. Our findings bring important new understanding of the immune-targeted action of roscovitine and have significant therapeutic implications for safely targeting inflammation in CF.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Neutrófilos , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Oxidases Duais , Camundongos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Roscovitina/farmacologia , Roscovitina/uso terapêutico , Peixe-Zebra
3.
J Antimicrob Chemother ; 77(12): 3496-3503, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36253948

RESUMO

BACKGROUND: Mycobacterium abscessus (Mabs), a rapidly growing Mycobacterium species, is considered an MDR organism. Among the standard antimicrobial multi-drug regimens against Mabs, amikacin is considered as one of the most effective. Parenteral amikacin, as a consequence of its inability to penetrate inside the cells, is only active against extracellular mycobacteria. The use of inhaled liposomal amikacin may yield improved intracellular efficacy by targeting Mabs inside the cells, while reducing its systemic toxicity. OBJECTIVES: To evaluate the colocalization of an amikacin liposomal inhalation suspension (ALIS) with intracellular Mabs, and then to measure its intracellular anti-Mabs activity. METHODS: We evaluated the colocalization of ALIS with Mabs in eukaryotic cells such as macrophages (THP-1 and J774.2) or pulmonary epithelial cells (BCi-NS1.1 and MucilAir), using a fluorescent ALIS and GFP-expressing Mabs, to test whether ALIS reaches intracellular Mabs. We then evaluated the intracellular anti-Mabs activity of ALIS inside macrophages using cfu and/or luminescence. RESULTS: Using confocal microscopy, we demonstrated fluorescent ALIS and GFP-Mabs colocalization in macrophages and epithelial cells. We also showed that ALIS was active against intracellular Mabs at a concentration of 32 to 64 mg/L, at 3 and 5 days post-infection. Finally, ALIS intracellular activity was confirmed when tested against 53 clinical Mabs isolates, showing intracellular growth reduction for nearly 80% of the isolates. CONCLUSIONS: Our experiments demonstrate the intracellular localization and intracellular contact between Mabs and ALIS, and antibacterial activity against intracellular Mabs, showing promise for its future use for Mabs pulmonary infections.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium , Humanos , Amicacina/farmacologia , Células Eucarióticas , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Lipossomos , Testes de Sensibilidade Microbiana
4.
PLoS Pathog ; 15(11): e1008069, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31703112

RESUMO

Free-living amoebae are thought to represent an environmental niche in which amoeba-resistant bacteria may evolve towards pathogenicity. To get more insights into factors playing a role for adaptation to intracellular life, we characterized the transcriptomic activities of the emerging pathogen Mycobacterium abscessus in amoeba and murine macrophages (Mϕ) and compared them with the intra-amoebal transcriptome of the closely related, but less pathogenic Mycobacterium chelonae. Data on up-regulated genes in amoeba point to proteins that allow M. abscessus to resist environmental stress and induce defense mechanisms, as well as showing a switch from carbohydrate carbon sources to fatty acid metabolism. For eleven of the most upregulated genes in amoeba and/or Mϕ, we generated individual gene knock-out M. abscessus mutant strains, from which ten were found to be attenuated in amoeba and/or Mϕ in subsequence virulence analyses. Moreover, transfer of two of these genes into the genome of M. chelonae increased the intra-Mϕ survival of the recombinant strain. One knock-out mutant that had the gene encoding Eis N-acetyl transferase protein (MAB_4532c) deleted, was particularly strongly attenuated in Mϕ. Taken together, M. abscessus intra-amoeba and intra-Mϕ transcriptomes revealed the capacity of M. abscessus to adapt to an intracellular lifestyle, with amoeba largely contributing to the enhancement of M. abscessus intra-Mϕ survival.


Assuntos
Amoeba/genética , Macrófagos/metabolismo , Infecções por Mycobacterium não Tuberculosas/genética , Mycobacterium abscessus/patogenicidade , Transcriptoma , Fatores de Virulência/genética , Virulência/genética , Amoeba/crescimento & desenvolvimento , Amoeba/microbiologia , Animais , Proteínas de Bactérias/genética , Macrófagos/microbiologia , Camundongos , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/isolamento & purificação
5.
Proc Natl Acad Sci U S A ; 115(43): E10147-E10156, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30301802

RESUMO

Mycobacterium abscessus is a peculiar rapid-growing Mycobacterium (RGM) capable of surviving within eukaryotic cells thanks to an arsenal of virulence genes also found in slow-growing mycobacteria (SGM), such as Mycobacterium tuberculosis A screen based on the intracellular survival in amoebae and macrophages (MΦ) of an M. abscessus transposon mutant library revealed the important role of MAB_0855, a yet uncharacterized Mycobacterial membrane protein Large (MmpL). Large-scale comparisons with SGM and RGM genomes uncovered MmpL12 proteins as putative orthologs of MAB_0855 and a locus-scale synteny between the MAB_0855 and Mycobacterium chelonae mmpL8 loci. A KO mutant of the MAB_0855 gene, designated herein as mmpL8MAB , had impaired adhesion to MΦ and displayed a decreased intracellular viability. Despite retaining the ability to block phagosomal acidification, like the WT strain, the mmpL8MAB mutant was delayed in damaging the phagosomal membrane and in making contact with the cytosol. Virulence attenuation of the mutant was confirmed in vivo by impaired zebrafish killing and a diminished propensity to induce granuloma formation. The previously shown role of MmpL in lipid transport prompted us to investigate the potential lipid substrates of MmpL8MAB Systematic lipid analysis revealed that MmpL8MAB was required for the proper expression of a glycolipid entity, a glycosyl diacylated nonadecyl diol (GDND) alcohol comprising different combinations of oleic and stearic acids. This study shows the importance of MmpL8MAB in modifying interactions between the bacteria and phagocytic cells and in the production of a previously unknown glycolipid family.


Assuntos
Proteínas de Bactérias/metabolismo , Glicolipídeos/metabolismo , Mycobacterium abscessus/metabolismo , Fatores de Virulência/metabolismo , Virulência/fisiologia , Amoeba/microbiologia , Animais , Transporte Biológico/fisiologia , Linhagem Celular , Citosol/metabolismo , Humanos , Lipídeos , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Camundongos , Fagossomos/microbiologia , Peixe-Zebra/microbiologia
6.
Proc Natl Acad Sci U S A ; 115(5): E1002-E1011, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29343644

RESUMO

Mycobacterium abscessus, a rapidly growing mycobacterium (RGM) and an opportunistic human pathogen, is responsible for a wide spectrum of clinical manifestations ranging from pulmonary to skin and soft tissue infections. This intracellular organism can resist the bactericidal defense mechanisms of amoebae and macrophages, an ability that has not been observed in other RGM. M. abscessus can up-regulate several virulence factors during transient infection of amoebae, thereby becoming more virulent in subsequent respiratory infections in mice. Here, we sought to identify the M. abscessus genes required for replication within amoebae. To this end, we constructed and screened a transposon (Tn) insertion library of an M. abscessus subspecies massiliense clinical isolate for attenuated clones. This approach identified five genes within the ESX-4 locus, which in M. abscessus encodes an ESX-4 type VII secretion system that exceptionally also includes the ESX conserved EccE component. To confirm the screening results and to get further insight into the contribution of ESX-4 to M. abscessus growth and survival in amoebae and macrophages, we generated a deletion mutant of eccB4 that encodes a core structural element of ESX-4. This mutant was less efficient at blocking phagosomal acidification than its parental strain. Importantly, and in contrast to the wild-type strain, it also failed to damage phagosomes and showed reduced signs of phagosome-to-cytosol contact, as demonstrated by a combination of cellular and immunological assays. This study attributes an unexpected and genuine biological role to the underexplored mycobacterial ESX-4 system and its substrates.


Assuntos
Amoeba/microbiologia , Mycobacterium abscessus/patogenicidade , Fagossomos/microbiologia , Sistemas de Secreção Tipo IV/genética , Fatores de Virulência/genética , Proteínas de Bactérias/genética , Caspase 1/metabolismo , Cromatografia em Camada Fina , Citosol/metabolismo , Ativação Enzimática , Citometria de Fluxo , Galectina 3/metabolismo , Deleção de Genes , Genômica , Humanos , Lipídeos/química , Macrófagos/microbiologia , Mutação , Mycobacterium abscessus/genética , Mycobacterium tuberculosis/patogenicidade , Células THP-1 , Virulência
7.
Artigo em Inglês | MEDLINE | ID: mdl-32253217

RESUMO

Mycobacterium abscessus lung infections remain difficult to treat. Recent studies have recognized the power of new combinations of antibiotics, such as bedaquiline and imipenem, although in vitro data have questioned this combination. We report that the efficacy of bedaquiline-imipenem combination treatment relies essentially on the activity of bedaquiline in a C3HeB/FeJ mice model of infection with a rough variant of M. abscessus The addition of imipenem contributed to clearing the infection in the spleen.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Animais , Antibacterianos/farmacologia , Diarilquinolinas , Imipenem/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico
8.
Proc Natl Acad Sci U S A ; 113(29): E4228-37, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27385830

RESUMO

Mycobacterium abscessus (Mabs) is a rapidly growing Mycobacterium and an emerging pathogen in humans. Transitioning from a smooth (S) high-glycopeptidolipid (GPL) producer to a rough (R) low-GPL producer is associated with increased virulence in zebrafish, which involves the formation of massive serpentine cords, abscesses, and rapid larval death. Generating a cord-deficient Mabs mutant would allow us to address the contribution of cording in the physiopathological signs of the R variant. Herein, a deletion mutant of MAB_4780, encoding a dehydratase, distinct from the ß-hydroxyacyl-ACP dehydratase HadABC complex, was constructed in the R morphotype. This mutant exhibited an alteration of the mycolic acid composition and a pronounced defect in cording. This correlated with an extremely attenuated phenotype not only in wild-type but also in immunocompromised zebrafish embryos lacking either macrophages or neutrophils. The abolition of granuloma formation in embryos infected with the dehydratase mutant was associated with a failure to replicate in macrophages, presumably due to limited inhibition of the phagolysosomal fusion. Overall, these results indicate that MAB_4780 is required for Mabs to successfully establish acute and lethal infections. Therefore, targeting MAB_4780 may represent an attractive antivirulence strategy to control Mabs infections, refractory to most standard chemotherapeutic interventions. The combination of a dehydratase assay with a high-resolution crystal structure of MAB_4780 opens the way to identify such specific inhibitors.


Assuntos
Hidroliases/fisiologia , Infecções por Mycobacterium/enzimologia , Mycobacterium/patogenicidade , Proteínas de Peixe-Zebra/fisiologia , Animais , Linhagem Celular , Embrião não Mamífero/enzimologia , Embrião não Mamífero/imunologia , Embrião não Mamífero/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Infecções por Mycobacterium/microbiologia , Neutrófilos/imunologia , Virulência , Peixe-Zebra/imunologia , Peixe-Zebra/metabolismo , Peixe-Zebra/microbiologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-28096155

RESUMO

Mycobacterium abscessus pulmonary infections are treated with a macrolide (clarithromycin or azithromycin), an aminoglycoside (amikacin), and a ß-lactam (cefoxitin or imipenem). The triple combination is used without any ß-lactamase inhibitor, even though Mabscessus produces the broad-spectrum ß-lactamase BlaMab We determine whether inhibition of BlaMab by avibactam improves the activity of imipenem against M. abscessus The bactericidal activity of drug combinations was assayed in broth and in human macrophages. The in vivo efficacy of the drugs was tested by monitoring the survival of infected zebrafish embryos. The level of BlaMab production in broth and in macrophages was compared by quantitative reverse transcription-PCR and Western blotting. The triple combination of imipenem (8 or 32 µg/ml), amikacin (32 µg/ml), and avibactam (4 µg/ml) was bactericidal in broth (<0.1% survival), with 3.2- and 4.3-log10 reductions in the number of CFU being achieved at 72 h when imipenem was used at 8 and 32 µg/ml, respectively. The triple combination achieved significant intracellular killing, with the bacterial survival rates being 54% and 7% with the low (8 µg/ml) and high (32 µg/ml) dosages of imipenem, respectively. In vivo inhibition of BlaMab by avibactam improved the survival of zebrafish embryos treated with imipenem. Expression of the gene encoding BlaMab was induced (20-fold) in the infected macrophages. Inhibition of BlaMab by avibactam improved the efficacy of imipenem against M. abscessusin vitro, in macrophages, and in zebrafish embryos, indicating that this ß-lactamase inhibitor should be clinically evaluated. The in vitro evaluation of imipenem may underestimate the impact of BlaMab, since the production of the ß-lactamase is inducible in macrophages.


Assuntos
Antibacterianos/farmacologia , Compostos Azabicíclicos/farmacologia , Imipenem/farmacologia , Mycobacterium/efeitos dos fármacos , Inibidores de beta-Lactamases/farmacologia , beta-Lactamases/farmacologia , Amicacina/farmacologia , Animais , Western Blotting , Embrião não Mamífero/microbiologia , Humanos , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Mycobacterium/genética , Mycobacterium/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
10.
Infect Immun ; 84(10): 2895-903, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481243

RESUMO

Mycobacterium abscessus is an emerging pathogenic mycobacterium involved in pulmonary and mucocutaneous infections, presenting a serious threat for patients with cystic fibrosis (CF). The lack of an efficient treatment regimen and the emergence of multidrug resistance in clinical isolates require the development of new therapeutic strategies against this pathogen. Reverse genetics has revealed genes that are present in M. abscessus but absent from saprophytic mycobacteria and that are potentially involved in pathogenicity. Among them, MAB_3593 encodes MgtC, a known virulence factor involved in intramacrophage survival and adaptation to Mg(2+) deprivation in several major bacterial pathogens. Here, we demonstrated a strong induction of M. abscessus MgtC at both the transcriptional and translational levels when bacteria reside inside macrophages or upon Mg(2+) deprivation. Moreover, we showed that M. abscessus MgtC was recognized by sera from M. abscessus-infected CF patients. The intramacrophage growth (J774 or THP1 cells) of a M. abscessus knockout mgtC mutant was, however, not significantly impeded. Importantly, our results indicated that inhibition of MgtC in vivo through immunization with M. abscessus mgtC DNA, formulated with a tetrafunctional amphiphilic block copolymer, exerted a protective effect against an aerosolized M. abscessus challenge in CF (ΔF508 FVB) mice. The formulated DNA immunization was likely associated with the production of specific MgtC antibodies, which may stimulate a protective effect by counteracting MgtC activity during M. abscessus infection. These results emphasize the importance of M. abscessus MgtC in vivo and provide a basis for the development of novel therapeutic tools against pulmonary M. abscessus infections in CF patients.


Assuntos
Vacinas Bacterianas/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium/imunologia , Fatores de Virulência/imunologia , Animais , Proteínas de Bactérias/genética , Western Blotting , Fibrose Cística/complicações , Modelos Animais de Doenças , Feminino , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
Infect Immun ; 83(2): 780-91, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25486995

RESUMO

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium involved in pulmonary and cutaneo-mucous infections worldwide, to which cystic fibrosis patients are exquisitely susceptible. The analysis of the genome sequence of M. abscessus showed that this bacterium is endowed with the metabolic pathways typically found in environmental microorganisms that come into contact with soil, plants, and aquatic environments, where free-living amoebae are frequently present. M. abscessus also contains several genes that are characteristically found only in pathogenic bacteria. One of them is MAB_0555, encoding a putative phospholipase C (PLC) that is absent from most other rapidly growing mycobacteria, including Mycobacterium chelonae and Mycobacterium smegmatis. Here, we report that purified recombinant M. abscessus PLC is highly cytotoxic to mouse macrophages, presumably due to hydrolysis of membrane phospholipids. We further showed by constructing and using an M. abscessus PLC knockout mutant that loss of PLC activity is deleterious to M. abscessus intracellular survival in amoebae. The importance of PLC is further supported by the fact that M. abscessus PLC was found to be expressed only in amoebae. Aerosol challenge of mice with M. abscessus strains that were precultured in amoebae enhanced M. abscessus lung infectivity relative to M. abscessus grown in broth culture. Our study underlines the importance of PLC for the virulence of M. abscessus. Despite the difficulties of isolating M. abscessus from environmental sources, our findings suggest that M. abscessus has evolved in close contact with environmental protozoa, which supports the argument that amoebae may contribute to the virulence of opportunistic mycobacteria.


Assuntos
Amoeba/fisiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium/patogenicidade , Fosfolipases Tipo C/fisiologia , Amoeba/microbiologia , Animais , Sequência de Bases , Células Cultivadas , Técnicas de Cocultura , Fibrose Cística/microbiologia , Técnicas de Inativação de Genes , Genoma Bacteriano/genética , Macrófagos/imunologia , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium/enzimologia , Mycobacterium/genética , Infecções por Mycobacterium não Tuberculosas/microbiologia , Proteínas Recombinantes , Análise de Sequência de DNA , Fosfolipases Tipo C/genética , Fatores de Virulência/genética
12.
Antimicrob Agents Chemother ; 58(7): 4054-63, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24798271

RESUMO

Mycobacterium abscessus is responsible for a wide spectrum of clinical syndromes and is one of the most intrinsically drug-resistant mycobacterial species. Recent evaluation of the in vivo therapeutic efficacy of the few potentially active antibiotics against M. abscessus was essentially performed using immunocompromised mice. Herein, we assessed the feasibility and sensitivity of fluorescence imaging for monitoring the in vivo activity of drugs against acute M. abscessus infection using zebrafish embryos. A protocol was developed where clarithromycin and imipenem were directly added to water containing fluorescent M. abscessus-infected embryos in a 96-well plate format. The status of the infection with increasing drug concentrations was visualized on a spatiotemporal level. Drug efficacy was assessed quantitatively by measuring the index of protection, the bacterial burden (CFU), and the number of abscesses through fluorescence measurements. Both drugs were active in infected embryos and were capable of significantly increasing embryo survival in a dose-dependent manner. Protection from bacterial killing correlated with restricted mycobacterial growth in the drug-treated larvae and with reduced pathophysiological symptoms, such as the number of abscesses within the brain. In conclusion, we present here a new and efficient method for testing and compare the in vivo activity of two clinically relevant drugs based on a fluorescent reporter strain in zebrafish embryos. This approach could be used for rapid determination of the in vivo drug susceptibility profile of clinical isolates and to assess the preclinical efficacy of new compounds against M. abscessus.


Assuntos
Claritromicina/uso terapêutico , Imipenem/uso terapêutico , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Micobactérias não Tuberculosas/efeitos dos fármacos , Imagem Óptica/métodos , Animais , Abscesso Encefálico/tratamento farmacológico , Abscesso Encefálico/microbiologia , Farmacorresistência Bacteriana Múltipla , Quimioterapia Combinada , Larva/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/microbiologia , Peixe-Zebra/microbiologia
13.
J Immunol ; 189(3): 1500-9, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22730532

RESUMO

Upon LPS binding, TLR4 activates a MyD88-dependent pathway leading to the transcriptional activation of proinflammatory genes, as well as a MyD88-independent/TRIF-dependent pathway, responsible for the transcriptional induction of IFN-ß. Previous findings delineated that human neutrophils are unable to induce the transcription of IFN-ß in response to TLR4 stimulation. Because neutrophils do not express protein kinase C ε, a molecule recently reported as essential for initiating the MyD88-independent/TRIF-dependent pathway, we optimized an electroporation method to transfect PKCε into neutrophils with very high efficiency. By doing so, a significant IFN-ß mRNA expression was induced, in the absence of LPS stimulation, not only in PKCε-overexpressing neutrophils but also in cells transfected with a series of empty DNA plasmids; however, LPS further upregulated the IFN-ß transcript levels in plasmid-transfected neutrophils, regardless of PKCε overexpression. Phosphoimmunoblotting studies, as well as chromatin immunoprecipitation assays targeting the IFN-ß promoter, revealed that IFN-ß mRNA induction occurred through the cooperative action of IRF3, activated by transfected DNA, and NF-κB, activated by LPS. Additional immunoblotting and coimmunoprecipitation studies revealed that neutrophils constitutively express various cytosolic DNA sensors, including IFN-inducible protein 16, leucine-rich repeat (in Flightless I) interacting protein-1, and DDX41, as well as that IFN-inducible protein 16 is the intracellular receptor recognizing transfected DNA. Consistently, infection of neutrophils with intracellular pathogens, such as Bartonella henselae, Listeria monocytogenes, Legionella pneumophila, or adenovirus type 5, promoted a marked induction of IFN-ß mRNA expression. Taken together, these data raise questions about the role of PKCε in driving the MyD88-independent/TRIF-dependent response and indicate that human neutrophils are able to recognize and respond to microbial cytosolic DNA.


Assuntos
DNA/biossíntese , Interferon beta/biossíntese , Neutrófilos/imunologia , Plasmídeos/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/fisiologia , Ativação Transcricional/imunologia , Regulação para Cima/imunologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Bartonella henselae/genética , Bartonella henselae/imunologia , Células Cultivadas , Citosol/imunologia , DNA/genética , Células HEK293 , Humanos , Interferon beta/genética , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , Transfecção/métodos , Regulação para Cima/genética
14.
Front Microbiol ; 15: 1327723, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38784795

RESUMO

Six genes encoding putative high molecular weight penicillin-binding proteins (Pbp) are present in the genome of the ß-lactam-resistant strain Corynebacterium jeikeium K411. In this study, we show that pbp2c, one of these six genes, is present in resistant strains of Corynebacteriaceae but absent from sensitive strains. The molecular study of the pbp2c locus from C. jeikeium and its heterologous expression in Corynebacterium glutamicum allowed us to show that Pbp2c confers high levels of ß-lactam resistance to the host and is under the control of a ß-lactam-induced regulatory system encoded by two adjacent genes, jk0410 and jk0411. The detection of this inducible resistance may require up to 48 h of incubation, particularly in Corynebacterium amycolatum. Finally, the Pbp2c-expressing strains studied were resistant to all the ß-lactam antibiotics tested, including carbapenems, ceftaroline, and ceftobiprole.

15.
Microbiol Spectr ; 10(3): e0019222, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35583329

RESUMO

The immunoglobulin A (IgA) status of cystic fibrosis (CF) patients, presenting with or without a non-tuberculous mycobacterial (NTM) infection, has to date not been fully elucidated toward two antigenic preparations previously described. We have chosen to determine the clinical values of an IgA ELISA for the diagnosis of NTM and/or Mycobacterium abscessus infections in CF patients. One hundred and 73 sera from CF patients, comprising 33 patients with M. abscessus positive cultures, and 31 non-CF healthy controls were assessed. IgA levels were evaluated by indirect ELISAs using a surface antigenic extract named TLR2eF for TLR2 positive extract and a recombinant protein, the phospholipase C (rMAB_0555 or rPLC). These assays revealed a sensitivity of 52.6% (95% CI = 35.8% to 69%) and 42.1% (95% CI = 26.3% to 59.2%) using TLR2eF and rPLC, respectively, and respective specificities of 92.6% (95% CI = 87.5% to 96.1%) and 92% (95% CI = 86.7% to 95.7%) for samples culture positive for M. abscessus. Overall sensitivity and specificity of 66.7% and 85.4%, respectively, were calculated for IgA detection in M. abscessus-culture positive CF patients, when we combine the results of the two used antigens, thus demonstrating the efficiency in detection of positive cases for these two antigens with IgA isotype. CF patients with a positive culture for M. abscessus had the highest IgA titers against TLR2eF and rPLC. The diagnosis of NTM infections, including those due to M. abscessus, can be improved by the addition of an IgA serological assay, especially when cultures, for example, are negative. Based on these promising results, a serological follow-up of a larger number of patients should be performed to determine if the IgA response may be correlated with an active/acute infection state or a very recent infection. IMPORTANCE Mycobacterium abscessus is currently the most frequently isolated rapid growing mycobacterium in human pathology and the major one involved in lung infections. It has recently emerged as responsible for severe pulmonary infections in patients with cystic fibrosis (CF) or those who have undergone lung transplantation. In addition, it represents the most antibiotic resistant mycobacterial species. However, despite its increasing clinical importance, very little is known about the use of M. abscessus parietal compounds and the host response. This has led to the development of serological tests to measure the antibody response in infected patients, and potentially to link this to the culture of respiratory samples. Herein, we describe an important analysis of the serological IgA response from CF patients, and we demonstrate the full diagnostic usefulness of this assay in the diagnosis of NTM infections, and more particularly M. abscessus, in CF patients.


Assuntos
Fibrose Cística , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Humanos , Imunoglobulina A , Infecções por Mycobacterium não Tuberculosas/diagnóstico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/fisiologia , Micobactérias não Tuberculosas
16.
J Cyst Fibros ; 21(2): 353-360, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34511392

RESUMO

BACKGROUND: Culture conditions sometimes make it difficult to detect non-tuberculous mycobacteria (NTM), particularly Mycobacterium abscessus, an emerging cystic fibrosis (CF) pathogen. The diagnosis of NTM positive cases not detected by classical culture methods might benefit from the development of a serological assay. METHODS: As part of a diagnostic accuracy study, a total of 173 sera CF-patients, including 33 patients with M. abscessus positive cultures, and 31 non-CF healthy controls (HC) were evaluated. Four M. abscessus antigens were used separately, comprising two surface extracts (Interphase (INP) and a TLR2 positive extract (TLR2eF)) and two recombinant proteins (rMAB_2545c and rMAB_0555 also known as the phospholipase C (rPLC)). RESULTS: TLR2eF and rPLC were the most efficient antigens to discriminate NTM-culture positive CF-patients from NTM-culture negative CF-patients. The best clinical values were obtained for the detection of M. abscessus-culture positive CF-patients; with sensitivities for the TLR2eF and rPLC of 81.2% (95% CI:65.7-92.3%) and 87.9% (95% CI:71.9-95.6%) respectively, and specificities of 88.9% (95% CI:85.3-94.8%) and 84.8% (95% CI:80.6-91.5%) respectively. When considering as positive all sera, giving a positive response in at least one of the two tests, and, as negative, all sera negative for both tests, we obtained a sensitivity of 93.9% and a specificity of 80.7% for the detection of M. abscessus-culture positive CF-patients. CONCLUSION: High antibody titers against TLR2eF and rPLC were obtained in M. abscessus-culture positive CF-patients, allowing us to consider these serological markers as potential tools in the detection of CF-patients infected with M. abscessus.


Assuntos
Fibrose Cística , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Biomarcadores , Fibrose Cística/complicações , Fibrose Cística/diagnóstico , Humanos , Infecções por Mycobacterium não Tuberculosas/diagnóstico , Micobactérias não Tuberculosas
17.
J Immunol ; 181(9): 6563-73, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18941247

RESUMO

Neutrophils, historically known for their involvement in acute inflammation, are also targets for infection by many different DNA and RNA viruses. However, the mechanisms by which they recognize and respond to viral components are poorly understood. Polyinosinic:polycytidylic acid (poly(I:C)) is a synthetic mimetic of viral dsRNA that is known to interact either with endosomal TLR3 (not expressed by human neutrophils) or with cytoplasmic RNA helicases such as melanoma differentiation-associated gene 5 (MDA5) and retinoic acid-inducible gene I (RIG-I). In this study, we report that intracellularly administered poly(I:C) stimulates human neutrophils to specifically express elevated mRNA levels encoding type I IFNs, immunoregulatory cytokines, and chemokines, such as TNF-alpha, IL-12p40, CXCL10, CXCL8, CCL4, and CCL20, as well as classical IFN-responsive genes (IRG), including IFIT1 (IFN-induced protein with tetratricopeptide repeats 1)/IFN-stimulated gene (ISG)56, G1P2/ISG15, PKR (dsRNA-dependent protein kinase), and IFN-regulatory factor (IRF)7. Investigations into the mechanisms whereby transfected poly(I:C) promotes gene expression in neutrophils uncovered a crucial involvement of the MAPK-, PKR-, NF-kappaB-, and TANK (TNF receptor-associated NF-kappaB kinase)-binding kinase (TBK1)/IRF3-signaling transduction pathways, as illustrated by the use of specific pharmacological inhibitors. Consistent with the requirement of the cytoplasmic dsRNA pathway for antiviral signaling, human neutrophils were found to constitutively express significant levels of both MDA5 and RIG-I, but not TLR3. Accordingly, neutrophils isolated from MDA5-deficient mice had a partial impairment in the production of IFN-beta and TNF-alpha upon infection with encephalomyocarditis virus. Taken together, our data demonstrate that neutrophils are able to activate antiviral responses via helicase recognition, thus acting at the frontline of immunity against viruses.


Assuntos
Regulação Viral da Expressão Gênica/imunologia , Genes Virais/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Neutrófilos/virologia , Poli I-C/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Humanos , Fatores Reguladores de Interferon/genética , Interferon beta/genética , Líquido Intracelular/enzimologia , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Camundongos , Camundongos Knockout , Ativação de Neutrófilo/genética , Neutrófilos/enzimologia , Poli I-C/administração & dosagem , RNA Helicases/fisiologia , Transfecção
18.
Artigo em Inglês | MEDLINE | ID: mdl-32984067

RESUMO

Mycobacterium abscessus is a prevalent pathogenic mycobacterium in cystic fibrosis (CF) patients and one of the most highly drug resistant mycobacterial species to antimicrobial agents. It possesses the property to transition from a smooth (S) to a rough (R) morphotype, thereby influencing the host innate immune response. This transition from the S to the R morphotype takes place in patients with an exacerbation of the disease and a persistence of M. abscessus. We have previously shown that the exacerbation of the Toll-like receptor 2 (TLR2)-mediated inflammatory response, following this S to R transition, is essentially due to overproduction of bacilli cell envelope surface compounds, which we were able to extract by mechanical treatment and isolation by solvent partition in a fraction called interphase. Here, we set up a purification procedure guided by bioactivity to isolate a fraction from the R variant of M. abscessus cells which exhibits a high TLR2 stimulating activity, referred to as TLR2-enriched fraction (TLR2eF). As expected, TLR2eF was found to contain several lipoproteins and proteins known to be stimuli for TLR2. Vaccination with TLR2eF showed no protection toward an M. abscessus aerosol challenge, but provided mild protection in ΔF508 mice and their FVB littermates when intravenously challenged by M. abscessus. Interestingly however, antibodies against TLR2eF compounds were detected during disease in CF patients. In conclusion, we show the potential for compounds in TLR2eF as vaccine and diagnostic candidates, in order to enhance diagnosis, prevent and/or treat M. abscessus-related infections.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium , Vacinas , Animais , Humanos , Camundongos , Infecções por Mycobacterium não Tuberculosas/diagnóstico , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Receptor 2 Toll-Like
19.
ChemistryOpen ; 9(3): 351-365, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32211280

RESUMO

Non-tuberculous mycobacterium (NTM) infections, such as those caused by Mycobacterium abscessus, are increasing globally. Due to their intrinsic drug resistance, M. abscessus pulmonary infections are often difficult to cure using standard chemotherapy. We previously demonstrated that a piperidinol derivative, named PIPD1, is an efficient molecule both against M. abscessus and Mycobacterium tuberculosis, the agent of tuberculosis, by targeting the mycolic acid transporter MmpL3. These results prompted us to design and synthesize a series of piperidinol derivatives and to determine the biological activity against M. abscessus. Structure-activity relationship (SAR) studies pointed toward specific sites on the scaffold that can tolerate slight modifications. Overall, these results identified FMD-88 as a new promising active analogue against M. abscessus. Also, we determined the pharmacokinetics properties of PIPD1 and showed that intraperitoneal administration of this compound resulted in promising serum concentration and an elimination half-life of 3.2 hours.


Assuntos
Antituberculosos/química , Mycobacterium abscessus/efeitos dos fármacos , Tuberculose/tratamento farmacológico , Antituberculosos/farmacocinética , Transporte Biológico , Humanos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Ácidos Micólicos/metabolismo , Relação Estrutura-Atividade
20.
Mol Immunol ; 45(9): 2499-507, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18289677

RESUMO

We have recently expressed and characterized the product of the gene Rv2108 of Mycobacterium tuberculosis (MT), the p27 protein. Here, we investigated the immune responses against the p27 protein in the context of different pathogen associated molecular patterns (PAMPs). Different immunization protocols were used. BALB/c mice were immunized either with the p27 recombinant protein in Freund's adjuvant or in phosphate saline buffer (PBS), with a pcDNA3 plasmid containing the gene encoding the p27 protein, or with the Escherichia coli bacteria expressing the p27 protein genetically fused into the flagellin. We found that p27 expressed into the flagellin led to the strongest cellular responses, where we obtained the highest production of IFN-gamma and cell proliferation, an indication of specific Th1-like orientation of the immune response. We confirmed the role of flagellin in this response by using different immunization combinations.


Assuntos
Adjuvantes Imunológicos , Antígenos de Bactérias/metabolismo , Flagelina/imunologia , Lipoproteínas/imunologia , Mycobacterium tuberculosis/imunologia , Células Th1/imunologia , Animais , Antígenos de Bactérias/imunologia , Feminino , Adjuvante de Freund , Imunização , Interferon gama/biossíntese , Interferon gama/imunologia , Lipoproteínas/isolamento & purificação , Lipoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Células Th1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA