Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34725157

RESUMO

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Fímbrias Bacterianas/fisiologia , Neisseria meningitidis/fisiologia , Animais , Anticorpos/uso terapêutico , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Fímbrias Bacterianas/química , Fímbrias Bacterianas/ultraestrutura , Humanos , Infecções Meningocócicas/tratamento farmacológico , Camundongos SCID
2.
Respirology ; 28(6): 525-532, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36889358

RESUMO

BACKGROUND AND OBJECTIVE: Dynamic hyperinflation (DH) is a major marker of exertional dyspnoea in severe emphysema. We hypothesized that bronchoscopic lung volume reduction (BLVR) using endobronchial valves (EBVs) decreases DH. METHODS: In this prospective bi-centre study from both Toulouse and Limoges Hospitals, we assessed DH during an incremental cycle ergometry before and 3 months after EBVs treatment. The primary objective was to observe the change in inspiratory capacity (IC) at isotime. Target lobe volume reduction (TLVR) and changes in residual volume (RV), forced expiratory volume in one-second (FEV1 ), mMRC, 6 minutes walking distance (6MWD), BODE and other dynamic measures like tele-expiratory volume (EELV) were also analysed. RESULTS: Thirty-nine patients were included, of whom thirty-eight presented DH. IC and EELV at isotime significantly improved (+214 mL, p = 0.004; -713 mL, p Ë‚ 0.001, respectively). Mean changes were +177 mL for FEV1 (+19%, p < 0.001), -600 mL for RV (p < 0.0001), +33 m for 6MWD (p < 0.0001), respectively. Patients who responded on RV (>430 mL decrease) and FEV1 (>12% gain) had better improvements compared to non-responders (+368 mL vs. +2 mL; +398 mL vs. -40 mL IC isotime, respectively). On the opposite, in patients who responded on DH (>200 mL IC isotime increase), changes in TLV (-1216 mL vs. -576 mL), FEV1 (+261 mL vs. +101 mL), FVC (+496 mL vs. +128 mL) and RV (-805 mL vs. -418 mL) were greater compared to non-responders. CONCLUSIONS: DH decreases after EBVs treatment, and this improvement is correlated with static changes.


Assuntos
Pneumonectomia , Enfisema Pulmonar , Humanos , Pneumonectomia/métodos , Estudos Prospectivos , Enfisema Pulmonar/cirurgia , Medidas de Volume Pulmonar , Volume Expiratório Forçado , Resultado do Tratamento , Broncoscopia/métodos
3.
J Infect Dis ; 226(7): 1276-1285, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-35524969

RESUMO

BACKGROUND: Staphylococcus aureus dominates the lung microbiota of children with cystic fibrosis (CF) and persistent clones are able to establish chronic infection for years, having a direct deleterious impact on lung function. However, in this context, the exact contribution of S. aureus to the decline in respiratory function in children with CF is not elucidated. METHODS: To investigate the contribution of persistent S. aureus clones in CF disease, we undertook the analysis of sequential isogenic isolates recovered from 15 young CF patients. RESULTS: Using an air-liquid infection model, we observed a strong correlation between S. aureus adaption in the lung (late isolates), low toxicity, and proinflammatory cytokine secretion. Conversely, early isolates appeared to be highly cytotoxic but did not promote cytokine secretion. We found that cytokine secretion was dependent on staphylococcal protein A (Spa), which was selectively expressed in late compared to early isolates as a consequence of dysfunctional agr quorum-sensing system. Finally, we demonstrated the involvement of TNF-α receptor 1 signaling in the inflammatory response of airway epithelial cells to these lung-adapted S. aureus isolates. CONCLUSIONS: Our results suggest an unexpected direct role of bacterial lung adaptation in the progression of chronic lung disease by promoting a proinflammatory response through acquired agr dysfunction.


Assuntos
Fibrose Cística , Infecções Estafilocócicas , Criança , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Humanos , Pulmão/metabolismo , Infecções Estafilocócicas/microbiologia , Proteína Estafilocócica A , Staphylococcus aureus/fisiologia , Fator de Necrose Tumoral alfa
4.
J Infect Dis ; 220(12): 1967-1976, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31420648

RESUMO

Staphylococcus aureus is a leading cause of both acute and chronic infections in humans. The importance of the pentose phosphate pathway (PPP) during S. aureus infection is currently largely unexplored. In the current study, we focused on one key PPP enzyme, transketolase (TKT). We showed that inactivation of the unique gene encoding TKT activity in S. aureus USA300 (∆tkt) led to drastic metabolomic changes. Using time-lapse video imaging and mice infection, we observed a major defect of the ∆tkt strain compared with wild-type strain in early intracellular proliferation and in the ability to colonize kidneys. Transcriptional activity of the 2 master regulators sigma B and RpiRc was drastically reduced in the ∆tkt mutant during host cells invasion. The concomitant increased RNAIII transcription suggests that TKT-or a functional PPP-strongly influences the ability of S. aureus to proliferate within host cells by modulating key transcriptional regulators.


Assuntos
Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Estresse Fisiológico , Transcetolase/metabolismo , Animais , Carbono/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Regulação Bacteriana da Expressão Gênica , Inativação Gênica , Genes Bacterianos , Humanos , Rim/metabolismo , Rim/microbiologia , Metabolômica/métodos , Camundongos , Mutação , Fenótipo , Transdução de Sinais , Staphylococcus aureus/enzimologia , Estresse Fisiológico/genética , Transcetolase/genética
5.
Clin Infect Dis ; 69(11): 1937-1945, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30753350

RESUMO

BACKGROUND: Chronic lung infection in cystic fibrosis (CF) patients by Staphylococcus aureus is a well-established epidemiological fact. Indeed, S. aureus is the most commonly identified pathogen in the lungs of CF patients. Improving our understanding of the mechanisms associated with the persistence of S. aureus is therefore an important issue. METHODS: We selected pairs of sequential S. aureus isolates from 3 patients with CF and from 1 patient with non-CF chronic lung disease. We used a combination of genomic, proteomic, and metabolomic approaches with functional assays for in-depth characterization of S. aureus long-term persistence. RESULTS: In this study, we show that late S. aureus isolates from CF patients have an increased ability for intracellular survival in CF bronchial epithelial-F508del cells compared to ancestral early isolates. Importantly, the increased ability to persist intracellularly was confirmed for S. aureus isolates within the own-patient F508del epithelial cells. An increased ability to form biofilm was also demonstrated. Furthermore, we identified the underlying genetic modifications that induce altered protein expression profiles and notable metabolic changes. These modifications affect several metabolic pathways and virulence regulators that could constitute therapeutic targets. CONCLUSIONS: Our results strongly suggest that the intracellular environment might constitute an important niche of persistence and relapse necessitating adapted antibiotic treatments.


Assuntos
Staphylococcus aureus/efeitos dos fármacos , Adaptação Fisiológica/efeitos dos fármacos , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Cromatografia Líquida , Humanos , Proteogenômica/métodos , Proteômica/métodos , Espectrometria de Massas em Tandem
6.
Int J Mol Sci ; 20(12)2019 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242705

RESUMO

In women, oral menopausal hormonal therapy (MHT) is associated with adverse effects including an increased incidence of thromboembolic events, classically attributed to an increase in several liver-derived coagulation factors due to hepatic first pass. While platelets are central players in thrombus constitution, their implication in women treated with estrogens remains incompletely characterized. Platelets and their medullar progenitors, megakaryocytes, express estrogen receptors (ER) that may explain, at least in part, a sensitivity to hormonal changes. The purpose of this review is to summarize our current knowledge of estrogen actions on platelets and megakaryocytes in mice following in vivo administration and in women using MHT.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Estrogênios/farmacologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Animais , Estrogênios/uso terapêutico , Feminino , Humanos , Ativação Plaquetária/efeitos dos fármacos , Fatores Sexuais , Trombopoese/efeitos dos fármacos
7.
Respiration ; 96(6): 500-506, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29428946

RESUMO

BACKGROUND: Cardiac output (CO) is a prognostic factor in pulmonary hypertension (PH). Right heart catheterisation using the direct Fick method or thermodilution (TD) is the reference technique for CO measurement. Impedance cardiography (IPc) is a known non-invasive method of measuring CO. OBJECTIVES: In our study, we assume that the measurement of CO by IPc using the PHYSIOFLOW® system is as accurate as TD or using the direct Fick method in patients with PH in group 1 or group 4. METHODS: A total of 75 patients were enrolled in a prospective study carried out at the hypertension reference centre of Toulouse University Hospital. Right heart catheterisation was performed for the diagnosis or follow-up of the disease. CO was measured using the Fick method, TD, and IPc simultaneously. A Bland-Altman analysis was plotted. RESULTS: CO was 5.7 ± 1.9 L/min as measured by the Fick method, 5.4 ± 1.5 L/min by TD, and 5.5 ± 1.7 L/min by IPc. The bias between CO measurements by IPc and the direct Fick method was 0.149 L/min (95% CI, -0.298 to 0.596). The bias between CO measurements by IPc and the TD method was -0.153 L/min (95% CI, -0.450 to 0.153). The correlation decreased with the more extreme CO values (< 3 L/min or > 7 L/min). A few factors changed the agreement between measurements (BMI or membership in group 4). CONCLUSION: To conclude, this study shows that the measurement of CO by IPc in PH patients is reliable compared to the direct Fick method and TD obtained by right heart catheterisation. This accuracy decreases for extreme CO values.


Assuntos
Débito Cardíaco , Cardiografia de Impedância , Hipertensão Pulmonar/fisiopatologia , Idoso , Feminino , Humanos , Hipertensão Pulmonar/diagnóstico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Termodiluição
8.
Mol Cell Proteomics ; 14(4): 870-81, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25616868

RESUMO

Upon entry into mammalian host cells, the pathogenic bacterium Francisella must import host cell arginine to multiply actively in the host cytoplasm. We identified and functionally characterized an arginine transporter (hereafter designated ArgP) whose inactivation considerably delayed bacterial phagosomal escape and intracellular multiplication. Intramacrophagic growth of the ΔargP mutant was fully restored upon supplementation of the growth medium with excess arginine, in both F. tularensis subsp. novicida and F. tularensis subsp. holarctica LVS, demonstrating the importance of arginine acquisition in these two subspecies. High-resolution mass spectrometry revealed that arginine limitation reduced the amount of most of the ribosomal proteins in the ΔargP mutant. In response to stresses such as nutritional limitation, repression of ribosomal protein synthesis has been observed in all kingdoms of life. Arginine availability may thus contribute to the sensing of the intracellular stage of the pathogen and to trigger phagosomal egress. All MS data have been deposited in the ProteomeXchange database with identifier PXD001584 (http://proteomecentral.proteomexchange.org/dataset/PXD001584).


Assuntos
Arginina/metabolismo , Francisella/metabolismo , Interações Hospedeiro-Patógeno , Fagossomos/microbiologia , Proteínas Ribossômicas/metabolismo , Animais , Autofagia , Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/imunologia , Análise por Conglomerados , Citosol/metabolismo , Feminino , Francisella/patogenicidade , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/ultraestrutura , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Modelos Biológicos , Mutação/genética , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Transporte Proteico , Proteoma/metabolismo , Estresse Fisiológico , Frações Subcelulares/metabolismo , Virulência
9.
Mol Microbiol ; 98(3): 518-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26192619

RESUMO

Intracellular multiplication and dissemination of the infectious bacterial pathogen Francisella tularensis implies the utilization of multiple host-derived nutrients. Here, we demonstrate that gluconeogenesis constitutes an essential metabolic pathway in Francisella pathogenesis. Indeed, inactivation of gene glpX, encoding the unique fructose 1,6-bisphosphatase of Francisella, severely impaired bacterial intracellular multiplication when cells were supplemented by gluconeogenic substrates such as glycerol or pyruvate. The ΔglpX mutant also showed a severe virulence defect in the mouse model, confirming the importance of this pathway during the in vivo life cycle of the pathogen. Isotopic profiling revealed the major role of the Embden-Meyerhof (glycolysis) pathway in glucose catabolism in Francisella and confirmed the importance of glpX in gluconeogenesis. Altogether, the data presented suggest that gluconeogenesis allows Francisella to cope with the limiting glucose availability it encounters during its infectious cycle by relying on host amino acids. Hence, targeting the gluconeogenic pathway might constitute an interesting therapeutic approach against this pathogen.


Assuntos
Francisella tularensis/metabolismo , Animais , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Genes Bacterianos , Gluconeogênese , Células Hep G2 , Humanos , Espectrometria de Massas , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos BALB C , Tularemia/microbiologia , Virulência
11.
PLoS Pathog ; 10(1): e1003893, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453979

RESUMO

Intracellular bacterial pathogens have developed a variety of strategies to avoid degradation by the host innate immune defense mechanisms triggered upon phagocytocis. Upon infection of mammalian host cells, the intracellular pathogen Francisella replicates exclusively in the cytosolic compartment. Hence, its ability to escape rapidly from the phagosomal compartment is critical for its pathogenicity. Here, we show for the first time that a glutamate transporter of Francisella (here designated GadC) is critical for oxidative stress defense in the phagosome, thus impairing intra-macrophage multiplication and virulence in the mouse model. The gadC mutant failed to efficiently neutralize the production of reactive oxygen species. Remarkably, virulence of the gadC mutant was partially restored in mice defective in NADPH oxidase activity. The data presented highlight links between glutamate uptake, oxidative stress defense, the tricarboxylic acid cycle and phagosomal escape. This is the first report establishing the role of an amino acid transporter in the early stage of the Francisella intracellular lifecycle.


Assuntos
Ciclo do Ácido Cítrico , Francisella tularensis/metabolismo , Ácido Glutâmico/metabolismo , Macrófagos/microbiologia , Fagossomos/metabolismo , Tularemia/metabolismo , Sistema X-AG de Transporte de Aminoácidos/genética , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Ácido Glutâmico/genética , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mutação , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Fagossomos/genética , Fagossomos/microbiologia , Fagossomos/patologia , Tularemia/genética
12.
Cell Microbiol ; 16(3): 434-49, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24134488

RESUMO

In order to develop a successful infectious cycle, intracellular bacterial pathogens must be able to adapt their metabolism to optimally utilize the nutrients available in the cellular compartments and tissues where they reside. Francisella tularensis, the agent of the zoonotic disease tularaemia, is a highly infectious bacterium for a large number of animal species. This bacterium replicates in its mammalian hosts mainly in the cytosol of infected macrophages. We report here the identification of a novel amino acid transporter of the major facilitator superfamily of secondary transporters that is required for bacterial intracellular multiplication and systemic dissemination. We show that inactivation of this transporter does not affect phagosomal escape but prevents multiplication in the cytosol of all cell types tested. Remarkably, the intracellular growth defect of the mutant was fully and specifically reversed by addition of asparagine or asparagine-containing dipeptides as well as by simultaneous addition of aspartic acid and ammonium. Importantly, bacterial virulence was also restored in vivo, in the mouse model, by asparagine supplementation. This work unravels thus, for the first time, the importance of asparagine for cytosolicmultiplication of Francisella. Amino acid transporters are likely to constitute underappreciated players in bacterial intracellular parasitism.


Assuntos
Sistemas de Transporte de Aminoácidos/genética , Asparagina/metabolismo , Proteínas de Bactérias/genética , Francisella tularensis/crescimento & desenvolvimento , Compostos de Amônio/farmacologia , Animais , Asparagina/farmacologia , Ácido Aspártico/metabolismo , Ácido Aspártico/farmacologia , Proteínas de Bactérias/farmacocinética , Linhagem Celular Tumoral , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidade , Células Hep G2 , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fagossomos/microbiologia , Tularemia/microbiologia
13.
Mol Cell Proteomics ; 12(8): 2278-92, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23669032

RESUMO

Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularemia. In vivo, this facultative intracellular bacterium survives and replicates mainly in the cytoplasm of infected cells. We have recently identified a genetic locus, designated moxR that is important for stress resistance and intramacrophage survival of F. tularensis. In the present work, we used tandem affinity purification coupled to mass spectrometry to identify in vivo interacting partners of three proteins encoded by this locus: the MoxR-like ATPase (FTL_0200), and two proteins containing motifs predicted to be involved in protein-protein interactions, bearing von Willebrand A (FTL_0201) and tetratricopeptide (FTL_0205) motifs. The three proteins were designated here for simplification, MoxR, VWA1, and TPR1, respectively. MoxR interacted with 31 proteins, including various enzymes. VWA1 interacted with fewer proteins, but these included the E2 component of 2-oxoglutarate dehydrogenase and TPR1. The protein TPR1 interacted with one hundred proteins, including the E1 and E2 subunits of both oxoglutarate and pyruvate dehydrogenase enzyme complexes, and their common E3 subunit. Remarkably, chromosomal deletion of either moxR or tpr1 impaired pyruvate dehydrogenase and oxoglutarate dehydrogenase activities, supporting the hypothesis of a functional role for the interaction of MoxR and TPR1 with these complexes. Altogether, this work highlights possible links between stress resistance and metabolism in F. tularensis virulence.


Assuntos
Proteínas de Bactérias/metabolismo , Ciclo do Ácido Cítrico/fisiologia , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidade , Estresse Fisiológico , Sequência de Aminoácidos , Animais , DNA Bacteriano/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sequência de DNA , Tularemia
14.
Med Sci (Paris) ; 39(1): 58-63, 2023 Jan.
Artigo em Francês | MEDLINE | ID: mdl-36692321

RESUMO

Increased life expectancy in cystic fibrosis has made transition from pediatric to adult cystic fibrosis centers a crucial step for patients, their families and caregivers. This transition must be gradual and carefully prepared. A formalized process, early discussion with patients and families about transition, patient's empowerment prior to transfer, and close links between pediatric and adult teams are key points to succeed. Therapeutic education, validated questionnaires, personalized action plans or connected tools can help. Transfer will take place at the appropriate time for each patient, ideally during a period of disease stability, in a progressive manner, with joint or alternating consultations between pediatric and adult cystic fibrosis center teams. Other chronic respiratory diseases with pediatric onset may benefit from similar transition processes.


Title: Transition des patients atteints d'une maladie respiratoire chronique depuis la pédiatrie vers les services pour adultes - L'exemple de la mucoviscidose. Abstract: Du fait de l'augmentation de l'espérance de vie des individus atteints de mucoviscidose, la transition de la pédiatrie vers les services de médecine pour adultes est devenue une étape essentielle pour les patients, les aidants et les soignants. Cette transition doit être progressive et longuement préparée. Avoir un processus établi et formalisé, aborder tôt le thème de la transition avec le patient et sa famille, autonomiser le patient avant le transfert et établir des liens étroits entre les structures médicales pédiatriques et pour adultes sont des éléments importants pour la réussite de cette étape. L'éducation thérapeutique ainsi que l'utilisation de questionnaires validés, d'un plan d'action personnalisé ou d'outils connectés peuvent aider. Le transfert se fera au moment le mieux adapté pour le patient, idéalement en période de stabilité de la maladie, de manière progressive, avec des consultations conjointes ou alternées entre les équipes de pédiatrie et de médecine pour adultes. D'autres maladies respiratoires chroniques débutant dans l'enfance ou à l'adolescence pourront bénéficier d'un processus de transition similaire.


Assuntos
Fibrose Cística , Transtornos Respiratórios , Transição para Assistência do Adulto , Humanos , Adulto , Criança , Fibrose Cística/terapia , Encaminhamento e Consulta , Pacientes
15.
Nat Commun ; 14(1): 8135, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38065959

RESUMO

Staphylococcus aureus is a predominant cause of chronic lung infections. While the airway environment is rich in highly sialylated mucins, the interaction of S. aureus with sialic acid is poorly characterized. Using S. aureus USA300 as well as clinical isolates, we demonstrate that quorum-sensing dysfunction, a hallmark of S. aureus adaptation, correlates with a greater ability to consume free sialic acid, providing a growth advantage in an air-liquid interface model and in vivo. Furthermore, RNA-seq experiment reveals that free sialic acid triggers transcriptional reprogramming promoting S. aureus chronic lifestyle. To support the clinical relevance of our results, we show the co-occurrence of S. aureus, sialidase-producing microbiota and free sialic acid in the airway of patients with cystic fibrosis. Our findings suggest a dual role for sialic acid in S. aureus airway infection, triggering virulence reprogramming and driving S. aureus adaptive strategies through the selection of quorum-sensing dysfunctional strains.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Percepção de Quorum/genética , Ácido N-Acetilneuramínico , Sistema Respiratório , Proteínas de Bactérias
16.
ERJ Open Res ; 8(2)2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35415190

RESUMO

Bilateral endobronchial valves treatment can lead to additional significant benefits in patients who respond to the first procedure and have a clear target lobe on both lungs but increases the rate of complications https://bit.ly/3J0mZ8h.

17.
Infect Immun ; 79(4): 1428-39, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21245269

RESUMO

Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularemia. This facultative intracellular bacterium replicates in vivo mainly inside macrophages and therefore has developed strategies to resist this stressful environment. Here, we identified a novel genetic locus that is important for stress resistance and intracellular survival of F. tularensis. In silico and transcriptional analyses suggest that this locus (genes FTL_0200 to FTL_0209 in the live vaccine strain [LVS]) constitutes an operon controlled by the alternative sigma factor σ³². The first gene, FTL_0200, encodes a putative AAA+ ATPase of the MoxR subfamily. Insertion mutagenesis into genes FTL_0200, FTL_0205, and FTL_0206 revealed a role for the locus in both intracellular multiplication and in vivo survival of F. tularensis. Deletion of gene FTL_0200 led to a mutant bacterium with increased vulnerability to various stress conditions, including oxidative and pH stresses. Proteomic analyses revealed a pleiotropic impact of the ΔFTL_0200 deletion, supporting a role as a chaperone for FTL_0200. This is the first report of a role for a MoxR family member in bacterial pathogenesis. This class of proteins is remarkably conserved among pathogenic species and may thus constitute a novel player in bacterial virulence.


Assuntos
Francisella tularensis/genética , Francisella tularensis/patogenicidade , Genes Bacterianos/genética , Chaperonas Moleculares/genética , Estresse Fisiológico/genética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Southern Blotting , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tularemia/genética , Tularemia/metabolismo , Virulência/genética
18.
PLoS Pathog ; 5(1): e1000284, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19158962

RESUMO

Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularemia. Its ability to multiply and survive in macrophages is critical for its virulence. By screening a bank of HimarFT transposon mutants of the F. tularensis live vaccine strain (LVS) to isolate intracellular growth-deficient mutants, we selected one mutant in a gene encoding a putative gamma-glutamyl transpeptidase (GGT). This gene (FTL_0766) was hence designated ggt. The mutant strain showed impaired intracellular multiplication and was strongly attenuated for virulence in mice. Here we present evidence that the GGT activity of F. tularensis allows utilization of glutathione (GSH, gamma-glutamyl-cysteinyl-glycine) and gamma-glutamyl-cysteine dipeptide as cysteine sources to ensure intracellular growth. This is the first demonstration of the essential role of a nutrient acquisition system in the intracellular multiplication of F. tularensis. GSH is the most abundant source of cysteine in the host cytosol. Thus, the capacity this intracellular bacterial pathogen has evolved to utilize the available GSH, as a source of cysteine in the host cytosol, constitutes a paradigm of bacteria-host adaptation.


Assuntos
Cisteína/metabolismo , Dipeptídeos/metabolismo , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidade , Glutationa/metabolismo , Viabilidade Microbiana , Animais , Linhagem Celular , Citosol/metabolismo , Escherichia coli/genética , Feminino , Francisella tularensis/genética , Genes Bacterianos , Teste de Complementação Genética , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Plasmídeos , Virulência , gama-Glutamiltransferase/genética , gama-Glutamiltransferase/metabolismo
19.
Int J Chron Obstruct Pulmon Dis ; 16: 1957-1965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234426

RESUMO

Background: Since successful development of endobronchial valves (EBV) as treatment for severe emphysema, its main complication, pneumothorax, remains an important concern. Objective: We hypothesized that a two-step EBV implantation, during two distinct iterative procedures could lead to a more progressive target lobe volume reduction (TLVR) and thus ipsilateral lobe re-expansion, resulting in a significant decrease in the pneumothorax rate. Methods: This retrospective bi-center study carried out by Limoges and Toulouse University Hospitals included patients following the inclusion criteria established by the BLVR expert panel. All patients were treated by two distinct procedures: first, EBVs were placed in all but the most proximal segment or sub-segment. The remaining segment was treated subsequently. All patients had a complete evaluation before treatment, and 3 months after the second procedure. Results: Out of 58 patients included, only 4 pneumothoraxes (7%) occurred during the study. The other complications were pneumonia and severe COPD exacerbation (8.6% and 13.7% of patients, respectively). Significant improvement was found for FEV1 (+19.6 ± 25%), RV (-468 ± 960mL), 6MWD (30 ± 85m), BODE Index (-1.4 ± 1.8 point) and TLVR (50.6 ± 35.1%). Significant TLVR (MCID) was obtained in 74.1% of patients (43/58). Conclusion: This new approach using EBV could reduce the incidence of pneumothorax without increasing other complication rates. Clinical and physiological outcomes are similar to those reported in studies using the conventional single-step treatment.


Assuntos
Pneumotórax , Doença Pulmonar Obstrutiva Crônica , Enfisema Pulmonar , Broncoscopia , Volume Expiratório Forçado , Humanos , Pneumonectomia/efeitos adversos , Pneumotórax/diagnóstico por imagem , Pneumotórax/etiologia , Pneumotórax/prevenção & controle , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/cirurgia , Enfisema Pulmonar/diagnóstico por imagem , Enfisema Pulmonar/cirurgia , Estudos Retrospectivos , Resultado do Tratamento
20.
ESC Heart Fail ; 8(4): 2513-2526, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34102018

RESUMO

AIMS: Optimizing medical cardiac treatment for sleep apnoea (SA) in patients with chronic heart failure and reduced ejection fraction (HFrEF) is an expert Grade C recommendation based on six studies encompassing a total of 67 patients only. Whether sacubitril-valsartan (SV), a cornerstone of HFrEF medical treatment, impacts SA is unknown and requires evaluation. METHODS AND RESULTS: The ENTRESTO-SAS trial is a six-centre, prospective, open-label real-life cohort study (NCT02916160). Ambulatory patients eligible for SV (i.e. HFrEF adults who remain symptomatic despite optimal treatment) were evaluated before and after 3 months of SV (including nocturnal ventilatory polygraphy); 118 patients were final analysed [median age was 66 (IQ25-75 : 56-73) years, 81.4% male, 36.5% New York Heart Association III-IV, N-terminal pro-B-type natriuretic peptide level of 1564 (701-3376) ng/L, left ventricular ejection fraction of 30 (25-34)%, 60.7% ischaemic HFrEF, 97.5% initially treated with angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers, 83.9% with beta-blockers, 64.4% with mineralocorticoid receptor antagonists, and 74.6% with diuretics]. Three groups were defined according to initial central/obstructive apnoea-hypopnoea indices (AHIs): G1 (n = 49, AHIcentral  ≥ 5/h and AHIobstructive  < 15/h); G2 (n = 27, AHIobstructive  ≥ 15/h); and G3 (n = 42, AHIcentral  < 5/h and AHIobstructive  < 15/h). At 3 months, the AHI (main predefined outcome) decreased significantly by -7.10/h (IQ25-75 : -16.10 to 0.40; P < 0.001) in G1 + G2 without positive airway pressure treatment (45 patients, median initial AHI of 24.20 (IQ25-75 : 16.40-43.50)/h). Of these, 24.4% presented an AHI decrease ≥50% and 37.78% had a final AHI < 15/h (tendency for improvement from an initial value of 20%: P = 0.0574). For G1 patients (n = 37), AHI significantly decreased from a median of 22.90 (16.00-43.50)/h to 19.20 (12.70-31.10)/h (P = 0.002). For G2 patients (n = 8), AHI decreased from a median of 30.10 (26.40-47.60)/h to 22.75 (14.60-36.90)/h (statistically non-significant, P = 0.059). CONCLUSIONS: In this real-life population, SV treatment for 3 months in SA patients is associated with a significant decrease in AHI. These results support the current guidelines that recommend first an optimization of the HFrEF treatment in patients with HFrEF and central SA. A potential positive airway pressure sparing effect merits further investigation.


Assuntos
Insuficiência Cardíaca , Síndromes da Apneia do Sono , Idoso , Aminobutiratos , Compostos de Bifenilo , Estudos de Coortes , Combinação de Medicamentos , Feminino , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Masculino , Estudos Prospectivos , Síndromes da Apneia do Sono/tratamento farmacológico , Síndromes da Apneia do Sono/epidemiologia , Volume Sistólico , Valsartana , Função Ventricular Esquerda
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa