Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 7797, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565565

RESUMO

Bacterial pathogens adapt and replicate within host cells, while host cells develop mechanisms to eliminate them. Using a dual proteomic approach, we characterized the intra-macrophage proteome of the facultative intracellular pathogen, Francisella novicida. More than 900 Francisella proteins were identified in infected macrophages after a 10-h infection. Biotin biosynthesis-related proteins were upregulated, emphasizing the role of biotin-associated genes in Francisella replication. Conversely, proteins encoded by the Francisella pathogenicity island (FPI) were downregulated, supporting the importance of the F. tularensis Type VI Secretion System for vacuole escape, not cytosolic replication. In the host cell, over 300 proteins showed differential expression among the 6200 identified during infection. The most upregulated host protein was cis-aconitate decarboxylase IRG1, known for itaconate production with antimicrobial properties in Francisella. Surprisingly, disrupting IRG1 expression did not impact Francisella's intracellular life cycle, suggesting redundancy with other immune proteins or inclusion in larger complexes. Over-representation analysis highlighted cell-cell contact and actin polymerization in macrophage deregulated proteins. Using flow cytometry and live cell imaging, we demonstrated that merocytophagy involves diverse cell-to-cell contacts and actin polymerization-dependent processes. These findings lay the groundwork for further exploration of merocytophagy and its molecular mechanisms in future research.Data are available via ProteomeXchange with identifier PXD035145.


Assuntos
Francisella tularensis , Tularemia , Animais , Francisella tularensis/genética , Actinas/metabolismo , Biotina/metabolismo , Proteômica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Macrófagos/metabolismo , Estágios do Ciclo de Vida , Tularemia/microbiologia , Ilhas Genômicas
2.
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
3.
Microbes Infect ; 25(6): 105124, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36871931

RESUMO

Staphylococcus aureus is the predominant pathogen in children with cystic fibrosis (CF) in France and, around 80% of them harbored S. aureus in their lungs. This study investigated virulence and antimicrobial resistance-associated genes and within-host evolution polymorphisms in 14 S. aureus persistent clones from 14 chronically infected CF children. For each of the 14 patients, we compared genomes of two isogenic sequential isolates separated by 2-9 years. All isolates were methicillin-sensitive and harbored the immune evasion gene cluster, whereas half of them harbored the enterotoxin gene cluster. Most clones were capsule type 8 (8/14) and accessory gene regulator (agr)-specificity group 1 (9/14). We identified convergent mutations in genes involved in carbohydrate metabolism, cell wall metabolism, genetic information processing and adhesion, which are likely to play important role in intracellular invasion and persistence. Further explorations relying notably on proteomics will contribute to improve our understanding of the mechanisms at play in the striking long-term persistence ability of S. aureus.


Assuntos
Fibrose Cística , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Criança , Humanos , Staphylococcus aureus/genética , Fibrose Cística/complicações , Pulmão , Proteômica , Antibacterianos , Staphylococcus aureus Resistente à Meticilina/genética , Testes de Sensibilidade Microbiana
4.
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
5.
PLoS Pathog ; 17(8): e1009326, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34339477

RESUMO

Metabolic pathways are now considered as intrinsic virulence attributes of pathogenic bacteria and thus represent potential targets for antibacterial strategies. Here we focused on the role of the pentose phosphate pathway (PPP) and its connections with other metabolic pathways in the pathophysiology of Francisella novicida. The involvement of the PPP in the intracellular life cycle of Francisella was first demonstrated by studying PPP inactivating mutants. Indeed, we observed that inactivation of the tktA, rpiA or rpe genes severely impaired intramacrophage multiplication during the first 24 hours. However, time-lapse video microscopy demonstrated that rpiA and rpe mutants were able to resume late intracellular multiplication. To better understand the links between PPP and other metabolic networks in the bacterium, we also performed an extensive proteo-metabolomic analysis of these mutants. We show that the PPP constitutes a major bacterial metabolic hub with multiple connections to glycolysis, the tricarboxylic acid cycle and other pathways, such as fatty acid degradation and sulfur metabolism. Altogether our study highlights how PPP plays a key role in the pathogenesis and growth of Francisella in its intracellular niche.


Assuntos
Proteínas de Bactérias/metabolismo , Drosophila melanogaster/metabolismo , Francisella/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Metaboloma , Via de Pentose Fosfato , Proteoma , Animais , Proteínas de Bactérias/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/microbiologia , Francisella/metabolismo , Regulação Bacteriana da Expressão Gênica , Glicólise , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação
6.
Front Immunol ; 10: 2461, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31708919

RESUMO

Mitochondria are essential organelles that act as metabolic hubs and signaling platforms within the cell. Numerous mitochondrial functions, including energy metabolism, lipid synthesis, and autophagy regulation, are intimately linked to mitochondrial dynamics, which is shaped by ongoing fusion and fission events. Recently, several intracellular bacterial pathogens have been shown to modulate mitochondrial functions to maintain their replicative niche. Through selected examples of human bacterial pathogens, we will discuss how infection induces mitochondrial changes in infected macrophages, triggering modifications of the host metabolism that lead to important immunological reprogramming.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , Macrófagos/imunologia , Mitocôndrias/fisiologia , Animais , Humanos , Ativação de Macrófagos , Macrófagos/microbiologia
7.
Mol Cell Proteomics ; 18(12): 2418-2432, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31578219

RESUMO

The bacterial pathogen Francisella tularensis possesses a noncanonical type VI secretion system (T6SS) that is required for phagosomal escape in infected macrophages. KCl stimulation has been previously used to trigger assembly and secretion of the T6SS in culture. By differential proteomics, we found here that the amounts of the T6SS proteins remained unchanged upon KCl stimulation, suggesting involvement of post-translational modifications in T6SS assembly. A phosphoproteomic analysis indeed identified a unique phosphorylation site on IglB, a key component of the T6SS sheath. Substitutions of Y139 with alanine or phosphomimetics prevented T6SS formation and abolished phagosomal escape whereas substitution with phenylalanine delayed but did not abolish phagosomal escape in J774-1 macrophages. Altogether our data demonstrated that the Y139 site of IglB plays a critical role in T6SS biogenesis, suggesting that sheath phosphorylation could participate to T6SS dynamics.Data are available via ProteomeXchange with identifier PXD013619; and on MS-Viewer, key lkaqkllxwx.


Assuntos
Francisella tularensis/metabolismo , Sistemas de Secreção Tipo VI/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Processamento Eletrônico de Dados , Francisella tularensis/genética , Francisella tularensis/ultraestrutura , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Macrófagos/microbiologia , Estrutura Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Cloreto de Potássio/farmacologia , Processamento de Proteína Pós-Traducional , Proteômica , Espectrometria de Massas em Tandem , Sistemas de Secreção Tipo VI/química , Sistemas de Secreção Tipo VI/efeitos dos fármacos , Sistemas de Secreção Tipo VI/genética
8.
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
9.
J Exp Med ; 215(8): 2035-2053, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29959173

RESUMO

To what extent immune responses against the gut flora are compartmentalized within mucosal tissues in homeostatic conditions remains a much-debated issue. We describe here, based on an inducible AID fate-mapping mouse model, that systemic memory B cell subsets, including mainly IgM+ B cells in spleen, together with IgA+ plasma cells in spleen and bone marrow, are generated in mice in the absence of deliberate immunization. While the IgA component appears dependent on the gut flora, IgM memory B cells are still generated in germ-free mice, albeit to a reduced extent. Clonal relationships and renewal kinetics after anti-CD20 treatment reveal that this long-lasting splenic population is mainly sustained by output of B cell clones persisting in mucosal germinal centers. IgM-secreting hybridomas established from splenic IgM memory B cells showed reactivity against various bacterial isolates and endogenous retroviruses. Ongoing activation of B cells in gut-associated lymphoid tissues thus generates a diversified systemic compartment showing long-lasting clonal persistence and protective capacity against systemic bacterial infections.


Assuntos
Antibacterianos/imunologia , Imunidade nas Mucosas , Imunoglobulina M/metabolismo , Memória Imunológica , Baço/imunologia , Envelhecimento/imunologia , Animais , Antígenos CD/metabolismo , Linfócitos B/imunologia , Proteínas de Bactérias/metabolismo , Medula Óssea/metabolismo , Citidina Desaminase/metabolismo , Microbioma Gastrointestinal , Vida Livre de Germes , Centro Germinativo/citologia , Imunização , Imunoglobulina A/metabolismo , Cinética , Proteínas Luminescentes/metabolismo , Camundongos , Mutação/genética , Plasmócitos/citologia , Transdução de Sinais , Linfócitos T/metabolismo , Receptores Toll-Like/metabolismo
10.
Nat Commun ; 8(1): 853, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29021545

RESUMO

The enzyme fructose-bisphosphate aldolase occupies a central position in glycolysis and gluconeogenesis pathways. Beyond its housekeeping role in metabolism, fructose-bisphosphate aldolase has been involved in additional functions and is considered as a potential target for drug development against pathogenic bacteria. Here, we address the role of fructose-bisphosphate aldolase in the bacterial pathogen Francisella novicida. We demonstrate that fructose-bisphosphate aldolase is important for bacterial multiplication in macrophages in the presence of gluconeogenic substrates. In addition, we unravel a direct role of this metabolic enzyme in transcription regulation of genes katG and rpoA, encoding catalase and an RNA polymerase subunit, respectively. We propose a model in which fructose-bisphosphate aldolase participates in the control of host redox homeostasis and the inflammatory immune response.The enzyme fructose-bisphosphate aldolase (FBA) plays central roles in glycolysis and gluconeogenesis. Here, Ziveri et al. show that FBA of the pathogen Francisella novicida acts, in addition, as a transcriptional regulator and is important for bacterial multiplication in macrophages.


Assuntos
Francisella tularensis/enzimologia , Frutose-Bifosfato Aldolase/metabolismo , Regulação Bacteriana da Expressão Gênica , Animais , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Frutose-Bifosfato Aldolase/genética , Gluconeogênese , Glucose/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Metabolômica , Camundongos Endogâmicos BALB C , Estresse Oxidativo
11.
Mol Ther ; 25(10): 2309-2322, 2017 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-28720467

RESUMO

Recombinant adeno-associated viral (rAAV) vectors exhibit interesting properties as vaccine carriers for their ability to induce long-lasting antibody responses. However, rAAV-based vaccines have been suggested to trigger functionally impaired long-term memory CD8+ T cell responses, in part due to poor dendritic cell (DC) transduction. Such results, albeit limited to intramuscular immunization, undermined the use of rAAV as vaccine vehicles against intracellular pathogens. We report here that intradermal immunization with a model rAAV2/1-based vaccine drives the development of bona fide long-term memory CD8+ T cell responses. The intradermal route of immunization and the presence of potent major histocompatibility complex (MHC) class II responses showed synergistic effects on the overall quantity and quality of systemic long-term effector memory transgene-specific CD8+ T cells being generated against the transgene. Of key interest, we found that the induction of memory cytotoxic T lymphocytes (CTLs) following intradermal immunization was solely dependent on the cross-presentation of skin-expressed transgene products, which appeared highly enhanced as compared to muscle-expressed transgene products. Overall our results highlight key tissue-specific differences in transgene presentation pathway requirements of importance for the design of rAAV-based T cell-inducing vaccines.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/metabolismo , Dependovirus/genética , Animais , Linfócitos T CD4-Positivos/metabolismo , Feminino , Citometria de Fluxo , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Artigo em Inglês | MEDLINE | ID: mdl-28401066

RESUMO

Francisella tularensis is a highly infectious Gram-negative bacterium and the causative agent of the zoonotic disease tularemia. This bacterial pathogen can infect a broad variety of animal species and can be transmitted to humans in numerous ways with various clinical outcomes. Although, Francisella possesses the capacity to infect numerous mammalian cell types, the macrophage constitutes the main intracellular niche, used for in vivo bacterial dissemination. To survive and multiply within infected macrophages, Francisella must imperatively escape from the phagosomal compartment. In the cytosol, the bacterium needs to control the host innate immune response and adapt its metabolism to this nutrient-restricted niche. Our laboratory has shown that intracellular Francisella mainly relied on host amino acid as major gluconeogenic substrates and provided evidence that the host metabolism was also modified upon Francisella infection. We will review here our current understanding of how Francisella copes with the available nutrient sources provided by the host cell during the course of infection.


Assuntos
Adaptação Fisiológica , Francisella/metabolismo , Francisella/patogenicidade , Interações Hospedeiro-Patógeno , Tularemia/metabolismo , Adaptação Fisiológica/genética , Aminoácidos/metabolismo , Animais , Metabolismo dos Carboidratos , Citosol/metabolismo , Citosol/microbiologia , Francisella/genética , Glicólise , Imunidade Inata , Macrófagos/metabolismo , Macrófagos/microbiologia , Fagossomos/metabolismo , Fagossomos/microbiologia , Tularemia/imunologia , Tularemia/microbiologia , Fatores de Virulência/metabolismo , Zoonoses/microbiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-28377902

RESUMO

Francisella tularensis is able to invade, survive and replicate inside a variety of cell types. However, in vivo F. tularensis preferentially enters host macrophages where it rapidly escapes to the cytosol to avoid phagosomal stresses and to multiply to high numbers. We previously showed that human monocyte infection by F. tularensis LVS triggered deglycosylation of the glutamine transporter SLC1A5. However, this deglycosylation, specifically induced by Francisella infection, was not restricted to SLC1A5, suggesting that host protein deglycosylation processes in general might contribute to intracellular bacterial adaptation. Indeed, we later found that Francisella infection modulated the transcription of numerous glycosidase and glycosyltransferase genes in human macrophages and analysis of cell extracts revealed an important increase of N and O-protein glycosylation. In eukaryotic cells, glycosylation has significant effects on protein folding, conformation, distribution, stability, and activity and dysfunction of protein glycosylation may lead to development of diseases like cancer and pathogenesis of infectious diseases. Pathogenic bacteria have also evolved dedicated glycosylation machineries and have notably been shown to use these glycoconjugates as ligands to specifically interact with the host. In this review, we will focus on Francisella and summarize our current understanding of the importance of these post-translational modifications on its intracellular niche adaptation.


Assuntos
Francisella tularensis/patogenicidade , Regulação da Expressão Gênica , Glicosídeo Hidrolases/metabolismo , Glicosiltransferases/metabolismo , Interações Hospedeiro-Patógeno , Animais , Glicosilação , Humanos , Macrófagos/microbiologia
14.
PLoS Pathog ; 12(9): e1005821, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27602570

RESUMO

The virulence of Francisella tularensis, the etiological agent of tularemia, relies on an atypical type VI secretion system (T6SS) encoded by a genomic island termed the Francisella Pathogenicity Island (FPI). While the importance of the FPI in F. tularensis virulence is clearly established, the precise role of most of the FPI-encoded proteins remains to be deciphered. In this study, using highly virulent F. tularensis strains and the closely related species F. novicida, IglG was characterized as a protein featuring a unique α-helical N-terminal extension and a domain of unknown function (DUF4280), present in more than 250 bacterial species. Three dimensional modeling of IglG and of the DUF4280 consensus protein sequence indicates that these proteins adopt a PAAR-like fold, suggesting they could cap the T6SS in a similar way as the recently described PAAR proteins. The newly identified PAAR-like motif is characterized by four conserved cysteine residues, also present in IglG, which may bind a metal atom. We demonstrate that IglG binds metal ions and that each individual cysteine is required for T6SS-dependent secretion of IglG and of the Hcp homologue, IglC and for the F. novicida intracellular life cycle. In contrast, the Francisella-specific N-terminal α-helical extension is not required for IglG secretion, but is critical for F. novicida virulence and for the interaction of IglG with another FPI-encoded protein, IglF. Altogether, our data suggest that IglG is a PAAR-like protein acting as a bi-modal protein that may connect the tip of the Francisella T6SS with a putative T6SS effector, IglF.


Assuntos
Proteínas de Bactérias/metabolismo , Francisella tularensis/genética , Ilhas Genômicas/genética , Tularemia/microbiologia , Sistemas de Secreção Tipo VI/genética , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Regulação Bacteriana da Expressão Gênica , Macrófagos/metabolismo , Modelos Moleculares , Alinhamento de Sequência , Deleção de Sequência , Tularemia/imunologia , Sistemas de Secreção Tipo VI/metabolismo , Virulência , Fatores de Virulência/genética
15.
Cell Microbiol ; 18(12): 1763-1781, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27185209

RESUMO

Protein glycosylation processes play a crucial role in most physiological functions, including cell signalling, cellular differentiation and adhesion. We previously demonstrated that rapid deglycosylation of membrane proteins was specifically triggered after infection of human macrophages by the bacterial pathogen Francisella tularensis. Using a glycan processing gene microarray, we found here that Francisella infection modulated expression of numerous glycosidase and glycosyltransferase genes. Furthermore, analysis of cell extracts from infected macrophages by Lectin and Western blotting revealed an important increase of N- and O-protein glycosylation. We chose to focus in the present work on one of the O-glycosylated proteins identified by mass spectrometry, the multifunctional endoplasmic reticulum chaperone BiP (HSPA5/GRP78). We demonstrate that BiP expression is modulated upon Francisella infection and is required to support its intracellular multiplication. Moreover, we show that Francisella differentially modulates the BiP-dependent activation of three key proteins of the unfolded protein response (UPR), IRE1, PERK and ATF6. The effects exerted on human cells by Francisella may thus constitute a novel excample of UPR manipulation contributing to intracellular bacterial adaptation.


Assuntos
Proteínas de Bactérias/genética , Francisella tularensis/genética , Proteínas de Choque Térmico/genética , Interações Hospedeiro-Patógeno , Macrófagos/microbiologia , Resposta a Proteínas não Dobradas , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Chaperona BiP do Retículo Endoplasmático , Endorribonucleases/genética , Endorribonucleases/metabolismo , Francisella tularensis/crescimento & desenvolvimento , Regulação da Expressão Gênica , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
16.
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
17.
Artigo em Inglês | MEDLINE | ID: mdl-25705612

RESUMO

Francisella tularensis, the agent of the zoonotic disease tularemia, is a highly infectious bacterium for a large number of animal species and can be transmitted to humans by various means. The bacterium is able to infect a variety of cell types but replicates in mammalian hosts mainly in the cytosol of infected macrophages. In order to resist the stressful and nutrient-restricted intracellular environments, it encounters during its systemic dissemination, Francisella has developed dedicated stress resistance mechanisms and adapted its metabolic and nutritional needs. Recent data form our laboratory and from several other groups have shown that Francisella simultaneously relies on multiple host amino acid sources during its intracellular life cycle. This review will summarize how intracellular Francisella use different amino acid sources, and their role in phagosomal escape and/or cytosolic multiplication and systemic dissemination. We will first summarize the data that we have obtained on two amino acid transporters involved in Francisella phagosomal escape and cytosolic multiplication i.e., the glutamate transporter GadC and the asparagine transporter AnsP, respectively. The specific contribution of glutamate and asparagine to the physiology of the bacterium will be evoked. Then, we will discuss how Francisella has adapted to obtain and utilize host amino acid resources, and notably the contribution of host transporters and autophagy process in the establishment of a nutrient-replete intracellular niche.


Assuntos
Aminoácidos/metabolismo , Francisella tularensis/metabolismo , Macrófagos/microbiologia , Tularemia/microbiologia , Animais , Francisella , Francisella tularensis/genética , Humanos , Macrófagos/metabolismo , Tularemia/metabolismo
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA