Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 212(1): 69-80, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37982695

RESUMEN

Staphylococcus aureus is a significant cause of morbidity and mortality in pulmonary infections. Patients with autosomal-dominant hyper-IgE syndrome due to STAT3 deficiency are particularly susceptible to acquiring staphylococcal pneumonia associated with lung tissue destruction. Because macrophages are involved in both pathogen defense and inflammation, we investigated the impact of murine myeloid STAT3 deficiency on the macrophage phenotype in vitro and on pathogen clearance and inflammation during murine staphylococcal pneumonia. Murine bone marrow-derived macrophages (BMDM) from STAT3 LysMCre+ knockout or Cre- wild-type littermate controls were challenged with S. aureus, LPS, IL-4, or vehicle control in vitro. Pro- and anti-inflammatory responses as well as polarization and activation markers were analyzed. Mice were infected intratracheally with S. aureus, bronchoalveolar lavage and lungs were harvested, and immunohistofluorescence was performed on lung sections. S. aureus infection of STAT3-deficient BMDM led to an increased proinflammatory cytokine release and to enhanced upregulation of costimulatory MHC class II and CD86. Murine myeloid STAT3 deficiency did not affect pathogen clearance in vitro or in vivo. Matrix metalloproteinase 9 was upregulated in Staphylococcus-treated STAT3-deficient BMDM and in lung tissues of STAT3 knockout mice infected with S. aureus. Moreover, the expression of miR-155 was increased. The enhanced inflammatory responses and upregulation of matrix metalloproteinase 9 and miR-155 expression in murine STAT3-deficient as compared with wild-type macrophages during S. aureus infections may contribute to tissue damage as observed in STAT3-deficient patients during staphylococcal pneumonia.


Asunto(s)
Síndrome de Job , MicroARNs , Neumonía Estafilocócica , Infecciones Estafilocócicas , Humanos , Ratones , Animales , Staphylococcus aureus/metabolismo , Activación de Macrófagos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Inflamación/genética , Ratones Noqueados , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(23): e2122386119, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35648835

RESUMEN

Pneumococcal conjugate vaccines (PCVs) used in childhood vaccination programs have resulted in replacement of vaccine-type with nonvaccine-type pneumococci in carriage and invasive pneumococcal disease (IPD). A vaccine based on highly conserved and protective pneumococcal antigens is urgently needed. Here, we performed intranasal immunization of mice with pneumococcal membrane particles (MPs) to mimic natural nasopharyngeal immunization. MP immunization gave excellent serotype-independent protection against IPD that was antibody dependent but independent of the cytotoxin pneumolysin. Using Western blotting, immunoprecipitation, mass spectrometry, and different bacterial mutants, we identified the conserved lipoproteins MalX and PrsA as the main antigens responsible for cross-protection. Additionally, we found that omitting the variable surface protein and vaccine candidate PspA from MPs enhanced protective immune responses to the conserved proteins. Our findings suggest that MPs containing MalX and PrsA could serve as a platform for pneumococcal vaccine development targeting the elderly and immunocompromised.


Asunto(s)
Proteínas Bacterianas , Lipoproteínas , Proteínas de la Membrana , Proteínas de Transporte de Membrana , Infecciones Neumocócicas , Vacunas Neumococicas , Administración Intranasal , Animales , Proteínas Bacterianas/inmunología , Membrana Celular/inmunología , Secuencia Conservada , Reacciones Cruzadas , Humanos , Inmunización/métodos , Lipoproteínas/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Transporte de Membrana/inmunología , Ratones , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/inmunología , Serogrupo , Streptococcus pneumoniae/inmunología
3.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34785593

RESUMEN

Emerging antibiotic resistance demands identification of novel antibacterial compound classes. A bacterial whole-cell screen based on pneumococcal autolysin-mediated lysis induction was developed to identify potential bacterial cell wall synthesis inhibitors. A hit class comprising a 1-amino substituted tetrahydrocarbazole (THCz) scaffold, containing two essential amine groups, displayed bactericidal activity against a broad range of gram-positive and selected gram-negative pathogens in the low micromolar range. Mode of action studies revealed that THCz inhibit cell envelope synthesis by targeting undecaprenyl pyrophosphate-containing lipid intermediates and thus simultaneously inhibit peptidoglycan, teichoic acid, and polysaccharide capsule biosynthesis. Resistance did not readily develop in vitro, and the ease of synthesizing and modifying these small molecules, as compared to natural lipid II-binding antibiotics, makes THCz promising scaffolds for development of cell wall-targeting antimicrobials.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Pared Celular/química , Pared Celular/efectos de los fármacos , Lípidos/química , Antibacterianos/química , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , N-Acetil Muramoil-L-Alanina Amidasa , Peptidoglicano/biosíntesis , Fosfatos de Poliisoprenilo , Streptococcus pneumoniae/efectos de los fármacos , Ácidos Teicoicos/química , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados
4.
Cell ; 133(4): 574-6, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18485865

RESUMEN

The chaperone/usher pathway is responsible for the assembly of adhesive pili on the surface of gram-negative pathogenic bacteria. In this issue, Remaut et al. (2008) present the crystal structure of the PapC usher translocation domain and images of the FimD usher bound to a pilus translocation intermediate. These new structures provide the first detailed view of a translocase in action.


Asunto(s)
Vías Biosintéticas , Escherichia coli/metabolismo , Fimbrias Bacterianas/metabolismo , Escherichia coli/química , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas Fimbrias/química , Proteínas Fimbrias/metabolismo , Porinas/química , Porinas/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(49): 31386-31397, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229573

RESUMEN

Influenza A virus (IAV)-related mortality is often due to secondary bacterial infections, primarily by pneumococci. Here, we study how IAV-modulated changes in the lungs affect bacterial replication in the lower respiratory tract (LRT). Bronchoalveolar lavages (BALs) from coinfected mice showed rapid bacterial proliferation 4 to 6 h after pneumococcal challenge. Metabolomic and quantitative proteomic analyses demonstrated capillary leakage with efflux of nutrients and antioxidants into the alveolar space. Pneumococcal adaptation to IAV-induced inflammation and redox imbalance increased the expression of the pneumococcal chaperone/protease HtrA. Presence of HtrA resulted in bacterial growth advantage in the IAV-infected LRT and protection from complement-mediated opsonophagocytosis due to capsular production. Absence of HtrA led to growth arrest in vitro that was partially restored by antioxidants. Pneumococcal ability to grow in the IAV-infected LRT depends on the nutrient-rich milieu with increased levels of antioxidants such as ascorbic acid and its ability to adapt to and cope with oxidative damage and immune clearance.


Asunto(s)
Antioxidantes/metabolismo , Capilares/patología , Gripe Humana/microbiología , Infecciones Neumocócicas/microbiología , Sistema Respiratorio/microbiología , Sistema Respiratorio/virología , Streptococcus pneumoniae/crecimiento & desarrollo , Animales , Proteínas Bacterianas/metabolismo , Glucosa/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Infecciones por Orthomyxoviridae/microbiología , Oxidación-Reducción , Estrés Oxidativo , Fagocitosis , Sistema Respiratorio/patología
6.
Mol Microbiol ; 113(3): 650-658, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32185835

RESUMEN

The Gram-positive bacterium Streptococcus pneumoniae, the pneumococcus, is an important commensal resident of the human nasopharynx. Carriage is usually asymptomatic, however, S. pneumoniae can become invasive and spread from the upper respiratory tract to the lungs causing pneumonia, and to other organs to cause severe diseases such as bacteremia and meningitis. Several pneumococcal proteins important for its disease-causing capability have been described and many are expressed on the bacterial surface. The surface located pneumococcal type-1 pilus has been associated with virulence and the inflammatory response, and it is present in 20%-30% of clinical isolates. Its tip protein RrgA has been shown to be a major adhesin to human cells and to promote invasion through the blood-brain barrier. In this review we discuss recent findings of the impact of RrgA on bacterial colonization of the upper respiratory tract and on pneumococcal virulence, and use epidemiological data and genome-mining to suggest trade-off mechanisms potentially explaining the rather low prevalence of pilus-1 expressing pneumococci in humans.


Asunto(s)
Proteínas Fimbrias/metabolismo , Streptococcus pneumoniae/metabolismo , Factores de Virulencia/metabolismo , Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Fimbrias/fisiología , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/fisiología , Unión Proteica , Streptococcus pneumoniae/patogenicidad , Virulencia/genética , Factores de Virulencia/fisiología
7.
Cell Microbiol ; 21(11): e13077, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31251447

RESUMEN

Streptococcus pneumoniae (the pneumococcus) is a human respiratory tract pathogen and a major cause of morbidity and mortality globally. Although the pneumococcus is a commensal bacterium that colonizes the nasopharynx, it also causes lethal diseases such as meningitis, sepsis, and pneumonia, especially in immunocompromised patients, in the elderly, and in young children. Due to the acquisition of antibiotic resistance and the emergence of nonvaccine serotypes, the pneumococcus has been classified as one of the priority pathogens for which new antibacterials are urgently required by the World Health Organization, 2017. Understanding molecular mechanisms behind the pathogenesis of pneumococcal infections and bacterial interactions within the host is crucial to developing novel therapeutics. Previously considered to be an extracellular pathogen, it is becoming evident that pneumococci may also occasionally establish intracellular niches within the body to escape immune surveillance and spread within the host. Intracellular survival within host cells also enables pneumococci to resist many antibiotics. Within the host cell, the bacteria exist in unique vacuoles, thereby avoiding degradation by the acidic lysosomes, and modulate the expression of its virulence genes to adapt to the intracellular environment. To invade and survive intracellularly, the pneumococcus utilizes a combination of virulence factors such as pneumolysin (PLY), pneumococcal surface protein A (PspA), pneumococcal adhesion and virulence protein B (PavB), the pilus-1 adhesin RrgA, pyruvate oxidase (SpxB), and metalloprotease (ZmpB). In this review, we discuss recent findings showing the intracellular persistence of Streptococcus pneumoniae and its underlying mechanisms.


Asunto(s)
Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Células Dendríticas/inmunología , Farmacorresistencia Microbiana , Corazón/microbiología , Corazón/fisiopatología , Humanos , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Miocardio/metabolismo , Miocardio/patología , Nasofaringe/microbiología , Sistema Respiratorio/inmunología , Sistema Respiratorio/microbiología , Bazo/citología , Bazo/microbiología , Bazo/patología , Streptococcus pneumoniae/inmunología , Factores de Virulencia/metabolismo
8.
Bioessays ; 37(4): 426-35, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640084

RESUMEN

Transport of DNA across bacterial membranes involves complex DNA uptake systems. In Gram-positive bacteria, the DNA uptake machinery shares fundamental similarities with type IV pili and type II secretion systems. Although dedicated pilus structures, such as type IV pili in Gram-negative bacteria, are necessary for efficient DNA uptake, the role of similar structures in Gram-positive bacteria is just beginning to emerge. Recently two essentially very different pilus structures composed of the same major pilin protein ComGC were proposed to be involved in transformation of the Gram-positive bacterium Streptococcus pneumoniae - one is a long, thin, type IV pilus-like fiber with DNA binding capacity and the other one is a pilus structure that was thicker, much shorter and not able to bind DNA. Here we discuss how competence induced pili, either by pilus retraction or by a transient pilus-related opening in the cell wall, may mediate DNA uptake in S. pneumoniae.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Streptococcus pneumoniae/metabolismo , Transformación Bacteriana , Fimbrias Bacterianas/genética , Streptococcus pneumoniae/genética
9.
Proc Natl Acad Sci U S A ; 111(7): E758-65, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550320

RESUMEN

Streptococcus pneumoniae is a major human pathogen that successfully adapts to the host environment via an efficient uptake system for free DNA liberated from other organisms in the upper respiratory tract, facilitating immune evasion and drug resistance. Although the initial signaling events leading to pneumococcal competence for DNA transformation and the fate of DNA when it has been taken up have been extensively studied, the actual mechanism by which DNA in the environment may traverse the thick capsular and cell wall layers remains unknown. Here we visualize that induction of competence results in the formation of a native morphologically distinct pilus structure on the bacterial surface. This plaited pilus is encoded by the competence (com)G locus, and, after assembly, it is rapidly released into the surrounding medium. Heterologous pneumococcal pilus expression in Escherichia coli was obtained by replacing the pulE-K putative pilin genes of the Klebsiella oxytoca type II secretion system with the complete comG locus. In the pneumococcus, the coordinated secretion of pili from the cells correlates to DNA transformation. A model for DNA transformation is proposed whereby pilus assembly "drills" a channel across the thick cell wall that becomes transiently open by secretion of the pilus, providing the entry port for exogenous DNA to gain access to DNA receptors associated with the cytoplasmic membrane.


Asunto(s)
Sistemas de Secreción Bacterianos/fisiología , Competencia de la Transformación por ADN/genética , ADN/metabolismo , Fimbrias Bacterianas/metabolismo , Streptococcus pneumoniae/metabolismo , Transformación Bacteriana/fisiología , Electroforesis en Gel de Poliacrilamida , Fimbrias Bacterianas/ultraestructura , Microscopía Electrónica de Transmisión , Transformación Bacteriana/genética , Ácido Tricloroacético
10.
Proc Natl Acad Sci U S A ; 110(50): 20230-5, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24191013

RESUMEN

Virulence factor secretion and assembly occurs at spatially restricted foci in some Gram-positive bacteria. Given the essentiality of the general secretion pathway in bacteria and the contribution of virulence factors to disease progression, the foci that coordinate these processes are attractive antimicrobial targets. In this study, we show in Enterococcus faecalis that SecA and Sortase A, required for the attachment of virulence factors to the cell wall, localize to discrete domains near the septum or nascent septal site as the bacteria proceed through the cell cycle. We also demonstrate that cationic human ß-defensins interact with E. faecalis at discrete septal foci, and this exposure disrupts sites of localized secretion and sorting. Modification of anionic lipids by multiple peptide resistance factor, a protein that confers antimicrobial peptide resistance by electrostatic repulsion, renders E. faecalis more resistant to killing by defensins and less susceptible to focal targeting by the cationic antimicrobial peptides. These data suggest a paradigm in which focal targeting by antimicrobial peptides is linked to their killing efficiency and to disruption of virulence factor assembly.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Enterococcus faecalis/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Factores de Virulencia/biosíntesis , beta-Defensinas/metabolismo , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Canales de Translocación SEC , Proteína SecA
11.
PLoS Genet ; 9(10): e1003868, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130509

RESUMEN

Streptococcus pneumoniae of serotype 3 possess a mucoid capsule and cause disease associated with high mortality rates relative to other pneumococci. Phylogenetic analysis of a complete reference genome and 81 draft sequences from clonal complex 180, the predominant serotype 3 clone in much of the world, found most sampled isolates belonged to a clade affected by few diversifying recombinations. However, other isolates indicate significant genetic variation has accumulated over the clonal complex's entire history. Two closely related genomes, one from the blood and another from the cerebrospinal fluid, were obtained from a patient with meningitis. The pair differed in their behaviour in a mouse model of disease and in their susceptibility to antimicrobials, with at least some of these changes attributable to a mutation that up-regulated the patAB efflux pump. This indicates clinically important phenotypic variation can accumulate rapidly through small alterations to the genotype.


Asunto(s)
Genoma Bacteriano , Mutación , Filogenia , Streptococcus pneumoniae/genética , Animales , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Meningitis/sangre , Meningitis/líquido cefalorraquídeo , Meningitis/microbiología , Ratones , Serotipificación , Streptococcus pneumoniae/patogenicidad
12.
13.
J Infect Dis ; 210(1): 4-13, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24443543

RESUMEN

BACKGROUND: Streptococcus pneumoniae serotype 1 has a high likelihood of causing invasive disease. Serotype 1 isolates belonging to CC228 are associated with low mortality, while CC217 isolates exhibit high mortality in patients. METHODS: Clinical pneumococcal isolates and mutants were evaluated in wild-type C57BL/6 mice, macrophage-depleted mice, neutrophil-depleted mice, and SIGN-R1 knockout mice. In vitro models included binding and phagocytosis by THP-1 cells, capsule measurements, hydrogen peroxide production, and viability assays. RESULTS: During early systemic infection in mice with serotype 1, large-colony variants appeared in blood. Similar large colonies were found in blood specimens from patients with invasive disease. Large morphotypes contained higher numbers of viable bacteria, grew faster, produced no or little hydrogen peroxide, and contained mutations in the spxB gene. spxB mutants were considerably more virulent in wild-type mice, less susceptible to early host clearance than wild-type strains after intravenous infection, but impaired in colonization. spxB mutants were less efficiently phagocytosed by macrophages than wild-type bacteria, which, in contrast to spxB mutants, caused more-severe disease when macrophages or SIGN-R1 were depleted. CONCLUSIONS: Hypervirulent spxB mutants are selected in both mice and patients and are resistant to early macrophage-mediated clearance.


Asunto(s)
Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Animales , Proteínas Bacterianas/genética , Técnicas de Tipificación Bacteriana , Línea Celular , Humanos , Huésped Inmunocomprometido , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Proteínas Mutantes/genética , Fagocitosis , Infecciones Neumocócicas/clasificación , Serotipificación , Virulencia
14.
J Infect Dis ; 209(3): 377-88, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24009156

RESUMEN

BACKGROUND: Pneumococcal serotypes are represented by a varying number of clonal lineages with different genetic contents, potentially affecting invasiveness. However, genetic variation within the same genetic lineage may be larger than anticipated. METHODS: A total of 715 invasive and carriage isolates from children in the same region and during the same period were compared using pulsed-field gel electrophoresis (PFGE) and multilocus sequence typing. Bacterial genome sequencing, functional assays, and in vivo virulence mice studies were performed. RESULTS: Clonal types of the same serotype but also intraclonal variants within clonal complexes (CCs) showed differences in invasive-disease potential. CC138, a common CC, was divided into several PFGE patterns, partly explained by number, location, and type of temperate bacteriophages. Whole-genome sequencing of 4 CC138 isolates representing PFGE clones with different invasive-disease potentials revealed intraclonal sequence variations of the virulence-associated proteins pneumococcal surface protein A (PspA) and pneumococcal choline-binding protein C (PspC). A carrier isolate lacking PcpA exhibited decreased virulence in mice, and there was a differential binding of human factor H, depending on invasiveness. CONCLUSIONS: Pneumococcal clonal types but also intraclonal variants exhibited different invasive-disease potentials in children. Intraclonal variants, reflecting different prophage contents, showed differences in major surface antigens. This suggests ongoing immune selection, such as that due to PspC-mediated complement resistance through varied human factor H binding, that may affect invasiveness in children.


Asunto(s)
Variación Genética , Infecciones Neumocócicas/epidemiología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/genética , Adolescente , Animales , Antígenos Bacterianos/análisis , Portador Sano/epidemiología , Portador Sano/microbiología , Niño , Preescolar , Modelos Animales de Enfermedad , Electroforesis en Gel de Campo Pulsado , Femenino , Genoma Bacteriano , Genotipo , Humanos , Lactante , Masculino , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Tipificación Molecular , Infecciones Neumocócicas/microbiología , Profagos/genética , Análisis de Secuencia de ADN , Fagos de Streptococcus/genética , Streptococcus pneumoniae/aislamiento & purificación , Virulencia
15.
Cell Microbiol ; 15(8): 1385-400, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23421931

RESUMEN

Secondary infections with Streptococcus pneumoniae (SP) are frequently observed following influenza A virus (IAV) infection and have a substantial impact on global health. Despite this, the basis for the disease progression is incompletely understood. To investigate the effect of co-infection on human monocyte-derived dendritic cells (MDDCs) we analysed the expression of clinically important pro-inflammatory and immune-modulatory cytokines. IAV infection or treatment with supernatants from IAV-infected cell cultures resulted in priming of the DCs which subsequently influenced the production of IL-12p70, as well as IL-6, following SP infection. Co-infection of the same cell was not required but this effect was dependent on the time, dose and duration of the infections, as well as pathogen viability, bacterial uptake and endosome acidification. Bacterially infected cells were characterized as the main producers of IL-12p70. Finally, we showed that type I interferons were primarily responsible for the priming of IL-12p70 that was observed by infection with IAV. These results provide a probable mechanism for the elevated levels of particular cytokines observed in IAV and SP co-infected cell cultures with implications for the pathogenic outcome observed during in vivo infection.


Asunto(s)
Coinfección/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Virus de la Influenza A/patogenicidad , Gripe Humana/metabolismo , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/patogenicidad , Células Cultivadas , Comorbilidad , Células Dendríticas/microbiología , Células Dendríticas/virología , Humanos , Virus de la Influenza A/fisiología , Gripe Humana/epidemiología , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Infecciones Neumocócicas/epidemiología , Transducción de Señal/fisiología , Streptococcus pneumoniae/fisiología , Factores de Tiempo
16.
Microlife ; 5: uqae008, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665235

RESUMEN

On September 20-22 September 2023, the international conference 'Microbiology 2023: from single cell to microbiome and host' convened microbiologists from across the globe for a very successful symposium, showcasing cutting-edge research in the field. Invited lecturers delivered exceptional presentations covering a wide range of topics, with a major emphasis on phages and microbiomes, on the relevant bacteria within these ecosystems, and their multifaceted roles in diverse environments. Discussions also spanned the intricate analysis of fundamental bacterial processes, such as cell division, stress resistance, and interactions with phages. Organized by four renowned Academies, the German Leopoldina, the French Académie des sciences, the Royal Society UK, and the Royal Swedish Academy of Sciences, the symposium provided a dynamic platform for experts to share insights and discoveries, leaving participants inspired and eager to integrate new knowledge into their respective projects. The success of Microbiology 2023 prompted the decision to host the next quadrennial academic meeting in Sweden. This choice underscores the commitment to fostering international collaboration and advancing the frontiers of microbiological knowledge. The transition to Sweden promises to be an exciting step in the ongoing global dialogue and specific collaborations on microbiology, a field where researchers will continue to push the boundaries of knowledge, understanding, and innovation not only in health and disease but also in ecology.

17.
J Bacteriol ; 195(19): 4484-95, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23913319

RESUMEN

Enterococci commonly cause hospital-acquired infections, such as infective endocarditis and catheter-associated urinary tract infections. In animal models of these infections, a long hairlike extracellular protein fiber known as the endocarditis- and biofilm-associated (Ebp) pilus is an important virulence factor for Enterococcus faecalis. For Ebp and other sortase-assembled pili, the pilus-associated sortases are essential for fiber formation as they create covalent isopeptide bonds between the sortase recognition motif and the pilin-like motif of the pilus subunits. However, the molecular requirements governing the incorporation of the three pilus subunits (EbpA, EbpB, and EbpC) have not been investigated in E. faecalis. Here, we show that a Lys residue within the pilin-like motif of the EbpC subunit was necessary for EbpC polymerization. However, incorporation of EbpA into the pilus fiber only required its sortase recognition motif (LPXTG), while incorporation of EbpB only required its pilin-like motif. Only the sortase recognition motif would be required for incorporation of the pilus tip subunit, while incorporation of the base subunit would only require the pilin recognition motif. Thus, these data support a model with EbpA at the tip and EbpB at the base of an EbpC polymer. In addition, the housekeeping sortase, SrtA, was found to process EbpB and its predicted catalytic Cys residue was required for efficient cell wall anchoring of mature Ebp pili. Thus, we have defined molecular interactions involved in fiber polymerization, minor subunit organization, and pilus subcellular compartmentalization in the E. faecalis Ebp pilus system. These studies advance our understanding of unique molecular mechanisms of sortase-assembled pilus biogenesis.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Enterococcus faecalis/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/fisiología , Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Pared Celular , Cisteína Endopeptidasas/genética , Enterococcus faecalis/genética , Enterococcus faecalis/fisiología , Proteínas Fimbrias/genética , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Mutación
18.
J Biol Chem ; 287(14): 11018-29, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334685

RESUMEN

The pneumococcal autolysin LytA is a virulence factor involved in autolysis as well as in fratricidal- and penicillin-induced lysis. In this study, we used biochemical and molecular biological approaches to elucidate which factors control the cytoplasmic translocation and lytic activation of LytA. We show that LytA is mainly localized intracellularly, as only a small fraction was found attached to the extracellular cell wall. By manipulating the extracellular concentration of LytA, we found that the cells were protected from lysis during exponential growth, but not in the stationary phase, and that a defined threshold concentration of extracellular LytA dictates the onset of autolysis. Stalling growth through nutrient depletion, or the specific arrest of cell wall synthesis, sensitized cells for LytA-mediated lysis. Inhibition of cell wall association via the choline binding domain of an exogenously added enzymatically inactive form of LytA revealed a potential substrate for the amidase domain within the cell wall where the formation of nascent peptidoglycan occurs.


Asunto(s)
Bacteriólisis , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/biosíntesis , Peptidoglicano/metabolismo , Streptococcus pneumoniae/citología , Streptococcus pneumoniae/enzimología , Antibacterianos/farmacología , Bacteriólisis/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Ácido Desoxicólico/farmacología , Detergentes/farmacología , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/farmacología , Señales de Clasificación de Proteína , Transporte de Proteínas/efectos de los fármacos , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/metabolismo
19.
Nat Commun ; 14(1): 3170, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37264013

RESUMEN

The bacterial cell envelope consists of multiple layers, including the peptidoglycan cell wall, one or two membranes, and often an external layer composed of capsular polysaccharides (CPS) or other components. How the synthesis of all these layers is precisely coordinated remains unclear. Here, we identify a mechanism that coordinates the synthesis of CPS and peptidoglycan in Streptococcus pneumoniae. We show that CPS synthesis initiates from the division septum and propagates along the long axis of the cell, organized by the tyrosine kinase system CpsCD. CpsC and the rest of the CPS synthesis complex are recruited to the septum by proteins associated with the divisome (a complex involved in septal peptidoglycan synthesis) but not the elongasome (involved in peripheral peptidoglycan synthesis). Assembly of the CPS complex starts with CpsCD, then CpsA and CpsH, the glycosyltransferases, and finally CpsJ. Remarkably, targeting CpsC to the cell pole is sufficient to reposition CPS synthesis, leading to diplococci that lack CPS at the septum. We propose that septal CPS synthesis is important for chain formation and complement evasion, thereby promoting bacterial survival inside the host.


Asunto(s)
Peptidoglicano , Streptococcus pneumoniae , Streptococcus pneumoniae/metabolismo , Peptidoglicano/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Polisacáridos/metabolismo , Membrana Celular/metabolismo , Cápsulas Bacterianas/metabolismo , Polisacáridos Bacterianos/metabolismo
20.
Oncogene ; 41(15): 2173-2186, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35197571

RESUMEN

Increasing evidence highlights the role of bacteria in the physiopathology of cancer. However, the underlying molecular mechanisms remains poorly understood. Several cancer-associated bacteria have been shown to produce toxins which interfere with the host defense against tumorigenesis. Here, we show that lipopolysaccharides from Klebsiella pneumoniae and other Enterobacteria strongly inhibit the host tumor suppressor p53 pathway through a novel mechanism of p53 regulation. We found that lipopolysaccharides destabilize TP53 mRNA through a TLR4-NF-κB-mediated inhibition of the RNA-binding factor Wig-1. Importantly, we show that K. pneumoniae disables two major tumor barriers, oncogene-induced DNA damage signaling and senescence, by impairing p53 transcriptional activity upon DNA damage and oncogenic stress. Furthermore, we found an inverse correlation between the levels of TLR4 and p53 mutation in colorectal tumors. Hence, our data suggest that the repression of p53 by Enterobacteria via TLR4 alleviates the selection pressure for p53 oncogenic mutations and shapes the genomic evolution of cancer.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Enterobacteriaceae/genética , Enterobacteriaceae/metabolismo , Humanos , Lipopolisacáridos/farmacología , ARN Mensajero/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA