Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1384072, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39376663

RESUMEN

There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1ß and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.


Asunto(s)
Encéfalo , Caspasa 1 , Modelos Animales de Enfermedad , Meningitis Meningocócica , Neisseria meningitidis , Piroptosis , Animales , Neisseria meningitidis/patogenicidad , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Ratones , Meningitis Meningocócica/microbiología , Caspasa 1/metabolismo , Encéfalo/patología , Encéfalo/microbiología , Encéfalo/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Caspasas/metabolismo , Caspasas Iniciadoras/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas de Unión a Fosfato/genética , Interleucina-1beta/metabolismo , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Sistemas de Secreción Bacterianos/genética , Femenino , Interleucina-18/metabolismo , Gasderminas
2.
Fluids Barriers CNS ; 18(1): 53, 2021 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-34863201

RESUMEN

BACKGROUND: The Gram-negative bacterium Neisseria meningitidis (Nm) can cause meningitis in humans, but the host signalling pathways manipulated by Nm during central nervous system (CNS) entry are not completely understood. METHODS: We investigate the role of the mitogen-activated protein kinases (MAPK) Erk1/2 and p38 in an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with Nm serogroup B (NmB) and serogroup C (NmC) strains. A transcriptome analysis of HIBCPP cells following infection with Nm by massive analysis of cDNA ends (MACE) was done to further characterize the cellular response to infection of the barrier. RESULTS: Interestingly, whereas NmB and NmC wild type strains required active Erk1/2 and p38 pathways for infection, invasion by capsule-deficient mutants was independent of Erk1/2 and, in case of the NmB strain, of p38 activity. The transcriptome analysis of HIBCPP cells following infection with Nm demonstrated specific regulation of genes involved in the immune response dependent on Erk1/2 signalling. Gene ontology (GO) analysis confirmed loss of MAPK signalling after Erk1/2 inhibition and revealed an additional reduction of cellular responses including NFκB and JAK-STAT signalling. Interestingly, GO terms related to TNF signalling and production of IL6 were lost specifically following Erk1/2 inhibition during infection with wild type Nm, which correlated with the reduced infection rates by the wild type in absence of Erk1/2 signalling. CONCLUSION: Our data point towards a role of MAPK signalling during infection of the CP epithelium by Nm, which is strongly influenced by capsule expression, and affects infection rates as well as the host cell response.


Asunto(s)
Barrera Hematoencefálica , Líquido Cefalorraquídeo , Plexo Coroideo , Células Epiteliales , Interacciones Huésped-Patógeno/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neisseria meningitidis/patogenicidad , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Línea Celular Tumoral , Líquido Cefalorraquídeo/inmunología , Líquido Cefalorraquídeo/metabolismo , Líquido Cefalorraquídeo/microbiología , Plexo Coroideo/inmunología , Plexo Coroideo/metabolismo , Plexo Coroideo/microbiología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos
3.
Pathog Dis ; 79(7)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34410374

RESUMEN

Neisseria meningitis (Nm) is a human-specific bacterial pathogen that can cause sepsis and meningitis. To cause meningitis Nm must enter the central nervous system (CNS) across one of the barriers between the blood and the brain. We have previously shown that a capsule-depleted Serogroup B strain of Nm displays enhanced invasion into human choroid plexus (CP) epithelial papilloma (HIBCPP) cells, which represent an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB). Still, the processes involved during CNS invasion by Nm, especially the role of host cell actin cytoskeleton remodeling, are not investigated in detail. Here, we demonstrate that invasion into CP epithelial cells by encapsulated and capsule-depleted Nm is mediated by distinct host cell pathways. Whereas a Serogroup B wild-type strain enters HIBCPP cells by a possibly dynamin-independent, but actin related protein 2/3 (Arp2/3)-dependent mechanism, invasion by a capsule-depleted mutant is reduced by the dynamin inhibitor dynasore and Arp2/3-independent. Both wild-type and mutant bacteria require Src kinase activity for entry into HIBCPP cells. Our data show that Nm can employ different mechanisms for invasion into the CP epithelium dependent on the presence of a capsule.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Cápsulas/metabolismo , Dinaminas/metabolismo , Células Epiteliales/microbiología , Infecciones Meningocócicas/metabolismo , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/metabolismo , Actinas/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Células Cultivadas , Plexo Coroideo/metabolismo , Plexo Coroideo/microbiología , Endocitosis , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/microbiología , Interacciones Huésped-Patógeno , Humanos , Neisseria meningitidis/patogenicidad , Transducción de Señal , Virulencia , Familia-src Quinasas/metabolismo
4.
BMC Microbiol ; 20(1): 92, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32295520

RESUMEN

BACKGROUND: Neisseria meningitidis serogroups W and Y are the most common serogroups causing invasive meningococcal disease in Sweden. The majority of cases are caused by the serogroup W UK 2013 strain of clonal complex (cc) 11, and subtype 1 of the serogroup Y, YI strain of cc23. In this study, virulence factors of several lineages within cc11 and cc23 were investigated in transgenic BALB/c mice expressing human transferrin. Transgenic mice were infected intraperitoneally with serogroup W and Y isolates. Levels of bacteria and the proinflammatory cytokine CXCL1 were determined in blood collected 3 h and 24 h post-infection. Apoptosis was investigated in immune cells from peritoneal washes of infected mice. Adhesion and induction of apoptosis in human epithelial cells were also scored. RESULTS: The levels of bacteraemia, CXCL1, and apoptosis were higher in serogroup W infected mice than in serogroup Y infected mice. Serogroup W isolates also induced higher levels of apoptosis and adhesion in human epithelial cells. No significant differences were observed between different lineages within cc11 and cc23. CONCLUSIONS: N. meningitidis Serogroup W displayed a higher virulence in vivo in transgenic mice, compared to serogroup Y. This was reflected by higher bacteremia, proinflammatory activity, and ability to induce apoptosis in mouse immune cells and human epithelial cells.


Asunto(s)
Bacteriemia/microbiología , Quimiocina CXCL1/sangre , Infecciones Meningocócicas/inmunología , Neisseria meningitidis/patogenicidad , Transferrina/genética , Animales , Apoptosis , Bacteriemia/inmunología , Adhesión Celular , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Infecciones Meningocócicas/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neisseria meningitidis/genética , Neisseria meningitidis/aislamiento & purificación , Serogrupo , Suecia
5.
PLoS Pathog ; 16(3): e1008372, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32208456

RESUMEN

It is increasingly being recognised that the interplay between commensal and pathogenic bacteria can dictate the outcome of infection. Consequently, there is a need to understand how commensals interact with their human host and influence pathogen behaviour at epithelial surfaces. Neisseria meningitidis, a leading cause of sepsis and meningitis, exclusively colonises the human nasopharynx and shares this niche with several other Neisseria species, including the commensal Neisseria cinerea. Here, we demonstrate that during adhesion to human epithelial cells N. cinerea co-localises with molecules that are also recruited by the meningococcus, and show that, similar to N. meningitidis, N. cinerea forms dynamic microcolonies on the cell surface in a Type four pilus (Tfp) dependent manner. Finally, we demonstrate that N. cinerea colocalises with N. meningitidis on the epithelial cell surface, limits the size and motility of meningococcal microcolonies, and impairs the effective colonisation of epithelial cells by the pathogen. Our data establish that commensal Neisseria can mimic and affect the behaviour of a pathogen on epithelial cell surfaces.


Asunto(s)
Adhesión Bacteriana , Células Epiteliales/microbiología , Fimbrias Bacterianas/metabolismo , Neisseria cinerea/crecimiento & desarrollo , Neisseria meningitidis/crecimiento & desarrollo , Células A549 , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Neisseria cinerea/patogenicidad , Neisseria meningitidis/patogenicidad
6.
Biomed Environ Sci ; 33(2): 114-122, 2020 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-32131958

RESUMEN

OBJECTIVE: To compare the pathogenicity of isolates of sequence type 7 (ST-7) Neisseria meningitidis( N. meningitidis) belonging to four different serogroups (A, B, C, and X). METHODS: Four ST-7 N. meningitidis isolates serogrouped as A, B, C, and X and characterized by different capsule structures, were examined for their adhesion and invasion properties, and their ability to induce cytokine release and apoptosis in the host cell (the A549 cell line). RESULTS: Among the four ST-7 N. meningitidis isolates, the serogroup A isolate possessed the strongest adhesion and invasion ability. This isolate also induced the release of the highest levels of the pro-inflammatory mediators interleukin-6, interleukin-1ß, and interferon, and the highest apoptosis rate in the host cells. However, there was no significant difference in interleukin-8 and tumor necrosis factor-α secretion between the four isolates. Based on the findings, the serogroup X N. meningitidis isolate had the weakest pathogenicity, whereas there was almost no difference in the pathogenicity of the isolates from serogroups B and C. CONCLUSIONS: The differences in the capsular structure of the four isolates of ST-7 N. meningitidis affected their pathogenic capacities. The findings also imply that the hyperinvasive ST-7 N. meningitidis lineage may include hypoinvasive isolates.


Asunto(s)
Neisseria meningitidis/patogenicidad , Neisseria meningitidis/genética , Serogrupo , Virulencia
7.
Infect Immun ; 88(4)2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-31964742

RESUMEN

Neisseria meningitidis, a common cause of sepsis and bacterial meningitis, infects the meninges and central nervous system (CNS), primarily via paracellular traversal across the blood-brain barrier (BBB) or blood-cerebrospinal fluid barrier. N. meningitidis is often present asymptomatically in the nasopharynx, and the nerves extending between the nasal cavity and the brain constitute an alternative route by which the meningococci may reach the CNS. To date, the cellular mechanisms involved in nerve infection are not fully understood. Peripheral nerve glial cells are phagocytic and are capable of eliminating microorganisms, but some pathogens may be able to overcome this protection mechanism and instead infect the glia, causing cell death or pathology. Here, we show that N. meningitidis readily infects trigeminal Schwann cells (the glial cells of the trigeminal nerve) in vitro in both two-dimensional and three-dimensional cell cultures. Infection of trigeminal Schwann cells may be one mechanism by which N. meningitidis is able to invade the CNS. Infection of the cells led to multinucleation and the appearance of atypical nuclei, with the presence of horseshoe nuclei and the budding of nuclei increasing over time. Using sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics followed by bioinformatics pathway analysis, we showed that N. meningitidis induced protein alterations in the glia that were associated with altered intercellular signaling, cell-cell interactions, and cellular movement. The analysis also suggested that the alterations in protein levels were consistent with changes occurring in cancer. Thus, infection of the trigeminal nerve by N. meningitidis may have ongoing adverse effects on the biology of Schwann cells, which may lead to pathology.


Asunto(s)
Interacciones Huésped-Patógeno , Neisseria meningitidis/crecimiento & desarrollo , Neisseria meningitidis/patogenicidad , Células de Schwann/microbiología , Células de Schwann/patología , Nervio Trigémino/citología , Animales , Células Cultivadas , Ratones Transgénicos , Proteoma/análisis , Proteómica
8.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570554

RESUMEN

Serotype 4821 (ST-4821) clonal complex (cc4821) Neisseria meningitidis strains are divided into two groups (groups I and II) according to the core genome-based phylogenetic analysis. Group I contains the greater number of invasive disease isolates. However, the differences in pathogenicity between the two groups are unclear. In this study, the pathogenicity of cc4821 isolates (n = 28) belonging to group I and group II (each containing eight invasive isolates and six isolates from healthy carriers) was investigated, including adhesion, invasion, and induction of interleukin-6 (IL-6) and interleukin-8 (IL-8) release from host cells (Hep2 and A549). The invasive isolates had higher adhesion and invasion capabilities than the carried isolates in both groups. The carried cc4821 isolates in group I had stronger invasion capability than those in group II. Invasive isolates induced more IL-6 and IL-8 secretion than carried isolates in both groups. The carried cc4821 isolates stimulated higher levels of IL-8 in group I than in group II. The isolates were defined as hyperadherent and hypoadherent groups according to their adhesion ability and as hyperinvasive and hypoinvasive groups based on their invasion ability. The hyperadherent and hyperinvasive isolates mediated more IL-6 and IL-8 release than the hypoadherent and hypoinvasive isolates. There was no difference in the level of cytokine release when cc4821 isolates lost their adhesion and invasion capability after lysis. The results revealed that differences in pathogenicity existed between the two groups and that the differences were mainly determined by differences in adhesion and invasion capabilities.


Asunto(s)
Adhesión Bacteriana/fisiología , Interleucina-6/inmunología , Interleucina-8/inmunología , Meningitis Meningocócica/patología , Neisseria meningitidis/patogenicidad , Células A549 , Línea Celular , China , Humanos , Meningitis Meningocócica/microbiología , Neisseria meningitidis/aislamiento & purificación , Serogrupo , Virulencia
9.
Virulence ; 10(1): 677-694, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31274379

RESUMEN

The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.


Asunto(s)
Infecciones Meningocócicas/inmunología , Neisseria meningitidis/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Anafilatoxinas/inmunología , Animales , Quimiocinas/inmunología , Citocinas/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neisseria meningitidis/patogenicidad , Neutrófilos/inmunología , Neutrófilos/microbiología , Receptor de Anafilatoxina C5a/genética , Receptores de Complemento/genética , Sepsis
10.
Cell Microbiol ; 21(11): e13063, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31167044

RESUMEN

Neisseria meningitidis is a Gram-negative bacterium that asymptomatically colonises the nasopharynx of humans. For an unknown reason, N. meningitidis can cross the nasopharyngeal barrier and invade the bloodstream where it becomes one of the most harmful extracellular bacterial pathogen. This infectious cycle involves the colonisation of two different environments. (a) In the nasopharynx, N. meningitidis grow on the top of mucus-producing epithelial cells surrounded by a complex microbiota. To survive and grow in this challenging environment, the meningococcus expresses specific virulence factors such as polymorphic toxins and MDAΦ. (b) Meningococci have the ability to survive in the extra cellular fluids including blood and cerebrospinal fluid. The interaction of N. meningitidis with human endothelial cells leads to the formation of typical microcolonies that extend overtime and promote vascular injury, disseminated intravascular coagulation, and acute inflammation. In this review, we will focus on the interplay between N. meningitidis and these two different niches at the cellular and molecular level and discuss the use of inhibitors of piliation as a potent therapeutic approach.


Asunto(s)
Infecciones Meningocócicas/microbiología , Nasofaringe/microbiología , Neisseria meningitidis/patogenicidad , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Vasos Sanguíneos/microbiología , Células Endoteliales/patología , Células Epiteliales/patología , Interacciones Microbiota-Huesped , Humanos , Inovirus/crecimiento & desarrollo , Inovirus/patogenicidad , Infecciones Meningocócicas/sangre , Infecciones Meningocócicas/líquido cefalorraquídeo , Neisseria meningitidis/metabolismo , Factores de Virulencia
11.
Proc Natl Acad Sci U S A ; 116(17): 8481-8486, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30948644

RESUMEN

Despite the availability of antibiotics and vaccines, Neisseria meningitidis remains a major cause of meningitis and sepsis in humans. Due to its extracellular lifestyle, bacterial adhesion to host cells constitutes an attractive therapeutic target. Here, we present a high-throughput microscopy-based approach that allowed the identification of compounds able to decrease type IV pilus-mediated interaction of bacteria with endothelial cells in the absence of bacterial or host cell toxicity. Compounds specifically inhibit the PilF ATPase enzymatic activity that powers type IV pilus extension but remain inefficient on the ATPase that promotes pilus retraction, thus leading to rapid pilus disappearance from the bacterial surface and loss of pili-mediated functions. Structure activity relationship of the most active compound identifies specific moieties required for the activity of this compound and highlights its specificity. This study therefore provides compounds targeting pilus biogenesis, thereby inhibiting bacterial adhesion, and paves the way for a novel therapeutic option for meningococcal infections.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Fimbrias Bacterianas , Adenosina Trifosfatasas/antagonistas & inhibidores , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Células Cultivadas , Fimbrias Bacterianas/efectos de los fármacos , Fimbrias Bacterianas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neisseria meningitidis/enzimología , Neisseria meningitidis/patogenicidad
12.
Methods Mol Biol ; 1969: 123-133, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30877674

RESUMEN

Neisseria meningitidis and Neisseria gonorrhoeae are obligate pathogens of the human host. Due to their adaptation to the human host, many factors required for infection are specialized for the human host to the point that natural infection processes are difficult to replicate in animal models. Immortalized human cell lines have been used to identify the host factors necessary for successful colonization of human mucosal surfaces. One such model is the Detroit 562 pharyngeal immortalized cell monolayer model which is used to measure the rate of attachment to and invasion of N. meningitidis and N. gonorrhoeae into epithelial cells. The methodology of this assay, as well as the maintenance of Detroit 562 cells necessary for the experiment, will be described.


Asunto(s)
Células Epiteliales/microbiología , Gonorrea/microbiología , Infecciones Meningocócicas/microbiología , Modelos Biológicos , Neisseria gonorrhoeae/patogenicidad , Neisseria meningitidis/patogenicidad , Faringe/microbiología , Células Cultivadas , Humanos
13.
Malays J Pathol ; 41(3): 351-354, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31901921

RESUMEN

INTRODUCTION: Neisseria meningitidis infections often cause severe meningitis as well as bacteraemia. However, cellulitis in meningococcal diseases have rarely been described. Here, we report a case of right lower limb cellulitis caused by N. meningitidis. CASE REPORT: A 69-year-old woman presented with fever and lower limb swelling. She had diabetes mellitus, hypertension, dyslipidaemia and a history of surgical resection of vulvar carcinoma. N. meningitidis was isolated from her blood culture. DISCUSSION: This report provides additional evidence in support of N. meningitidis as a cause of cellulitis.


Asunto(s)
Celulitis (Flemón)/patología , Infecciones Meningocócicas/patología , Neisseria meningitidis/patogenicidad , Anciano , Celulitis (Flemón)/diagnóstico , Femenino , Humanos , Extremidad Inferior/microbiología , Extremidad Inferior/patología , Infecciones Meningocócicas/diagnóstico
14.
Microb Cell Fact ; 17(1): 157, 2018 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-30285743

RESUMEN

BACKGROUND: Outer membrane vesicles (OMVs) are nanoparticles released by Gram-negative bacteria and can be used as vaccines. Often, detergents are used to promote release of OMVs and to remove the toxic lipopolysaccharides. Lipopolysaccharides can be detoxified by genetic modification such that vesicles spontaneously produced by bacteria can be directly used as vaccines. The use of spontaneous OMVs has the advantage that no separate extraction step is required in the purification process. However, the productivity of spontaneous OMVs by bacteria at optimal growth conditions is low. One of many methods for increasing OMV formation is to reduce the linkage of the outer membrane to the peptidoglycan layer by knocking out the rmpM gene. A previous study showed that for Neisseria meningitidis this resulted in release of more OMVs. Furthermore, cysteine depletion was found to trigger OMV release and at the same time cause reduced growth and oxidative stress responses. Here we study the effect of growth rate and oxidative stress on OMV release. RESULTS: First, we identified using chemostat and accelerostat cultures of N. meningitidis that increasing the growth rate from 0.03 to 0.18 h-1 has a limited effect on OMV productivity. Thus, we hypothesized that oxidative stress is the trigger for OMV release and that oxidative stress can be introduced directly by increasing the dissolved oxygen tension of bacterial cultures. Slowly increasing oxygen concentrations in a N. meningitidis changestat showed that an increase from 30 to 150% air saturation improved OMV productivity four-fold. Batch cultures controlled at 100% air saturation increased OMV productivity three-fold over batch cultures controlled at 30% air saturation. CONCLUSION: Increased dissolved oxygen tension induces the release of outer membrane vesicles in N. meningitidis cultures. Since oxygen concentration is a well-controlled process parameter of bacterial cultures, this trigger can be applied as a convenient process parameter to induce OMV release in bacterial cultures. Improved productivity of OMVs not only improves the production costs of OMVs as vaccines, it also facilitates the use of OMVs as adjuvants, enzyme carriers, or cell-specific drug delivery vehicles.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Neisseria meningitidis/patogenicidad , Oxígeno/metabolismo , Estrés Oxidativo
15.
Med. infant ; 24(4): 320-324, dic. 2017. tab, ilus
Artículo en Español | LILACS | ID: biblio-878278

RESUMEN

Introducción: Las meningitis bacterianas en niños son causa de importante morbimortalidad. Los principales agentes etiológicos son Neisseria meningitidis, Streptococcus pneumoniae y Haemophilus influenzae. En los últimos años, luego de la introducción sucesiva de vacunas conjugadas al calendario nacional de inmunizaciones, se ha visto un cambio en la epidemiología de estas infecciones. Objetivo: Describir las características clínicas, epidemiológicas y evolutivas de los niños hospitalizados con meningitis bacteriana confirmada microbiológicamente entre 2011 y 2016 en un hospital de tercer nivel de complejidad. Materiales y métodos: Cohorte retrospectiva. Se incluyeron niños entre 1 mes de vida y 17 años con cuadro clínico compatible con meningitis bacteriana y cultivo positivo y/o PCR en líquido cefalorraquídeo y/o hemocultivos positivos para Neisseria meningitidis, Streptococcus pneumoniae y Haemophilus influenzae b. Se registraron las características demográficas, clínicas y evolutivas hasta los 30 días del egreso. Se utilizó mediana y rango intercuartilo (RIC) para variables continuas y porcentaje para variables categóricas. Se utilizó Stata 10. Resultados: n=65. Edad: mediana de 9 meses (RIC 4-35). Varones: 58% (n=38). Se identificó Neisseria meningitidis en un 48% (n=31), Haemophilus influenzae b en un 26% (n=17) y Streptococcus pneumoniae en un 26% (n=17). El 26% (n=17) de los pacientes presentaba alguna comorbilidad. Tuvieron hemocultivos positivos el 62% (n = 40) de los pacientes y 86% (n=55) de los líquidos cefalorraquídeos. Todos los pacientes recibieron tratamiento antimicrobiano con ceftriaxona tanto como tratamiento empírico como definitivo y 92% (n=60) recibieron corticoides empíricos. La mediana de días de internación fue de 11 (RIC 8-17). El 28% (n=18) requirió cuidados intensivos, y el 8% (n=5) falleció. Durante el período de estudio se observó que la frecuencia de meningitis por Streptococcus pneumoniae disminuyó en el final del estudio (9% en 2016 vs 60% en 2011), mientras que la frecuencia de meningitis por Neisseria meningitidis en 2016 fue mayor que al inicio del período (64% en 2016 vs. 40% en 2011). La frecuencia de identificación de Haemophilus influenzae b se mantuvo estable. Conclusiones: Las meningitis bacterianas confirmadas por Neisseria meningitidis, Streptococcus pneumoniae y Haemophilus influenzae b prevalecieron en niños menores de 12 meses. En esta cohorte se observó un predominio de las infecciones por Neisseria meningitidis en los últimos años, y una disminución en la frecuencia de meningitis por Streptococcus pneumoniae en el período post introducción de la vacuna conjugada 13 valente al calendario nacional de inmunizaciones. (AU)


Introduction: In children, bacterial meningitis is an important cause of morbidity and mortality. The main etiological agents are Neisseria meningitidis, Streptococcus pneumoniae, and Haemophilus influenzae. Over the last years, the successive introduction of conjugated vaccines in the national immunization calendar has led to a change in the epidemiology of these infections. Objective: To describe the clinical and epidemiological features and outcome of children admitted because of microbiologically confirmed meningitis seen between 2011 and 2016 at a tertiary care hospital. Material and methods: A retrospective cohort study was conducted. Children between 1 month of life and 17 years of age with clinical features compatible with bacterial meningitis and positive cultures and/or PCR in cerebrospinal fluid (CSF) and/or positive blood cultures for Neisseria meningitidis, Streptococcus pneumoniae, and Haemophilus influenzae b were included. Demographic, clinical, and outcome features were recorded until 30 days after discharge. Median and interquartile range (IQR) were calculated for continuous variables and percentages for categorical variables. The Stata 10 program was used. Results: n=65. Age: median was 9 months (IQR 4-35). Boys: 58% (n=38). Neisseria meningitidis was identified in 48% (n=31), Haemophilus influenzae b in 26% (n=17), and Streptococcus pneumoniae in 26% (n=17). Overall, 26% (n=17) of the patients presented with comorbidities. Positive blood cultures were found in 62% (n = 40) and positive CSF cultures in 86% (n=55) of the patients. All patients received antimicrobial treatment with ceftriaxone both empirically and as final treatment and corticosteroids were empirically started in 92% (n=60). Median hospital stay was 11 days (IQR 8-17). Overall, 28% (n=18) required intensive care and 8% (n=5) of the patients died. The incidence of meningitis due to Streptococcus pneumoniae was observed to diminish at the end of the study period (9% in 2016 vs 60% in 2011), while the incidence of meningitis due to Neisseria meningitidis in 2016 was higher than at the end of the study period (64% in 2016 vs. 40% in 2011). The frequency of identification of Haemophilus influenzae b remained stable. Conclusions: Confirmed bacterial infections due to Neisseria meningitidis, Streptococcus pneumoniae, and Haemophilus influenzae b were prevalent in infants younger than 12 months of age in this cohort of patients. Infections due to Neisseria meningitidis predominated over the last years and the incidence of meningitis due to Streptococcus pneumoniae diminished after the introduction of the 13 valent conjugated vaccine was introduced in the national immunization calendar.(AU)


Asunto(s)
Humanos , Lactante , Preescolar , Niño , Adolescente , Streptococcus pneumoniae/patogenicidad , Meningitis Bacterianas/diagnóstico , Meningitis Bacterianas/líquido cefalorraquídeo , Meningitis Bacterianas/prevención & control , Meningitis Bacterianas/epidemiología , Haemophilus influenzae tipo b/patogenicidad , Neisseria meningitidis/patogenicidad
16.
PLoS Pathog ; 13(7): e1006495, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704569

RESUMEN

Neisseria meningitidis is a commensal of human nasopharynx. In some circumstances, this bacteria can invade the bloodstream and, after crossing the blood brain barrier, the meninges. A filamentous phage, designated MDAΦ for Meningococcal Disease Associated, has been associated with invasive disease. In this work we show that the prophage is not associated with a higher virulence during the bloodstream phase of the disease. However, looking at the interaction of N. meningitidis with epithelial cells, a step essential for colonization of the nasopharynx, we demonstrate that the presence of the prophage, via the production of viruses, increases colonization of encapsulated meningococci onto monolayers of epithelial cells. The analysis of the biomass covering the epithelial cells revealed that meningococci are bound to the apical surface of host cells by few layers of heavily piliated bacteria, whereas, in the upper layers, bacteria are non-piliated but surrounded by phage particles which (i) form bundles of filaments, and/or (ii) are in some places associated with bacteria. The latter are likely to correspond to growing bacteriophages during their extrusion through the outer membrane. These data suggest that, as the biomass increases, the loss of piliation in the upper layers of the biomass does not allow type IV pilus bacterial aggregation, but is compensated by a large production of phage particles that promote bacterial aggregation via the formation of bundles of phage filaments linked to the bacterial cell walls. We propose that MDAΦ by increasing bacterial colonization in the mucosa at the site-of-entry, increase the occurrence of diseases.


Asunto(s)
Inovirus/fisiología , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/patogenicidad , Neisseria meningitidis/virología , Animales , Adhesión Bacteriana , Células Epiteliales/microbiología , Femenino , Fimbrias Bacterianas/fisiología , Humanos , Ratones , Ratones SCID , Nasofaringe/microbiología , Neisseria meningitidis/crecimiento & desarrollo , Neisseria meningitidis/fisiología , Profagos/fisiología , Virulencia
17.
Infect Disord Drug Targets ; 17(2): 81-85, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28460613

RESUMEN

BACKGROUND: Toxin antitoxin systems is the one of the important elements among pathogenic bacteria which have proven roles such as biofilm formation, cell programmed death and persister cells formation. Neisseria meningitidis causing serious diseases in humans must be highlighted. OBJECTIVE: The current study aimed to identify the mazEF and relBE TA systems in N. meningitidis. METHOD: The potential TA loci were searched in RASTA database by bioinformatics analysis and then, experimental analysis was performed by PCR assay. PCR products were confirmed by sequencing. RESULTS: Our findings demonstrated that mazEF and relBE TA systems were positive by PCR assay and results of sequencing confirmed the PCR results. CONCLUSION: Notably, our highlighted findings are the first report of mazEF and RelBE TA loci in N. meningitides. Finally, we strongly recommended that laboratory experiments should be performed to identify the roles of these TA loci in N. meningitides.


Asunto(s)
Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Sistemas Toxina-Antitoxina/genética , Proteínas Bacterianas/metabolismo , Biología Computacional , ADN Bacteriano/análisis , ADN Bacteriano/genética , Humanos , Neisseria meningitidis/patogenicidad , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo
18.
PLoS Pathog ; 13(4): e1006251, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28384279

RESUMEN

The development of meningococcal disease, caused by the human pathogen Neisseria meningitidis, is preceded by the colonization of the epithelial layer in the nasopharynx. After initial adhesion to host cells meningococci form aggregates, through pilus-pilus interactions, termed microcolonies from which the bacteria later detach. Dispersal from microcolonies enables access to new colonization sites and facilitates the crossing of the cell barrier; however, this process is poorly understood. In this study, we used live-cell imaging to investigate the process of N. meningitidis microcolony dispersal. We show that direct contact with host cells is not required for microcolony dispersal, instead accumulation of a host-derived effector molecule induces microcolony dispersal. By using a host-cell free approach, we demonstrated that lactate, secreted from host cells, initiate rapid dispersal of microcolonies. Interestingly, metabolic utilization of lactate by the bacteria was not required for induction of dispersal, suggesting that lactate plays a role as a signaling molecule. Furthermore, Neisseria gonorrhoeae microcolony dispersal could also be induced by lactate. These findings reveal a role of host-secreted lactate in microcolony dispersal and virulence of pathogenic Neisseria.


Asunto(s)
Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Ácido Láctico/metabolismo , Infecciones Meningocócicas/metabolismo , Neisseria meningitidis/patogenicidad , Fimbrias Bacterianas/microbiología , Humanos , Neisseria gonorrhoeae/patogenicidad , Virulencia/fisiología
19.
BMC Genomics ; 18(1): 282, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28388876

RESUMEN

BACKGROUND: Commensal bacteria like Neisseria meningitidis sometimes cause serious disease. However, genomic comparison of hyperinvasive and apathogenic lineages did not reveal unambiguous hints towards indispensable virulence factors. Here, in a systems biological approach we compared gene expression of the invasive strain MC58 and the carriage strain α522 under different ex vivo conditions mimicking commensal and virulence compartments to assess the strain-specific impact of gene regulation on meningococcal virulence. RESULTS: Despite indistinguishable ex vivo phenotypes, both strains differed in the expression of over 500 genes under infection mimicking conditions. These differences comprised in particular metabolic and information processing genes as well as genes known to be involved in host-damage such as the nitrite reductase and numerous LOS biosynthesis genes. A model based analysis of the transcriptomic differences in human blood suggested ensuing metabolic flux differences in energy, glutamine and cysteine metabolic pathways along with differences in the activation of the stringent response in both strains. In support of the computational findings, experimental analyses revealed differences in cysteine and glutamine auxotrophy in both strains as well as a strain and condition dependent essentiality of the (p)ppGpp synthetase gene relA and of a short non-coding AT-rich repeat element in its promoter region. CONCLUSIONS: Our data suggest that meningococcal virulence is linked to transcriptional buffering of cryptic genetic variation in metabolic genes including global stress responses. They further highlight the role of regulatory elements for bacterial virulence and the limitations of model strain approaches when studying such genetically diverse species as N. meningitidis.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Variación Genética , Neisseria meningitidis/genética , Transcriptoma , Virulencia/genética , Técnicas de Tipificación Bacteriana , Biomarcadores , Análisis por Conglomerados , Biología Computacional/métodos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Genes Reguladores , Genoma Bacteriano , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Meningitis Meningocócica/sangre , Meningitis Meningocócica/metabolismo , Meningitis Meningocócica/microbiología , Anotación de Secuencia Molecular , Neisseria meningitidis/clasificación , Neisseria meningitidis/patogenicidad , Fenotipo , Regiones Promotoras Genéticas
20.
Mil Med ; 182(3): e1713-e1718, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28290948

RESUMEN

INTRODUCTION: The Mortality Surveillance Division (MSD) of the U.S. Armed Forces Medical Examiner System was established in 1998 to improve surveillance for all military deaths although emphasizing deaths from infectious diseases. Establishment of the MSD was part of the 1997 Department of Defense initiative to improve surveillance and response for emerging infectious diseases. Before 1998, mortality surveillance was limited to compiling information from death certificates, a system that provided limited useful information and lacked the timeliness needed to take meaningful action to address emerging infectious disease threats. MATERIALS AND METHODS: The MSD was tasked to quickly identify all infectious disease deaths and the infecting agents. The system developed by the MSD staff identified deaths in near real-time and immediately notified military Public Health authorities of situations that warranted an investigation. Autopsy, medical, and investigative reports were collected. Testing specimens for agent identification was encouraged. The data and information collected were archived in the MSD-developed Medical Mortality Registry (MMR), a database that included all active duty Service Member deaths and contained manner and cause of death with medical, demographic, circumstantial, and diagnostic information. The MMR was the only comprehensive, autopsy-based source for mortality information on active duty military deaths. RESULTS: During 1998-2013, 217 (1.3%) infectious disease deaths were identified among 16,192 noncombat deaths. Of the 217 deaths, 29.5% were classified as respiratory, 18.0% cardiac, 15.2% blood borne, 12.9% nervous system, and 12.4% sepsis. A pathogen was identified for 64.5%. Agents of military interest identified included Neisseria meningitidis, influenza viruses, adenoviruses, and malaria. Neisseria meningitidis was identified in 10 fatal cases; grouping of the agent was done for eight cases. Four were group B, two were C, and two were Y. All eight had been immunized with a quadrivalent meningococcal vaccine. The most commonly detected respiratory agent was influenza virus (nine deaths), three of which were the 2009 pandemic H1N1 influenza virus. Adenoviruses were identified as the infectious agents in a total of nine deaths. Two deaths resulted from Plasmodium falciparum malaria infections acquired in Africa during military deployments. An important but unexplained finding was that Black Service Members made up only16.3% of all military personnel but accounted for 28.6% of all infectious disease deaths. CONCLUSION: The time lag between death and notification of the MSD at the start of this surveillance program was 24 to 48 hours. The lag at the end of the reported surveillance period was 8 to 24 hours. The MSD surveillance system identified an agent in 140 of 217 (64.5%) uniformed deaths. In a similar program by the Centers for Disease Control and Prevention, in 122 cases with specimens, an agent could be identified in 34 (28%). MMR data and information provided strong support for re-establishing the military recruit adenovirus vaccination program, which ceased in 1999 and was finally re-established in 2011. MMR data and information also assisted in monitoring the military meningococcal vaccine program, helped to describe the virulence of circulating influenza viruses, and identified areas where deadly malaria infections were not being prevented.


Asunto(s)
Causas de Muerte/tendencias , Enfermedades Transmisibles/mortalidad , Personal Militar/estadística & datos numéricos , Vigilancia de la Población/métodos , United States Department of Defense/estadística & datos numéricos , Infecciones por Adenoviridae/epidemiología , Médicos Forenses/estadística & datos numéricos , Humanos , Gripe Humana/epidemiología , Malaria/epidemiología , Neisseria meningitidis/patogenicidad , Infecciones por Neisseriaceae/epidemiología , Estados Unidos/epidemiología , United States Department of Defense/organización & administración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA