Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 327
Filtrar
1.
Front Cell Infect Microbiol ; 13: 1113528, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37065199

RESUMEN

The Gram-negative bacterium Neisseria meningitidis, which causes meningitis in humans, has been demonstrated to manipulate or alter host signalling pathways during infection of the central nervous system (CNS). However, these complex signalling networks are not completely understood. We investigate the phosphoproteome of an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with the N. meningitidis serogroup B strain MC58 in presence and absence of the bacterial capsule. Interestingly, our data demonstrates a stronger impact on the phosphoproteome of the cells by the capsule-deficient mutant of MC58. Using enrichment analyses, potential pathways, molecular processes, biological processes, cellular components and kinases were determined to be regulated as a consequence of N. meningitidis infection of the BCSFB. Our data highlight a variety of protein regulations that are altered during infection of CP epithelial cells with N. meningitidis, with the regulation of several pathways and molecular events only being detected after infection with the capsule-deficient mutant. Mass spectrometry proteomics data are available via ProteomeXchange with identifier PXD038560.


Asunto(s)
Neisseria meningitidis , Humanos , Neisseria meningitidis/fisiología , Plexo Coroideo/microbiología , Células Epiteliales/microbiología , Barrera Hematoencefálica/microbiología , Línea Celular Tumoral
2.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34725157

RESUMEN

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


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/fisiología , Fimbrias Bacterianas/fisiología , Neisseria meningitidis/fisiología , Animales , Anticuerpos/uso terapéutico , Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Línea Celular , Evaluación Preclínica de Medicamentos , Femenino , Fimbrias Bacterianas/química , Fimbrias Bacterianas/ultraestructura , Humanos , Infecciones Meningocócicas/tratamiento farmacológico , Ratones SCID
3.
Curr Opin Infect Dis ; 34(1): 34-39, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33278178

RESUMEN

PURPOSE OF REVIEW: Neisseria meningitidis (Nm) is primarily associated with asymptomatic nasopharyngeal carriage and invasive meningococcal disease (sepsis and meningitis), but like N. gonorrhoea (Ng), Nm can colonize urogenital and rectal mucosal surfaces and cause disease. First noted in 2015, but with origins in 2011, male urethritis clusters caused by a novel Nm clade were reported in the USA (the US_NmUC). This review describes research developments that characterize this urogenital-tropic Nm. RECENT FINDINGS: The US_NmUC evolved from encapsulated Nm serogroup C strains. Loss of capsule expression, lipooligosaccharide (LOS) sialylation, genetic acquisition of gonococcal alleles (including the gonococcal anaerobic growth aniA/norB cassette), antimicrobial peptide heteroresistance and high surface expression of a unique factor-H-binding protein, can contribute to the urethra-tropic phenotype. Loss-of-function mutations in mtrC are overrepresented in clade isolates. Similar to Ng, repeat US_NmUC urethritis episodes can occur. The US_NmUC is now circulating in the UK and Southeast Asia. Genomic sequencing has defined the clade and rapid diagnostic tests are being developed for surveillance. SUMMARY: The US_NmUC emerged as a cause of urethritis due to acquisition of gonococcal genetic determinants and phenotypic traits that facilitate urogenital tract infection. The epidemiology and pathogenesis of this urogenital-tropic pathogen continues to be defined.


Asunto(s)
Enfermedades Transmisibles Emergentes/microbiología , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/fisiología , Uretritis/microbiología , Humanos , Neisseria meningitidis/clasificación , Neisseria meningitidis/genética , Neisseria meningitidis/aislamiento & purificación
4.
Semin Immunol ; 50: 101433, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33309166

RESUMEN

Outer Membrane Vesicles (OMV) have received increased attention in recent years as a vaccine platform against bacterial pathogens. OMV from Neisseria meningitidis serogroup B have been extensively explored. Following the success of the MeNZB OMV vaccine in controlling an outbreak of N. meningitidis B in New Zealand, additional research and development resulted in the licensure of the OMV-containing four-component 4CMenB vaccine, Bexsero. This provided broader protection against multiple meningococcal B strains. Advances in the field of genetic engineering have permitted further improvements in the platform resulting in increased yields, reduced endotoxicity and decoration with homologous and heterologous antigens to enhance immuno genicity and provide broader protection. The OMV vaccine platform has been extended to many other pathogens. In this review, we discuss progress in the development of the OMV vaccine delivery platform, highlighting successful applications, together with potential challenges and gaps.


Asunto(s)
Membrana Externa Bacteriana/inmunología , Vacunas Bacterianas/inmunología , Infecciones Meningocócicas/inmunología , Neisseria meningitidis/fisiología , Animales , Ingeniería Genética , Humanos , Inmunidad Heteróloga , Inmunogenicidad Vacunal
5.
Int J Mol Sci ; 21(22)2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33233688

RESUMEN

The Gram-negative diplococcus Neisseria meningitidis, also called meningococcus, exclusively infects humans and can cause meningitis, a severe disease that can lead to the death of the afflicted individuals. To cause meningitis, the bacteria have to enter the central nervous system (CNS) by crossing one of the barriers protecting the CNS from entry by pathogens. These barriers are represented by the blood-brain barrier separating the blood from the brain parenchyma and the blood-cerebrospinal fluid (CSF) barriers at the choroid plexus and the meninges. During the course of meningococcal disease resulting in meningitis, the bacteria undergo several interactions with host cells, including the pharyngeal epithelium and the cells constituting the barriers between the blood and the CSF. These interactions are required to initiate signal transduction pathways that are involved during the crossing of the meningococci into the blood stream and CNS entry, as well as in the host cell response to infection. In this review we summarize the interactions and pathways involved in these processes, whose understanding could help to better understand the pathogenesis of meningococcal meningitis.


Asunto(s)
Barrera Hematoencefálica , Interacciones Huésped-Patógeno , Meningitis Meningocócica/microbiología , Neisseria meningitidis/fisiología , Transducción de Señal , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Plexo Coroideo/metabolismo , Plexo Coroideo/microbiología , Humanos , Meninges/metabolismo , Meninges/microbiología
6.
Allergy Asthma Proc ; 41(5): 386-388, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32867893

RESUMEN

Background: Terminal complement component deficiencies are risk factors for neisserial infections. Objective: To review the clinical characteristics, the diagnosis and the management of patients with a terminal complement component deficiency. Methods: Pertinent articles were selected and reviewed in relation to a case presentation of C6 deficiency. Results: A case of a 56-year old patient with a history of meningitis, chronic rash, and C6 deficiency was presented, followed by discussion of clinical characteristics, diagnosis, and management of terminal complement component deficiencies. Clinical pearls and pitfalls were reviewed for the practicing allergist/immunologist and fellow-in-training. Conclusion: C6 deficiency is the most common terminal complement component deficiency and can present later in age with N. meningitidis infections. Patients can be screened for terminal complement component deficiency by checking CH50.


Asunto(s)
Envejecimiento/fisiología , Complemento C6/deficiencia , Complemento C6/genética , Enfermedades por Deficiencia de Complemento Hereditario/diagnóstico , Meningitis Meningocócica/diagnóstico , Vacunas Meningococicas/inmunología , Neisseria meningitidis/fisiología , Profilaxis Antibiótica , Ensayo de Actividad Hemolítica de Complemento , Femenino , Fibronectinas/análisis , Enfermedades por Deficiencia de Complemento Hereditario/complicaciones , Humanos , Meningitis Meningocócica/etiología , Meningitis Meningocócica/prevención & control , Persona de Mediana Edad , Proteínas Recombinantes/análisis
7.
J Vis Exp ; (161)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32744533

RESUMEN

Meningococcal meningitis is a life-threatening infection that occurs when Neisseria meningitidis (meningococcus, Nm) can gain access to the central nervous system (CNS) by penetrating highly specialized brain endothelial cells (BECs). As Nm is a human-specific pathogen, the lack of robust in vivo model systems makes study of the host-pathogen interactions between Nm and BECs challenging and establishes a need for a human based model that mimics native BECs. BECs possess tighter barrier properties when compared to peripheral endothelial cells characterized by complex tight junctions and elevated trans-endothelial electrical resistance (TEER). However, many in vitro models, such as primary BECs and immortalized BECs, either lack or rapidly lose their barrier properties after removal from the native neural microenvironment. Recent advances in human stem-cell technologies have developed methods for deriving brain-like endothelial cells from induced pluripotent stem-cells (iPSCs) that better phenocopy BECs when compared to other in vitro human models. The use of iPSC-derived BECs (iPSC-BECs) to model Nm-BEC interaction has the benefit of using human cells that possess BEC barrier properties, and can be used to examine barrier destruction, innate immune activation, and bacterial interaction. Here we demonstrate how to derive iPSC-BECs from iPSCs in addition to bacterial preparation, infection, and sample collection for analysis.


Asunto(s)
Encéfalo/citología , Células Endoteliales/citología , Células Endoteliales/microbiología , Células Madre Pluripotentes Inducidas/citología , Neisseria meningitidis/fisiología , Humanos
8.
Front Immunol ; 11: 485, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32269571

RESUMEN

Inflammatory bowel disease (IBD), which comprises ulcerative colitis and Crohn's disease, is an immune-mediated, chronic-relapsing, disabling disorder which is associated with increased mortality and poor patients' quality of life. Patients with IBD are at increased risk of infections for many reasons. In fact, IBD often requires a lifelong immunosuppressive and/or biologic therapy, both commonly associated with respiratory and opportunistic infections, but also gastrointestinal, urinary tract infections, and sepsis. Moreover, impaired spleen function has been found in a considerable proportion of IBD patients, further increasing the risk of developing infections sustained by encapsulated bacteria, such as S. pneumoniae, H. influenzae, and N. meningitidis. Finally, comorbidities and surgery represent additional risk factors for these patients. Despite the availability of vaccinations against the most common serotypes of encapsulated bacteria, uncertainties still exist regarding a proper vaccination strategy and the actual effectiveness of vaccinations in this particular setting. Aim of this narrative review is to focus on the broad topic of vaccinations against encapsulated bacteria in IBD patients, discussing the clinical impact of infections, predisposing factors, vaccinations strategies, and unmet research and clinical needs.


Asunto(s)
Infecciones Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Haemophilus influenzae/fisiología , Enfermedades Inflamatorias del Intestino/inmunología , Neisseria meningitidis/fisiología , Infecciones Oportunistas/inmunología , Streptococcus pneumoniae/fisiología , Animales , Infecciones Bacterianas/etiología , Infecciones Bacterianas/prevención & control , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Infecciones Oportunistas/etiología , Infecciones Oportunistas/prevención & control , Vacunación
9.
Microb Pathog ; 139: 103890, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31765768

RESUMEN

Neisseria meningitidis is a human-restricted bacterium that can invade the bloodstream and cross the blood-brain barrier resulting in life-threatening sepsis and meningitis. Meningococci express a cytoplasmic peroxiredoxin-glutaredoxin (Prx5-Grx) hybrid protein that has also been identified on the bacterial surface. Here, recombinant Prx5-Grx was confirmed as a plasminogen (Plg)-binding protein, in an interaction which could be inhibited by the lysine analogue ε-aminocapronic acid. rPrx5-Grx derivatives bearing a substituted C-terminal lysine residue (rPrx5-GrxK244A), but not the active site cysteine residue (rPrx5-GrxC185A) or the sub-terminal rPrx5-GrxK230A lysine residue, exhibited significantly reduced Plg-binding. The absence of Prx5-Grx did not significantly reduce the ability of whole meningococcal cells to bind Plg, but under hydrogen peroxide-mediated oxidative stress, the N. meningitidis Δpxn5-grx mutant survived significantly better than the wild-type or complemented strains. Significantly, using human whole blood as a model of meningococcal bacteremia, it was found that the N. meningitidis Δpxn5-grx mutant had a survival defect compared with the parental or complemented strain, confirming an important role for Prx5-Grx in meningococcal pathogenesis.


Asunto(s)
Glutarredoxinas/metabolismo , Interacciones Huésped-Patógeno , Infecciones Meningocócicas/metabolismo , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/fisiología , Peroxirredoxinas/metabolismo , Plasminógeno/metabolismo , Ensayo de Inmunoadsorción Enzimática , Glutarredoxinas/química , Glutarredoxinas/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Infecciones Meningocócicas/diagnóstico , Infecciones Meningocócicas/mortalidad , Mutación , Peroxirredoxinas/química , Peroxirredoxinas/genética , Plasminógeno/química , Pronóstico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
10.
Cell Microbiol ; 22(1): e13132, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31658405

RESUMEN

The skull, spine, meninges, and cellular barriers at the blood-brain and the blood-cerebrospinal fluid interfaces well protect the brain and meningeal spaces against microbial invasion. However, once in the bloodstream, a range of pathogenic bacteria is able to reach the brain and cause meningitis. Despite advances in antibacterial therapy, bacterial meningitis remains one of the most important infectious diseases worldwide. The most common causative bacteria in children and adults are Streptococcus pneumoniae and Neisseria meningitidis associated with high morbidity and mortality, while among neonates, most cases of bacterial meningitis are due to group B Streptococcus and Escherichia coli. Here we summarise our current knowledge on the strategies used by these bacterial pathogens to survive in the bloodstream, to colonise the brain vasculature and to cross the blood-brain barrier.


Asunto(s)
Bacterias/patogenicidad , Barrera Hematoencefálica/microbiología , Animales , Transporte Biológico , Encéfalo/microbiología , Células Endoteliales/microbiología , Humanos , Inflamación , Neisseria meningitidis/patogenicidad , Neisseria meningitidis/fisiología , Streptococcus pneumoniae/patogenicidad , Streptococcus pneumoniae/fisiología , Factores de Virulencia
11.
Clin Immunol ; 210: 108269, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683054

RESUMEN

Genetic studies have led to identification of an increasing number of monogenic primary immunodeficiency disorders. Monoallelic pathogenic gain-of-function (GOF) variants in NFKBIA, the gene encoding IκBα, result in an immunodeficiency disorder, typically accompanied by anhidrotic ectodermal dysplasia (EDA). So far, 14 patients with immunodeficiency due to NFKBIA GOF mutations have been reported. In this study we report three patients from the same family with immunodeficiency, presenting with recurrent respiratory tract infections, bronchiectasis and viral skin conditions due to a novel pathogenic NFKBIA variant (c.106 T > G, p.Ser36Ala), which results in reduced IκBα degradation. Immunological investigations revealed inadequate antibody responses against vaccine antigens, despite hypergammaglobulinemia. Interestingly, none of the studied patients displayed features of EDA. Therefore, missense NFKBIA variants substituting serine 36 of IκBα, differ from the rest of pathogenic GOF NFKBIA variants in that they cause combined immunodeficiency, even in the absence of EDA.


Asunto(s)
Mutación con Ganancia de Función/genética , Síndromes de Inmunodeficiencia/diagnóstico , Leucocitos Mononucleares/inmunología , Meningitis Bacterianas/diagnóstico , Inhibidor NF-kappaB alfa/genética , Neisseria meningitidis/fisiología , Papillomaviridae/fisiología , Infecciones por Pseudomonas/diagnóstico , Pseudomonas aeruginosa/fisiología , Virosis/inmunología , Adulto , Artritis Juvenil , Azitromicina/uso terapéutico , Bronquiectasia , Proliferación Celular , Células Cultivadas , Niño , Displasia Ectodérmica , Gentamicinas/uso terapéutico , Humanos , Síndromes de Inmunodeficiencia/genética , Masculino , Meningitis Bacterianas/tratamiento farmacológico , Linaje , Infecciones por Pseudomonas/tratamiento farmacológico , Virosis/diagnóstico , Verrugas , Adulto Joven
12.
Nat Commun ; 10(1): 4752, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31628314

RESUMEN

Meningococcus utilizes ß-arrestin selective activation of endothelial cell ß2 adrenergic receptor (ß2AR) to cause meningitis in humans. Molecular mechanisms of receptor activation by the pathogen and of its species selectivity remained elusive. We report that ß2AR activation requires two asparagine-branched glycan chains with terminally exposed N-acetyl-neuraminic acid (sialic acid, Neu5Ac) residues located at a specific distance in its N-terminus, while being independent of surrounding amino-acid residues. Meningococcus triggers receptor signaling by exerting direct and hemodynamic-promoted traction forces on ß2AR glycans. Similar activation is recapitulated with beads coated with Neu5Ac-binding lectins, submitted to mechanical stimulation. This previously unknown glycan-dependent mode of allosteric mechanical activation of a G protein-coupled receptor contributes to meningococcal species selectivity, since Neu5Ac is only abundant in humans due to the loss of CMAH, the enzyme converting Neu5Ac into N-glycolyl-neuraminic acid in other mammals. It represents an additional mechanism of evolutionary adaptation of a pathogen to its host.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neisseria meningitidis/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Línea Celular , Membrana Celular/metabolismo , Fimbrias Bacterianas/genética , Células HEK293 , Humanos , Lectinas/metabolismo , Microscopía Confocal , Neisseria meningitidis/fisiología , Polisacáridos/metabolismo , Receptores Adrenérgicos beta 2/genética , Homología de Secuencia de Aminoácido , beta-Arrestinas/metabolismo
13.
EMBO J ; 38(22): e102145, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31609039

RESUMEN

Type IV pili (TFP) are multifunctional micrometer-long filaments expressed at the surface of many prokaryotes. In Neisseria meningitidis, TFP are crucial for virulence. Indeed, these homopolymers of the major pilin PilE mediate interbacterial aggregation and adhesion to host cells. However, the mechanisms behind these functions remain unclear. Here, we simultaneously determined regions of PilE involved in pilus display, auto-aggregation, and adhesion by using deep mutational scanning and started mining this extensive functional map. For auto-aggregation, pili must reach a minimum length to allow pilus-pilus interactions through an electropositive cluster of residues centered around Lys140. For adhesion, results point to a key role for the tip of the pilus. Accordingly, purified pili interacting with host cells initially bind via their tip-located major pilin and then along their length. Overall, these results identify functional domains of PilE and support a direct role of the major pilin in TFP-dependent aggregation and adhesion.


Asunto(s)
Adhesión Bacteriana , Agregación Celular , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/fisiología , Mutación , Neisseria meningitidis/fisiología , Proteínas Fimbrias/química , Regulación Bacteriana de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mutagénesis Sitio-Dirigida
14.
PLoS One ; 14(8): e0203234, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31369555

RESUMEN

Neisserial Heparin Binding Antigen (NHBA) is a surface-exposed lipoprotein of Neisseria meningitidis and a component of the Bexsero vaccine. NHBA is characterized by the presence of a highly conserved Arg-rich region involved in binding to heparin and heparan sulphate proteoglycans present on the surface of host epithelial cells, suggesting a possible role of NHBA during N. meningitidis colonization. NHBA has been shown to be cleaved by the meningococcal protease NalP and by human lactoferrin (hLF), a host protease presents in different body fluids (saliva, breast milk and serum). Cleavage occurs upstream or downstream the Arg-rich region. Since the human nasopharynx is the only known reservoir of infection, we further investigated the susceptibility of NHBA to human proteases present in the saliva to assess whether proteolytic cleavage could happen during the initial steps of colonization. Here we show that human saliva proteolytically cleaves NHBA, and identified human kallikrein 1 (hK1), a serine protease, as responsible for this cleavage. Kallikrein-related peptidases (KLKs) have a distinct domain structure and exist as a family of 15 genes which are differentially expressed in many tissues and in the central nervous system. They are present in plasma, lymph, urine, saliva, pancreatic juices, and other body fluids where they catalyze the proteolysis of several human proteins. Here we report the characterization of NHBA cleavage by the tissue kallikrein, expressed in saliva and the identification of the cleavage site on NHBA both, as recombinant protein or as native protein, when expressed on live bacteria. Overall, these findings provide new insights on NHBA as target of host proteases, highlights thepotential role of NHBA in the Neisseria meningitidis nasopharyngeal colonization, and of kallikrein as a defensive agent against meningococcal infection.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Infecciones Meningocócicas/microbiología , Proteolisis , Saliva/química , Calicreínas de Tejido/metabolismo , Secuencia de Aminoácidos , Humanos , Neisseria meningitidis/fisiología , Proteómica
15.
FASEB J ; 33(11): 12099-12111, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31442074

RESUMEN

The 4 component meningococcus B vaccine (4CMenB) vaccine is the first vaccine containing recombinant proteins licensed for the prevention of invasive meningococcal disease caused by meningococcal serogroup B strains. 4CMenB contains 3 main recombinant proteins, including the Neisseria meningitidis factor H binding protein (fHbp), a lipoprotein able to bind the human factor H. To date, over 1000 aa sequences of fHbp have been identified, and they can be divided into variant groups 1, 2, and 3, which are usually not crossprotective. Nevertheless, previous characterizations of a small set (n = 10) of mAbs generated in humans after 4CMenB immunization revealed 2 human Fabs (huFabs) (1A12, 1G3) with some crossreactivity for variants 1, 2, and 3. This unexpected result prompted us to examine a much larger set of human mAbs (n = 110), with the aim of better understanding the extent and nature of crossreactive anti-fHbp antibodies. In this study, we report an analysis of the human antibody response to fHbp, by the characterization of 110 huFabs collected from 3 adult vaccinees during a 6-mo study. Although the 4CMenB vaccine contains fHbp variant 1, 13 huFabs were also found to be crossreactive with variants 2 and 3. The crystal structure of the crossreactive huFab 1E6 in complex with fHbp variant 3 was determined, revealing a novel, highly conserved epitope distinct from the epitopes recognized by 1A12 or 1G3. Further, functional characterization shows that human mAb 1E6 is able to elicit rabbit, but not human, complement-mediated bactericidal activity against meningococci displaying fHbp from any of the 3 different variant groups. This functional and structural information about the human antibody response upon 4CMenB immunization contributes to further unraveling the immunogenic properties of fHbp. Knowledge gained about the epitope profile recognized by the human antibody repertoire could guide future vaccine design.-Bianchi, F., Veggi, D., Santini, L., Buricchi, F., Bartolini, E., Lo Surdo, P., Martinelli, M., Finco, O., Masignani, V., Bottomley, M. J., Maione, D., Cozzi, R. Cocrystal structure of meningococcal factor H binding protein variant 3 reveals a new crossprotective epitope recognized by human mAb 1E6.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Factor H de Complemento/inmunología , Epítopos/inmunología , Vacunas Meningococicas/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Cristalografía por Rayos X , Epítopos/genética , Epítopos/metabolismo , Variación Genética , Humanos , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/administración & dosificación , Modelos Moleculares , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/inmunología , Neisseria meningitidis/fisiología , Unión Proteica , Conformación Proteica
16.
Neurosci Lett ; 708: 134334, 2019 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-31238130

RESUMEN

Glia are key regulators of inflammatory responses within the central nervous system (CNS) following infection or trauma. We have previously demonstrated the ability of activated astrocytes to rapidly produce pro-inflammatory mediators followed by a transition to an anti-inflammatory cytokine production profile that includes the immunosuppressive cytokine interleukin (IL)-10 and the closely related cytokines IL-19 and IL-24. IL-20, another member of the IL-10 family, is known to modulate immune cell activity in the periphery and we have previously demonstrated that astrocytes constitutively express the cognate receptors for this cytokine. However, the ability of glia to produce IL-20 remains unclear and the effects of this pleiotropic cytokine on glial immune functions have not been investigated. In this study, we report that primary murine and human astrocytes are not an appreciable source of IL-20 following challenge with disparate bacterial species or their components. Importantly, we have determined that astrocyte are responsive to the immunomodulatory actions of this cytokine by showing that recombinant IL-20 administration upregulates microbial pattern recognition receptor expression and induces release of the inflammatory mediator IL-6 by these cells. Taken together, these data suggest that IL-20 acts in a dissimilar manner to other IL-10 family members to augment the inflammatory responses of astrocytes.


Asunto(s)
Astrocitos/metabolismo , Interleucinas/metabolismo , Animales , Astrocitos/microbiología , Células Cultivadas , Humanos , Inmunomodulación , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucinas/farmacología , Ratones Endogámicos C57BL , Neisseria meningitidis/fisiología , Neuroglía/metabolismo , Neuroglía/microbiología , Proteínas Recombinantes/farmacología , Staphylococcus aureus/fisiología , Streptococcus pneumoniae/fisiología , Receptores Toll-Like/metabolismo
17.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31160362

RESUMEN

Acid sphingomyelinase (ASM) is a lipid hydrolase that converts sphingomyelin to ceramide and that can be activated by various cellular stress mechanisms, including bacterial pathogens. Vesicle transportation or trafficking of ASM from the lysosomal compartment to the cell membrane is a prerequisite for its activation in response to bacterial infections; however, the effectors and mechanisms of ASM translocation and activation are poorly defined. Our recent work documented the key importance of ASM for Neisseria meningitidis uptake into human brain microvascular endothelial cells (HBMEC). We clearly identified OpcA to be one bacterial effector promoting ASM translocation and activity, though it became clear that additional bacterial components were involved, as up to 80% of ASM activity and ceramide generation was retained in cells infected with an opcA-deficient mutant. We hypothesized that N. meningitidis might use pilus components to promote the translocation of ASM into HBMEC. Indeed, we found that both live, piliated N. meningitidis and pilus-enriched fractions trigger transient ASM surface display, followed by the formation of ceramide-rich platforms (CRPs). By using indirect immunocytochemistry and direct stochastic optical reconstruction microscopy, we show that the overall number of CRPs with a size of ∼80 nm in the plasma membrane is significantly increased after exposure to pilus-enriched fractions. Infection with live bacteria as well as exposure to pilus-enriched fractions transiently increased cytosolic Ca2+ levels in HBMEC, and this was found to be important for ASM surface display mediated by lysosomal exocytosis, as depletion of cytosolic Ca2+ resulted in a significant decrease in ASM surface levels, ASM activity, and CRP formation.


Asunto(s)
Calcio/fisiología , Ceramidas/metabolismo , Fimbrias Bacterianas/fisiología , Lisosomas/metabolismo , Neisseria meningitidis/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Células Cultivadas , Humanos
18.
J Immunol Res ; 2019: 6193186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31198794

RESUMEN

Meningococcal disease such as sepsis and meningitidis is hallmarked by an excessive inflammatory response. The causative agent, Neisseria meningitidis, expresses the endotoxin lipooligosaccharide (LOS) that is responsible for activation of immune cells and the release of proinflammatory cytokines. One of the most potent proinflammatory cytokines, interleukin-1ß (IL-1ß), is activated following caspase-1 activity in the intracellular multiprotein complex called inflammasome. Inflammasomes are activated by a number of microbial factors as well as danger molecules by a two-step mechanism-priming and licensing of inflammasome activation-but there are no data available regarding a role for inflammasome activation in meningococcal disease. The aim of this study was to investigate if N. meningitidis activates the inflammasome and, if so, the role of bacterial LOS in this activation. Cells were subjected to N. meningitidis, both wild-type (FAM20) and its LOS-deficient mutant (lpxA), and priming as well as licensing of inflammasome activation was investigated. The wild-type LOS-expressing parental FAM20 serogroup C N. meningitidis (FAM20) strain significantly enhanced the caspase-1 activity in human neutrophils and monocytes, whereas lpxA was unable to induce caspase-1 activity as well as to induce IL-1ß release. While the lpxA mutant induced a priming response, measured as increased expression of NLRP3 and IL1B, the LOS-expressing FAM20 further increased this priming. We conclude that although non-LOS components of N. meningitidis contribute to the priming of the inflammasome activity, LOS per se is to be considered as the central component of N. meningitidis virulence, responsible for both priming and licensing of inflammasome activation.


Asunto(s)
Antígenos Bacterianos/metabolismo , Inflamasomas/metabolismo , Lipopolisacáridos/metabolismo , Meningitis Meningocócica/inmunología , Neisseria meningitidis/fisiología , Neutrófilos/inmunología , Factores de Virulencia/metabolismo , Antígenos Bacterianos/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Humanos , Inmunidad Innata , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neisseria meningitidis/patogenicidad , Neutrófilos/microbiología , Transducción de Señal , Células THP-1
19.
Int J Infect Dis ; 85: 37-48, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31085314

RESUMEN

OBJECTIVE: To evaluate the epidemiological profile of invasive meningococcal disease (IMD), meningococcal meningitis, and Neisseria meningitidis carriers in Latin America. METHODS: A systematic review was conducted to identify and analyze studies published in 2008-2018. Incidence rates, case fatality rates (CFRs), and the relative distribution of cases per serogroup by country were assessed. RESULTS: Meningococcal surveillance in Latin America differs among countries, and most systems are based on passive sentinel surveillance. Thirty-nine studies were selected. In 2006, the incidence rate of IMD per 100 000 inhabitants was highest in Brazil (1.9), followed by Uruguay (1.3), Chile (0.8), Argentina (0.7), Colombia and Venezuela (0.3 each), and Mexico (0.06). Brazil reported the highest CFR (20%), followed by Uruguay (15%), Chile (11%), and Venezuela and Argentina (10% each). In 2012, the CFR in Chile increased to approximately 27%. The most frequent serogroups among IMD cases were C in Brazil (2007-2010) and Mexico (2005-2016), W in Chile (2012-2018), and B in Argentina (2012-2015). However, the true burden of IMD in Latin America is probably underestimated due to underreporting of cases. CONCLUSIONS: Improvements in IMD notification, IMD registration, national surveillance programs (including active surveillance systems), diagnostic tools, and characterization of isolates may better elucidate the true epidemiological burden of IMD in Latin America.


Asunto(s)
Infecciones Meningocócicas/epidemiología , Humanos , Incidencia , América Latina/epidemiología , Meningitis Meningocócica/epidemiología , Infecciones Meningocócicas/economía , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/genética , Neisseria meningitidis/aislamiento & purificación , Neisseria meningitidis/fisiología
20.
Arch Immunol Ther Exp (Warsz) ; 67(4): 237-248, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31030218

RESUMEN

The present studies prove that conjugation of meningococcal lipooligosaccharides through their non-reducing terminus conserves their inner epitopes resulting in conjugates potent to induce a protective immune response. Four different oligosaccharides were obtained by specific degradations of the same L7 lipooligosaccharide (L7-LOS), and each was linked to tetanus toxoid by direct reductive amination. Two were truncated oligosaccharides with incomplete inner epitopes and were obtained by mild acid hydrolysis of lipooligosaccharide. The terminal galactose of one oligosaccharide was additionally enzymatically oxidized. These oligosaccharides were conjugated through a newly exposed terminal Kdo in reducing end or through oxidized galactose localized at non-reducing end of the core, respectively. The third was a full-length oligosaccharide obtained by O-deacylation of the L7-LOS and subsequent enzymatic removal of phosphate substituents from its lipid A moiety. The fourth one was also a full-length O-deacylated lipooligosaccharide, but treated with galactose oxidase. This allowed direct conjugation to tetanus toxoid through terminal 2-N-acyl-2-deoxy-D-glucopyranose or through oxidized galactose, respectively. Comparison of the immune performance of four conjugates in mice revealed, that while each was able to induce significant level of L7-LOS-specific IgG antibody, the conjugates made with the full-length saccharides were able to induce antibodies with increased bactericidal activity against homologous meningococci. Only full-length oligosaccharides were good inhibitors of the binding of L7-LOS to the bactericidal antiserum. Moreover, induction of the significant level of the L7-LOS-specific antibody by full-length lipooligosaccharide conjugated from non-reducing end, provided also the direct evidence that internal core epitopes are fully responsible for the immunorecognition and immunoreactivity.


Asunto(s)
Inmunogenicidad Vacunal , Lipopolisacáridos/inmunología , Meningitis Meningocócica/inmunología , Vacunas Meningococicas/inmunología , Neisseria meningitidis/fisiología , Toxoide Tetánico/inmunología , Vacunas Conjugadas/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Epítopos , Femenino , Galactosa/química , Humanos , Inmunidad Humoral , Inmunización , Lipopolisacáridos/química , Vacunas Meningococicas/química , Ratones , Ratones Endogámicos , Oxidación-Reducción , Toxoide Tetánico/química , Vacunas Conjugadas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA