Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Curr Opin Infect Dis ; 32(5): 490-496, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31356239

RESUMO

PURPOSE OF REVIEW: Nonpathogenic commensal Neisseria are rarely considered in the clinical setting despite evidence that they can cause invasive opportunistic infections. In contrast, they may offer protection against pathogenic Neisseria, and such relationships are being actively explored in experimental studies. RECENT FINDINGS: Recent case reports are presented of invasive infection caused by nonpathogenic Neisseria in patients on novel biologic therapies. On the other hand, Neisseria lactamica, a nonpathogenic commensal, has been shown in human challenge studies to inhibit colonization by Neisseria meningitidis. Experimental mouse models have also explored the inhibitory effects of nonpathogenic Neisseria on Neisseria gonnhoreae infection. Cutting-edge advances in metagenomics and microbiomics are being used to understand the mechanisms underpinning these effects. SUMMARY: Clinicians should have increased awareness of nonpathogenic Neisseria. First, as new immunomodulating therapies become licenced, the interactions that maintain balance between commensals and their human hosts may be altered. Second, these bacteria are showing promise in their capacity to exclude pathogenic Neisseria species from their anatomical niches.


Assuntos
Hospedeiro Imunocomprometido , Neisseria/imunologia , Neisseria/patogenicidade , Infecções por Neisseriaceae/microbiologia , Infecções por Neisseriaceae/prevenção & controle , Animais , Modelos Animais de Doenças , Humanos
2.
Methods Mol Biol ; 1997: 403-412, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31119636

RESUMO

We have developed a natural mouse model to study persistent colonization by commensal Neisseria. The system couples the ordinary lab mouse with Neisseria musculi (Nmus), a commensal in the oral cavity and gut of the wild mouse, Mus musculus. The pairing of Nmus with its natural reservoir circumvents host restriction barriers that have impeded previous studies of Neisseria in vivo behavior. The model allows, for the first time, for the dissection of host and neisserial determinants of asymptomatic colonization. Inoculation procedures are noninvasive and susceptibility to Nmus colonization varies with host genetic background. In colonized mice, bacterial burdens are detectable up to 1-year post inoculation, making it an ideal model for the study of persistence. As Nmus encodes several Neisseria gonorrhoeae (and Neisseria meningitidis) host interaction factors, the system can be used to query the in vivo functions of these commonly held genes and factors. Nmus also encodes many pathogenic Neisseria vaccine targets including a polysaccharide capsule, making the model potentially useful for vaccine development. The ease of genetic manipulation of Nmus enhances the feasibility of such studies.


Assuntos
Modelos Animais de Doenças , Gonorreia/microbiologia , Neisseria/patogenicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Microbioma Gastrointestinal/imunologia , Gonorreia/terapia , Humanos , Camundongos/microbiologia , Mucosa Bucal/imunologia , Mucosa Bucal/microbiologia , Neisseria/genética , Neisseria/imunologia , Simbiose/imunologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
3.
J Mol Biol ; 431(16): 3010-3014, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30986425

RESUMO

The 10 human-restricted Neisseria species all colonize mucosal surfaces, but show a spectrum of pathogenicity. The commensal Neisseria do not normally cause pathology, while the two pathogenic species, Neisseria meningitidis and Neisseria gonorrhoeae, straddle the border between commensalism and pathogenicity. Why the pathogenic Neisseria continue to mediate host damage after thousands of years of co-evolution with their human host, and why the commensal species have not acquired the ability to damage the host, if this capability provides a selective advantage, is not understood. One way the pathogenic species are different from the commensal species is by their ability to induce PMN inflammation, which is dependent on the site of colonization. I discuss how the site of colonization dictates whether copious inflammation occurs with both pathogenic species. I put forth a model that posits that an ancestor of both pathogenic species changed colonization site from the oral cavity to the genital tract of a human or humanoid and had to evolve multiple, new traits - to induce PMN inflammation and avoid adaptive immunity - to allow efficient sexual transmission. This model predicts that PMN inflammation produces the serious sequelae of gonorrhea and increases the probability that N. meningitidis might exit the oral cavity to produce systemic disease. In both cases, the pathology produced by these host-adapted species is an unintended by product of the inflammation but host damage does not provide any selective advantage for these organisms.


Assuntos
Neisseria/imunologia , Neisseria/patogenicidade , Neutrófilos/metabolismo , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Neisseria/classificação , Neisseria/fisiologia , Neisseria gonorrhoeae/imunologia , Neisseria gonorrhoeae/patogenicidade , Neisseria meningitidis/imunologia , Neisseria meningitidis/patogenicidade , Simbiose
4.
Immunobiology ; 223(12): 750-760, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30055864

RESUMO

BACKGROUND: The Neisseria meningitidis bacterium is a Gram-negative diplococcus that can be classified into different serogroups according to the capsular structure. Six of them (A, B, C, W, X, Y) are responsible for causing Invasive Meningococcal Disease (IMD). The strategies for the development of a vaccine for serogroup B have been directed to the use of outer membrane vesicles (OMVs). The aim of this study was to evaluate the immunogenicity of antigenic determinants from OMVs of N. meningitidis B complexed with two different adjuvants: DODAB-BF and aluminum hydroxide (alum), comparing the evaluation of intranasal and subcutaneous route of immunization. METHODS: We used prime-boost immunization for the first time in outbred neonatal mice evaluating the cellular and humoral immune response. RESULTS: Immunoblot, ELISA DOT-ELISA and ELISpot were used universal methods of antibody detection, in order to detect the humoral and cellular immune response in male and female mice. Immunoblot analyzes the specificity of antibodies with the homologous N. meningitidis strain. ELISA served to quantify and compare the titers of antibodies in the serum of mice immunized with DODAB-BF + OMVs and alum + OMVs for IgG, IgG1, and IgG2a. Intranasal immunization produced a mixed response in the T helper cells Th1 and Th2, while subcutaneous immunization exhibited a Th1 profile. The DOT-ELISA identified cross-reactivity with DODAB-BF to different serogroups of N. meningitidis (B, C, W, and Y) that was not observed with alum. ELISpot analyzed IFN-γ- and IL-4 and the results showed the response directly to Th1 and Th2 profile. CONCLUSION: Our findings indicate that DODAB-BF can be an alternative adjuvant for mucosal cell activation with OMVs of N. meningitidis B and that DODAB-BF was similar to aluminum hydroxide as an adjuvant for subcutaneous immunization.


Assuntos
Adjuvantes Imunológicos , Imunização , Neisseria meningitidis/imunologia , Neisseria/imunologia , Compostos de Amônio Quaternário , Administração Intranasal , Animais , Anticorpos Antibacterianos/imunologia , Afinidade de Anticorpos/imunologia , Antígenos de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Imunidade Humoral , Imunização/métodos , Isotipos de Imunoglobulinas/imunologia , Injeções Subcutâneas , Masculino , Camundongos
5.
Sci Rep ; 8(1): 6512, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29695781

RESUMO

Neisseria meningitidis is a Gram-negative bacterial pathogen that causes life threatening meningitis and septicemia. Neisseria Heparin Binding Antigen (NHBA) is an outer membrane protein that binds heparin and heparan sulfate and DNA. This protein is one of the four antigens in the meningococcal serogroup B vaccine Bexsero. In the current study, we sought to define the full glycan-binding repertoire of NHBA to better understand its role in meningococcal pathogenesis and vaccine efficacy. Glycan array analysis revealed binding to 28 structures by recombinant NHBA. Surface plasmon resonance was used to confirm the binding phenotype and to determine the affinity of the interactions. These studies revealed that the highest affinity binding of NHBA was with chondroitin sulfate (KD = 5.2 nM). This affinity is 10-fold higher than observed for heparin. Analysis of binding with well-defined disaccharides of the different chondroitin sulfate types demonstrated that the most preferred ligand has a sulfate at the 2 position of the GlcA/IdoA and 6 position of the GalNAc, which is an equivalent structure to chondroitin sulfate D. Chondroitin sulfate is widely expressed in human tissues, while chondroitin sulfate D is predominantly expressed in the brain and may constitute a new receptor structure for meningococci.


Assuntos
Antígenos de Bactérias/imunologia , Sulfatos de Condroitina/imunologia , Vacinas Meningocócicas/imunologia , Neisseria meningitidis/imunologia , Neisseria/imunologia , Anticorpos Antibacterianos/imunologia , Proteínas de Transporte/imunologia , Humanos , Sorogrupo
6.
Immunohorizons ; 2(9): 305-313, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31022695

RESUMO

Neisseria musculi, isolated from the oral cavity of wild-caught mice, does not colonize most inbred mouse strains. N. musculi does weakly (50%) colonize C57BL/6J (B6) mice but readily colonizes CAST/EiJ (CAST) mice. In this study, we examined whether differences in the CAST and B6 host response could elucidate mechanisms governing N. musculi colonization. In vivo stimulation of B6 or CAST splenocytes with wild type (WT) Neisseria or Escherichia coli LPS showed that CAST mice had a blunted inflammatory response, producing significantly lower levels of IL-6 than B6 mice. The use of specific genetic knockouts highlighted a need for an intact innate immune system to prevent colonization. B6-RAG-1-/- mice were colonized at a similar rate as WT B6 mice, whereas B6-MyD88-/- and TLR4-/- mice were readily colonized like CAST (100%) mice. Sequence analysis revealed a unique point mutation in TLR4 in CAST mice. However, crosses to TLR4-/- mice and analysis of recombinant inbred Collaborative Cross mice showed that TLR4 from CAST mice was not sufficient to allow Neisseria colonization. In vitro stimulation of B6 bone marrow-derived macrophages or splenocytes with WT Neisseria yielded low levels of IL-6 compared with LPS stimulation. Surprisingly, UV-inactivated Neisseria induced high levels of IL-6, suggesting suppression of IL-6 production is an active bacterial process. Consistent with a critical role for IL-6 in preventing colonization, mice deficient for the IL-6 receptor were efficiently colonized, indicating host IL-6 production plays a critical role in determining host colonization susceptibility.


Assuntos
Infecções por Bactérias Gram-Negativas/imunologia , Neisseria/imunologia , Imunidade Adaptativa , Animais , Escherichia coli , Interações entre Hospedeiro e Microrganismos , Imunidade Inata , Interleucina-6/deficiência , Interleucina-6/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Simbiose/imunologia , Receptor 4 Toll-Like
7.
Biotechnol Lett ; 39(3): 407-413, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27888365

RESUMO

OBJECTIVES: A Neissaria bacterial pilus sugar, bacillosamine, was synthesized and, for the first time, used as a probe to screen a single-chain variable fragment (scFv). RESULTS: Four Neisseria, Neisseria gonorrhoeae, Neisseria meningitidis, Neisseria sicca and Neisseria subflava, and two negative controls, Streptococcus pneumoniae and Escherichia coli, were tested through ELISA, immunostaining and gold nanoparticle immunological assay. All results indicated that the selected scFv is feasible for the specific detection of Neisseria species via the recognition of bacillosamine. CONCLUSIONS: The recombinant scFv could detect Neisseria strains at 106 CFU/ml.


Assuntos
Anticorpos Antibacterianos/imunologia , Neisseria/imunologia , Engenharia de Proteínas/métodos , Especificidade de Anticorpos/imunologia , Eletroforese em Gel de Poliacrilamida , Ouro/química , Nanopartículas/química , Anticorpos de Cadeia Única/metabolismo , Especificidade da Espécie
8.
Immunobiology ; 221(10): 1110-23, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27297292

RESUMO

Novel therapies are urgently needed to combat the global threat of multidrug-resistant pathogens. Complement forms an important arm of innate defenses against infections. In physiological conditions, complement activation is tightly controlled by soluble and membrane-associated complement inhibitors, but must be selectively activated on invading pathogens to facilitate microbial clearance. Many pathogens, including Neisseria gonorrhoeae and N. meningitidis, express glycans, including N-acetylneuraminic acid (Neu5Ac), that mimic host structures to evade host immunity. Neu5Ac is a negatively charged 9-cabon sugar that inhibits complement, in part by enhancing binding of the complement inhibitor factor H (FH) through C-terminal domains (19 and 20) on FH. Other microbes also bind FH, in most instances through FH domains 6 and 7 or 18-20. Here we describe two strategies to target complement activation on Neisseriae. First, microbial binding domains of FH were fused to IgG Fc to create FH18-20/Fc (binds gonococci) and FH6,7/Fc (binds meningococci). A point mutation in FH domain 19 eliminated hemolysis caused by unmodified FH18-20, but retained binding to gonococci. FH18-20/Fc and FH6,7/Fc mediated complement-dependent killing in vitro and showed efficacy in animal models of gonorrhea and meningococcal bacteremia, respectively. The second strategy utilized CMP-nonulosonate (CMP-NulO) analogs of sialic acid that were incorporated into LOS and prevented complement inhibition by physiologic CMP-Neu5Ac and resulted in attenuated gonococcal infection in mice. While studies to establish the safety of these agents are needed, enhancing complement activation on microbes may represent a promising strategy to treat antimicrobial resistant organisms.


Assuntos
Antibacterianos/farmacologia , Bactérias/imunologia , Infecções Bacterianas/imunologia , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Desenho de Fármacos , Fatores Imunológicos/farmacologia , Animais , Antibacterianos/uso terapêutico , Antígenos de Bactérias/imunologia , Bactérias/química , Bactérias/metabolismo , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/microbiologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Fatores Imunológicos/uso terapêutico , Mimetismo Molecular/imunologia , Neisseria/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Ácidos Siálicos/imunologia
9.
FASEB J ; 30(1): 93-101, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26304221

RESUMO

Neisseria adhesin A (NadA) is one of the antigens of Bexsero, the recently licensed multicomponent vaccine against serogroup B Neisseria meningitidis (MenB). NadA belongs to the class of oligomeric coiled-coil adhesins and is able to mediate adhesion and invasion of human epithelial cells. As a vaccine antigen, NadA has been shown to induce high levels of bactericidal antibodies; however, the domains important for protective response are still unknown. In order to further investigate its immunogenic properties, we have characterized the murine IgG1 mAb (6E3) that was able to recognize the 2 main antigenic variants of NadA on the surface of MenB strains. The epitope targeted by mAb 6E3 was mapped by hydrogen-deuterium exchange mass spectrometry and shown to be located on the coiled-coil stalk region of NadA (aa 206-249). Although no serum bactericidal activity was observed for murine IgG1 mAb 6E3, functional activity was restored when using chimeric antibodies in which the variable regions of the murine mAb 6E3 were fused to human IgG3 constant regions, thus confirming the protective nature of the mAb 6E3 epitope. The use of chimeric antibody molecules will enable future investigations of complement-mediated antibody functionality independently of the Fc-mediated differences in complement activation.


Assuntos
Adesinas Bacterianas/imunologia , Anticorpos Antibacterianos/imunologia , Epitopos/imunologia , Infecções Meningocócicas/imunologia , Vacinas Meningocócicas/imunologia , Neisseria/imunologia , Animais , Medição da Troca de Deutério/métodos , Mapeamento de Epitopos/métodos , Humanos , Camundongos
10.
Proc Natl Acad Sci U S A ; 112(51): E7128-37, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26621750

RESUMO

This paper reviews the developments that have occurred in the field of human genetics of infectious diseases from the second half of the 20th century onward. In particular, it stresses and explains the importance of the recently described monogenic inborn errors of immunity underlying resistance or susceptibility to specific infections. The monogenic component of the genetic theory provides a plausible explanation for the occurrence of severe infectious diseases during primary infection. Over the last 20 y, increasing numbers of life-threatening infectious diseases striking otherwise healthy children, adolescents, and even young adults have been attributed to single-gene inborn errors of immunity. These studies were inspired by seminal but neglected findings in plant and animal infections. Infectious diseases typically manifest as sporadic traits because human genotypes often display incomplete penetrance (most genetically predisposed individuals remain healthy) and variable expressivity (different infections can be allelic at the same locus). Infectious diseases of childhood, once thought to be archetypal environmental diseases, actually may be among the most genetically determined conditions of mankind. This nascent and testable notion has interesting medical and biological implications.


Assuntos
Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Infecções/genética , Infecções/imunologia , Adolescente , Candidíase Mucocutânea Crônica/genética , Candidíase Mucocutânea Crônica/imunologia , Criança , Proteínas do Sistema Complemento/genética , Encefalite por Herpes Simples/genética , Encefalite por Herpes Simples/imunologia , Epidermodisplasia Verruciforme/genética , Epidermodisplasia Verruciforme/imunologia , Predisposição Genética para Doença , Humanos , Influenza Humana/genética , Influenza Humana/imunologia , Interferon gama/genética , Interferon gama/imunologia , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/imunologia , Malária/genética , Malária/imunologia , Modelos Genéticos , Modelos Imunológicos , Infecções por Mycobacterium/genética , Infecções por Mycobacterium/imunologia , Neisseria/imunologia , Neisseria/patogenicidade , Infecções Pneumocócicas/genética , Infecções Pneumocócicas/imunologia , Tinha/genética , Tinha/imunologia , Adulto Jovem
11.
PLoS One ; 9(1): e86704, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24466206

RESUMO

Antigenic variation occurs in a broad range of species. This process resembles gene conversion in that variant DNA is unidirectionally transferred from partial gene copies (or silent loci) into an expression locus. Previous studies of antigenic variation have involved the amplification and sequencing of individual genes from hundreds of colonies. Using the pilE gene from Neisseria gonorrhoeae we have demonstrated that it is possible to use PCR amplification, followed by high-throughput DNA sequencing and a novel assembly process, to detect individual antigenic variation events. The ability to detect these events was much greater than has previously been possible. In N. gonorrhoeae most silent loci contain multiple partial gene copies. Here we show that there is a bias towards using the copy at the 3' end of the silent loci (copy 1) as the donor sequence. The pilE gene of N. gonorrhoeae and some strains of Neisseria meningitidis encode class I pilin, but strains of N. meningitidis from clonal complexes 8 and 11 encode a class II pilin. We have confirmed that the class II pili of meningococcal strain FAM18 (clonal complex 11) are non-variable, and this is also true for the class II pili of strain NMB from clonal complex 8. In addition when a gene encoding class I pilin was moved into the meningococcal strain NMB background there was no evidence of antigenic variation. Finally we investigated several members of the opa gene family of N. gonorrhoeae, where it has been suggested that limited variation occurs. Variation was detected in the opaK gene that is located close to pilE, but not at the opaJ gene located elsewhere on the genome. The approach described here promises to dramatically improve studies of the extent and nature of antigenic variation systems in a variety of species.


Assuntos
Variação Antigênica , Antígenos de Bactérias/genética , Neisseria/genética , Antígenos de Bactérias/imunologia , Biologia Computacional , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Neisseria/classificação , Neisseria/imunologia
12.
mBio ; 4(3): e00163-13, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23760461

RESUMO

Neisseria meningitidis, one of the major causes of bacterial meningitis and sepsis, is a member of the genus Neisseria, which includes species that colonize the mucosae of many animals. Three meningococcal proteins, factor H-binding protein (fHbp), neisserial heparin-binding antigen (NHBA), and N. meningitidis adhesin A (NadA), have been described as antigens protective against N. meningitidis of serogroup B, and they have been employed as vaccine components in preclinical and clinical studies. In the vaccine formulation, fHbp and NHBA were fused to the GNA2091 and GNA1030 proteins, respectively, to enhance protein stability and immunogenicity. To determine the possible impact of vaccination on commensal neisseriae, we determined the presence, distribution, and conservation of these antigens in the available genome sequences of the genus Neisseria, finding that fHbp, NHBA, and NadA were conserved only in species colonizing humans, while GNA1030 and GNA2091 were conserved in many human and nonhuman neisseriae. Sequence analysis showed that homologous recombination contributed to shape the evolution and distribution of both NHBA and fHbp, three major variants of which have been defined. fHbp variant 3 was probably the ancestral form of meningococcal fHbp, while fHbp variant 1 from N. cinerea was introduced into N. meningitidis by a recombination event. fHbp variant 2 was the result of a recombination event inserting a stretch of 483 bp from variant 1 into the variant 3 background. These data indicate that a high rate of exchange of genetic material between neisseriae that colonize the human upper respiratory tract exists. IMPORTANCE The upper respiratory tract of healthy individuals is a complex ecosystem colonized by many bacterial species. Among these, there are representatives of the genus Neisseria, including Neisseria meningitidis, a major cause of bacterial meningitis and sepsis. Given the close relationship between commensal and pathogenic species, a protein-based vaccine against N. meningitidis has the potential to impact the other commensal species of Neisseria. For this reason, we have studied the distribution and evolutionary history of the antigen components of a recombinant vaccine, 4CMenB, that recently received approval in Europe under the commercial name of Bexsero®. We found that fHbp, NHBA, and NadA can be found in some of the human commensal species and that the evolution of these antigens has been essentially shaped by the high rate of genetic exchange that occurs between strains of neisseriae that cocolonize the same environment.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Sequência Conservada , Neisseria/genética , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Análise por Conglomerados , Evolução Molecular , Humanos , Neisseria/imunologia , Infecções por Neisseriaceae/microbiologia , Recombinação Genética , Homologia de Sequência de Aminoácidos
13.
Immunobiology ; 218(10): 1304-10, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23743184

RESUMO

Inherited deficiency states of the terminal complement component C5 are rare and often associated with increased risk of recurrent Neisseria infections. More than 50 cases with primary C5 deficiency have been reported. In spite of this, the molecular basis has only been documented in a few cases. In the present study we investigated two unrelated Caucasian probands with C5 deficiency originating from Norway and Denmark, respectively, and found three previously undescribed mutations. With these data, thirteen mutations associated with C5 deficiency have been described. By genetic screening of the family of the Norwegian patient, previously diagnosed as homozygous C5 deficient and suffering four Neisseria infections, an additional case of C5 deficiency was discovered, who had experienced one episode of Neisseria infections. Detailed review of the clinical history of the patients and their healthy relatives did not reveal any differences between C5 deficient and sufficient individuals with regard to clinical presentation, apart from the susceptibility to Neisseria infections. Of note, one of the patients described here, and several C5 deficient patients from the literature had Neisseria meningitidis serotype B infections, which is not covered by the current vaccines. These data support the clinical guidelines for patients treated with C5 inhibitors, who are functional C5 deficient by the treatment.


Assuntos
Complemento C5/genética , Infecções por Bactérias Gram-Negativas/imunologia , Síndromes de Imunodeficiência/imunologia , Neisseria/imunologia , Adolescente , Adulto , Criança , Análise Mutacional de DNA , Dinamarca , Feminino , Infecções por Bactérias Gram-Negativas/genética , Humanos , Síndromes de Imunodeficiência/genética , Pessoa de Meia-Idade , Mutação/genética , Noruega , Linhagem , Guias de Prática Clínica como Assunto , Risco , População Branca
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(4): 414-7, 2013 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-23643173

RESUMO

OBJECTIVE: To construct recombinant protein of Neisserial surface protein A (NspA) and determine the anti-NspA antibody levels using it as antigen in patients at the recovery stage of Neisseria meningitides, so as to explore the possibility of using NspA protein as the antigen for designing a novel vaccine for epidemic Neisseria meningitides. METHODS: NspA gene was cloned from the isolated pathogen of patients with Neisseria meningitides to construct the prokaryotic expression vector. NspA protein was expressed in the form of soluble protein in E.coli and purified by GSTrap FF affinity chromatography. The antibody titers of recovery-stage patients with Neisseria meningitides were determined using the purified recombinant NspA. RESULTS: Functional NspA was successfully expressed with Mr; being 44 000. The purified NspA had a good biological function. Antibodies against NspA could be detected in the sera of patients at the recovery stage of Neisseria meningitides using NspA protein. Positive rate reached 90% when titer was 1:40. CONCLUSION: Recombinant NspA protein was expressed successfully and it could be used to detect the anti-NspA antibodies in the sera of patients with Neisseria meningitides at recovery stage.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/genética , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/biossíntese , Escherichia coli/genética , Escherichia coli/imunologia , Humanos , Neisseria/genética , Neisseria/imunologia , Neisseria meningitidis/genética , Neisseria meningitidis/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
15.
Infect Immun ; 80(11): 4014-26, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22949553

RESUMO

The interaction of the immune system with Neisseria commensals remains poorly understood. We have previously shown that phosphoethanolamine on the lipid A portion of lipooligosaccharide (LOS) plays an important role in Toll-like receptor 4 (TLR4) signaling. For pathogenic Neisseria, phosphoethanolamine is added to lipid A by the phosphoethanolamine transferase specific for lipid A, which is encoded by lptA. Here, we report that Southern hybridizations and bioinformatics analyses of genomic sequences from all eight commensal Neisseria species confirmed that lptA was absent in 15 of 17 strains examined but was present in N. lactamica. Mass spectrometry of lipid A and intact LOS revealed the lack of both pyrophosphorylation and phosphoethanolaminylation in lipid A of commensal species lacking lptA. Inflammatory signaling in human THP-1 monocytic cells was much greater with pathogenic than with commensal Neisseria strains that lacked lptA, and greater sensitivity to polymyxin B was consistent with the absence of phosphoethanolamine. Unlike the other commensals, whole bacteria of two N. lactamica commensal strains had low inflammatory potential, whereas their lipid A had high-level pyrophosphorylation and phosphoethanolaminylation and induced high-level inflammatory signaling, supporting previous studies indicating that this species uses mechanisms other than altering lipid A to support commensalism. A meningococcal lptA deletion mutant had reduced inflammatory potential, further illustrating the importance of lipid A pyrophosphorylation and phosphoethanolaminylation in the bioactivity of LOS. Overall, our results indicate that lack of pyrophosphorylation and phosphoethanolaminylation of lipid A contributes to the immune privilege of most commensal Neisseria strains by reducing the inflammatory potential of LOS.


Assuntos
Inflamação/imunologia , Lipídeo A/metabolismo , Neisseria/imunologia , Southern Blotting , Células Cultivadas , Biologia Computacional , Humanos , Lipídeo A/imunologia , Neisseria/patogenicidade , Fosforilação , Transdução de Sinais , Espectrometria de Massas em Tandem
16.
PLoS Pathog ; 8(4): e1002597, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496641

RESUMO

LPS-activated neutrophils secrete IL-1ß by activation of TLR-4. Based on studies in macrophages, it is likely that ROS and lysosomal destabilization regulated by Syk activation may also be involved. Since neutrophils have abundant expression of the ITIM-containing co-receptor CEACAM1 and Gram-negative bacteria such as Neisseria utilize CEACAM1 as a receptor that inhibits inflammation, we hypothesized that the overall production of IL-1ß in LPS treated neutrophils may be negatively regulated by CEACAM1. We found that LPS treated neutrophils induced phosphorylation of Syk resulting in the formation of a complex including TLR4, p-Syk, and p-CEACAM1, which in turn, recruited the inhibitory phosphatase SHP-1. LPS treatment leads to ROS production, lysosomal damage, caspase-1 activation and IL-1ß secretion in neutrophils. The absence of this regulation in Ceacam1⁻/⁻ neutrophils led to hyper production of IL-1ß in response to LPS. The hyper production of IL-1ß was abrogated by in vivo reconstitution of wild type but not ITIM-mutated CEACAM1 bone marrow stem cells. Blocking Syk activation by kinase inhibitors or RNAi reduced Syk phosphorylation, lysosomal destabilization, ROS production, and caspase-1 activation in Ceacam1⁻/⁻ neutrophils. We conclude that LPS treatment of neutrophils triggers formation of a complex of TLR4 with pSyk and pCEACAM1, which upon recruitment of SHP-1 to the ITIMs of pCEACAM1, inhibits IL-1ß production by the inflammasome. Thus, CEACAM1 fine-tunes IL-1ß production in LPS treated neutrophils, explaining why the additional utilization of CEACAM1 as a pathogen receptor would further inhibit inflammation.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Complexos Multiproteicos/metabolismo , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Caspase 1/genética , Caspase 1/imunologia , Caspase 1/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lisossomos/genética , Lisossomos/imunologia , Lisossomos/metabolismo , Lisossomos/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Neisseria/imunologia , Neisseria/metabolismo , Infecções por Neisseriaceae/genética , Infecções por Neisseriaceae/imunologia , Infecções por Neisseriaceae/metabolismo , Infecções por Neisseriaceae/patologia , Ativação de Neutrófilo/genética , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Neutrófilos/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo
18.
J Bacteriol ; 192(11): 2816-29, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20363948

RESUMO

Bacterial capsular polysaccharides and lipopolysaccharides are well-established ligands of innate and adaptive immune effectors and often exhibit structural and antigenic variability. Although many surface-localized glycoproteins have been identified in bacterial pathogens and symbionts, it not clear if and how selection impacts associated glycoform structure. Here, a systematic approach was devised to correlate gene repertoire with protein-associated glycoform structure in Neisseria species important to human health and disease. By manipulating the protein glycosylation (pgl) gene content and assessing the glycan structure by mass spectrometry and reactivity with monoclonal antibodies, it was established that protein-associated glycans are antigenically variable and that at least nine distinct glycoforms can be expressed in vitro. These studies also revealed that in addition to Neisseria gonorrhoeae strain N400, one other gonococcal strain and isolates of Neisseria meningitidis and Neisseria lactamica exhibit broad-spectrum O-linked protein glycosylation. Although a strong correlation between pgl gene content, glycoform expression, and serological profile was observed, there were significant exceptions, particularly with regard to levels of microheterogeneity. This work provides a technological platform for molecular serotyping of neisserial protein glycans and for elucidating pgl gene evolution.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Glicoproteínas/química , Glicoproteínas/imunologia , Neisseria/metabolismo , Polissacarídeos/química , Polissacarídeos/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Eletroforese em Gel de Poliacrilamida , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilação , Humanos , Immunoblotting , Microscopia Eletrônica de Transmissão , Neisseria/genética , Neisseria/imunologia , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/imunologia , Neisseria gonorrhoeae/metabolismo , Neisseria meningitidis , Polissacarídeos/genética , Polissacarídeos/metabolismo , Coelhos , Espectrometria de Massas por Ionização por Electrospray
19.
Cell Microbiol ; 12(3): 372-85, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19888989

RESUMO

Gram-negative bacterial peptidoglycan is specifically recognized by the host intracellular sensor NOD1, resulting in the generation of innate immune responses. Although epithelial cells are normally refractory to external stimulation with peptidoglycan, these cells have been shown to respond in a NOD1-dependent manner to Gram-negative pathogens that can either invade or secrete factors into host cells. In the present work, we report that Gram-negative bacteria can deliver peptidoglycan to cytosolic NOD1 in host cells via a novel mechanism involving outer membrane vesicles (OMVs). We purified OMVs from the Gram-negative mucosal pathogens: Helicobacter pylori, Pseudomonas aeruginosa and Neisseria gonorrhoea and demonstrated that these peptidoglycan containing OMVs upregulated NF-kappaB and NOD1-dependent responses in vitro. These OMVs entered epithelial cells through lipid rafts thereby inducing NOD1-dependent responses in vitro. Moreover, OMVs delivered intragastrically to mice-induced innate and adaptive immune responses via a NOD1-dependent but TLR-independent mechanism. Collectively, our findings identify OMVs as a generalized mechanism whereby Gram-negative bacteria deliver peptidoglycan to cytosolic NOD1. We propose that OMVs released by bacteria in vivo may promote inflammation and pathology in infected hosts.


Assuntos
Células Epiteliais/imunologia , Bactérias Gram-Negativas/imunologia , NF-kappa B/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Peptidoglicano/metabolismo , Vesículas Secretórias/metabolismo , Animais , Feminino , Células HeLa , Helicobacter pylori/imunologia , Humanos , Masculino , Camundongos , Neisseria/imunologia , Peptidoglicano/imunologia , Pseudomonas aeruginosa/imunologia , Vesículas Secretórias/imunologia
20.
Blood ; 113(26): 6522-7, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19372253

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare clonal blood disorder that manifests with hemolytic anemia, bone marrow failure, and thrombosis. Many of the clinical manifestations of the disease result from complement-mediated intravascular hemolysis. Allogeneic bone marrow transplantation is the only curative therapy for PNH. Eculizumab, a monoclonal antibody that blocks terminal complement activation, is highly effective in reducing hemolysis, improving quality of life, and reducing the risk for thrombosis in PNH patients. Insights into the relevance of detecting PNH cells in PNH and other bone marrow failure disorders are highlighted, and indications for treating PNH patients with bone marrow transplantation and eculizumab are explored.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hemoglobinúria Paroxística/terapia , Anticorpos Monoclonais Humanizados , Anticoagulantes/administração & dosagem , Anticoagulantes/uso terapêutico , Vacinas Bacterianas/administração & dosagem , Transfusão de Sangue , Medula Óssea/patologia , Transplante de Medula Óssea , Células Clonais/patologia , Terapia Combinada , Ativação do Complemento/efeitos dos fármacos , Complemento C5/antagonistas & inibidores , Gerenciamento Clínico , Feminino , Ácido Fólico/administração & dosagem , Ácido Fólico/uso terapêutico , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/genética , Humanos , Recém-Nascido , Ferro/administração & dosagem , Ferro/uso terapêutico , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Neisseria/imunologia , Infecções por Neisseriaceae/prevenção & controle , Gravidez , Complicações Hematológicas na Gravidez/terapia , Trombose/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA