Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.142
Filtrar
Más filtros

Publication year range
1.
PLoS Pathog ; 20(7): e1012369, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38976720

RESUMEN

Neisseria gonorrhoeae (the gonococcus, Gc) causes the sexually transmitted infection gonorrhea. Gc is a prominent threat to human health by causing severe lifelong sequelae, including infertility and chronic pelvic pain, which is amplified by the emergence of "superbug" strains resistant to all current antibiotics. Gc is highly adapted to colonize human mucosal surfaces, where it survives despite initiating a robust inflammatory response and influx of polymorphonuclear leukocytes (PMNs, neutrophils) that typically clear bacteria. Here, dual-species RNA-sequencing was used to define Gc and PMN transcriptional profiles alone and after infection. Core host and bacterial responses were assessed for two strains of Gc and three human donors' PMNs. Comparative analysis of Gc transcripts revealed overlap between Gc responses to PMNs, iron, and hydrogen peroxide; 98 transcripts were differentially expressed across both Gc strains in response to PMN co-culture, including iron-responsive and oxidative stress response genes. We experimentally determined that the iron-dependent TbpB is suppressed by PMN co-culture, and iron-limited Gc have a survival advantage when cultured with PMNs. Analysis of PMN transcripts modulated by Gc infection revealed differential expression of genes driving cell adhesion, migration, inflammatory responses, and inflammation resolution pathways. Production of pro-inflammatory cytokines, including IL1B and IL8, the adhesion factor ICAM1, and prostaglandin PGE2 were induced in PMNs in response to Gc. Together, this study represents a comprehensive and experimentally validated dual-species transcriptomic analysis of two isolates of Gc and primary human PMNs that gives insight into how this bacterium survives innate immune onslaught to cause disease.


Asunto(s)
Gonorrea , Neisseria gonorrhoeae , Neutrófilos , Transcriptoma , Humanos , Neisseria gonorrhoeae/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Gonorrea/inmunología , Gonorrea/microbiología
2.
Infect Immun ; 92(5): e0000424, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38563734

RESUMEN

Neisseria gonorrhoeae, a human restricted pathogen, releases inflammatory peptidoglycan (PG) fragments that contribute to the pathophysiology of pelvic inflammatory disease. The genus Neisseria is also home to multiple species of human- or animal-associated Neisseria that form part of the normal microbiota. Here we characterized PG release from the human-associated nonpathogenic species Neisseria lactamica and Neisseria mucosa and animal-associated Neisseria from macaques and wild mice. An N. mucosa strain and an N. lactamica strain were found to release limited amounts of the proinflammatory monomeric PG fragments. However, a single amino acid difference in the PG fragment permease AmpG resulted in increased PG fragment release in a second N. lactamica strain examined. Neisseria isolated from macaques also showed substantial release of PG monomers. The mouse colonizer Neisseria musculi exhibited PG fragment release similar to that seen in N. gonorrhoeae with PG monomers being the predominant fragments released. All the human-associated species were able to stimulate NOD1 and NOD2 responses. N. musculi was a poor inducer of mouse NOD1, but ldcA mutation increased this response. The ability to genetically manipulate N. musculi and examine effects of different PG fragments or differing amounts of PG fragments during mouse colonization will lead to a better understanding of the roles of PG in Neisseria infections. Overall, we found that only some nonpathogenic Neisseria have diminished release of proinflammatory PG fragments, and there are differences even within a species as to types and amounts of PG fragments released.


Asunto(s)
Neisseria , Proteína Adaptadora de Señalización NOD1 , Proteína Adaptadora de Señalización NOD2 , Peptidoglicano , Animales , Humanos , Ratones , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas de Transporte de Membrana , Neisseria/genética , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Peptidoglicano/metabolismo
3.
Sex Transm Dis ; 51(6): 381-387, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38403294

RESUMEN

BACKGROUND: Gonorrhea's rapid development of antimicrobial resistance underscores the importance of new prevention modalities. Recent evidence suggests that a serogroup B meningococcal vaccine may be partially effective against gonococcal infection. However, the viability of vaccination and the role it should play in gonorrhea prevention are an open question. METHODS: We modeled the transmission of gonorrhea over a 10-year period in a heterosexual population to find optimal patterns of year-over-year investment of a fixed budget in vaccination and screening programs. Each year, resources could be allocated to vaccinating people or enrolling them in a quarterly screening program. Stratifying by mode (vaccination vs. screening), sex (male vs. female), and enrollment venue (background screening vs. symptomatic visit), we consider 8 different ways of controlling gonorrhea. We then found the year-over-year pattern of investment among those 8 controls that most reduced the incidence of gonorrhea under different assumptions. A compartmental transmission model was parameterized from existing literature in the US context. RESULTS: Vaccinating men with recent symptomatic infection, which selected for higher sexual activity, was optimal for population-level gonorrhea control. Given a prevention budget of $3 per capita, 9.5% of infections could be averted ($299 per infection averted), decreasing gonorrhea sequelae and associated antimicrobial use by similar percentages. These results were consistent across sensitivity analyses that increased the budget, prioritized incidence or prevalence reductions in women, or lowered screening costs. Under a scenario where only screening was implemented, just 5.5% of infections were averted. CONCLUSIONS: A currently available vaccine, although only modestly effective, may be superior to frequent testing for population-level gonorrhea control.


Asunto(s)
Gonorrea , Tamizaje Masivo , Vacunación , Humanos , Gonorrea/prevención & control , Gonorrea/epidemiología , Gonorrea/economía , Masculino , Femenino , Tamizaje Masivo/economía , Vacunación/economía , Neisseria gonorrhoeae/inmunología , Análisis Costo-Beneficio , Estados Unidos/epidemiología , Incidencia , Adulto , Vacunas Meningococicas/administración & dosificación , Vacunas Meningococicas/economía , Heterosexualidad
4.
Infect Immun ; 91(12): e0024523, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37916806

RESUMEN

Virus-like particles (VLPs) are promising nanotools for the development of subunit vaccines due to high immunogenicity and safety. Herein, we explored the versatile and effective Tag/Catcher-AP205 capsid VLP (cVLP) vaccine platform to address the urgent need for the development of an effective and safe vaccine against gonorrhea. The benefits of this clinically validated cVLP platform include its ability to facilitate unidirectional, high-density display of complex/full-length antigens through an effective split-protein Tag/Catcher conjugation system. To assess this modular approach for making cVLP vaccines, we used a conserved surface lipoprotein, SliC, that contributes to the Neisseria gonorrhoeae defense against human lysozyme, as a model antigen. This protein was genetically fused at the N- or C-terminus to the small peptide Tag enabling their conjugation to AP205 cVLP, displaying the complementary Catcher. We determined that SliC with the N-terminal SpyTag, N-SliC, retained lysozyme-blocking activity and could be displayed at high density on cVLPs without causing aggregation. In mice, the N-SliC-VLP vaccines, adjuvanted with AddaVax or CpG, induced significantly higher antibody titers compared to controls. In contrast, similar vaccine formulations containing monomeric SliC were non-immunogenic. Accordingly, sera from N-SliC-VLP-immunized mice also had significantly higher human complement-dependent serum bactericidal activity. Furthermore, the N-SliC-VLP vaccines administered subcutaneously with an intranasal boost elicited systemic and vaginal IgG and IgA, whereas subcutaneous delivery alone failed to induce vaginal IgA. The N-SliC-VLP with CpG (10 µg/dose) induced the most significant increase in total serum IgG and IgG3 titers, vaginal IgG and IgA, and bactericidal antibodies.


Asunto(s)
Neisseria gonorrhoeae , Vacunas de Partículas Similares a Virus , Animales , Femenino , Humanos , Ratones , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Cápside , Inmunoglobulina A , Inmunoglobulina G , Ratones Endogámicos BALB C , Muramidasa , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/inmunología , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología
5.
PLoS Pathog ; 16(12): e1008602, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33290434

RESUMEN

There is a pressing need for a gonorrhea vaccine due to the high disease burden associated with gonococcal infections globally and the rapid evolution of antibiotic resistance in Neisseria gonorrhoeae (Ng). Current gonorrhea vaccine research is in the stages of antigen discovery and the identification of protective immune responses, and no vaccine has been tested in clinical trials in over 30 years. Recently, however, it was reported in a retrospective case-control study that vaccination of humans with a serogroup B Neisseria meningitidis (Nm) outer membrane vesicle (OMV) vaccine (MeNZB) was associated with reduced rates of gonorrhea. Here we directly tested the hypothesis that Nm OMVs induce cross-protection against gonorrhea in a well-characterized female mouse model of Ng genital tract infection. We found that immunization with the licensed Nm OMV-based vaccine 4CMenB (Bexsero) significantly accelerated clearance and reduced the Ng bacterial burden compared to administration of alum or PBS. Serum IgG and vaginal IgA and IgG that cross-reacted with Ng OMVs were induced by 4CMenB vaccination by either the subcutaneous or intraperitoneal routes. Antibodies from vaccinated mice recognized several Ng surface proteins, including PilQ, BamA, MtrE, NHBA (known to be recognized by humans), PorB, and Opa. Immune sera from both mice and humans recognized Ng PilQ and several proteins of similar apparent molecular weight, but MtrE was only recognized by mouse serum. Pooled sera from 4CMenB-immunized mice showed a 4-fold increase in serum bactericidal50 titers against the challenge strain; in contrast, no significant difference in bactericidal activity was detected when sera from 4CMenB-immunized and unimmunized subjects were compared. Our findings directly support epidemiological evidence that Nm OMVs confer cross-species protection against gonorrhea, and implicate several Ng surface antigens as potentially protective targets. Additionally, this study further defines the usefulness of murine infection model as a relevant experimental system for gonorrhea vaccine development.


Asunto(s)
Protección Cruzada/inmunología , Vacunas Meningococicas/farmacología , Neisseria gonorrhoeae/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Estudios de Casos y Controles , Reacciones Cruzadas/inmunología , Femenino , Gonorrea/inmunología , Humanos , Sueros Inmunes/inmunología , Inmunización/métodos , Masculino , Infecciones Meningocócicas/microbiología , Vacunas Meningococicas/inmunología , Vacunas Meningococicas/metabolismo , Ratones , Ratones Endogámicos BALB C , Neisseria meningitidis/inmunología , Neisseria meningitidis Serogrupo B/inmunología , Estudios Retrospectivos , Serogrupo , Vacunación/métodos
6.
Annu Rev Microbiol ; 71: 665-686, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28886683

RESUMEN

Gonorrhea, an obligate human infection, is on the rise worldwide and gonococcal strains resistant to many antibiotics are emerging. Appropriate antimicrobial treatment and prevention, including effective vaccines, are urgently needed. To guide investigation, an experimental model of genital tract infection has been developed in female mice to study mechanisms by which Neisseria gonorrhoeae evades host-derived antimicrobial factors and to identify protective and immunosuppressive pathways. Refinements of the animal model have also improved its use as a surrogate host of human infection and accelerated the testing of novel therapeutic and prophylactic compounds against gonococcal infection. Reviewed herein are the (a) history of antibiotic usage and resistance against gonorrhea and the consequences of resistance mechanisms that may increase gonococcal fitness and therefore the potential for spread, (b) use of gonococcal infection in the animal model system to study mechanisms of pathogenesis and host defenses, and


Asunto(s)
Antibacterianos/uso terapéutico , Vacunas Bacterianas/inmunología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Gonorrea/tratamiento farmacológico , Gonorrea/prevención & control , Neisseria gonorrhoeae/efectos de los fármacos , Animales , Antibacterianos/farmacología , Vacunas Bacterianas/aislamiento & purificación , Descubrimiento de Drogas , Femenino , Gonorrea/microbiología , Ratones , Neisseria gonorrhoeae/inmunología
7.
PLoS Biol ; 17(6): e3000323, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31216278

RESUMEN

Multidrug-resistant Neisseria gonorrhoeae is a global health problem. Monoclonal antibody (mAb) 2C7 recognizes a gonococcal lipooligosaccharide epitope that is expressed by >95% of clinical isolates and hastens gonococcal vaginal clearance in mice. Chimeric mAb 2C7 (human immunoglobulin G1 [IgG1]) with an E430G Fc modification that enhances Fc:Fc interactions and hexamerization following surface-target binding and increases complement activation (HexaBody technology) showed significantly greater C1q engagement and C4 and C3 deposition compared to mAb 2C7 with wild-type Fc. Greater complement activation by 2C7-E430G Fc translated to increased bactericidal activity in vitro and, consequently, enhanced efficacy in mice, compared with "Fc-unmodified" chimeric 2C7. Gonococci bind the complement inhibitors factor H (FH) and C4b-binding protein (C4BP) in a human-specific manner, which dampens antibody (Ab)-mediated complement-dependent killing. The variant 2C7-E430G Fc overcame the barrier posed by these inhibitors in human FH/C4BP transgenic mice, for which a single 1 µg intravenous dose cleared established infection. Chlamydia frequently coexists with and exacerbates gonorrhea; 2C7-E430G Fc also proved effective against gonorrhea in gonorrhea/chlamydia-coinfected mice. Complement activation alone was necessary and sufficient for 2C7 function, evidenced by the fact that (1) "complement-inactive" Fc modifications that engaged Fc gamma receptor (FcγR) rendered 2C7 ineffective, nonetheless; (2) 2C7 was nonfunctional in C1q-/- mice, when C5 function was blocked, or in C9-/- mice; and (3) 2C7 remained effective in neutrophil-depleted mice and in mice treated with PMX205, a C5a receptor (C5aR1) inhibitor. We highlight the importance of complement activation for antigonococcal Ab function in the genital tract. Elucidating the correlates of protection against gonorrhea will inform the development of Ab-based gonococcal vaccines and immunotherapeutics.


Asunto(s)
Activación de Complemento/inmunología , Gonorrea/inmunología , Neisseria gonorrhoeae/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos Bacterianos , Proteína de Unión al Complemento C4b/inmunología , Factor H de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Epítopos/inmunología , Femenino , Voluntarios Sanos , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neisseria gonorrhoeae/patogenicidad
8.
J Infect Dis ; 224(12 Suppl 2): S152-S160, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34396408

RESUMEN

Murine models of Neisseria gonorrhoeae lower reproductive tract infection are valuable systems for studying N. gonorrhoeae adaptation to the female host and immune responses to infection. These models have also accelerated preclinical testing of candidate therapeutic and prophylactic products against gonorrhea. However, because N. gonorrhoeae infection is restricted to the murine cervicovaginal region, there is a need for an in vivo system for translational work on N. gonorrhoeae pelvic inflammatory disease (PID). Here we discuss the need for well-characterized preclinical upper reproductive tract infection models for developing candidate products against N. gonorrhoeae PID, and report a refinement of the gonorrhea mouse model that supports sustained upper reproductive tract infection. To establish this new model for vaccine testing, we also tested the licensed meningococcal 4CMenB vaccine, which cross-protects against murine N. gonorrhoeae lower reproductive tract infection, for efficacy against N. gonorrhoeae in the endometrium and oviducts following transcervical or vaginal challenge.


Asunto(s)
Antiinfecciosos/administración & dosificación , Gonorrea/prevención & control , Enfermedad Inflamatoria Pélvica/prevención & control , Infecciones del Sistema Genital/microbiología , Animales , Modelos Animales de Enfermedad , Femenino , Gonorrea/tratamiento farmacológico , Ratones , Neisseria gonorrhoeae/inmunología , Enfermedad Inflamatoria Pélvica/microbiología
9.
PLoS Pathog ; 15(8): e1007937, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31369630

RESUMEN

Neisseria gonorrhoeae causes the sexually-transmitted infection gonorrhea, a global disease that is difficult to treat and for which there is no vaccine. This pathogen employs an arsenal of conserved outer membrane proteins called TonB-dependent transporters (TdTs) that allow the gonococcus to overcome nutritional immunity, the host strategy of sequestering essential nutrients away from invading bacteria to handicap infectious ability. N. gonorrhoeae produces eight known TdTs, of which four are utilized for acquisition of iron or iron chelates from host-derived proteins or xenosiderophores produced by other bacteria. Of the remaining TdTs, two of them, TdfH and TdfJ, facilitate zinc uptake. TdfH was recently shown to bind Calprotectin, a member of the S100 protein family, and subsequently extract its zinc, which is then internalized by N. gonorrhoeae. Like Calprotectin, other S100s are also capable of binding transition metals such as zinc and copper, and thus have demonstrated growth suppression of numerous other pathogens via metal sequestration. Considering the functional and structural similarities of the TdTs and of the S100s, as well as the upregulation in response to Zn limitation shown by TdfH and TdfJ, we sought to evaluate whether other S100s have the ability to support gonococcal growth by means of zinc acquisition and to frame this growth in the context of the TdTs. We found that both S100A7 and S10012 are utilized by N. gonorrhoeae as a zinc source in a mechanism that depends on the zinc transport system ZnuABC. Moreover, TdfJ binds directly to S100A7, from which it internalizes zinc. This interaction is restricted to the human version of S100A7, and zinc presence in S100A7 is required to fully support gonococcal growth. These studies highlight how gonococci co-opt human nutritional immunity, by presenting a novel interaction between TdfJ and human S100A7 for overcoming host zinc restriction.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Gonorrea/microbiología , Interacciones Huésped-Patógeno , Neisseria gonorrhoeae/metabolismo , Proteína A7 de Unión a Calcio de la Familia S100/metabolismo , Zinc/metabolismo , Secuencia de Aminoácidos , Animales , Gonorrea/patología , Humanos , Ratones , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/patogenicidad
10.
J Appl Microbiol ; 130(5): 1770-1777, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32770820

RESUMEN

AIM: Gonorrhoea remains a leading public health burden and the development of vaccine against gonorrhoea becomes more urgent. Here, a novel Neisseria gonorrhoeae DNA vaccine delivered by Salmonella enteritidis ghosts was developed and the immune responses of the vaccine candidate were evaluated. METHODS AND RESULTS: Neisseria gonorrhoeae nspA gene was cloned into the pVAX1 vector. The constructed recombinant plasmid pVAX1-nspA was loaded into the lyophilized SE ghosts to produce SE ghosts (pVAX1-nspA). Then, the immune responses induced by SE ghosts (pVAX1-nspA) alone and co-administrated with SE ghosts (pVAX1-porB) were evaluated in mouse model. Co-administered SE ghosts (pVAX1-nspA) and SE ghosts (pVAX1-porB) could elicited significantly higher levels of specific IgG antibody responses and lymphocyte proliferative responses than the control groups. Furthermore, the group co-administered SE ghosts (pVAX1-nspA) and SE ghosts (pVAX1-porB) had the highest bactericidal antibody titres. CONCLUSIONS: Co-administration of SE ghosts (pVAX1-nspA) and SE ghosts (pVAX1-porB) elicited significant specific humoral and cellular immune responses. SIGNIFICANCE AND IMPACT OF THE STUDY: This study demonstrates the potential of co-administration of SE ghosts (pVAX1-nspA) and SE ghosts (pVAX1-porB) as an attractive vaccination regimen for gonorrhoea.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Neisseria gonorrhoeae/inmunología , Salmonella enteritidis/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antibacterianos , Formación de Anticuerpos , Proteínas de la Membrana Bacteriana Externa/genética , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/microbiología , ADN Bacteriano , Femenino , Gonorrea/prevención & control , Humanos , Inmunidad Celular , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Neisseria gonorrhoeae/genética , Plásmidos/genética , Salmonella enteritidis/genética , Vacunación , Vacunas de ADN/administración & dosificación
11.
Proc Natl Acad Sci U S A ; 115(51): 12911-12919, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30559203

RESUMEN

We discuss the need to make economic evaluations of vaccines antimicrobial resistance (AMR)-sensitive and ways to do so. Such AMR-sensitive evaluations can play a role in value-for-money comparisons of different vaccines within a national immunization program, or in comparisons of vaccine-centric and non-vaccine-centric technologies within an anti-AMR program. In general terms, incremental cost-effectiveness ratios and rates of return and their associated decision rules are unaltered by consideration of AMR-related value. The decision metrics need to have their various health, cost, and socioeconomic terms disaggregated into resistance-related subcategories, which in turn have to be measured carefully before they are reaggregated. The fundamental scientific challenges lie primarily in quantifying the causal impact of health technologies on resistance-related health outcomes, and secondarily in ascertaining the economic value of those outcomes. We emphasize the importance of evaluating vaccines in the context of other potentially complementary and substitutable nonvaccine technologies. Complementarity implies that optimal spending on each set of interventions is positive, and substitutability implies that the ratio of spending will depend on relative value for money. We exemplify this general point through a qualitative discussion of the complementarities and (especially the) substitutability between pneumococcal conjugate vaccines and antimicrobial stewardship and between research and development (R&D) of a gonorrhea vaccine versus R&D of a gonorrhea antibiotic. We propose a roadmap for future work, which includes quantifying the causal effects of vaccination and other health technologies on short-term and long-term resistance-related outcomes, measuring the health-sector costs and broader socioeconomic consequences of resistance-related mortality and morbidity, and evaluating vaccines in the context of nonvaccine complements and substitutes.


Asunto(s)
Antibacterianos/uso terapéutico , Vacunas Bacterianas/uso terapéutico , Farmacorresistencia Microbiana , Gonorrea/prevención & control , Vacunación/economía , Antibacterianos/economía , Vacunas Bacterianas/economía , Análisis Costo-Beneficio , Técnicas de Apoyo para la Decisión , Gonorrea/tratamiento farmacológico , Costos de la Atención en Salud , Humanos , Programas de Inmunización , Neisseria gonorrhoeae/efectos de los fármacos , Neisseria gonorrhoeae/inmunología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/uso terapéutico , Investigación/economía , Factores Socioeconómicos
12.
Biochemistry ; 59(10): 1104-1112, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32078293

RESUMEN

Neisseria gonorrhoeae possesses a programmed recombination system that allows the bacteria to alter the major subunit of the type IV pilus, pilin or PilE. An alternate DNA structure known as a guanine quadruplex (G4) is required for pilin antigenic variation (pilin Av). The G-C base pairs within the G4 motif are required for pilin Av, but simple mutation of the loop bases does not affect pilin Av. We show that more substantial changes to the loops, in both size and nucleotide composition, with the core guanines unchanged, decrease or abrogate pilin Av. We investigated why these loop changes might influence the efficiency of pilin Av. RecA is a recombinase required for pilin Av that can bind the pilE G4 in vitro. RecA binds different G4 structures with altered loops with varied affinities. However, changes in RecA binding affinities to the loop mutants do not absolutely correlate with the pilin Av phenotypes. Interestingly, the yeast RecA ortholog, Rad51, also binds the pilE G4 structure with a higher affinity than it binds single-stranded DNA, suggesting that RecA G4 binding is conserved in eukaryotic orthologs. The thermal stability the pilE G4 structure and its loop mutants showed that the parental G4 structure had the highest melting temperature, and the melting temperature of the loop mutants correlated with pilin Av phenotype. These results suggest that the folding kinetics and stability of G4 structures are important for the efficiency of pilin Av.


Asunto(s)
Proteínas Fimbrias/metabolismo , Neisseria gonorrhoeae/genética , Variación Antigénica/genética , Emparejamiento Base/genética , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , G-Cuádruplex , Guanina/metabolismo , Mutación/genética , Neisseria gonorrhoeae/inmunología , Unión Proteica/genética , Recombinación Genética/genética
13.
Infect Immun ; 88(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31818965

RESUMEN

The sialylatable lacto-N-neotetraose (LNnT; Gal-GlcNAc-Gal-Glc) moiety from heptose I (HepI) of the lipooligosaccharide (LOS) of Neisseria gonorrhoeae undergoes positive selection during human infection. Lactose (Gal-Glc) from HepII, although phase variable, is commonly expressed in humans; loss of HepII lactose compromises gonococcal fitness in mice. Anti-LOS monoclonal antibody (MAb) 2C7, a promising antigonococcal immunotherapeutic that elicits complement-dependent bactericidal activity and attenuates gonococcal colonization in mice, recognizes an epitope comprised of lactoses expressed simultaneously from HepI and HepII. Glycan extensions beyond lactose on HepI modulate binding and function of MAb 2C7 in vitro Here, four gonococcal LOS mutants, each with lactose from HepII but fixed (unable to phase-vary) LOS HepI glycans extended beyond the lactose substitution of HepI (lactose alone, Gal-lactose, LNnT, or GalNAc-LNnT), were used to define how HepI glycan extensions affect (i) mouse vaginal colonization and (ii) efficacy in vitro and in vivo of a human IgG1 chimeric derivative of MAb 2C7 (2C7-Ximab) with a complement-enhancing E-to-G Fc mutation at position 430 (2C7-Ximab-E430G). About 10-fold lower 2C7-Ximab-E430G concentrations achieved similar complement-dependent killing of three gonococcal mutants with glycan extensions beyond lactose-substituted HepI (lactose alone, LNnT, or GalNAc-LNnT) as 2C7-Ximab (unmodified Fc). The fourth mutant (Gal-lactose) resisted direct complement-dependent killing but was killed approximately 70% by 2C7-Ximab-E430G in the presence of polymorphonuclear leukocytes and complement. Only mutants with (sialylatable) LNnT from HepI colonized mice for >3 days, reiterating the importance of LNnT sialylation for infection. 2C7-Ximab-E430G significantly attenuated colonization caused by the virulent mutants.


Asunto(s)
Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Gonorrea/terapia , Lipopolisacáridos/inmunología , Neisseria gonorrhoeae/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Resultado del Tratamiento , Vagina/microbiología
14.
PLoS Pathog ; 14(7): e1007080, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29975775

RESUMEN

The bacterial pathogen Neisseria gonorrhoeae (Gc) infects mucosal sites rich in antimicrobial proteins, including the bacterial cell wall-degrading enzyme lysozyme. Certain Gram-negative bacteria produce protein inhibitors that bind to and inhibit lysozyme. Here, we identify Ng_1063 as a new inhibitor of lysozyme in Gc, and we define its functions in light of a second, recently identified lysozyme inhibitor, Ng_1981. In silico analyses indicated that Ng_1063 bears sequence and structural homology to MliC-type inhibitors of lysozyme. Recombinant Ng_1063 inhibited lysozyme-mediated killing of a susceptible mutant of Gc and the lysozyme-sensitive bacterium Micrococcus luteus. This inhibitory activity was dependent on serine 83 and lysine 103 of Ng_1063, which are predicted to interact with lysozyme's active site residues. Lysozyme co-immunoprecipitated with Ng_1063 and Ng_1981 from intact Gc. Ng_1063 and Ng_1981 protein levels were also increased in Gc exposed to lysozyme. Gc lacking both ng1063 and ng1981 was significantly more sensitive to killing by lysozyme than wild-type or single mutant bacteria. When exposed to human tears or saliva, in which lysozyme is abundant, survival of Δ1981Δ1063 Gc was significantly reduced compared to wild-type, and survival was restored upon addition of recombinant Ng_1981. Δ1981Δ1063 mutant Gc survival was additionally reduced in the presence of human neutrophils, which produce lysozyme. We found that while Ng_1063 was exposed on the surface of Gc, Ng_1981 was both in an intracellular pool and extracellularly released from the bacteria, suggesting that Gc employs these two proteins at multiple spatial barriers to fully neutralize lysozyme activity. Together, these findings identify Ng_1063 and Ng_1981 as critical components for Gc defense against lysozyme. These proteins may be attractive targets for antimicrobial therapy aimed to render Gc susceptible to host defenses and/or for vaccine development, both of which are urgently needed against drug-resistant gonorrhea.


Asunto(s)
Proteínas Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , Neisseria gonorrhoeae/patogenicidad , Gonorrea/inmunología , Humanos , Muramidasa/antagonistas & inhibidores , Muramidasa/inmunología , Neisseria gonorrhoeae/inmunología
15.
J Immunol ; 201(9): 2700-2709, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30266769

RESUMEN

Novel therapeutics against multidrug-resistant Neisseria gonorrhoeae are urgently needed. Gonococcal lipooligosaccharide often expresses lacto-N-neotetraose (LNnT), which becomes sialylated in vivo, enhancing factor H (FH) binding and contributing to the organism's ability to resist killing by complement. We previously showed that FH domains 18-20 (with a D-to-G mutation at position 1119 in domain 19) fused to Fc (FHD1119G/Fc) displayed complement-dependent bactericidal activity in vitro and attenuated gonococcal vaginal colonization of mice. Gonococcal lipooligosaccharide phase variation can result in loss of LNnT expression. Loss of sialylated LNnT, although associated with a considerable fitness cost, could decrease efficacy of FHD1119G/Fc. Similar to N. meningitidis, gonococci also bind FH domains 6 and 7 through Neisserial surface protein A (NspA). In this study, we show that a fusion protein comprising FH domains 6 and 7 fused to human IgG1 Fc (FH6,7/Fc) bound to 15 wild-type antimicrobial resistant isolates of N. gonorrhoeae and to each of six lgtA gonococcal deletion mutants. FH6,7/Fc mediated complement-dependent killing of 8 of the 15 wild-type gonococcal isolates and effectively reduced the duration and burden of vaginal colonization of three gonococcal strains tested in wild-type mice, including two strains that resisted complement-dependent killing but on which FH6,7/Fc enhanced C3 deposition. FH/Fc lost efficacy when Fc was mutated to abrogate C1q binding and in C1q-/- mice, highlighting the requirement of the classical pathway for its activity. Targeting gonococci with FH6,7/Fc provides an additional immunotherapeutic approach against multidrug-resistant gonorrhea.


Asunto(s)
Gonorrea , Fragmentos Fc de Inmunoglobulinas , Inmunoterapia/métodos , Proteínas Recombinantes de Fusión/farmacología , Animales , Factor H de Complemento , Humanos , Inmunoglobulina G , Ratones , Neisseria gonorrhoeae/inmunología
16.
J Immunol ; 200(8): 2941-2956, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29531169

RESUMEN

Sexually transmitted infections with Chlamydia trachomatis and/or Neisseria gonorrhoeae and rates of pelvic inflammatory disease (PID) in women continue to rise, with reinfection being common because of poor adaptive immunity. Diagnosis remains imprecise, and pathogenesis data are derived primarily from monoinfection of mice with C. trachomatis or N. gonorrhoeae By comparing blood mRNA responses of women with C. trachomatis- and/or N. gonorrhoeae-induced PID and histologic endometritis with those from women with C. trachomatis and/or N. gonorrhoeae infection limited to their cervix and asymptomatic uninfected women determined via microarray, we discovered important pathogenic mechanisms in PID and response differences that provide a pathway to biomarker discovery. Women with N. gonorrhoeae- and/or C. trachomatis-induced PID exhibit overexpression of myeloid cell genes and suppression of protein synthesis, mitochondrial oxidative phosphorylation, and T cell-specific genes. Coinfected women exhibited the greatest activation of cell death pathways and suppression of responses essential for adaptive immunity. Women solely infected with C. trachomatis expressed elevated levels of type I and type II IFN genes, and enhanced type I IFN-induced chemokines in cervical secretions were associated with ascension of C. trachomatis to the endometrium. Blood microarrays reveal discrete pathobiological endotypes in women with PID that are driven by pathogen invasion of the upper genital tract.


Asunto(s)
Infecciones por Chlamydia/inmunología , Gonorrea/inmunología , Enfermedad Inflamatoria Pélvica/sangre , Enfermedad Inflamatoria Pélvica/etiología , Enfermedad Inflamatoria Pélvica/inmunología , Inmunidad Adaptativa/inmunología , Adolescente , Adulto , Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis/inmunología , Coinfección , Femenino , Gonorrea/complicaciones , Humanos , Neisseria gonorrhoeae/inmunología , Adulto Joven
17.
J Infect Dis ; 219(7): 1130-1137, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30346576

RESUMEN

BACKGROUND: Meningococcal outer membrane vesicle (OMV) vaccines are prepared with detergents to remove endotoxin, which also remove desirable antigens such as factor H binding protein (FHbp). Native OMV (NOMV) vaccines with genetically attenuated endotoxin do not require detergent treatment and elicit broader serum bactericidal antibody (SBA) responses than OMV or recombinant FHbp (rFHbp) vaccines. METHODS: We measured human complement-mediated SBA responses in mice immunized with NOMV with overexpressed FHbp subfamily B (NOMV-FHbp), NOMV with FHbp genetically inactivated (NOMV-KO), and/or a control rFHbp vaccine against meningococcal and gonococcal strains. RESULTS: Despite having 36-fold less FHbp per dose, the NOMV-FHbp vaccine elicited a ≥3-fold higher serum IgG anti-FHbp geometric mean titer than control vaccines containing rFHbp (P ≤ .003). Against 2 meningococcal outbreak strains with mismatched PorA and heterologous FHbp subfamily B sequence variants, the NOMV-FHbp vaccine produced ≥30-fold higher SBA titers than control vaccines. Mice immunized with NOMV-FHbp and NOMV-KO vaccines also elicited SBA against a gonococcal strain (P < .0001 vs the adjuvant-only control group). In contrast, 2 licensed meningococcal serogroup B vaccines, including one containing detergent-extracted OMV, did not produce gonococcal SBA in humans. CONCLUSIONS: A meningococcal NOMV vaccine elicits SBA against gonococci and with overexpressed FHbp elicits SBA against meningococci.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/inmunología , Vacunas Meningococicas/inmunología , Neisseria gonorrhoeae/inmunología , Neisseria meningitidis/inmunología , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Endotoxinas/genética , Femenino , Técnicas de Inactivación de Genes , Humanos , Inmunoglobulina G/sangre , Ratones , Vacunas Atenuadas/inmunología
18.
J Infect Dis ; 219(1): 133-144, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29688440

RESUMEN

The bacterial pathogen Neisseria gonorrhoeae is able to transmigrate across the mucosal epithelia following the intracellular route and cause disseminated infections. It is currently unknown whether the autophagy pathway is able target intracellular N. gonorrhoeae for destruction in autolysosomes or whether this bacterium is able to escape autophagy-mediated killing. In this study, we demonstrate that during the early stage of epithelial cell invasion, N. gonorrhoeae is targeted by the autophagy pathway and sequestered into double-membrane autophagosomes that subsequently fuse with lysosomes for destruction. However, a subpopulation of the intracellular gonococci is able to escape early autophagy-mediated killing. N. gonorrhoeae is subsequently able to inhibit this pathway, allowing intracellular survival and exocytosis. During this stage, N. gonorrhoeae activates the autophagy repressor mammalian target of rapamycin complex 1 and inhibits autophagosome maturation and lysosome fusion. Thus, our results provide novel insight into the interactions between N. gonorrhoeae and the autophagy pathway during invasion and transcytosis of epithelial cells.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Neisseria gonorrhoeae/metabolismo , Autofagosomas/microbiología , Autofagosomas/ultraestructura , Autofagia/inmunología , Células Epiteliales/citología , Gentamicinas/farmacología , Gonorrea/metabolismo , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Lisosomas/metabolismo , Viabilidad Microbiana , Neisseria gonorrhoeae/inmunología
19.
J Biol Chem ; 293(28): 11218-11229, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29752412

RESUMEN

Neisseria gonorrhoeae is an exclusive human pathogen that evades the host immune system through multiple mechanisms. We have shown that N. gonorrhoeae suppresses the capacity of antigen-presenting cells to induce CD4+ T cell proliferation. In this study, we sought to determine the gonococcal factors involved in this adaptive immune suppression. We show that suppression of the capacity of antigen-pulsed dendritic cells to induce T cell proliferation is recapitulated by administration of a high-molecular-weight fraction of conditioned medium from N. gonorrhoeae cultures, which includes outer membrane vesicles that are shed during growth of the bacteria. N. gonorrhoeae PorB is the most abundant protein in N. gonorrhoeae-derived vesicles, and treatment of dendritic cells with purified recombinant PorB inhibited the capacity of the cells to stimulate T cell proliferation. This immunosuppressive feature of purified PorB depended on proper folding of the protein. PorB from N. gonorrhoeae, as well as other Neisseria species and other Gram-negative bacterial species, are known to activate host Toll-like receptor 2 (TLR2) signaling. Published studies have demonstrated that purified Neisseria PorB forms proteinacious nanoparticles, termed proteosomes, when detergent micelles are removed. Unlike folded, detergent-solubilized PorB, PorB proteosomes stimulate immune responses. We now demonstrate that the formation of PorB proteosomes from structurally intact PorB eliminates the immunosuppressive property of the protein while enhancing TLR2 stimulation. These findings suggest that gonococcal PorB present in shed outer membrane vesicles plays a role in suppression of adaptive immune responses to this immune-evasive pathogen.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Gonorrea/inmunología , Neisseria gonorrhoeae/inmunología , Porinas/química , Pliegue de Proteína , Linfocitos T CD4-Positivos/microbiología , Células Cultivadas , Células Dendríticas/microbiología , Gonorrea/microbiología , Humanos , Activación de Linfocitos , Porinas/metabolismo , Transducción de Señal , Receptor Toll-Like 2/metabolismo
20.
Infect Immun ; 87(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30510105

RESUMEN

Neisseria gonorrhoeae, the causative agent of gonorrhea, has evolved several mechanisms to subvert complement, including binding of the complement inhibitor factor H (FH). We previously reported FH binding to N. gonorrhoeae independently of lipooligosaccharide (LOS) sialylation. Here we report that factor H-like protein 1 (FHL-1), which contains FH domains 1 through 7 and possesses complement-inhibitory activity, also binds to N. gonorrhoeae The ligand for both FH and FHL-1 was identified as neisserial surface protein A (NspA), which has previously been identified as a ligand for these molecules on Neisseria meningitidis As with N. meningitidis NspA (Nm-NspA), N. gonorrhoeae NspA (Ng-NspA) bound FH/FHL-1 through FH domains 6 and 7. Binding of FH/FHL-1 to NspA was human specific; the histidine (H) at position 337 of domain 6 contributed to human-specific FH binding to both Ng- and Nm-NspA. FH/FHL-1 bound Nm-NspA better than Ng-NspA; introducing Q at position 73 (loop 2, present in Ng-NspA) or replacing V and D at positions 112 and 113 in Nm-NspA loop 3 with A and H (Ng-NspA), respectively, reduced FH/FHL-1 binding. The converse Ng-NspA to Nm-NspA mutations increased FH/FHL-1 binding. Binding of FH/FHL-1 through domains 6 and 7 to N. gonorrhoeae increased with truncation of the heptose I (HepI) chain of LOS and decreased with LOS sialylation. Loss of NspA significantly decreased serum resistance of N. gonorrhoeae with either wild-type or truncated LOS. This report highlights the role for NspA in enabling N. gonorrhoeae to subvert complement despite LOS phase variation. Knowledge of FH-NspA interactions will inform the design of vaccines and immunotherapies against the global threat of multidrug-resistant gonorrhea.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Inactivadoras del Complemento C3b/inmunología , Factor H de Complemento/inmunología , Gonorrea/inmunología , Neisseria gonorrhoeae/inmunología , Neisseria meningitidis/inmunología , Adhesión Bacteriana/inmunología , Células Cultivadas , Humanos , Neisseria gonorrhoeae/patogenicidad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda