Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(3): e1011249, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36961851

RESUMEN

Pasteurella multocida can infect a multitude of wild and domesticated animals, with infections in cattle resulting in hemorrhagic septicemia (HS) or contributing to bovine respiratory disease (BRD) complex. Current cattle vaccines against P. multocida consist of inactivated bacteria, which only offer limited and serogroup specific protection. Here, we describe a newly identified surface lipoprotein, PmSLP, that is present in nearly all annotated P. multocida strains isolated from cattle. Bovine associated variants span three of the four identified phylogenetic clusters, with PmSLP-1 and PmSLP-2 being restricted to BRD associated isolates and PmSLP-3 being restricted to isolates associated with HS. Recombinantly expressed, soluble PmSLP-1 (BRD-PmSLP) and PmSLP-3 (HS-PmSLP) vaccines were both able to provide full protection in a mouse sepsis model against the matched P. multocida strain, however no cross-protection and minimal serum IgG cross-reactivity was identified. Full protection against both challenge strains was achieved with a bivalent vaccine containing both BRD-PmSLP and HS-PmSLP, with serum IgG from immunized mice being highly reactive to both variants. Year-long stability studies with lyophilized antigen stored under various temperatures show no appreciable difference in biophysical properties or loss of efficacy in the mouse challenge model. PmSLP-1 and PmSLP-3 vaccines were each evaluated for immunogenicity in two independent cattle trials involving animals of different age ranges and breeds. In all four trials, vaccination with PmSLP resulted in an increase in antigen specific serum IgG over baseline. In a blinded cattle challenge study with a recently isolated HS strain, the matched HS-PmSLP vaccine showed strong efficacy (75-87.5% survival compared to 0% in the control group). Together, these data suggest that cattle vaccines composed of PmSLP antigens can be a practical and effective solution for preventing HS and BRD related P. multocida infections.


Asunto(s)
Septicemia Hemorrágica , Infecciones por Pasteurella , Pasteurella multocida , Bovinos , Animales , Ratones , Filogenia , Vacunología , Vacunas Bacterianas , Septicemia Hemorrágica/microbiología , Septicemia Hemorrágica/prevención & control , Septicemia Hemorrágica/veterinaria , Modelos Animales de Enfermedad , Inmunoglobulina G , Infecciones por Pasteurella/microbiología , Infecciones por Pasteurella/prevención & control , Infecciones por Pasteurella/veterinaria
2.
J Immunol ; 211(6): 981-993, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37493438

RESUMEN

Current vaccine efforts to combat SARS-CoV-2 are focused on the whole spike protein administered as mRNA, viral vector, or protein subunit. However, the SARS-CoV-2 receptor-binding domain (RBD) is the immunodominant portion of the spike protein, accounting for 90% of serum neutralizing activity. In this study, we constructed several versions of RBD and together with aluminum hydroxide or DDA (dimethyldioctadecylammonium bromide)/TDB (d-(+)-trehalose 6,6'-dibehenate) adjuvant evaluated immunogenicity in mice. We generated human angiotensin-converting enzyme 2 knock-in mice to evaluate vaccine efficacy in vivo following viral challenge. We found that 1) subdomain (SD)1 was essential for the RBD to elicit maximal immunogenicity; 2) RBDSD1 produced in mammalian HEK cells elicited better immunogenicity than did protein produced in insect or yeast cells; 3) RBDSD1 combined with the CD4 Th1 adjuvant DDA/TDB produced higher neutralizing Ab responses and stronger CD4 T cell responses than did aluminum hydroxide; 4) addition of monomeric human Fc receptor to RBDSD1 (RBDSD1Fc) significantly enhanced immunogenicity and neutralizing Ab titers; 5) the Beta version of RBDSD1Fc provided a broad range of cross-neutralization to multiple antigenic variants of concern, including Omicron; and 6) the Beta version of RBDSD1Fc with DDA/TDB provided complete protection against virus challenge in the knock-in mouse model. Thus, we have identified an optimized RBD-based subunit vaccine suitable for clinical trials.


Asunto(s)
COVID-19 , Vacunas Virales , Humanos , Animales , Ratones , SARS-CoV-2 , Vacunas contra la COVID-19 , Hidróxido de Aluminio , Glicoproteína de la Espiga del Coronavirus , Vacunas de Subunidad , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Mamíferos
3.
J Biol Chem ; 298(10): 102448, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36063991

RESUMEN

Mapping the self-organization and spatial distribution of membrane proteins is key to understanding their function. Developing methods that can provide insight into correlations between membrane protein colocalization and interactions is challenging. We report here on a correlated stochastic optical reconstruction microscopy/homoFRET imaging approach for resolving the nanoscale distribution and oligomeric state of membrane proteins. Using live cell homoFRET imaging of carcinoembryonic antigen-related cellular adhesion molecule 1, a cell-surface receptor known to exist in a complex equilibrium between monomer and dimer/oligomer states, we revealed highly heterogeneous diffraction-limited structures on the surface of HeLa cells. Furthermore, correlated super-resolved stochastic optical reconstruction microscopy imaging showed that these structures comprised a complex mixture and spatial distribution of self-associated carcinoembryonic antigen-related cellular adhesion molecule 1 molecules. In conclusion, this correlated approach provides a compelling strategy for addressing challenging questions about the interplay between membrane protein concentration, distribution, interaction, clustering, and function.


Asunto(s)
Microscopía , Humanos , Células HeLa , Proteínas de la Membrana , Microscopía/métodos , Imagen Óptica , Técnicas Citológicas
4.
Retrovirology ; 19(1): 18, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986377

RESUMEN

BACKGROUND: The generation of over 69 spliced HIV-1 mRNAs from one primary transcript by alternative RNA splicing emphasizes the central role that RNA processing plays in HIV-1 replication. Control is mediated in part through the action of host SR proteins whose activity is regulated by multiple SR kinases (CLK1-4, SRPKs). METHODS: Both shRNA depletion and small molecule inhibitors of host SR kinases were used in T cell lines and primary cells to evaluate the role of these factors in the regulation of HIV-1 gene expression. Effects on virus expression were assessed using western blotting, RT-qPCR, and immunofluorescence. RESULTS: The studies demonstrate that SR kinases play distinct roles; depletion of CLK1 enhanced HIV-1 gene expression, reduction of CLK2 or SRPK1 suppressed it, whereas CLK3 depletion had a modest impact. The opposing effects of CLK1 vs. CLK2 depletion were due to action at distinct steps; reduction of CLK1 increased HIV-1 promoter activity while depletion of CLK2 affected steps after transcript initiation. Reduced CLK1 expression also enhanced the response to several latency reversing agents, in part, by increasing the frequency of responding cells, consistent with a role in regulating provirus latency. To determine whether small molecule modulation of SR kinase function could be used to control HIV-1 replication, we screened a GSK library of protein kinase inhibitors (PKIS) and identified several pyrazolo[1,5-b] pyridazine derivatives that suppress HIV-1 gene expression/replication with an EC50 ~ 50 nM. The compounds suppressed HIV-1 protein and viral RNA accumulation with minimal impact on cell viability, inhibiting CLK1 and CLK2 but not CLK3 function, thereby selectively altering the abundance of individual CLK and SR proteins in cells. CONCLUSIONS: These findings demonstrate the unique roles played by individual SR kinases in regulating HIV-1 gene expression, validating the targeting of these functions to either enhance latency reversal, essential for "Kick-and-Kill" strategies, or to silence HIV protein expression for "Block-and-Lock" strategies.


Identifying cellular factors that regulate HIV-1 RNA processing provides important insights into novel strategies to control this infection. Different members of the SR kinase family have distinct roles in regulating virus expression because they affect distinct steps of transcription/RNA processing. We identify inhibitors of these kinases that suppress HIV-1 gene expression and replication in multiple assay systems at nanomolar concentrations with limited or no cytotoxicity. Our results highlight the therapeutic potential of targeting the post-integration stage of the HIV-1 lifecycle to selectively enhance or reverse provirus latency. A greater understanding of the molecular mechanisms underlying the effects observed will facilitate the development of more targeted approaches to modulate HIV-1 latency on the path toward a "functional" cure for this infection.


Asunto(s)
VIH-1 , Empalme Alternativo , Expresión Génica , VIH-1/fisiología , Inhibidores de Proteínas Quinasas/farmacología , ARN Viral/genética , Latencia del Virus
5.
J Infect Dis ; 224(12 Suppl 2): S56-S63, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34396410

RESUMEN

While infection by Neisseria gonorrhoeae is often asymptomatic in women, undetected infections can ascend into the upper genital tract to elicit an inflammatory response that manifests as pelvic inflammatory disease, with the outcomes depending on the intensity and duration of inflammation and whether it is localized to the endometrial, fallopian tube, ovarian, and/or other tissues. This review examines the contribution of N. gonorrhoeae versus other potential causes of pelvic inflammatory disease by considering new insights gained through molecular, immunological, and microbiome-based analyses, and the current epidemiological burden of infection, with an aim to highlighting key areas for future study.


Asunto(s)
Infecciones por Chlamydia/epidemiología , Gonorrea/epidemiología , Neisseria gonorrhoeae/aislamiento & purificación , Enfermedad Inflamatoria Pélvica/epidemiología , Infecciones por Chlamydia/complicaciones , Endometritis/microbiología , Endometrio/microbiología , Endometrio/patología , Trompas Uterinas/microbiología , Femenino , Gonorrea/diagnóstico , Humanos , Enfermedad Inflamatoria Pélvica/diagnóstico , Enfermedad Inflamatoria Pélvica/microbiología
6.
PLoS Pathog ; 15(12): e1008136, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31790511

RESUMEN

Sexually transmitted infections are a critical public health issue. However, the mechanisms underlying sexually transmitted infections in women and the link between the infection mechanism and the wide range of clinical outcomes remain elusive due to a lack of research models mimicking human infection in vivo. We established a human cervical tissue explant model to mimic local Neisseria gonorrhoeae (GC) infections. We found that GC preferentially colonize the ectocervix by activating integrin-ß1, which inhibits epithelial shedding. GC selectively penetrate into the squamocolumnar junction (TZ) and endocervical epithelia by inducing ß-catenin phosphorylation, which leads to E-cadherin junction disassembly. Epithelial cells in various cervical regions differentially express carcinoembryonic antigen-related cell adhesion molecules (CEACAMs), the host receptor for GC opacity-associated proteins (OpaCEA). Relatively high levels were detected on the luminal membrane of ecto/endocervical epithelial cells but very low levels intracellularly in TZ epithelial cells. CEACAM-OpaCEA interaction increased ecto/endocervical colonization and reduced endocervical penetration by increasing integrin-ß1 activation and inhibiting ß-catenin phosphorylation respectively, through CEACAM downstream signaling. Thus, the intrinsic properties of cervical epithelial cells and phase-variation of bacterial surface molecules both play a role in controlling GC infection mechanisms and infectivity, preferential colonization or penetration, potentially leading to asymptomatic or symptomatic infection.


Asunto(s)
Adhesión Bacteriana/fisiología , Cuello del Útero/microbiología , Gonorrea/microbiología , Membrana Mucosa/microbiología , Neisseria gonorrhoeae/patogenicidad , Cuello del Útero/metabolismo , Femenino , Humanos , Membrana Mucosa/metabolismo , Técnicas de Cultivo de Órganos
7.
CMAJ ; 192(31): E871-E874, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32646870

RESUMEN

BACKGROUND: Provision of pasteurized donor human milk, as a bridge to mother's own milk, is the standard of care for very low-birth-weight infants in hospital. The aim of this research was to confirm that Holder pasteurization (62.5°C for 30 min) would be sufficient to inactivate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in donated human milk samples. METHODS: We spiked frozen milk samples from 10 donors to the Rogers Hixon Ontario Human Milk Bank with SARS-CoV-2 to achieve a final concentration of 1 × 107 TCID50/mL (50% of the tissue culture infectivity dose per mL). We pasteurized samples using the Holder method or held them at room temperature for 30 minutes and plated serial dilutions on Vero E6 cells for 5 days. We included comparative controls in the study using milk samples from the same donors without addition of virus (pasteurized and unpasteurized) as well as replicates of Vero E6 cells directly inoculated with SARS-CoV-2. We reported cytopathic effects as TCID50/mL. RESULTS: We detected no cytopathic activity in any of the SARS-CoV-2-spiked milk samples that had been pasteurized using the Holder method. In the SARS-CoV-2-spiked milk samples that were not pasteurized but were kept at room temperature for 30 minutes, we observed a reduction in infectious viral titre of about 1 log. INTERPRETATION: Pasteurization of human milk by the Holder method (62.5°C for 30 min) inactivates SARS-CoV-2. Thus, in the event that donated human milk contains SARS-CoV-2 by transmission through the mammary gland or by contamination, this method of pasteurization renders milk safe for consumption and handling by care providers.


Asunto(s)
Betacoronavirus/crecimiento & desarrollo , Infecciones por Coronavirus/prevención & control , Bancos de Leche Humana , Leche Humana/virología , Pandemias/prevención & control , Pasteurización/métodos , Neumonía Viral/prevención & control , Inactivación de Virus , COVID-19 , Calor , Humanos , Leche Humana/química , Ontario , SARS-CoV-2 , Factores de Tiempo , Ensayo de Placa Viral
8.
J Immunol ; 201(8): 2385-2391, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30224513

RESUMEN

d-Glycero-ß-d-manno-heptose 1,7-biphosphate (ß-HBP) is a novel microbial-associated molecular pattern that triggers inflammation and thus has the potential to act as an immune modulator in many therapeutic contexts. To better understand the structure-activity relationship of this molecule, we chemically synthesized analogs of ß-HBP and tested their ability to induce canonical TIFA-dependent inflammation in human embryonic kidney cells (HEK 293T) and colonic epithelial cells (HCT 116). Of the analogs tested, only d-glycero-ß-d-manno-heptose 1-phosphate (ß-HMP) induced TIFA-dependent NF-κB activation and cytokine production in a manner similar to ß-HBP. This finding expands the spectrum of metabolites from the Gram-negative ADP-heptose biosynthesis pathway that can function as innate immune agonists and provides a more readily available agonist of the TIFA-dependent inflammatory pathway that can be easily produced by synthetic methods.


Asunto(s)
Bacterias Gramnegativas/fisiología , Heptosas/inmunología , Inmunidad Innata , Factores Inmunológicos/inmunología , Inflamación/inmunología , Manosa/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fosfatos/inmunología , Piranos/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células HEK293 , Heptosas/síntesis química , Humanos , Inmunización , Factores Inmunológicos/síntesis química , Inflamación/inducido químicamente , Manosa/síntesis química , Fosfatos/síntesis química , Piranos/síntesis química , Transducción de Señal , Relación Estructura-Actividad , Especificidad por Sustrato
9.
Cell Microbiol ; 20(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28886618

RESUMEN

The neutrophil-specific innate immune receptor CEACAM3 functions as a decoy to capture Gram-negative pathogens, such as Neisseria gonorrhoeae, that exploit CEACAM family members to adhere to the epithelium. Bacterial binding to CEACAM3 results in their efficient engulfment and triggers activation of an nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-dependent inflammatory response by human neutrophils. Herein, we report that CEACAM3 cross-linking is not sufficient for induction of cytokine production and show that the inflammatory response induced by Neisseria gonorrhoeae infection is elicited by an integration of signals from CEACAM3 and toll-like receptors. Using neutrophils from a human CEACAM-expressing mouse line (CEABAC), we use a genetic approach to reveal a molecular bifurcation of the CEACAM3-mediated antimicrobial and inflammatory responses. Ex vivo experiments with CEABAC-Rac2-/- , CEABAC-Bcl10-/- , and CEABAC-Malt1-/- neutrophils indicate that these effectors are not necessary for gonococcal engulfment, yet all 3 effectors contribute to CEACAM3-mediated cytokine production. Interestingly, although Bcl10 and Malt1 are often inextricably linked, Bcl10 enabled synergy between toll-like receptor 4 and CEACAM3, whereas Malt1 did not. Together, these findings reveal an integration of the specific innate immune receptor CEACAM3 into the network of more conventional pattern recognition receptors, providing a mechanism by which the innate immune system can unleash its response to a relentless pathogen.


Asunto(s)
Proteína 10 de la LLC-Linfoma de Células B/genética , Antígeno Carcinoembrionario/inmunología , Gonorrea/inmunología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Neisseria gonorrhoeae/inmunología , Neutrófilos/inmunología , Receptor Toll-Like 4/inmunología , Animales , Antígenos CD/inmunología , Proteína 10 de la LLC-Linfoma de Células B/inmunología , Moléculas de Adhesión Celular/inmunología , Células Cultivadas , Proteínas Ligadas a GPI/inmunología , Gonorrea/microbiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/inmunología , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/inmunología , Proteína RCA2 de Unión a GTP
10.
Infect Immun ; 86(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29760215

RESUMEN

The gonococcal Opa proteins are an antigenically variable family of surface adhesins that bind human carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), CEACAM3, CEACAM5, and/or CEACAM6, cell surface glycoproteins that are differentially expressed on a broad spectrum of human cells and tissues. While they are presumed to be important for infection, the significance of various Opa-CEACAM-mediated cellular interactions in the context of the genital tract has remained unclear. Here, we observed that CEACAM1 and CEACAM5 are differentially expressed on epithelia lining the upper and lower portions of the human female genital tract, respectively. Using transgenic mouse lines expressing human CEACAMs in a manner that reflects this differential pattern, we considered the impact of Opa-CEACAM interactions during uncomplicated lower genital tract infections versus during pelvic inflammatory disease. Our results demonstrate that Opa-CEACAM5 binding on vaginal epithelia facilitates the long-term colonization of the lower genital tract, while Opa protein binding to CEACAM1 on uterine epithelia enhances gonococcal association and penetration into these tissues. While these Opa-dependent interactions with CEACAM-expressing epithelial surfaces promote infection, Opa binding by neutrophil-expressed CEACAMs counterbalances this by facilitating more effective gonococcal clearance. Furthermore, during uterine infections, CEACAM-dependent tissue invasion aggravates disease pathology by increasing the acute inflammatory response. Together, these findings demonstrate that the outcome of infection is determined by both the cell type-specific expression of human CEACAMs and the CEACAM specificity of the Opa variants expressed, which combine to determine the level of gonococcal association with the genital mucosa versus the extent of CEACAM-dependent inflammation and gonococcal clearance by neutrophils.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Genitales Femeninos/patología , Gonorrea/fisiopatología , Infecciones del Sistema Genital/fisiopatología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Perfilación de la Expresión Génica , Genitales Femeninos/microbiología , Gonorrea/microbiología , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Transgénicos , Neisseria gonorrhoeae/fisiología , Infecciones del Sistema Genital/microbiología , Resultado del Tratamiento , Útero/microbiología , Útero/patología , Vagina/microbiología , Vagina/patología
11.
BMC Genomics ; 19(1): 627, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30134832

RESUMEN

BACKGROUND: The emergence of fully antimicrobial resistant Neisseria gonorrhoeae has led global public health agencies to identify a critical need for next generation anti-gonococcal pharmaceuticals. The development and success of these compounds will rely upon valid pre-clinical models of gonorrhoeae infection. We recently developed and reported the first model of upper genital tract gonococcal infection. During initial characterization, we observed significant reproductive cycle-based variation in infection outcome. When uterine infection occurred in the diestrus phase, there was significantly greater pathology than during estrus phase. The aim of this study was to evaluate transcriptional profiles of infected uterine tissue from mice in either estrus or diestrus phase in order to elucidate possible mechanisms for these differences. RESULTS: Genes and biological pathways with phase-independent induction during infection showed a chemokine dominant cytokine response to Neisseria gonorrhoeae. Despite general induction being phase-independent, this common anti-gonococcal response demonstrated greater induction during diestrus phase infection. Greater activity of granulocyte adhesion and diapedesis regulators during diestrus infection, particularly in chemokines and diapedesis regulators, was also shown. In addition to a greater induction of the common anti-gonococcal response, Gene Set Enrichment Analysis identified a diestrus-specific induction of type-1 interferon signaling pathways. CONCLUSIONS: This transcriptional analysis of murine uterine gonococcal infection during distinct points in the natural reproductive cycle provided evidence for a common anti-gonococcal response characterized by significant induction of granulocyte chemokine expression and high proinflammatory mediators. The basic biology of this host response to N. gonorrhoeae in estrus and diestrus is similar at the pathway level but varies drastically in magnitude. Overlaying this, we observed type-1 interferon induction specifically in diestrus infection where greater pathology is observed. This supports recent work suggesting this pathway has a significant, possibly host-detrimental, function in gonococcal infection. Together these findings lay the groundwork for further examination of the role of interferons in gonococcal infection. Additionally, this work enables the implementation of the diestrus uterine infection model using the newly characterized host response as a marker of pathology and its prevention as a correlate of candidate vaccine efficacy and ability to protect against the devastating consequences of N. gonorrhoeae-associated sequelae.


Asunto(s)
Ciclo Estral/fisiología , Gonorrea/genética , Interacciones Huésped-Patógeno/genética , Inflamación/genética , Neisseria gonorrhoeae , Infecciones del Sistema Genital/genética , Transcriptoma , Animales , Modelos Animales de Enfermedad , Ciclo Estral/genética , Femenino , Perfilación de la Expresión Génica , Gonorrea/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/fisiopatología , Ratones , Análisis por Micromatrices , Neisseria gonorrhoeae/inmunología , Infecciones del Sistema Genital/inmunología , Infecciones del Sistema Genital/microbiología
12.
J Infect Dis ; 215(10): 1590-1598, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28368526

RESUMEN

Background: The multicomponent meningococcal serogroup B vaccine (4CMenB) is an outer membrane vesicle and recombinant protein-based vaccine licensed to protect against serogroup B meningococcal disease. It remains unknown whether this vaccine will prevent carriage or transmission, key aspects in long-term vaccine success and disease eradication. Methods: Using a "humanized" transgenic mouse model of nasal colonization, we took a systematic approach to estimate the potential for carriage prevention against antigenically diverse Neisseria meningitidis strains and to compare this protection to an invasive meningococcal disease challenge model. Results: The 4CMenB vaccine prevented morbidity and mortality after lethal invasive doses of all meningococcal strains tested. Immunization effectively prevented carriage with only 1 of 4 single antigen-matched strains but reduced or prevented nasal colonization by all 4 isolates with multiple cross-reacting antigens. Each immunized mouse had substantial immunoglobulin G targeting the challenge strains, indicating that antibody correlates with protection against sepsis but not nasal carriage. Conclusions: Immunization with the 4CMenB vaccine elicits a robust humoral response that correlates with protection against invasive challenge but not with prevention of asymptomatic colonization. This suggests that widespread use of this vaccine will reduce morbidity and mortality rates in immunized individuals, with the potential to contribute to herd protection against a subset of strains.


Asunto(s)
Portador Sano/inmunología , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/inmunología , Cavidad Nasal/microbiología , Neisseria meningitidis Serogrupo B/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones
13.
Nat Rev Immunol ; 6(6): 433-46, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16724098

RESUMEN

The carcinoembryonic-antigen-related cell-adhesion molecule (CEACAM) family of proteins has been implicated in various intercellular-adhesion and intracellular-signalling-mediated effects that govern the growth and differentiation of normal and cancerous cells. Recent studies show that there is an important role for members of the CEACAM family in modulating the immune responses associated with infection, inflammation and cancer. In this Review, we consider the evidence for CEACAM involvement in immunity, with a particular emphasis on CEACAM1, which functions as a regulatory co-receptor for both lymphoid and myeloid cell types.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Comunicación Celular/inmunología , Inmunidad Celular , Tejido Linfoide/inmunología , Células Mieloides/inmunología , Animales , Antígenos CD/biosíntesis , Antígenos CD/química , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/química , Humanos , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Tejido Linfoide/patología , Células Mieloides/citología , Células Mieloides/metabolismo , Células Mieloides/patología
14.
Clin Infect Dis ; 62(9): 1126-1132, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908785

RESUMEN

BACKGROUND: Streptococcus pneumoniaeis a major cause of community-acquired pneumonia and septicemia in adults. The global drug-susceptible capsular serotype 12F, clonal complex 218 caused several outbreaks in the United States between 1989 and 2008, as well as a recent large outbreak in Manitoba, Canada, that resulted in 36 cases of septicemia and 3 deaths. The evolutionary origin of the Canadian outbreak strain and its relationship to the historical US outbreak strains are not known. METHODS: Whole-genome deep sequencing was performed on isolates from the Canadian outbreak (n = 36), the US outbreaks (n = 9), and nonoutbreak surveys (n = 21). Phylogenomic analysis and comparative genomics were used to assess evolutionary relationships and to detect gene content differences between the isolates. RESULTS: The Canadian outbreak was closely related to sporadic cases that occurred preoutbreak in cross-border geographic regions in Manitoba, North Dakota, and Iowa. The emerging Canadian strain differed from US strains by acquisition of a cell-surface protein and macrolide resistance determinants via incorporation of a 5.3-kb mega cassette harboringmsrDandmefE Furthermore, during 11 months of transmission, this clone evolved rapidly and acquired fluoroquinolone resistance through precise stepwise mutations in bothparCandgyrA, and putative compensatory mutations inuraAorIMPDHunder drug selection. Alarmingly, this drug-resistant clone appears to have spread quickly to other regions of Canada and the United States, and replaced drug-susceptible strains. CONCLUSIONS: Whole-genome sequencing revealed an independent emergence and secondary adaptation of a new virulent and drug-resistant pneumococcal epidemic clone. Ongoing molecular surveillance is required, and measures to prevent its spread should be developed.


Asunto(s)
Evolución Biológica , Farmacorresistencia Bacteriana/genética , Genoma Bacteriano , Análisis de Secuencia de ADN , Serogrupo , Streptococcus pneumoniae/genética , Antibacterianos/uso terapéutico , Canadá , Brotes de Enfermedades , Humanos , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/genética , Streptococcus pneumoniae/efectos de los fármacos , Estados Unidos
15.
Int J Cancer ; 139(4): 841-53, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27037842

RESUMEN

The engraftment of circulating cancer cells at distal sites represents a key step in the metastatic cascade, yet remains an unexplored target for therapeutic intervention. In this study, we establish that a vaccination strategy yielding an antigen-specific TH 9 response induces long term host surveillance and prevents the engraftment of circulating cancer cells. Specifically, we show that vaccination with a recombinant CEA IgV-like N domain, formulated with the TLR3 ligand poly I:C, elicits a CEA-specific TH 9 response, wherein IL-9 secreting TH cells act in concert with CEA N domain-specific antibodies as well as activated mast cells in preventing tumor cell engraftment. The development of this immune response was dependent on TLR3, since interference with the TLR3-dsRNA complex formation led to a reduction in vaccine-imparted protection and a shift in the resulting immune response toward a TH 2 response. These findings point to the existence of an alternate tumor targeting immune mechanism that can be exploited for the purpose of developing vaccine therapies targeting tumor dissemination and engraftment.


Asunto(s)
Antígenos de Neoplasias/inmunología , Mastocitos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/inmunología , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Interleucina-9/biosíntesis , Mastocitos/metabolismo , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Receptor Toll-Like 3/metabolismo
16.
PLoS Pathog ; 10(9): e1004341, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25188454

RESUMEN

An overwhelming neutrophil-driven response causes both acute symptoms and the lasting sequelae that result from infection with Neisseria gonorrhoeae. Neutrophils undergo an aggressive opsonin-independent response to N. gonorrhoeae, driven by the innate decoy receptor CEACAM3. CEACAM3 is exclusively expressed by human neutrophils, and drives a potent binding, phagocytic engulfment and oxidative killing of Opa-expressing bacteria. In this study, we sought to explore the contribution of neutrophils to the pathogenic inflammatory process that typifies gonorrhea. Genome-wide microarray and biochemical profiling of gonococcal-infected neutrophils revealed that CEACAM3 engagement triggers a Syk-, PKCδ- and Tak1-dependent signaling cascade that results in the activation of an NF-κB-dependent transcriptional response, with consequent production of pro-inflammatory cytokines. Using an in vivo model of N. gonorrhoeae infection, we show that human CEACAM-expressing neutrophils have heightened migration toward the site of the infection where they may be further activated upon Opa-dependent binding. Together, this study establishes that the role of CEACAM3 is not restricted to the direct opsonin-independent killing by neutrophils, since it also drives the vigorous inflammatory response that typifies gonorrhea. By carrying the potential to mobilize increasing numbers of neutrophils, CEACAM3 thereby represents the tipping point between protective and pathogenic outcomes of N. gonorrhoeae infection.


Asunto(s)
Biomarcadores/metabolismo , Gonorrea/inmunología , Mediadores de Inflamación/metabolismo , Inflamación/etiología , Neisseria gonorrhoeae/patogenicidad , Neutrófilos/inmunología , Animales , Adhesión Bacteriana , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Gonorrea/metabolismo , Gonorrea/microbiología , Humanos , Inflamación/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Transgénicos , Neisseria gonorrhoeae/inmunología , Neutrófilos/microbiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo , Fagocitosis/fisiología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Quinasa Syk
17.
Proc Natl Acad Sci U S A ; 110(25): 10234-9, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23733950

RESUMEN

Clinical and epidemiological synergy exists between the globally important sexually transmitted infections, gonorrhea and HIV. Neisseria gonorrhoeae, which causes gonorrhea, is particularly adept at driving HIV-1 expression, but the molecular determinants of this relationship remain undefined. N. gonorrhoeae liberates a soluble factor that potently induces expression from the HIV-1 LTR in coinfected cluster of differentiation 4-positive (CD4(+)) T lymphocytes, but this factor is not a previously described innate effector. A genome-wide mutagenesis approach was undertaken to reveal which component(s) of N. gonorrhoeae induce HIV-1 expression in CD4(+) T lymphocytes. A mutation in the ADP-heptose biosynthesis gene, hldA, rendered the bacteria unable to induce HIV-1 expression. The hldA mutant has a truncated lipooligosaccharide structure, contains lipid A in its outer membrane, and remains bioactive in a TLR4 reporter-based assay but did not induce HIV-1 expression. Mass spectrometry analysis of extensively fractionated N. gonorrhoeae-derived supernatants revealed that the LTR-inducing fraction contained a compound having a mass consistent with heptose-monophosphate (HMP). Heptose is a carbohydrate common in microbes but is absent from the mammalian glycome. Although ADP-heptose biosynthesis is common among Gram-negative bacteria, and heptose is a core component of most lipopolysaccharides, N. gonorrhoeae is peculiar in that it effectively liberates HMP during growth. This N. gonorrhoeae-derived HMP activates CD4(+) T cells to invoke an NF-κB-dependent transcriptional response that drives HIV-1 expression and viral production. Our study thereby shows that heptose is a microbial-specific product that is sensed as an innate immune agonist and unveils the molecular link between N. gonorrhoeae and HIV-1.


Asunto(s)
Coinfección/inmunología , Gonorrea , Infecciones por VIH , VIH-1/enzimología , Heptosas/inmunología , Neisseria gonorrhoeae/enzimología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/virología , Femenino , Gonorrea/inmunología , Gonorrea/microbiología , Gonorrea/virología , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/virología , Duplicado del Terminal Largo de VIH/genética , VIH-1/inmunología , Heptosas/genética , Heptosas/metabolismo , Humanos , Células Jurkat , Masculino , Neisseria gonorrhoeae/inmunología , Receptor Toll-Like 5/inmunología
18.
Infect Immun ; 83(1): 354-63, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25368118

RESUMEN

Neisseria meningitidis asymptomatically colonizes the human upper respiratory tract but is also the cause of meningitis and severe septicemia. Carriage or disease evokes an immune response against the infecting strain. Hitherto, we have known little about the breadth of immunity induced by natural carriage of a single strain or its implications for subsequent infectious challenge. In this study, we establish that transgenic mice expressing human CEACAM1 support nasal colonization by a variety of strains of different capsular types. Next, we nasally challenged these mice with either of the N. meningitidis strains H44/76 (serogroup B, ST-32) and 90/18311 (serogroup C, ST-11), while following the induction of strain-specific immunoglobulin. When these antisera were tested for reactivity with a diverse panel of N. meningitidis strains, very low levels of antibody were detected against all meningococcal strains, yet a mutually exclusive "fingerprint" of high-level cross-reactivity toward certain strains became apparent. To test the efficacy of these responses for protection against subsequent challenge, CEACAM1-humanized mice exposed to strain 90/18311 were then rechallenged with different N. meningitidis strains. As expected, the mice were immune to challenge with the same strain and with a closely related ST-11 strain, 38VI, while H44/76 (ST-32) could still colonize these animals. Notably, however, despite the paucity of detectable humoral response against strain 196/87 (ST-32), this strain was unable to colonize the 90/18311-exposed mice. Combined, our data suggest that current approaches may underestimate the actual breadth of mucosal protection gained through natural exposure to N. meningitidis strains.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos CD/inmunología , Portador Sano/inmunología , Moléculas de Adhesión Celular/inmunología , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/prevención & control , Neisseria meningitidis/inmunología , Animales , Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Humanos , Ratones Transgénicos
19.
Infect Immun ; 83(4): 1372-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25605771

RESUMEN

Infections by Neisseria gonorrhoeae are increasingly common, are often caused by antibiotic-resistant strains, and can result in serious and lasting sequelae, prompting the reemergence of gonococcal disease as a leading global health concern. N. gonorrhoeae is a human-restricted pathogen that primarily colonizes urogenital mucosal surfaces. Disease progression varies greatly between the sexes: men usually present with symptomatic infection characterized by a painful purulent urethral discharge, while in women, the infection is often asymptomatic, with the most severe pathology occurring when the bacteria ascend from the lower genital tract into the uterus and fallopian tubes. Classical clinical studies demonstrated that clinically infectious strains uniformly express Opa adhesins; however, their specificities were unknown at the time. While in vitro studies have since identified CEACAM proteins as the primary target of Opa proteins, the gonococcal specificity for this human family of receptors has not been addressed in the context of natural infection. In this study, we characterize a collection of low-passage-number clinical-specimen-derived N. gonorrhoeae isolates for Opa expression and assess their CEACAM-binding profiles. We report marked in vivo selection for expression of phase-variable Opa proteins that bind CEACAM1 and CEACAM5 but selection against expression of Opa variants that bind to the neutrophil-restricted decoy receptor CEACAM3. This is the first study showing phenotypic selection for distinct CEACAM-binding phenotypes in vivo, and it supports the opposing functions of CEACAMs that facilitate infection versus driving inflammation within the genital tract.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Bacteriana/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Gonorrea/inmunología , Adhesinas Bacterianas/metabolismo , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Degranulación de la Célula/inmunología , Línea Celular , Cuello del Útero/microbiología , Femenino , Proteínas Ligadas a GPI/metabolismo , Gonorrea/microbiología , Humanos , Inflamación/inmunología , Masculino , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/aislamiento & purificación , Neutrófilos/inmunología , Unión Proteica , Isoformas de Proteínas/metabolismo , Uretra/microbiología
20.
PLoS Pathog ; 9(7): e1003509, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935487

RESUMEN

Neisseria meningitidis (Nme) asymptomatically colonizes the human nasopharynx, yet can initiate rapidly-progressing sepsis and meningitis in rare instances. Understanding the meningococcal lifestyle within the nasopharyngeal mucosa, a phase of infection that is prerequisite for disease, has been hampered by the lack of animal models. Herein, we compare mice expressing the four different human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) that can bind the neisserial Opa protein adhesins, and find that expression of human CEACAM1 is necessary and sufficient to establish intranasal colonization. During infection, in vivo selection for phase variants expressing CEACAM1-specific Opa proteins occurs, allowing mucosal attachment and entry into the subepithelial space. Consistent with an essential role for Opa proteins in this process, Opa-deficient meningococci were unable to colonize the CEACAM1-humanized mice. While simple Opa-mediated attachment triggered an innate response regardless of meningococcal viability within the inoculum, persistence of viable Opa-expressing bacteria within the CEACAM1-humanized mice was required for a protective memory response to be achieved. Parenteral immunization with a capsule-based conjugate vaccine led to the accumulation of protective levels of Nme-specific IgG within the nasal mucus, yet the sterilizing immunity afforded by natural colonization was instead conferred by Nme-specific IgA without detectable IgG. Considered together, this study establishes that the availability of CEACAM1 helps define the exquisite host specificity of this human-restricted pathogen, displays a striking example of in vivo selection for the expression of desirable Opa variants, and provides a novel model in which to consider meningococcal infection and immunity within the nasopharyngeal mucosa.


Asunto(s)
Adaptación Fisiológica , Infecciones Meningocócicas/microbiología , Nasofaringe/microbiología , Neisseria meningitidis/crecimiento & desarrollo , Mucosa Respiratoria/microbiología , Infecciones del Sistema Respiratorio/microbiología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Adhesión Bacteriana , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Escherichia coli/metabolismo , Células HeLa , Humanos , Inmunidad Mucosa , Infecciones Meningocócicas/inmunología , Infecciones Meningocócicas/metabolismo , Ratones , Ratones Transgénicos , Viabilidad Microbiana , Mutación , Nasofaringe/inmunología , Nasofaringe/metabolismo , Nasofaringe/patología , Neisseria meningitidis/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Neutrófilos/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA