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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Infect Dis ; 75(1): e705-e714, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34891165

RESUMEN

BACKGROUND: Following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination there is significant variability between individuals in protective antibody levels against SARS-CoV-2, and within individuals against different virus variants. However, host demographic or clinical characteristics that predict variability in cross-reactive antibody levels are not well-described. These data could inform clinicians, researchers, and policymakers on the populations most likely to require vaccine booster shots. METHODS: In an institutional review board-approved prospective observational cohort study of staff at St. Jude Children's Research Hospital, we identified participants with plasma samples collected after SARS-CoV-2 infection, after mRNA vaccination, and after vaccination following infection, and quantitated immunoglobulin G (IgG) levels by enzyme-linked immunosorbent assay to the spike receptor binding domain (RBD) from 5 important SARS-CoV-2 variants (Wuhan Hu-1, B.1.1.7, B.1.351, P.1, and B.1.617.2). We used regression models to identify factors that contributed to cross-reactive IgG against 1 or multiple viral variants. RESULTS: Following infection, a minority of the cohort generated cross-reactive antibodies, IgG antibodies that bound all tested variants. Those who did had increased disease severity, poor metabolic health, and were of a particular ancestry. Vaccination increased the levels of cross-reactive IgG levels in all populations, including immunocompromised, elderly, and persons with poor metabolic health. Younger people with a healthy weight mounted the highest responses. CONCLUSIONS: Our findings provide important new information on individual antibody responses to infection/vaccination that could inform clinicians on populations that may require follow-on immunization.


Asunto(s)
COVID-19 , SARS-CoV-2 , Adulto , Anciano , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Humanos , Inmunoglobulina G , Persona de Mediana Edad , Estudios Prospectivos , Glicoproteína de la Espiga del Coronavirus , Vacunación
2.
Nat Immunol ; 9(12): 1399-406, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18978793

RESUMEN

Toll-like receptor (TLR) signaling in macrophages is required for antipathogen responses, including the biosynthesis of nitric oxide from arginine, and is essential for immunity to Mycobacterium tuberculosis, Toxoplasma gondii and other intracellular pathogens. Here we report a 'loophole' in the TLR pathway that is advantageous to these pathogens. Intracellular pathogens induced expression of the arginine hydrolytic enzyme arginase 1 (Arg1) in mouse macrophages through the TLR pathway. In contrast to diseases dominated by T helper type 2 responses in which Arg1 expression is greatly increased by interleukin 4 and 13 signaling through the transcription factor STAT6, TLR-mediated Arg1 induction was independent of the STAT6 pathway. Specific elimination of Arg1 in macrophages favored host survival during T. gondii infection and decreased lung bacterial load during tuberculosis infection.


Asunto(s)
Arginasa/inmunología , Infecciones Bacterianas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Receptores Toll-Like/inmunología , Animales , Arginasa/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/inmunología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Factor de Transcripción STAT6/inmunología , Factor de Transcripción STAT6/metabolismo , Receptores Toll-Like/metabolismo
3.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308088

RESUMEN

Acute otitis media is one of the most common childhood infections worldwide. Currently licensed vaccines against the common otopathogen Streptococcus pneumoniae target the bacterial capsular polysaccharide and confer no protection against nonencapsulated strains or capsular types outside vaccine coverage. Mucosal infections such as acute otitis media remain prevalent, even those caused by vaccine-covered serotypes. Here, we report that a protein-based vaccine, a fusion construct of epitopes of CbpA to pneumolysin toxoid, confers effective protection against pneumococcal acute otitis media for non-PCV-13 serotypes and enhances protection for PCV-13 serotypes when coadministered with PCV-13. Having cross-reactive epitopes, the fusion protein also induces potent antibody responses against nontypeable Haemophilus influenzae and S. pneumoniae, engendering protection against acute otitis media caused by emerging unencapsulated otopathogens. These data suggest that augmenting capsule-based vaccination with conserved, cross-reactive protein-based vaccines broadens and enhances protection against acute otitis media.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Infecciones por Haemophilus/prevención & control , Haemophilus influenzae/inmunología , Otitis Media/prevención & control , Vacunas Neumococicas/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Streptococcus pneumoniae/inmunología , Enfermedad Aguda , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Protección Cruzada , Reacciones Cruzadas , Femenino , Expresión Génica , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/patogenicidad , Humanos , Inmunogenicidad Vacunal , Ratones , Ratones Endogámicos BALB C , Otitis Media/inmunología , Otitis Media/microbiología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/biosíntesis , Estreptolisinas/genética , Toxoides/biosíntesis , Toxoides/genética , Vacunación , Vacunas Sintéticas
4.
Proc Natl Acad Sci U S A ; 113(41): E6153-E6161, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671632

RESUMEN

Infection is the single greatest threat to survival during cancer chemotherapy because of depletion of bone marrow-derived immune cells. Phagocytes, especially neutrophils, are key effectors in immunity to extracellular pathogens, which has limited the development of new approaches to protect patients with cancer and chemotherapy-induced neutropenia. Using a model of vaccine-induced protection against lethal Pseudomonas aeruginosa pneumonia in the setting of chemotherapy-induced neutropenia, we found a population of resident lung macrophages in the immunized lung that mediated protection in the absence of neutrophils, bone marrow-derived monocytes, or antibodies. These vaccine-induced macrophages (ViMs) expanded after immunization, locally proliferated, and were closely related to alveolar macrophages (AMs) by surface phenotype and gene expression profiles. By contrast to AMs, numbers of ViMs were stable through chemotherapy, showed enhanced phagocytic activity, and prolonged survival of neutropenic mice from lethal P. aeruginosa pneumonia upon intratracheal adoptive transfer. Thus, induction of ViMs by tissue macrophage remodeling may become a framework for new strategies to activate immune-mediated reserves against infection in immunocompromised hosts.


Asunto(s)
Antineoplásicos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Resistencia a la Enfermedad/inmunología , Interacciones Huésped-Patógeno/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Animales , Biomarcadores , Médula Ósea/patología , Ciclo Celular/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Inmunofenotipificación , Macrófagos Alveolares/metabolismo , Ratones , Ratones Noqueados , Neutropenia/etiología , Fagocitosis/genética , Fagocitosis/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas/inmunología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/inmunología , Análisis de Supervivencia
5.
Clin Infect Dis ; 67(4): 541-548, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29518185

RESUMEN

Background: Myelosuppression-related infections remain important causes of morbidity and mortality in children with acute lymphoblastic leukemia (ALL). Methods: By analyzing fecal samples collected at diagnosis and after each of the initial 3 phases of chemotherapy, we evaluated the role of gut microbiota in predicting infections in 199 children with newly diagnosed ALL. The bacterial 16S rRNA gene was analyzed by high-depth sequencing to determine the diversity and composition of the microbiome. Results: After the induction and reinduction I phases of chemotherapy, microbial diversity decreased significantly relative to the prechemotherapy value. After chemotherapy, the relative abundance of certain bacterial taxa (eg, Bacteroidetes) decreased significantly, whereas that of other taxa (eg, Clostridiaceae and Streptococcaceae) increased. A baseline gut microbiome characterized by Proteobacteria predicted febrile neutropenia. Adjusting for the chemotherapy phase and ALL risk level, Enterococcaceae dominance (relative abundance ≥30%) predicted significantly greater risk of subsequent febrile neutropenia and diarrheal illness, whereas Streptococcaceae dominance predicted significantly greater risk of subsequent diarrheal illness. Conclusions: In children undergoing therapy for newly diagnosed ALL, the relative abundance of Proteobacteria before chemotherapy initiation predicts development of febrile neutropenia, and domination of the gut microbiota by Enterococcaceae or Streptococcaceae at any time during chemotherapy predicts infection in subsequent phases of chemotherapy. Clinical Trial Registration: NCT00549848.


Asunto(s)
Antineoplásicos/efectos adversos , Bacterias/efectos de los fármacos , Infecciones Bacterianas/complicaciones , Microbioma Gastrointestinal/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Adolescente , Antineoplásicos/uso terapéutico , Bacterias/clasificación , Niño , Preescolar , Heces/microbiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Valor Predictivo de las Pruebas , ARN Ribosómico 16S/genética , Factores de Riesgo , Análisis de Secuencia de ADN
6.
J Immunol ; 196(9): 3677-85, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27029587

RESUMEN

Intact, inactivated Streptococcus pneumoniae [including the unencapsulated S. pneumoniae, serotype 2 strain (R36A)] markedly inhibits the humoral immune response to coimmunized heterologous proteins, a property not observed with several other intact Gram-positive or Gram-negative bacteria. In this study, we determined the nature of this immunosuppressive property. Because phosphorylcholine (PC), a major haptenic component of teichoic acid in the S. pneumoniae cell wall, and lipoteichoic acid in the S. pneumoniae membrane were previously reported to be immunosuppressive when derived from filarial parasites, we determined whether R36A lacking PC (R36A(pc-)) was inhibitory. Indeed, although R36A(pc-) exhibited a markedly reduced level of inhibition of the IgG response to coimmunized chicken OVA (cOVA), no inhibition was observed when using several other distinct PC-expressing bacteria or a soluble, protein-PC conjugate. Further, treatment of R36A with periodate, which selectively destroys PC residues, had no effect on R36A-mediated inhibition. Because R36A(pc-) also lacks choline-binding proteins (CBPs) that require PC for cell wall attachment, and because treatment of R36A with trypsin eliminated its inhibitory activity, we incubated R36A in choline chloride, which selectively strips CBPs from its surface. R36A lacking CBPs lost most of its inhibitory property, whereas the supernatant of choline chloride-treated R36A, containing CBPs, was markedly inhibitory. Coimmunization studies using cOVA and various S. pneumoniae mutants, each genetically deficient in one of the CBPs, demonstrated that only S. pneumoniae lacking the CBP pneumococcal surface protein A lost its ability to inhibit the IgG anti-cOVA response. These results strongly suggest that PspA plays a major role in mediating the immunosuppressive property of S. pneumoniae.


Asunto(s)
Proteínas Bacterianas/inmunología , Tolerancia Inmunológica , Fosforilcolina/inmunología , Streptococcus pneumoniae/inmunología , Animales , Proteínas Bacterianas/genética , Inmunización , Inmunoglobulina G/inmunología , Inmunosupresores , Ratones , Mutación , Ovalbúmina/inmunología , Ácido Peryódico/farmacología , Fosforilcolina/metabolismo , Streptococcus pneumoniae/química , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Tripsina/metabolismo
7.
Microbiology (Reading) ; 163(8): 1198-1207, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28749326

RESUMEN

Neuraminidase A (NanA) is an important virulence factor that is anchored to the pneumococcal cell wall and cleaves sialic acid on host substrates. We noted that a secreted allele of NanA was over-represented in invasive pneumococcal isolates and promoted the development of meningitis when swapped into the genome of non-meningitis isolates replacing cell wall-anchored NanA. Both forms of recombinant NanA directly activated transforming growth factor (TGF)-ß, increased SMAD signalling and promoted loss of endothelial tight junction ZO-1. However, in assays using whole bacteria, only the cell-bound NanA decreased expression of ZO-1 and showed NanA dependence of bacterial invasion of endothelial cells. We conclude that NanA secretion versus retention on the cell surface does not influence neurotropism of clinical isolates. However, we describe a new NanA-TGF-ß signalling axis that leads to decreased blood-brain barrier integrity and enhances bacterial invasion.

8.
J Infect Dis ; 213(3): 407-10, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26068783

RESUMEN

BACKGROUND: The relationship between influenza virus infectivity and virus shedding, based on different diagnostic methods, has not been defined. METHODS: Three donor ferrets infected with 2009 pandemic influenza A(H1N1) underwent daily quantitative culture, antigen-detection testing, and real-time reverse transcription-polymerase chain reaction (RT-PCR). Eight contacts were sequentially cohoused with each of the donors for 24 hours during days 3-10 after inoculation. RESULTS: Transmission was observed until day 5 after inoculation, corresponding to high culture titers and positive results of antigen-detection tests. Real-time RT-PCR showed no relation to the cessation of transmission. CONCLUSIONS: Antigen-detection testing and virus culture but not real-time RT-PCR identified the end of the infectious period.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/virología , Esparcimiento de Virus/fisiología , Animales , Antígenos Virales , Hurones , Infecciones por Orthomyxoviridae/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sensibilidad y Especificidad , Cultivo de Virus
9.
PLoS Pathog ; 10(9): e1004383, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25232870

RESUMEN

Hospitalization of the elderly for invasive pneumococcal disease is frequently accompanied by the occurrence of an adverse cardiac event; these are primarily new or worsened heart failure and cardiac arrhythmia. Herein, we describe previously unrecognized microscopic lesions (microlesions) formed within the myocardium of mice, rhesus macaques, and humans during bacteremic Streptococcus pneumoniae infection. In mice, invasive pneumococcal disease (IPD) severity correlated with levels of serum troponin, a marker for cardiac damage, the development of aberrant cardiac electrophysiology, and the number and size of cardiac microlesions. Microlesions were prominent in the ventricles, vacuolar in appearance with extracellular pneumococci, and remarkable due to the absence of infiltrating immune cells. The pore-forming toxin pneumolysin was required for microlesion formation but Interleukin-1ß was not detected at the microlesion site ruling out pneumolysin-mediated pyroptosis as a cause of cell death. Antibiotic treatment resulted in maturing of the lesions over one week with robust immune cell infiltration and collagen deposition suggestive of long-term cardiac scarring. Bacterial translocation into the heart tissue required the pneumococcal adhesin CbpA and the host ligands Laminin receptor (LR) and Platelet-activating factor receptor. Immunization of mice with a fusion construct of CbpA or the LR binding domain of CbpA with the pneumolysin toxoid L460D protected against microlesion formation. We conclude that microlesion formation may contribute to the acute and long-term adverse cardiac events seen in humans with IPD.


Asunto(s)
Macaca/microbiología , Miocardio/patología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Adhesinas Bacterianas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Femenino , Inmunización , Interleucina-1beta/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocardio/inmunología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Infecciones Neumocócicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Laminina/metabolismo , Estreptolisinas/metabolismo
10.
Acta Neuropathol ; 131(2): 185-209, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26744349

RESUMEN

Bacterial meningitis is a devastating disease occurring worldwide with up to half of the survivors left with permanent neurological sequelae. Due to intrinsic properties of the meningeal pathogens and the host responses they induce, infection can cause relatively specific lesions and clinical syndromes that result from interference with the function of the affected nervous system tissue. Pathogenesis is based on complex host-pathogen interactions, some of which are specific for certain bacteria, whereas others are shared among different pathogens. In this review, we summarize the recent progress made in understanding the molecular and cellular events involved in these interactions. We focus on selected major pathogens, Streptococcus pneumonia, S. agalactiae (Group B Streptococcus), Neisseria meningitidis, and Escherichia coli K1, and also include a neglected zoonotic pathogen, Streptococcus suis. These neuroinvasive pathogens represent common themes of host-pathogen interactions, such as colonization and invasion of mucosal barriers, survival in the blood stream, entry into the central nervous system by translocation of the blood-brain and blood-cerebrospinal fluid barrier, and induction of meningeal inflammation, affecting pia mater, the arachnoid and subarachnoid spaces.


Asunto(s)
Encéfalo/inmunología , Interacciones Huésped-Patógeno/inmunología , Meningitis Bacterianas/fisiopatología , Animales , Encéfalo/microbiología , Humanos
11.
PLoS Pathog ; 9(11): e1003727, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244159

RESUMEN

Streptococcus pneumoniae infection is a leading cause of bacterial pneumonia, sepsis and meningitis and is associated with high morbidity and mortality. Type I interferon (IFN-I), whose contribution to antiviral and intracellular bacterial immunity is well established, is also elicited during pneumococcal infection, yet its functional significance is not well defined. Here, we show that IFN-I plays an important role in the host defense against pneumococci by counteracting the transmigration of bacteria from the lung to the blood. Mice that lack the type I interferon receptor (Ifnar1 (-/-)) or mice that were treated with a neutralizing antibody against the type I interferon receptor, exhibited enhanced development of bacteremia following intranasal pneumococcal infection, while maintaining comparable bacterial numbers in the lung. In turn, treatment of mice with IFNß or IFN-I-inducing synthetic double stranded RNA (poly(I:C)), dramatically reduced the development of bacteremia following intranasal infection with S. pneumoniae. IFNß treatment led to upregulation of tight junction proteins and downregulation of the pneumococcal uptake receptor, platelet activating factor receptor (PAF receptor). In accordance with these findings, IFN-I reduced pneumococcal cell invasion and transmigration across epithelial and endothelial layers, and Ifnar1 (-/-) mice showed overall enhanced lung permeability. As such, our data identify IFN-I as an important component of the host immune defense that regulates two possible mechanisms involved in pneumococcal invasion, i.e. PAF receptor-mediated transcytosis and tight junction-dependent pericellular migration, ultimately limiting progression from a site-restricted lung infection to invasive, lethal disease.


Asunto(s)
Barrera Alveolocapilar/metabolismo , Barrera Hematoencefálica/metabolismo , Interferón Tipo I/metabolismo , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Bacteriemia/genética , Bacteriemia/metabolismo , Barrera Alveolocapilar/microbiología , Barrera Alveolocapilar/patología , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Neumonía Neumocócica/genética , Neumonía Neumocócica/patología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Streptococcus pneumoniae/metabolismo
12.
J Infect Dis ; 209(7): 1116-25, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24041791

RESUMEN

BACKGROUND: Pneumococcus, meningococcus, and Haemophilus influenzae cause a similar spectrum of infections in the ear, lung, blood, and brain. They share cross-reactive antigens that bind to the laminin receptor of the blood-brain barrier as a molecular basis for neurotropism, and this step in pathogenesis was addressed in vaccine design. METHODS: Biologically active peptides derived from choline-binding protein A (CbpA) of pneumococcus were identified and then genetically fused to L460D pneumolysoid. The fusion construct was tested for vaccine efficacy in mouse models of nasopharyngeal carriage, otitis media, pneumonia, sepsis, and meningitis. RESULTS: The CbpA peptide-L460D pneumolysoid fusion protein was more broadly immunogenic than pneumolysoid alone, and antibodies were active in vitro against Streptococcus pneumoniae, Neisseria meningitidis, and H. influenzae. Passive and active immunization protected mice from pneumococcal carriage, otitis media, pneumonia, bacteremia, meningitis, and meningococcal sepsis. CONCLUSIONS: The CbpA peptide-L460D pneumolysoid fusion protein was broadly protective against pneumococcal infection, with the potential for additional protection against other meningeal pathogens.


Asunto(s)
Proteínas Bacterianas/inmunología , Portador Sano/prevención & control , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Estreptolisinas/inmunología , Toxoides/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/genética , Protección Cruzada , Modelos Animales de Enfermedad , Femenino , Haemophilus influenzae/inmunología , Ratones , Ratones Endogámicos BALB C , Neisseria meningitidis/inmunología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Streptococcus pneumoniae/inmunología , Estreptolisinas/genética , Toxoides/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
13.
PLoS Pathog ; 8(7): e1002788, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22807675

RESUMEN

Small noncoding RNAs (sRNAs) play important roles in gene regulation in both prokaryotes and eukaryotes. Thus far, no sRNA has been assigned a definitive role in virulence in the major human pathogen Streptococcus pneumoniae. Based on the potential coding capacity of intergenic regions, we hypothesized that the pneumococcus produces many sRNAs and that they would play an important role in pathogenesis. We describe the application of whole-genome transcriptional sequencing to systematically identify the sRNAs of Streptococcus pneumoniae. Using this approach, we have identified 89 putative sRNAs, 56 of which are newly identified. Furthermore, using targeted genetic approaches and Tn-seq transposon screening, we demonstrate that many of the identified sRNAs have important global and niche-specific roles in virulence. These data constitute the most comprehensive analysis of pneumococcal sRNAs and provide the first evidence of the extensive roles of sRNAs in pneumococcal pathogenesis.


Asunto(s)
Infecciones Neumocócicas/microbiología , ARN Bacteriano/genética , ARN Pequeño no Traducido/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Animales , Secuencia de Bases , Regulación Bacteriana de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Conformación de Ácido Nucleico , Infecciones Neumocócicas/patología , ARN Bacteriano/química , ARN Bacteriano/metabolismo , ARN Pequeño no Traducido/química , ARN Pequeño no Traducido/metabolismo , Análisis de Secuencia de ARN
14.
Blood ; 119(8): 1915-21, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22130804

RESUMEN

Sickle cell anemia is characterized by chronic hemolysis coupled with extensive vascular inflammation. This inflammatory state also mechanistically promotes a high risk of lethal, invasive pneumococcal infection. Current treatments to reduce vaso-occlusive complications include chronic hydroxyurea therapy to induce fetal hemoglobin. Because hydroxyurea also reduces leukocytosis, an understanding of the impact of this treatment on pneumococcal pathogenesis is needed. Using a sickle cell mouse model of pneumococcal pneumonia and sepsis, administration of hydroxyurea was found to significantly improve survival. Hydroxyurea treatment decreased neutrophil extravasation into the infected lung coincident with significantly reduced levels of E-selectin in serum and on pulmonary epithelia. The protective effect of hydroxyurea was abrogated in mice deficient in E-selectin. The decrease in E-selectin levels was also evident in human sickle cell patients receiving hydroxyurea therapy. These data indicate that in addition to induction of fetal hemoglobin, hydroxyurea attenuates leukocyte-endothelial interactions in sickle cell anemia, resulting in protection against lethal pneumococcal sepsis.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Selectina E/metabolismo , Hidroxiurea/uso terapéutico , Neumonía Neumocócica/prevención & control , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/metabolismo , Animales , Antidrepanocíticos/uso terapéutico , Niño , Modelos Animales de Enfermedad , Selectina E/sangre , Selectina E/genética , Femenino , Humanos , Inmunohistoquímica , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Neumonía Neumocócica/complicaciones , Análisis de Supervivencia
15.
Cell Chem Biol ; 31(2): 195-206, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38052216

RESUMEN

Streptococcus pneumoniae is a remarkably adaptable and successful human pathogen, playing dual roles of both asymptomatic carriage in the nasopharynx and invasive disease including pneumonia, bacteremia, and meningitis. Efficacious vaccines and effective antibiotic therapies are critical to mitigating morbidity and mortality. However, clinical interventions can be rapidly circumvented by the pneumococcus by its inherent proclivity for genetic exchange. This leads to an underappreciated interplay between vaccine and antibiotic pressures on pneumococcal populations. Circulating populations have undergone dramatic shifts due to the introduction of capsule-based vaccines of increasing valency imparting strong selective pressures. These alterations in population structure have concurrent consequences on the frequency of antibiotic resistance profiles in the population. This review will discuss the interactions of these two selective forces. Understanding and forecasting the drivers of antibiotic resistance and capsule switching are of critical importance for public health, particularly for such a genetically promiscuous pathogen as S. pneumoniae.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Humanos , Streptococcus pneumoniae/genética , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/prevención & control , Infecciones Neumocócicas/epidemiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Vacunas Neumococicas , Vacunación , Vacunas Conjugadas
16.
Microbiology (Reading) ; 159(Pt 11): 2333-2341, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23963302

RESUMEN

Systemic release of norepinephrine (NE) is a component of the acute host response to infection, and studies in the field of microbial endocrinology indicate generally that NE increases the bacterial growth rate and promotes invasive disease. However, NE attenuates experimental invasive pneumococcal disease. We determined that NE promoted pneumococcal growth but paradoxically decreased pneumococcal adhesion to host cells. This effect was independent of the classical adhesin CbpA. Microarray analysis indicated that the effect of NE involved two two-component regulatory systems that both regulate expression of the Piu iron uptake ABC transport operon. We propose that NE, a known siderophore, enhances iron availability to the bacteria, resulting in greater bacterial replication and decreased expression of Piu operon products. Downregulation of the operon includes decreased expression of the Piu-associated adhesin PiuD. Our results suggested that the iron-dependent inhibitory effect of NE on pneumococcal adherence is a mechanism underlying the amelioration of pneumococcal disease by NE.


Asunto(s)
Adhesión Bacteriana/efectos de los fármacos , Células Epiteliales/microbiología , Hierro/metabolismo , Norepinefrina/metabolismo , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/fisiología , Perfilación de la Expresión Génica , Análisis por Micromatrices , Transducción de Señal/efectos de los fármacos , Streptococcus pneumoniae/crecimiento & desarrollo
17.
Cell Host Microbe ; 31(5): 681-682, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37167946

RESUMEN

During meningitis, sensory neurons detect bacterial toxins and metabolites. Early activation of pain receptors suppresses host defense in the meninges while, later, taste receptors amplify inflammation in the spinal cord. Is a neuroimmune axis a clue to new treatments?


Asunto(s)
Meningitis Bacterianas , Gusto , Humanos , Meningitis Bacterianas/metabolismo , Meninges , Dolor/metabolismo , Médula Espinal/metabolismo , Células Receptoras Sensoriales/metabolismo
18.
mBio ; 14(3): e0051023, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37052506

RESUMEN

Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.


Asunto(s)
Proteínas Hedgehog , Neocórtex , Embarazo , Femenino , Humanos , Proteínas Hedgehog/metabolismo , Neocórtex/metabolismo , Receptor Toll-Like 2/metabolismo , Ligandos , Receptor Toll-Like 6/metabolismo
19.
PLoS Pathog ; 6(12): e1001240, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21203486

RESUMEN

Disruption of p53/Puma-mediated apoptosis protects against lethality due to DNA damage. Here we demonstrate the unexpected requirement of the pro-apoptotic p53-target gene Puma to mount a successful innate immune response to bacterial sepsis. Puma⁻/⁻ mice rapidly died when challenged with bacteria. While the immune response in Puma⁻/⁻ mice was unchanged in cell migration, phagocytosis and bacterial killing, sites of infection accumulated large abscesses and sepsis was progressive. Blocking p53/Puma-induced apoptosis during infection caused resistance to ROS-induced cell death in the CD49d+ neutrophil subpopulation, resulting in insufficient immune resolution. This study identifies a biological role for p53/Puma apoptosis in optimizing neutrophil lifespan so as to ensure the proper clearance of bacteria and exposes a counter-balance between the innate immune response to infection and survival from DNA damage.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Genes p53 , Neutrófilos/inmunología , Sepsis/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Infecciones Bacterianas/inmunología , Supervivencia Celular/inmunología , Inmunidad Innata , Ratones , Ratones Noqueados , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
20.
J Infect Dis ; 204(9): 1358-66, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21900488

RESUMEN

Host inflammatory responses contribute to the significant immunopathology that occurs during treatment of secondary bacterial pneumonia following influenza. We undertook the present study to determine the mechanisms underlying disparate outcomes in a mouse model with ß-lactam and macrolide antibiotics. Lysis of superinfecting bacteria by ampicillin caused an extensive influx of neutrophils into the lungs resulting in a consolidative pneumonia, necrotic lung damage, and significant mortality. This was mediated through Toll-like receptor (TLR) 2 and was independent of TLR4 and the Streptococcus pneumoniae cytotoxin pneumolysin. Treatment with azithromycin prevented neutrophil accumulation and rescued mice from subsequent mortality. This effect was independent of the antibacterial activity of this macrolide since dual therapy with ampicillin and azithromycin against an azithromycin-resistant strain also was able to cure secondary pneumonia. These data suggest that strategies for eliminating bacteria without lysis coupled with immunomodulation of inflammation should be pursued clinically.


Asunto(s)
Antibacterianos/administración & dosificación , Factores Inmunológicos/administración & dosificación , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/patología , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/inmunología , Animales , Femenino , Pulmón/inmunología , Pulmón/patología , Macrólidos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neumonía Neumocócica/tratamiento farmacológico , Neumonía Neumocócica/mortalidad , Streptococcus pneumoniae/patogenicidad , Resultado del Tratamiento , beta-Lactamas/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA