Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 14(3): e0051023, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37052506

RESUMO

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.


Assuntos
Proteínas Hedgehog , Neocórtex , Gravidez , Feminino , Humanos , Proteínas Hedgehog/metabolismo , Neocórtex/metabolismo , Receptor 2 Toll-Like/metabolismo , Ligantes , Receptor 6 Toll-Like/metabolismo
2.
mBio ; 8(5)2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928207

RESUMO

Obesity is a risk factor for developing severe disease following influenza virus infection; however, the comorbidity of obesity and secondary bacterial infection, a serious complication of influenza virus infections, is unknown. To fill this gap in knowledge, lean and obese C57BL/6 mice were infected with a nonlethal dose of influenza virus followed by a nonlethal dose of Streptococcus pneumoniae Strikingly, not only did significantly enhanced death occur in obese coinfected mice compared to lean controls, but also high mortality was seen irrespective of influenza virus strain, bacterial strain, or timing of coinfection. This result was unexpected, given that most influenza virus strains, especially seasonal human A and B viruses, are nonlethal in this model. Both viral and bacterial titers were increased in the upper respiratory tract and lungs of obese animals as early as days 1 and 2 post-bacterial infection, leading to a significant decrease in lung function. This increased bacterial load correlated with extensive cellular damage and upregulation of platelet-activating factor receptor, a host receptor central to pneumococcal invasion. Importantly, while vaccination of obese mice against either influenza virus or pneumococcus failed to confer protection, antibiotic treatment was able to resolve secondary bacterial infection-associated mortality. Overall, secondary bacterial pneumonia could be a widespread, unaddressed public health problem in an increasingly obese population.IMPORTANCE Worldwide obesity rates have continued to increase. Obesity is associated with increased severity of influenza virus infection; however, very little is known about respiratory coinfections in this expanding, high-risk population. Our studies utilized a coinfection model to show that obesity increases mortality from secondary bacterial infection following influenza virus challenge through a "perfect storm" of host factors that lead to excessive viral and bacterial outgrowth. In addition, we found that vaccination of obese mice against either virus or bacteria failed to confer protection against coinfection, but antibiotic treatment did alleviate mortality. Combined, these results represent an understudied and imminent public health concern in a weighty portion of the global population.


Assuntos
Coinfecção/etiologia , Vírus da Influenza A/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Obesidade/complicações , Infecções por Orthomyxoviridae/complicações , Vacinas Pneumocócicas/administração & dosagem , Animais , Coinfecção/microbiologia , Coinfecção/virologia , Comorbidade , Vírus da Influenza A/crescimento & desenvolvimento , Pulmão/microbiologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/microbiologia , Obesidade/virologia , Infecções por Orthomyxoviridae/microbiologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/prevenção & controle , Infecções Pneumocócicas/virologia , Falha de Tratamento , Vacinação
3.
Microbiology (Reading) ; 163(8): 1198-1207, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28749326

RESUMO

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.

4.
mBio ; 8(1)2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28049146

RESUMO

The Gram-positive bacterial cell wall (CW) peptidoglycan-teichoic acid complex is released into the host environment during bacterial metabolism or death. It is a highly inflammatory Toll-like receptor 2 (TLR2) ligand, and previous in vivo studies have demonstrated its ability to recapitulate pathological features of pneumonia and meningitis. We report that an actin-dependent pathway is involved in the internalization of the CW by epithelial and endothelial cells, in addition to the previously described platelet-activating factor receptor (PAFr)-dependent uptake pathway. Unlike the PAFr-dependent pathway, which is mediated by clathrin and dynamin and does not lead to signaling, the alternative pathway is sensitive to 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and engenders Rac1, Cdc42, and phosphatidylinositol 3-kinase (PI3K) signaling. Upon internalization by this macropinocytosis-like pathway, CW is trafficked to lysosomes. Intracellular CW trafficking is more complex than previously recognized and suggests multiple points of interaction with and without innate immune signaling. IMPORTANCE: Streptococcus pneumoniae is a major human pathogen infecting the respiratory tract and brain. It is an established model organism for understanding how infection injures the host. During infection or bacterial growth, bacteria shed their cell wall (CW) into the host environment and trigger inflammation. A previous study has shown that CW enters and crosses cell barriers by interacting with a receptor on the surfaces of host cells, termed platelet-activating factor receptor (PAFr). In the present study, by using cells that are depleted of PAFr, we identified a second pathway with features of macropinocytosis, which is a receptor-independent fluid uptake mechanism by cells. Each pathway contributes approximately the same amount of cell wall trafficking, but the PAFr pathway is silent, while the new pathway appears to contribute to the host inflammatory response to CW insult.


Assuntos
Actinas/metabolismo , Parede Celular/metabolismo , Endocitose , Células Endoteliais/microbiologia , Células Epiteliais/microbiologia , Bactérias Gram-Positivas/fisiologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Cultivadas , Humanos , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 113(41): E6153-E6161, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27671632

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Resistência à Doença/imunologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Animais , Biomarcadores , Medula Óssea/patologia , Ciclo Celular/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Imunofenotipagem , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Knockout , Neutropenia/etiologia , Fagocitose/genética , Fagocitose/imunologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Análise de Sobrevida
7.
Nat Commun ; 7: 10859, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26924467

RESUMO

Bacterial pathogens produce complex carbohydrate capsules to protect against bactericidal immune molecules. Paradoxically, the pneumococcal capsule sensitizes the bacterium to antimicrobial peptides found on epithelial surfaces. Here we show that upon interaction with antimicrobial peptides, encapsulated pneumococci survive by removing capsule from the cell surface within minutes in a process dependent on the suicidal amidase autolysin LytA. In contrast to classical bacterial autolysis, during capsule shedding, LytA promotes bacterial survival and is dispersed circumferentially around the cell. However, both autolysis and capsule shedding depend on the cell wall hydrolytic activity of LytA. Capsule shedding drastically increases invasion of epithelial cells and is the main pathway by which pneumococci reduce surface bound capsule during early acute lung infection of mice. The previously unrecognized role of LytA in removing capsule to combat antimicrobial peptides may explain why nearly all clinical isolates of pneumococci conserve this enzyme despite the lethal selective pressure of antibiotics.


Assuntos
Cápsulas Bacterianas/fisiologia , Células Epiteliais/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Streptococcus pneumoniae/metabolismo , Animais , Aderência Bacteriana/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica/fisiologia , Camundongos , Mutação , N-Acetil-Muramil-L-Alanina Amidase/genética , Infecções Pneumocócicas/microbiologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia
8.
Cell Host Microbe ; 19(3): 388-99, 2016 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-26962947

RESUMO

Maternal infection during pregnancy is associated with adverse outcomes for the fetus, including postnatal cognitive disorders. However, the underlying mechanisms are obscure. We find that bacterial cell wall peptidoglycan (CW), a universal PAMP for TLR2, traverses the murine placenta into the developing fetal brain. In contrast to adults, CW-exposed fetal brains did not show any signs of inflammation or neuronal death. Instead, the neuronal transcription factor FoxG1 was induced, and neuroproliferation leading to a 50% greater density of neurons in the cortical plate was observed. Bacterial infection of pregnant dams, followed by antibiotic treatment, which releases CW, yielded the same result. Neuroproliferation required TLR2 and was recapitulated in vitro with fetal neuronal precursor cells and TLR2/6, but not TLR2/1, ligands. The fetal neuroproliferative response correlated with abnormal cognitive behavior in CW-exposed pups following birth. Thus, the bacterial CW-TLR2 signaling axis affects fetal neurodevelopment and may underlie postnatal cognitive disorders.


Assuntos
Infecções Bacterianas/complicações , Encéfalo/patologia , Proliferação de Células/efeitos dos fármacos , Transtornos Cognitivos/fisiopatologia , Troca Materno-Fetal , Neurônios/efeitos dos fármacos , Peptidoglicano/metabolismo , Animais , Comportamento Animal , Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Feminino , Camundongos , Neurônios/fisiologia , Gravidez , Receptor 2 Toll-Like/metabolismo
9.
Bio Protoc ; 6(23)2016 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-28573167

RESUMO

Cell wall is a complex biopolymer on the surface of all Gram-positive bacteria. During infection, cell wall is recognized by the innate immune receptor Toll-like receptor 2 causing intense inflammation and tissue damage. In animal models, cell wall traffics from the blood stream to many organs in the body, including brain, heart, placenta and fetus. This protocol describes how to prepare purified cell wall from Streptococcus pneumoniae, detect its distribution in animal tissues, and study the tissue response using the placenta and fetal brain as examples.

10.
PLoS Pathog ; 11(12): e1005337, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26659062

RESUMO

Necroptosis is a highly pro-inflammatory mode of cell death regulated by RIP (or RIPK)1 and RIP3 kinases and mediated by the effector MLKL. We report that diverse bacterial pathogens that produce a pore-forming toxin (PFT) induce necroptosis of macrophages and this can be blocked for protection against Serratia marcescens hemorrhagic pneumonia. Following challenge with S. marcescens, Staphylococcus aureus, Streptococcus pneumoniae, Listeria monocytogenes, uropathogenic Escherichia coli (UPEC), and purified recombinant pneumolysin, macrophages pretreated with inhibitors of RIP1, RIP3, and MLKL were protected against death. Alveolar macrophages in MLKL KO mice were also protected during S. marcescens pneumonia. Inhibition of caspases had no impact on macrophage death and caspase-1 and -3/7 were determined to be inactive following challenge despite the detection of IL-1ß in supernatants. Bone marrow-derived macrophages from RIP3 KO, but not caspase-1/11 KO or caspase-3 KO mice, were resistant to PFT-induced death. We explored the mechanisms for PFT-induced necroptosis and determined that loss of ion homeostasis at the plasma membrane, mitochondrial damage, ATP depletion, and the generation of reactive oxygen species were together responsible. Treatment of mice with necrostatin-5, an inhibitor of RIP1; GW806742X, an inhibitor of MLKL; and necrostatin-5 along with co-enzyme Q10 (N5/C10), which enhances ATP production; reduced the severity of S. marcescens pneumonia in a mouse intratracheal challenge model. N5/C10 protected alveolar macrophages, reduced bacterial burden, and lessened hemorrhage in the lungs. We conclude that necroptosis is the major cell death pathway evoked by PFTs in macrophages and the necroptosis pathway can be targeted for disease intervention.


Assuntos
Toxinas Bacterianas/toxicidade , Macrófagos Alveolares/microbiologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia , Proteínas Citotóxicas Formadoras de Poros/toxicidade , Animais , Apoptose/fisiologia , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Knockout , Necrose , Proteínas Quinases/metabolismo , RNA Interferente Pequeno , Espécies Reativas de Oxigênio
11.
Clin Vaccine Immunol ; 22(10): 1079-89, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26245351

RESUMO

Immunization with the pneumococcal proteins pneumolysin (Ply), choline binding protein A (CbpA), or pneumococcal surface protein A (PspA) elicits protective responses against invasive pneumococcal disease in animal models. In this study, we used different mouse models to test the efficacy of a variety of multivalent protein-based vaccines that comprised various combinations of full-length or peptide regions of the immunogens Ply, CbpA, or PspA: Ply toxoid with the L460D substitution (referred to herein as L460D); L460D fused with protective peptide epitopes from CbpA (YPT-L460D-NEEK [YLN]); L460D fused with the CD2 peptide containing the proline-rich region (PRR) of PspA (CD2-L460D); a combination of L460D and H70 (L460D+H70), a slightly larger PspA-derived peptide containing the PRR and the SM1 region; H70+YLN; and other combinations. Each mouse was immunized either intraperitoneally (i.p.) or subcutaneously (s.c.) with three doses (at 2-week intervals) of the various antigen combinations in alum adjuvant and then challenged in mouse models featuring different infection routes with multiple Streptococcus pneumoniae strains. In the i.p. infection sepsis model, H70+YLN consistently provided significant protection against three different challenge strains (serotypes 1, 2, and 6A); the CD2+YLN and H70+L460D combinations also elicited significant protection. Protection against intravenous (i.v.) sepsis (type 3 and 6A challenge strains) was largely dependent on PspA-derived antigen components, and the most protection was elicited by H70 with or without L460D or YLN. In a type 4 intratracheal (i.t.) challenge model that results in progression to meningitis, antigen combinations that contained YLN elicited the strongest protection. Thus, the trivalent antigen combination of H70+YLN elicited the strongest and broadest protection in diverse pneumococcal challenge models.


Assuntos
Proteínas de Bactérias/imunologia , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/imunologia , Sepse/prevenção & controle , Streptococcus pneumoniae/imunologia , Estreptolisinas/imunologia , Animais , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Epitopos/genética , Epitopos/imunologia , Esquemas de Imunização , Imunoglobulina G/sangue , Meningite Pneumocócica/imunologia , Meningite Pneumocócica/microbiologia , Meningite Pneumocócica/prevenção & controle , Camundongos Endogâmicos BALB C , Infecções Pneumocócicas/imunologia , Vacinas Pneumocócicas/administração & dosagem , Vacinas Pneumocócicas/genética , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/prevenção & controle , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Sepse/microbiologia , Streptococcus pneumoniae/classificação , Toxoides/imunologia , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
12.
PLoS Pathog ; 10(9): e1004383, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25232870

RESUMO

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.


Assuntos
Macaca/microbiologia , Miocárdio/patologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/patogenicidade , Adesinas Bacterianas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Feminino , Imunização , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miocárdio/imunologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Infecções Pneumocócicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Laminina/metabolismo , Estreptolisinas/metabolismo
13.
EMBO Mol Med ; 6(1): 141-54, 2014 01.
Artigo em Inglês | MEDLINE | ID: mdl-24408968

RESUMO

Acute otitis media (AOM) caused by Streptococcus pneumoniae remains one of the most common infectious diseases worldwide despite widespread vaccination. A major limitation of the currently licensed pneumococcal vaccines is the lack of efficacy against mucosal disease manifestations such as AOM, acute bacterial sinusitis and pneumonia. We sought to generate a novel class of live vaccines that (1) retain all major antigenic virulence proteins yet are fully attenuated and (2) protect against otitis media. A live vaccine candidate based on deletion of the signal recognition pathway component ftsY induced potent, serotype-independent protection against otitis media, sinusitis, pneumonia and invasive pneumococcal disease. Protection was maintained in animals coinfected with influenza virus, but was lost if mice were depleted of CD4(+) T cells at the time of vaccination. The live vaccine induced a strong serum IgG2a and IgG2b response that correlated with CD4(+) T-cell mediated class switching. Deletion of genes required for microbial adaptation to the host environment is a novel live attenuated vaccine strategy yielding the first experimental vaccine effective against pneumococcal otitis media.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Otite Média/prevenção & controle , Vacinas Pneumocócicas/imunologia , Doença Aguda , Animais , Linfócitos T CD4-Positivos/citologia , Chinchila , Modelos Animais de Doenças , Switching de Imunoglobulina , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Otite Média/mortalidade , Otite Média/patologia , Sorotipagem , Sinusite/microbiologia , Sinusite/mortalidade , Sinusite/prevenção & controle , Streptococcus pneumoniae/metabolismo , Taxa de Sobrevida , Vacinas Atenuadas/imunologia , Fatores de Virulência/imunologia
14.
J Infect Dis ; 209(7): 1116-25, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24041791

RESUMO

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.


Assuntos
Proteínas de Bactérias/imunologia , Portador Sadio/prevenção & controle , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/imunologia , Estreptolisinas/imunologia , Toxoides/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Proteção Cruzada , Modelos Animais de Doenças , Feminino , Haemophilus influenzae/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neisseria meningitidis/imunologia , Vacinas Pneumocócicas/administração & dosagem , Vacinas Pneumocócicas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Streptococcus pneumoniae/imunologia , Estreptolisinas/genética , Toxoides/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
15.
Blood ; 119(8): 1915-21, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22130804

RESUMO

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.


Assuntos
Anemia Falciforme/tratamento farmacológico , Selectina E/metabolismo , Hidroxiureia/uso terapêutico , Pneumonia Pneumocócica/prevenção & controle , Anemia Falciforme/complicações , Anemia Falciforme/metabolismo , Animais , Antidrepanocíticos/uso terapêutico , Criança , Modelos Animais de Doenças , Selectina E/sangue , Selectina E/genética , Feminino , Humanos , Imuno-Histoquímica , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Pneumonia Pneumocócica/complicações , Análise de Sobrevida
16.
Mol Microbiol ; 72(1): 12-25, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19226324

RESUMO

The ability of bacteria to sense and respond to both environmental and intracellular metal concentrations plays an important role in pathogenesis. The acquisition of manganese is vital for the virulence of several bacterial species. Although manganese uptake systems have been well studied in bacteria, no manganese efflux system has yet been identified. In this study we have identified a cation diffusion facilitator (CDF) protein (Sp1552) of unknown substrate specificity that functions as a manganese export system in Streptococcus pneumoniae. We designated the gene for this manganese efflux system mntE and found that the mutant strain was highly sensitive to manganese stress. Although the mutant was more resistant to oxidative stress and produced more H(2)O(2) and pili, it had reduced virulence in a murine model of infection, indicating that manganese export plays a role in host pathogenesis. There was a distinct differential transcriptional response to extracellular and intracellular manganese accumulation. Our study indicates that manganese efflux is required for invasive disease and may provide a useful antimicrobial target to devise future therapeutics.


Assuntos
Proteínas de Bactérias/metabolismo , Difusão Facilitada , Manganês/metabolismo , Streptococcus pneumoniae/metabolismo , Animais , Proteínas de Bactérias/genética , Fímbrias Bacterianas/metabolismo , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Homeostase , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , RNA Bacteriano/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidade , Virulência
17.
Mol Microbiol ; 70(2): 435-44, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18761687

RESUMO

In dynamic environments, intracellular homeostasis is maintained by transport systems found in all cells. While bacterial influx systems for essential trace cations are known to contribute to pathogenesis, efflux systems have been characterized mainly in contaminated environmental sites. We describe that the high calcium concentrations in the normal human host were toxic to pneumococci and that bacterial survival in vivo depended on CaxP, the first Ca2+ exporter reported in bacteria. CaxP homologues were found in the eukaryotic sacroplasmic reticulum and in many bacterial genomes. A caxP- mutant accumulated intracellular calcium, a state that was used to reveal signalling networks responsive to changes in intracellular calcium concentration. Chemical inhibition of CaxP was bacteriostatic in physiological calcium concentrations, suggesting a new antibiotic target uncovered under conditions in the eukaryotic host.


Assuntos
Proteínas de Bactérias/metabolismo , Cálcio/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Viabilidade Microbiana , Streptococcus pneumoniae/metabolismo , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Sangue/microbiologia , Contagem de Colônia Microbiana , Inibidores Enzimáticos/farmacologia , Feminino , Perfilação da Expressão Gênica , Humanos , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos BALB C , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Análise de Sobrevida , Virulência
18.
Infect Genet Evol ; 7(6): 708-16, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17768094

RESUMO

In the United States, Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and invasive bacterial disease. As antimicrobial resistance increases, it will become critical to determine if strains circulating in a population are likely to cause invasive pneumococcal disease (IPD). This is possible by comparison of an isolate's genotype to strains known to be invasive. In this work, we compared pulse-field gel electrophoresis (PFGE) and multilocus sequence typing (MLST), comparative genomic hybridization (CGH) and multi-invasive-locus sequence typing (MILST) for their ability to distinguish between known IPD causing and carrier strains using phylogenetic analyses. In addition, we assess the ability of these techniques to determine true clones from highly related strains. The resulting trees suggest that despite similar overall topologies, the clearest picture of invasiveness and genetic relatedness can be viewed when typing methods are used collectively.


Assuntos
Infecções Estreptocócicas/microbiologia , Streptococcus pneumoniae/classificação , Portador Sadio , Estudos de Coortes , Eletroforese em Gel de Campo Pulsado , Genes Bacterianos , Humanos , Testes de Sensibilidade Microbiana , Epidemiologia Molecular , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , Infecções Estreptocócicas/epidemiologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidade , Streptococcus pneumoniae/fisiologia
19.
J Infect Dis ; 195(4): 581-4, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17230418

RESUMO

Children with sickle cell disease have a 600-fold increased incidence of invasive pneumococcal disease. Platelet-activating factor receptor (PAFr) mediates pneumococcal invasion, and up-regulation of PAFr on chronically activated endothelia could contribute to increased bacterial invasion. Mice transplanted with sickle cell bone marrow developed more extensive infection, and 57% died, compared with 16% of wild-type mice. Histopathological analysis revealed that sickle cell mice expressed significantly more PAFr on endothelia and epithelia. Pharmacological blockade or genetic deletion of PAFr protected sickle cell mice from mortality. We conclude that PAFr plays an important role in hypersusceptibility to pneumococcal infection in sickle cell disease.


Assuntos
Anemia Falciforme/complicações , Anemia Falciforme/metabolismo , Suscetibilidade a Doenças , Glicoproteínas da Membrana de Plaquetas/fisiologia , Infecções Pneumocócicas/etiologia , Infecções Pneumocócicas/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Animais , Modelos Animais de Doenças , Células Endoteliais/química , Endotélio Vascular/química , Células Epiteliais/química , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/química , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteínas da Membrana de Plaquetas/análise , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/biossíntese , Pneumonia Pneumocócica/etiologia , Pneumonia Pneumocócica/metabolismo , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/biossíntese , Regulação para Cima
20.
Infect Immun ; 74(7): 3783-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16790750

RESUMO

Neuronal dysfunction can occur in the course of sepsis without meningitis. Sepsis-associated neuronal damage (SAND) was observed in the hippocampus within hours in experimental pneumococcal bacteremia. Intravascular challenge with purified bacterial cell wall recapitulated SAND. SAND persisted in PAFr(-/-) mice but was partially mitigated in mice lacking cell wall recognition proteins TLR2 and Nod2 and in mice overexpressing interleukin-10 (IL-10) in macrophages. Thus, cell wall drives SAND through IL-10-repressible inflammatory events. Treatment with CDP-choline ameliorated SAND, suggesting that it may be an effective adjunctive therapy to increase survival and reduce organ damage in sepsis.


Assuntos
Parede Celular/fisiologia , Neurônios/patologia , Infecções Pneumocócicas/metabolismo , Sepse/metabolismo , Streptococcus pneumoniae/patogenicidade , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infecções Pneumocócicas/genética , Sepse/genética , Sepse/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...