Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
Open Forum Infect Dis ; 8(9): ofab420, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34557558

RESUMEN

The efficacy of coronavirus disease 2019 (COVID-19) vaccines administered after COVID-19-specific monoclonal antibody is unknown, and "antibody interference" might hinder immune responses leading to vaccine failure. In an institutional review board-approved prospective study, we found that an individual who received mRNA COVID-19 vaccination <40 days after COVID-19-specific monoclonal antibody therapy for symptomatic COVID-19 had similar postvaccine antibody responses to SARS-CoV-2 receptor binding domain (RBD) for 4 important SARS-CoV-2 variants (B.1, B.1.1.7, B.1.351, and P.1) as other participants who were also vaccinated following COVID-19. Vaccination against COVID-19 shortly after COVID-19-specific monoclonal antibody can boost and expand antibody protection, questioning the need to delay vaccination in this setting. TRIAL REGISTRATION: The St. Jude Tracking of Viral and Host Factors Associated with COVID-19 study; NCT04362995; https://clinicaltrials.gov/ct2/show/NCT04362995.

6.
Lancet Microbe ; 2(4): e159-e167, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-34355208

RESUMEN

BACKGROUND: Although antibiotic prophylaxis with levofloxacin can reduce the risk of serious infection in immunocompromised patients, the potential contribution of prophylaxis to antibiotic resistance is a major drawback. We aimed to identify the effects of levofloxacin prophylaxis, given to paediatric patients with acute lymphoblastic leukaemia to prevent infections during induction chemotherapy, on antibiotic resistance in gastrointestinal microbiota after completion of induction and consolidation therapy. METHODS: This prospective, single-centre (St Jude Children's Research Hospital, Memphis, TN, USA) cohort study included children (≤18 years) receiving therapy for newly diagnosed acute lymphoblastic leukaemia and who received either primary levofloxacin prophylaxis or no antibacterial prophylaxis (aside from Pneumocystis jirovecii prophylaxis with trimethoprim-sulfamethoxazole) and provided at least two stool samples, including one after completion of induction therapy. We used metagenomic sequencing to identify bacterial genes that confer resistance to fluoroquinolones, trimethoprim-sulfamethoxazole, or other antibiotics, and to identify point mutations in bacterial topoisomerases (gyrA, parC) that confer resistance to fluoroquinolones. We then used generalised linear mixed models to compare the prevalence and relative abundance of antibiotic resistance gene groups after completion of induction and consolidation therapy between participants who had received levofloxacin and those who received no prophylaxis. FINDINGS: Between Feb 1, 2012, and April 30, 2016, 118 stool samples (32 baseline, 49 after induction, and 37 after consolidation) were collected from 49 evaluable participants; of these participants, 31 (63%) received levofloxacin prophylaxis during induction therapy and 18 (37%) received no antibacterial prophylaxis. Over the course of induction therapy, there was an overall increase in the relative abundance of trimethoprim-sulfamethoxazole resistance genes (estimated mean fold change 5·9, 95% CI 3·6-9·6; p<0·0001), which was not modified by levofloxacin prophylaxis (p=0·46). By contrast, the prevalence of topoisomerase point mutations increased over the course of induction therapy in levofloxacin recipients (mean prevalence 10·4% [95% CI 3·2-25·4] after induction therapy vs 3·7% [0·2-22·5] at baseline) but not other participants (0% vs 0%; p<0·0001). There was no significant difference between prophylaxis groups with respect to changes in aminoglycoside, ß-lactam, vancomycin, or multidrug resistance genes after completion of induction or consolidation therapy. INTERPRETATION: Analysing the gastrointestinal resistome can provide insights into the effects of antibiotics on the risk of antibiotic-resistant infections. In this study, antibiotic prophylaxis with trimethoprim-sulfamethoxazole or levofloxacin during induction therapy for acute lymphoblastic leukaemia appeared to increase the short-term and medium-term risk of colonisation with bacteria resistant to these antibiotics, but not to other drugs. More research is needed to determine the longer-term effects of antibacterial prophylaxis on colonisation with antibiotic-resistant bacteria. FUNDING: Children's Infection Defense Center at St Jude Children's Research Hospital, American Lebanese Syrian Associated Charities, and National Institutes of Health.


Asunto(s)
Profilaxis Antibiótica , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antibacterianos/uso terapéutico , Niño , Estudios de Cohortes , Fluoroquinolonas/uso terapéutico , Humanos , Levofloxacino/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Estudios Prospectivos , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico
7.
Vaccines (Basel) ; 9(3)2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33801372

RESUMEN

Streptococcus pneumoniae (Spn) is a bacterial pathogen known to colonize the upper respiratory tract and cause serious opportunistic diseases such as pneumonia, bacteremia, sepsis and meningitis. As a consequence, millions of attributable deaths occur annually, especially among infants, the elderly and immunocompromised individuals. Although current vaccines, composed of purified pneumococcal polysaccharide in free form or conjugated to a protein carrier, are widely used and have been demonstrated to be effective in target groups, Spn has continued to colonize and cause life-threatening disease in susceptible populations. This lack of broad protection highlights the necessity of improving upon the current "gold standard" pneumococcal vaccines to increase protection both by decreasing colonization and reducing the incidence of sterile-site infections. Over the past century, most of the pneumococcal proteins that play an essential role in colonization and pathogenesis have been identified and characterized. Some of these proteins have the potential to serve as antigens in a multi-valent protein vaccine that confers capsule independent protection. This review seeks to summarize the benefits and limitations of the currently employed vaccine strategies, describes how leading candidate proteins contribute to pneumococcal disease development, and discusses the potential of these proteins as protective antigens-including as a hybrid construct.

9.
Front Cell Infect Microbiol ; 10: 588378, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194834

RESUMEN

During acute bacterial meningitis, recognition of the bacterial envelope by immune cells of the central nervous system (CNS) generates a robust response that is essential to clear bacteria. This response is further amplified during treatment when lytic antibiotics, required for cure, also generate a burst of highly inflammatory cell envelope debris. Different peptidoglycan (PG) subcomponents interact with neurons, glia, and the blood brain barrier resulting in the entire symptom complex of meningitis. Recently, this CNS-cell envelope signaling axis has been extended to non-inflammatory recognition of cell wall components circulating from endogenous bacteria to the brain resulting in both benefit and chronic damage. This review will describe the molecular details of a broad array of cell envelope-induced responses in the CNS and what current strategies can be implemented to improve clinical outcome.


Asunto(s)
Pared Celular , Peptidoglicano , Bacterias/metabolismo , Barrera Hematoencefálica , Membrana Celular/metabolismo , Pared Celular/metabolismo , Peptidoglicano/metabolismo
10.
Front Cell Infect Microbiol ; 10: 585791, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178633

RESUMEN

Highly successful invasive pathogens exploit host vulnerabilities by adapting tools to co-opt highly conserved host features. This is especially true when pathogens develop ligands to hijack trafficking routes or signaling patterns of host receptors. In this context, highly successful pathogens can be grouped together by the patterns of organs infected and diseases they cause. In the case of this perspective, the focus is on the historically most successful invasive bacterial pathogens of children that cause pneumonia, sepsis and meningitis: Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitidis. This triad shares a ligand to bind to PAF receptor to enter host cells despite early defenses by innate immunity. All three also target laminin receptor to cross endothelial barriers using a common set of molecular tools that may prove to be a design for a cross-protective vaccine.


Asunto(s)
Meningitis Bacterianas , Meningitis , Neisseria meningitidis , Niño , Haemophilus influenzae , Humanos , Lactante , Pediatras , Streptococcus pneumoniae
11.
Front Microbiol ; 11: 1543, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714314

RESUMEN

Streptococcus pneumoniae is an opportunistic pathogen responsible for widespread illness and is a major global health issue for children, the elderly, and the immunocompromised population. Pneumolysin (PLY) is a cholesterol-dependent cytolysin (CDC) and key pneumococcal virulence factor involved in all phases of pneumococcal disease, including transmission, colonization, and infection. In this review we cover the biology and cytolytic function of PLY, its contribution to S. pneumoniae pathogenesis, and its known interactions and effects on the host with regard to tissue damage and immune response. Additionally, we review statins as a therapeutic option for CDC toxicity and PLY toxoid as a vaccine candidate in protein-based vaccines.

12.
Cell Host Microbe ; 27(4): 489-490, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32272070

RESUMEN

Horizontal gene transfer (HGT) facilitates spead of antibiotic resistance elements. In this issue of Cell Host & Microbe, Domenech et al. discover that compounds disrupting proton motive force block natural competence (COM) and interrupt intraspecies HGT and exchange of antibiotic resistance. Such strategies might minimize clinical spread of antibiotic resistance.


Asunto(s)
Transferencia de Gen Horizontal , Fuerza Protón-Motriz , Bacterias/genética , Farmacorresistencia Microbiana , Plásmidos
13.
Pathogens ; 8(4)2019 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-31652741

RESUMEN

Pneumococcal vaccine development is driven by the achievement of high activity in a single gatekeeper assay: the bacterial opsonophagocytic killing (OPK) assay. New evidence challenges the dogma that anti-capsular antibodies have only a single function that predicts success. The emerging concept of multi-modal protection presents an array of questions that are fundamental to adopting a new vaccine design process. If antibodies have hidden non-opsonic functions that are protective, should these be optimized for better vaccines? What would protein antigens add to protective activity? Are cellular immune functions additive to antibodies for success? Do different organs benefit from different modes of protection? Can vaccine activities beyond OPK protect the immunocompromised host? This commentary raises these issues at a time when capsule-only OPK assay-based vaccines are increasingly seen as a limiting strategy.

14.
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
15.
ACS Infect Dis ; 5(12): 1987-1995, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31268283

RESUMEN

Bacterial meningitis is a medical emergency requiring highly bactericidal antibiotics to achieve cure. Many challenges exist to achieving optimal patient outcome. First, antibiotics must pass the blood brain barrier. Once in the subarachnoid space, achieving bactericidal therapy involves circumventing antibiotic resistance and, more commonly, antibiotic tolerance arising from the slow growth of bacteria in the nutrient poor cerebrospinal fluid. Finally, bactericidal therapy is most often bacteriolytic, and debris from lysis is highly inflammatory. Controlling damage from lytic products may require adjunctive therapy to prevent neuronal death. These challenges are an extreme example of the different requirements for treating infections in different body sites.


Asunto(s)
Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Meningitis Bacterianas/tratamiento farmacológico , Enfermedad Aguda/terapia , Barrera Hematoencefálica , Humanos , Neuronas/efectos de los fármacos , Neuronas/patología
16.
Microbiol Spectr ; 7(2)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30873934

RESUMEN

Streptococcus pneumoniae (the pneumoccus) is the leading cause of otitis media, community-acquired pneumonia, and bacterial meningitis. The success of the pneumococcus stems from its ability to persist in the population as a commensal and avoid killing by immune system. This chapter first reviews the molecular mechanisms that allow the pneumococcus to colonize and spread from one anatomical site to the next. Then, it discusses the mechanisms of inflammation and cytotoxicity during emerging and classical pneumococcal infections.


Asunto(s)
Inflamación/inmunología , Inflamación/microbiología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Sangre/inmunología , Sangre/microbiología , Encéfalo/inmunología , Encéfalo/microbiología , Pared Celular/inmunología , Infecciones Comunitarias Adquiridas/microbiología , Oído Medio/microbiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Feto/inmunología , Feto/microbiología , Corazón/microbiología , Humanos , Meningitis Bacterianas/microbiología , Nasofaringe/inmunología , Nasofaringe/microbiología , Otitis Media/microbiología , Infecciones Neumocócicas/microbiología , Neumonía/microbiología , Sistema Respiratorio/inmunología , Sistema Respiratorio/microbiología , Streptococcus pneumoniae/patogenicidad
17.
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
18.
J Pediatric Infect Dis Soc ; 7(4): 275-282, 2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-29106589

RESUMEN

BACKGROUND: Respiratory virus infections (RVIs) pose a threat to children undergoing hematopoietic stem cell transplantation (HSCT). In this era of sensitive molecular diagnostics, the incidence and outcome of HSCT recipients who are hospitalized with RVI (H-RVI) are not well described. METHODS: A retrospective observational cohort of pediatric HSCT recipients (between January 2010 and June 2013) was assembled from 9 US pediatric transplant centers. Their medical charts were reviewed for H-RVI events within 1 year after their transplant. An H-RVI diagnosis required respiratory signs or symptoms plus viral detection (human rhinovirus/enterovirus, human metapneumovirus, influenza, parainfluenza, coronaviruses, and/or respiratory syncytial virus). The incidence of H-RVI was calculated, and the association of baseline HSCT factors with subsequent pulmonary complications and death was assessed. RESULTS: Among 1560 HSCT recipients, 259 (16.6%) acquired at least 1 H-RVI within 1 year after their transplant. The median age of the patients with an H-RVI was lower than that of patients without an H-RVI (4.8 vs 7.1 years; P < .001). Among the patients with a first H-RVI, 48% required some respiratory support, and 14% suffered significant pulmonary sequelae. The all-cause and attributable case-fatality rates within 3 months of H-RVI onset were 11% and 5.4%, respectively. Multivariate logistic regression revealed that H-RVI onset within 60 days of HSCT, steroid use in the 7 days before H-RVI onset, and the need for respiratory support at H-RVI onset were associated with subsequent morbidity or death. CONCLUSION: Results of this multicenter cohort study suggest that H-RVIs are relatively common in pediatric HSCT recipients and contribute to significant morbidity and death. These data should help inform interventional studies specific to each viral pathogen.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Hospitalización/estadística & datos numéricos , Infecciones del Sistema Respiratorio/epidemiología , Virosis/epidemiología , Corticoesteroides/uso terapéutico , Niño , Preescolar , Femenino , Humanos , Incidencia , Masculino , Terapia Respiratoria , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/terapia , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos/epidemiología , Virosis/mortalidad , Virosis/terapia
19.
mBio ; 8(5)2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928207

RESUMEN

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.


Asunto(s)
Coinfección/etiología , Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/administración & dosificación , Obesidad/complicaciones , Infecciones por Orthomyxoviridae/complicaciones , Vacunas Neumococicas/administración & dosificación , Animales , Coinfección/microbiología , Coinfección/virología , Comorbilidad , Virus de la Influenza A/crecimiento & desarrollo , Pulmón/microbiología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/microbiología , Obesidad/virología , Infecciones por Orthomyxoviridae/microbiología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/prevención & control , Infecciones Neumocócicas/virología , Insuficiencia del Tratamiento , Vacunación
20.
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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA