Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Immunol ; 211(9): 1397-1405, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37721419

RESUMEN

High levels of IFN-γ are produced in the lung during an adaptive immune response to Pneumocystis, but the effects of this prototypical Th1 cytokine on fungal clearance and immunopathogenesis have not been fully defined. Therefore, Pneumocystis-infected immunodeficient mice were immune reconstituted and administered control or anti-IFN-γ neutralizing Ab to determine how IFN-γ regulates the balance between host defense and immune-mediated lung injury. Mice treated with anti-IFN-γ demonstrated an initial worsening of Pneumocystis pneumonia-related immunopathogenesis, with greater weight loss, heightened lung inflammation, and more severe pulmonary function deficits than control mice. However, IFN-γ neutralization also enhanced macrophage phagocytosis of Pneumocystis and accelerated fungal clearance. When anti-IFN-γ-treated mice were also given IL-4 and IL-13 to promote a Th2-biased lung environment, the accelerated fungal clearance was preserved, but the severity of immunopathogenesis was reduced, and a more rapid recovery was observed. A direct suppressive effect of IFN-γ on macrophages was required but was not solely responsible for delayed fungal clearance, suggesting that IFN-γ acts through multiple mechanisms that likely include modulation of both macrophage and Th polarization. Enhanced Pneumocystis clearance in anti-IFN-γ-treated and IFN-γR-deficient mice was associated with significantly elevated IL-17+ CD4+ T cells and IL-17 protein in the lungs. Furthermore, neutralization of IL-17, but not IL-4, signaling blocked the accelerated fungal clearance observed in anti-IFN-γ-treated mice. Together, these data demonstrate that although IFN-γ delays fungal clearance by suppressing the lung Th17 response, it also serves an important regulatory role that limits immunopathogenesis and preserves pulmonary function.


Asunto(s)
Pneumocystis , Neumonía por Pneumocystis , Animales , Ratones , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/patología , Interleucina-17 , Pulmón , Interferón gamma , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Infect Immun ; 91(4): e0049022, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36916933

RESUMEN

Pneumocystis is a respiratory fungal pathogen that is among the most frequent causes of life-threatening pneumonia (PcP) in immunocompromised hosts. Alveolar macrophages play an important role in host defense against Pneumocystis, and several studies have suggested that M2 polarized macrophages have anti-Pneumocystis effector activity. Our prior work found that the immunomodulatory drug sulfasalazine (SSZ) provides a dual benefit during PcP-related immune reconstitution inflammatory syndrome (IRIS) by concurrently suppressing immunopathogenesis while also accelerating macrophage-mediated fungal clearance. The benefits of SSZ were associated with heightened Th2 cytokine production and M2 macrophage polarization. Therefore, to determine whether SSZ improves the outcome of PcP through a mechanism that requires Th2-dependent M2 polarization, RAG2-/- mice lacking interleukin 4 receptor alpha chain (IL-4Rα) on macrophage lineage cells were generated. As expected, SSZ treatment dramatically reduced the severity of PcP-related immunopathogenesis and accelerated fungal clearance in immune-reconstituted RAG2-/- mice. Similarly, SSZ treatment was also highly effective in immune-reconstituted RAG2/IL-4Rα-/- and RAG2/gamma interferon receptor (IFN-γR)-/- mice, demonstrating that neither IL-4Rα-dependent M2 nor IFN-γR-dependent M1 macrophage polarization programs were required for the beneficial effects of SSZ. Despite the fact that macrophages from RAG2/IL-4Rα-/- mice could not respond to the Th2 cytokines IL-4 and IL-13, M2-biased alveolar macrophages were identified in the lungs following SSZ treatment. These data demonstrate that not only does SSZ enhance phagocytosis and fungal clearance in the absence of macrophage IL-4Rα signaling, but also that SSZ promotes M2 macrophage polarization in an IL-4Rα-independent manner. These findings could have implications for the treatment of PcP and other diseases in which M2 polarization is beneficial.


Asunto(s)
Pneumocystis , Neumonía por Pneumocystis , Ratones , Animales , Sulfasalazina/farmacología , Neumonía por Pneumocystis/tratamiento farmacológico , Antifúngicos/farmacología , Macrófagos , Macrófagos Alveolares/microbiología
3.
Infect Immun ; 88(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31611280

RESUMEN

The pulmonary immune response protects healthy individuals against Pneumocystis pneumonia (PcP). However, the immune response also drives immunopathogenesis in patients who develop severe PcP, and it is generally accepted that optimal treatment requires combination strategies that promote fungal killing and also provide effective immunomodulation. The anti-inflammatory drug sulfasalazine programs macrophages for enhanced Pneumocystis phagocytosis and also suppresses PcP-related immunopathogenesis. Anti-Pneumocystis antibody opsonizes Pneumocystis organisms for greater phagocytosis and may also mask antigens that drive immunopathogenesis. Thus, we hypothesized that combining antibody and sulfasalazine would have the dual benefit of enhancing fungal clearance while dampening immunopathogenesis and allow the rescue of severe PcP. To model a clinically relevant treatment scenario in mice, therapeutic interventions were withheld until clear symptoms of pneumonia were evident. When administered individually, both passive antibody and sulfasalazine improved pulmonary function and enhanced Pneumocystis clearance to similar degrees. However, combination treatment with antibody and sulfasalazine produced a more rapid improvement, with recovery of body weight, a dramatic improvement in pulmonary function, reduced lung inflammation, and the rapid clearance of the Pneumocystis organisms. Accelerated fungal clearance in the combination treatment group was associated with a significant increase in macrophage phagocytosis of Pneumocystis Both passive antibody and sulfasalazine resulted in the suppression of Th1 cytokines and a marked increase in lung macrophages displaying an alternatively activated phenotype, which were enhanced by combination treatment. Our data support the concept that passive antibody and sulfasalazine could be an effective and specific adjunctive therapy for PcP, with the potential to accelerate fungal clearance while attenuating PcP-associated immunopathogenesis.


Asunto(s)
Anticuerpos/inmunología , Hongos/efectos de los fármacos , Hongos/inmunología , Neumonía por Pneumocystis/tratamiento farmacológico , Neumonía por Pneumocystis/inmunología , Sulfasalazina/farmacología , Animales , Antiinflamatorios/farmacología , Citocinas/inmunología , Femenino , Factores Inmunológicos/inmunología , Inmunomodulación/efectos de los fármacos , Inmunomodulación/inmunología , Inmunoterapia/métodos , Inflamación/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Ratones , Ratones SCID , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología
4.
Front Immunol ; 9: 2131, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283457

RESUMEN

Invasive fungal infections, including Pneumocystis Pneumonia (PcP), remain frequent life-threatening conditions of patients with adaptive immune defects. While innate immunity helps control pathogen growth early during infection, it is typically not sufficient for complete protection against Pneumocystis and other human fungal pathogens. Alveolar macrophages (AM) possess pattern recognition molecules capable of recognizing antigenic and structural determinants of Pneumocystis. However, this pathogen effectively evades innate immunity to infect both immunocompetent and immunosuppressed hosts, albeit with differing outcomes. During our studies of mouse models of PcP, the FVB/N strain was identified as unique because of its ability to mount a protective innate immune response against Pneumocystis infection. In contrast to other immunocompetent strains, which become transiently infected prior to the onset of adaptive immunity, FVB/N mice rapidly eradicated Pneumocystis before an adaptive immune response was triggered. Furthermore, FVB/N mice remained highly resistant to infection even in the absence of functional T cells. The effector mechanism of innate protection required the action of functional alveolar macrophages, and the adoptive transfer of resistant FVB/N AMs, but not susceptible CB.17 AMs, conferred protection to immunodeficient mice. Macrophage IFNγ receptor signaling was not required for innate resistance, and FVB/N macrophages were found to display markers of alternative activation. IFNγ reprogrammed resistant FVB/N macrophages to a permissive M1 biased phenotype through a mechanism that required direct activation of the macrophage IFNγR. These results demonstrate that appropriately programmed macrophages provide protective innate immunity against this opportunistic fungal pathogen, and suggest that modulating macrophage function may represent a feasible therapeutic strategy to enhance antifungal host defense. The identification of resistant and susceptible macrophages provides a novel platform to study not only the mechanisms of macrophage-mediated antifungal defense, but also the mechanisms by which Pneumocystis evades innate immunity.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Pneumocystis/inmunología , Animales , Humanos , Inmunocompetencia , Huésped Inmunocomprometido , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Infecciones por Pneumocystis/microbiología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
Infect Immun ; 85(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28031260

RESUMEN

Pneumocystis pneumonia (PcP) is a life-threatening infection that affects immunocompromised individuals. Nearly half of all PcP cases occur in those prescribed effective chemoprophylaxis, suggesting that additional preventive methods are needed. To this end, we have identified a unique mouse Pneumocystis surface protein, designated Pneumocystis cross-reactive antigen 1 (Pca1), as a potential vaccine candidate. Mice were immunized with a recombinant fusion protein containing Pca1. Subsequently, CD4+ T cells were depleted, and the mice were exposed to Pneumocystis murina Pca1 immunization completely protected nearly all mice, similar to immunization with whole Pneumocystis organisms. In contrast, all immunized negative-control mice developed PcP. Unexpectedly, Pca1 immunization generated cross-reactive antibody that recognized Pneumocystis jirovecii and Pneumocystis carinii Potential orthologs of Pca1 have been identified in P. jirovecii Such cross-reactivity is rare, and our findings suggest that Pca1 is a conserved antigen and potential vaccine target. The evaluation of Pca1-elicited antibodies in the prevention of PcP in humans deserves further investigation.


Asunto(s)
Antígenos Fúngicos/inmunología , Proteínas Fúngicas/inmunología , Pneumocystis carinii/inmunología , Pneumocystis/inmunología , Neumonía por Pneumocystis/inmunología , Animales , Anticuerpos Antifúngicos/inmunología , Especificidad de Anticuerpos/inmunología , Antígenos Fúngicos/administración & dosificación , Antígenos Fúngicos/genética , Reacciones Cruzadas , Proteínas Fúngicas/administración & dosificación , Proteínas Fúngicas/genética , Vacunas Fúngicas/administración & dosificación , Vacunas Fúngicas/inmunología , Inmunización , Ratones , Pneumocystis/genética , Pneumocystis carinii/genética , Neumonía por Pneumocystis/prevención & control
6.
Infect Immun ; 83(12): 4594-603, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371121

RESUMEN

Pneumocystis is a respiratory fungal pathogen that causes pneumonia (Pneumocystis pneumonia [PcP]) in immunocompromised patients. Alveolar macrophages are critical effectors for CD4(+) T cell-dependent clearance of Pneumocystis, and previous studies found that alternative macrophage activation accelerates fungal clearance during PcP-related immune reconstitution inflammatory syndrome (IRIS). However, the requirement for either classically or alternatively activated macrophages for Pneumocystis clearance has not been determined. Therefore, RAG2(-/-) mice lacking either the interferon gamma (IFN-γ) receptor (IFN-γR) or interleukin 4 receptor alpha (IL-4Rα) were infected with Pneumocystis. These mice were then immune reconstituted with wild-type lymphocytes to preserve the normal T helper response while preventing downstream effects of Th1 or Th2 effector cytokines on macrophage polarization. As expected, RAG2(-/-) mice developed severe disease but effectively cleared Pneumocystis and resolved IRIS. Neither RAG/IFN-γR(-/-) nor RAG/IL-4Rα(-/-) mice displayed impaired Pneumocystis clearance. However, RAG/IFN-γR(-/-) mice developed a dysregulated immune response, with exacerbated IRIS and greater pulmonary function deficits than those in RAG2 and RAG/IL-4Rα(-/-) mice. RAG/IFN-γR(-/-) mice had elevated numbers of lung CD4(+) T cells, neutrophils, eosinophils, and NK cells but severely depressed numbers of lung CD8(+) T suppressor cells. Impaired lung CD8(+) T cell responses in RAG/IFN-γR(-/-) mice were associated with elevated lung IFN-γ levels, and neutralization of IFN-γ restored the CD8 response. These data demonstrate that restricting the ability of macrophages to polarize in response to Th1 or Th2 cytokines does not impair Pneumocystis clearance. However, a cell type-specific IFN-γ/IFN-γR-dependent mechanism regulates CD8(+) T suppressor cell recruitment, limits immunopathogenesis, preserves lung function, and enhances the resolution of PcP-related IRIS.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Síndrome Inflamatorio de Reconstitución Inmune/inmunología , Macrófagos Alveolares/inmunología , Pneumocystis/inmunología , Neumonía por Pneumocystis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Eosinófilos/inmunología , Eosinófilos/microbiología , Eosinófilos/patología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Síndrome Inflamatorio de Reconstitución Inmune/genética , Síndrome Inflamatorio de Reconstitución Inmune/microbiología , Síndrome Inflamatorio de Reconstitución Inmune/patología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Activación de Macrófagos , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Ratones SCID , Neutrófilos/inmunología , Neutrófilos/microbiología , Neutrófilos/patología , Pneumocystis/patogenicidad , Neumonía por Pneumocystis/genética , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/patología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/microbiología , Linfocitos T Colaboradores-Inductores/patología , Balance Th1 - Th2 , Receptor de Interferón gamma
7.
Hosp Pediatr ; 5(6): 293-300, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26034160

RESUMEN

OBJECTIVE: Describe the etiology of bacteremia among a geographically diverse sample of previously well infants with fever admitted for general pediatric care and to characterize demographic and clinical characteristics of infants with bacteremia according to bacterial etiology. We hypothesized that the epidemiology of bacteremia in febrile infants from a geographically diverse cohort would show similar results to smaller or single-center cohorts previously reported. METHODS: This was a retrospective review of positive, pathogenic blood cultures in previously healthy, febrile infants≤90 days old admitted to a general unit. In total, there were 17 participating sites from diverse geographic regions of the United States. Cultures were included if the results were positive for bacteria, obtained from an infant 90 days old or younger with a temperature≥38.0°C, analyzed using an automated detection system, and treated as pathogenic. RESULTS: Escherichia coli was the most prevalent species, followed by group B Streptococcus, Streptococcus viridans, and Staphylococcus aureus. Among the most prevalent bacteria, there was no association between gender and species (Ps>.05). Age at presentation was associated only with Streptococcus pneumoniae. There were no cases of Listeria monocytogenes. CONCLUSIONS: Our study confirms the data from smaller or single-center studies and suggests that the management of febrile well-appearing infants should change to reflect the current epidemiology of bacteremia. Further research is needed into the role of lumbar puncture, as well as the role of Listeria and Enterococcus species in infantile bacteremia.


Asunto(s)
Bacteriemia/epidemiología , Estudios Transversales , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Estudios Retrospectivos , Estados Unidos/epidemiología
8.
Cold Spring Harb Perspect Med ; 4(12): a019828, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25367973

RESUMEN

Since its initial misidentification as a trypanosome some 100 years ago, Pneumocystis has remained recalcitrant to study. Although we have learned much, we still do not have definitive answers to such basic questions as, where is the reservoir of infection, how does Pneumocystis reproduce, what is the mechanism of infection, and are there true species of Pneumocystis? The goal of this review is to provide the reader the most up to date information available about the biology of Pneumocystis and the disease it produces.


Asunto(s)
Infecciones por Pneumocystis/microbiología , Pneumocystis/fisiología , Humanos , Pneumocystis/genética , Pneumocystis/patogenicidad , Infecciones por Pneumocystis/fisiopatología , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/fisiopatología , Factores de Virulencia/genética
9.
J Pediatr ; 165(3): 625-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24961788

RESUMEN

We analyzed the practice of paired erythrocyte sedimentation rate and C-reactive protein testing when evaluating fever or inflammation. In our hospital, this resulted in additional charges of $250000-$400000/year without demonstrable added benefit to patient care. Extrapolating our results, we estimate reducing this practice could save up to $300000000 nationally.


Asunto(s)
Sedimentación Sanguínea , Proteína C-Reactiva/análisis , Fiebre/sangre , Fiebre/economía , Gastos en Salud , Inflamación/sangre , Inflamación/economía , Niño , Pruebas Hematológicas/economía , Humanos , Factores de Tiempo
10.
J Immunol ; 192(1): 282-92, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24293628

RESUMEN

The immune response protects against Pneumocystis infection but is also a key component of Pneumocystis pneumonia (PcP)-related immunopathogenesis. Signaling through myeloid differentiation factor 88 (MyD88) is critical for activation of immune pathways downstream of TLRs and IL-1R. To determine whether MyD88 regulates normal host defense against Pneumocystis, nonimmunosuppressed wild-type (WT) and MyD88-deficient mice were infected. MyD88(-/-) mice had higher early Pneumocystis burdens than did WT mice but mounted an effective adaptive immune response and cleared Pneumocystis similarly to WT. However, MyD88(-/-) mice displayed a more intense and prolonged pulmonary immune response than did WT mice. To determine the role of MyD88 in the development of PcP-related immunopathogenesis, WT and MyD88(-/-) mice were rendered susceptible to PcP by depletion of CD4(+) T cells. At 4 wk postinfection, CD4-depleted WT and MyD88(-/-) mice harbored similar organism burdens, but MyD88(-/-) mice were protected from the PcP-related respiratory impairment observed in WT mice. Improved pulmonary physiology in MyD88(-/-) mice correlated with lower lung CCL2 levels and reduced cell recruitment. However, by 5 wk postinfection, the overall health of MyD88(-/-) mice began to deteriorate rapidly relative to WT, with accelerated weight loss, impaired lung function, and exacerbated alveolar inflammation. This physiological decline of MyD88(-/-) mice was associated with increased TNF-α and IFN-γ in the lung, and by the inability to control Pneumocystis burden. Thus, MyD88 is not required for resistance to Pneumocystis infection, but limits the adaptive immune response in immunocompetent mice. In the setting of active PcP, MyD88 signaling contributes to both immunopathogenesis and control of fungal burden.


Asunto(s)
Factor 88 de Diferenciación Mieloide/metabolismo , Infecciones por Pneumocystis/inmunología , Infecciones por Pneumocystis/metabolismo , Pneumocystis/inmunología , Transducción de Señal , Animales , Células de la Médula Ósea/metabolismo , Quimiocinas/biosíntesis , Recuento de Colonia Microbiana , Citocinas/biosíntesis , Femenino , Hematopoyesis/genética , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Infecciones por Pneumocystis/genética , Infecciones por Pneumocystis/microbiología
11.
Pediatrics ; 132(6): 990-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24218461

RESUMEN

BACKGROUND: Fever in infants is a common clinical dilemma. The objective of this study was to present data from hospital systems across the northeast, southeast, mid-west, and western United States to identify the pathogens causing bacteremia in febrile infants admitted to general care units. METHODS: This was a retrospective review of positive blood culture results in febrile infants aged ≤90 days admitted to a general care unit across 6 hospital systems. Data were collected from January 1, 2006 through December 31, 2012 from emergency departments and general inpatient units. Cultures from ICUs, central lines, or infants who had complex comorbidities were excluded, as were repeat cultures positive for the same bacteria. Common contaminants were considered pathogens if they were treated as such. RESULTS: We identified 181 cases of bacteremia in 177 infants. The most common pathogen was Escherichia coli (42%), followed by group B Streptococcus (23%). Streptococcus pneumoniae was more likely in older infants (P = .01). Non-low-risk bacteremic infants were more likely to have E. coli or group B Streptococcus than low-risk bacteremic infants. We identified no cases of Listeria monocytogenes. Variation between sites was minimal. CONCLUSIONS: This is the largest and most geographically diverse study to date examining the epidemiology of bacteremia in infants. We suggest E. coli is the most common cause of bacteremia in previously healthy febrile infants admitted to a general inpatient unit. We identified no cases of L monocytogenes and question whether empirical therapy remains necessary for this pathogen.


Asunto(s)
Bacteriemia/epidemiología , Infecciones por Escherichia coli/epidemiología , Fiebre/microbiología , Infecciones Estreptocócicas/epidemiología , Factores de Edad , Bacteriemia/complicaciones , Bacteriemia/diagnóstico , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/diagnóstico , Femenino , Infecciones por Bacterias Gramnegativas/complicaciones , Infecciones por Bacterias Gramnegativas/diagnóstico , Infecciones por Bacterias Gramnegativas/epidemiología , Infecciones por Bacterias Grampositivas/complicaciones , Infecciones por Bacterias Grampositivas/diagnóstico , Infecciones por Bacterias Grampositivas/epidemiología , Humanos , Lactante , Recién Nacido , Listeriosis/complicaciones , Listeriosis/diagnóstico , Listeriosis/epidemiología , Masculino , Infecciones Neumocócicas/complicaciones , Infecciones Neumocócicas/diagnóstico , Infecciones Neumocócicas/epidemiología , Prevalencia , Estudios Retrospectivos , Factores de Riesgo , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/diagnóstico , Streptococcus agalactiae/aislamiento & purificación , Estados Unidos/epidemiología
12.
J Immunol ; 191(9): 4720-30, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078701

RESUMEN

Pneumocystis is an atypical fungal pathogen that causes severe, often fatal pneumonia in immunocompromised patients. Healthy humans and animals also encounter this pathogen, but they generate a protective CD4(+) T cell-dependent immune response that clears the pathogen with little evidence of disease. Pneumocystis organisms attach tightly to respiratory epithelial cells, and in vitro studies have demonstrated that this interaction triggers NF-κB-dependent epithelial cell responses. However, the contribution of respiratory epithelial cells to the normal host response to Pneumocystis remains unknown. IκB kinase 2 (IKK2) is the upstream kinase that is critical for inducible NF-κB activation. To determine whether IKK2-dependent lung epithelial cell (LEC) responses contribute to the anti-Pneumocystis immune response in vivo, transgenic mice with LEC-specific deletion of IKK2 (IKK2(ΔLEC)) were generated. Compared to wild-type mice, IKK2(ΔLEC) mice exhibited a delayed onset of Th17 and B cell responses in the lung and delayed fungal clearance. Importantly, delayed Pneumocystis clearance in IKK2(ΔLEC) mice was associated with an exacerbated immune response, impaired pulmonary function, and altered lung histology. These data demonstrate that IKK2-dependent LEC responses are important regulators of pulmonary adaptive immune responses and are required for optimal host defense against Pneumocystis infection. LECs likely set the threshold for initiation of the pulmonary immune response and serve to prevent exacerbated lung inflammation by promoting the rapid control of respiratory fungal infection.


Asunto(s)
Quinasa I-kappa B/metabolismo , Pneumocystis/inmunología , Neumonía por Pneumocystis/inmunología , Células Th17/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Pulmón/citología , Pulmón/inmunología , Activación de Linfocitos/inmunología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología
14.
J Pediatr ; 162(4): 857-61, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23092529

RESUMEN

OBJECTIVE: To perform a randomized controlled trial comparing moxifloxacin hydrochloride with polymyxin B-trimethoprim for the treatment of acute conjunctivitis. STUDY DESIGN: Patients ages 1-18 years old with acute conjunctivitis had cultures performed and were randomized to receive either moxifloxacin hydrochloride or polymyxin B-trimethoprim ophthalmic solution for 7 days. Response to treatment was determined by phone query on day 4-6 and by examination with post-treatment conjunctival culture on day 7-10. RESULTS: One hundred and twenty-four patients were enrolled. Eighty patients (65%) had recognized pathogens (55 Haemophilus influenzae, 22 Streptococcus pneumoniae, 4 Moraxella catarrhalis) isolated from their conjunctiva. One hundred fourteen (56/62 moxifloxacin and 58/62 polymyxin B-trimethoprim) completed the 4-6 day evaluation, with 43/56 (77%) of the moxifloxacin group and 42/58 (72%) of the polymyxin B-trimethoprim group clinically cured according to parents (noninferiority test P = .04). Eighty-nine (39/56 moxifloxacin and 50/58 polymyxin B-trimethoprim) patients completed the 7-10 day evaluation. Clinical cure was observed in 37/39 (95%) of the moxifloxacin and 49/51 (96%) of the polymyxin B-trimethoprim treated groups (noninferiority test P ≤ .01). Clinical cure rates for culture positive and negative conjunctivitis were not different. There was no statistically significant difference in bacteriologic cure rates between the 2 groups. CONCLUSIONS: Polymyxin B-trimethoprim continues to be an effective treatment for acute conjunctivitis with a clinical response rate that does not differ from moxifloxacin. Use of polymyxin B-trimethoprim for the treatment of conjunctivitis would result in significant cost savings compared with fluoroquinolones.


Asunto(s)
Compuestos Aza/uso terapéutico , Conjuntivitis Bacteriana/tratamiento farmacológico , Polimixina B/uso terapéutico , Quinolinas/uso terapéutico , Trimetoprim/uso terapéutico , Adolescente , Antibacterianos/uso terapéutico , Niño , Preescolar , Conjuntiva/microbiología , Quimioterapia Combinada , Femenino , Fluoroquinolonas , Haemophilus influenzae/metabolismo , Humanos , Lactante , Masculino , Moraxella catarrhalis/metabolismo , Moxifloxacino , Soluciones Oftálmicas/uso terapéutico , Método Simple Ciego , Streptococcus pneumoniae/metabolismo
16.
Infect Immun ; 80(11): 3912-20, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22927048

RESUMEN

Pneumocystis is an opportunistic fungal pathogen that causes pneumonia in a variety of clinical settings. An early step in Pneumocystis infection involves the attachment of organisms to alveolar epithelial cells (AECs). AECs produce chemokines in response to Pneumocystis stimulation, but the upstream host-pathogen interactions that activate AEC signaling cascades are not well-defined. MyD88 is an adaptor molecule required for activation of proinflammatory signaling cascades following Toll-like receptor (TLR)-dependent recognition of conserved molecular patterns on pathogens. To determine whether the TLR/MyD88 pathway is required for the AEC chemokine response to Pneumocystis, wild-type (WT) and MyD88-deficient AECs were incubated with Pneumocystis. As expected, WT AECs produced CCL2 and CXCL2 following Pneumocystis stimulation. In contrast, MyD88-deficient AECs were severely impaired in their ability to respond to Pneumocystis. MyD88-deficient AECs did not display Pneumocystis-induced Jun N-terminal protein kinase activation and produced much less chemokine than Pneumocystis-stimulated WT AECs. Using a panel of TLR agonists, primary murine AECs were found to respond vigorously to TLR2 and TLR4 agonists. However, the AEC chemokine response to Pneumocystis did not require TLR2 or TLR4. Surprisingly, the interleukin-1 receptor (IL-1R) was required for an AEC chemokine response to Pneumocystis. The role of MyD88 in early responses during Pneumocystis infection was supported by in vivo studies demonstrating that MyD88-deficient mice showed impaired Pneumocystis-stimulated chemokine production and impaired inflammatory cell recruitment. These data indicate an important role for MyD88 in the AEC inflammatory response to Pneumocystis.


Asunto(s)
Factor 88 de Diferenciación Mieloide/metabolismo , Pneumocystis/inmunología , Neumonía por Pneumocystis/metabolismo , Alveolos Pulmonares/citología , Receptores de Interleucina-1/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Pneumocystis/aislamiento & purificación , Neumonía por Pneumocystis/inmunología , Transducción de Señal
17.
Interdiscip Perspect Infect Dis ; 2011: 918038, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21904545

RESUMEN

Pneumocystis is an opportunistic fungal respiratory pathogen that causes life-threatening pneumonia (Pcp) in patients suffering from defects in cell-mediated immunity, including those with acquired immunodeficiency syndrome (AIDS) and immunosuppression secondary to chemotherapy or organ transplantation. Despite major advances in health care, the mortality associated with Pcp has changed little over the past 25 years. Pcp remains a leading cause of death among HIV infected patients, with mortality rates of 50% or higher for patients developing severe Pcp. In addition, as more potent immunosuppressive therapies are developed for chronic inflammatory diseases, more cases of Pcp are occurring in non-HIV patients and in previously unreported clinical settings. These features highlight the importance of developing a better understanding of the pathogenesis of this disease, and the need to search for new therapeutic strategies to improve the outcome of Pcp patients. Immune-mediated inflammatory responses play an important role in the pathogenesis of Pcp, and may be even more significant in determining the outcome of Pcp than direct damage due to the organism itself. In this review we will summarize the immunopathogenic mechanisms that contribute to Pcp-associated lung injury, and discuss the potential to target these pathways for adjunctive immune modulation therapy for Pcp.

18.
PLoS Pathog ; 6(8): e1001058, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20808846

RESUMEN

Although T cells are critical for host defense against respiratory fungal infections, they also contribute to the immunopathogenesis of Pneumocystis pneumonia (PcP). However, the precise downstream effector mechanisms by which T cells mediate these diverse processes are undefined. In the current study the effects of immune modulation with sulfasalazine were evaluated in a mouse model of PcP-related Immune Reconstitution Inflammatory Syndrome (PcP-IRIS). Recovery of T cell-mediated immunity in Pneumocystis-infected immunodeficient mice restored host defense, but also initiated the marked pulmonary inflammation and severe pulmonary function deficits characteristic of IRIS. Sulfasalazine produced a profound attenuation of IRIS, with the unexpected consequence of accelerated fungal clearance. To determine whether macrophage phagocytosis is an effector mechanism of T cell-mediated Pneumocystis clearance and whether sulfasalazine enhances clearance by altering alveolar macrophage phagocytic activity, a novel multispectral imaging flow cytometer-based method was developed to quantify the phagocytosis of Pneumocystis in vivo. Following immune reconstitution, alveolar macrophages from PcP-IRIS mice exhibited a dramatic increase in their ability to actively phagocytose Pneumocystis. Increased phagocytosis correlated temporally with fungal clearance, and required the presence of CD4(+) T cells. Sulfasalazine accelerated the onset of the CD4(+) T cell-dependent alveolar macrophage phagocytic response in PcP-IRIS mice, resulting in enhanced fungal clearance. Furthermore, sulfasalazine promoted a TH2-polarized cytokine environment in the lung, and sulfasalazine-enhanced phagocytosis of Pneumocystis was associated with an alternatively activated alveolar macrophage phenotype. These results provide evidence that macrophage phagocytosis is an important in vivo effector mechanism for T cell-mediated Pneumocystis clearance, and that macrophage phenotype can be altered to enhance phagocytosis without exacerbating inflammation. Immune modulation can diminish pulmonary inflammation while preserving host defense, and has therapeutic potential for the treatment of PcP-related immunopathogenesis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Inmunomodulación/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Neumonía por Pneumocystis/inmunología , Sulfasalazina/farmacología , Animales , Linfocitos T CD4-Positivos/inmunología , Separación Celular/métodos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo/métodos , Síndrome Inflamatorio de Reconstitución Inmune/inmunología , Inmunomodulación/inmunología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Ratones , Ratones SCID , Microscopía Confocal , Fagocitosis/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Immunol ; 184(1): 497-502, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19949093

RESUMEN

The T cell-mediated immune response elicited by Pneumocystis plays a key role in pulmonary damage and dysfunction during Pneumocystis carinii pneumonia (PcP). Mice depleted of CD4(+) and CD8(+) T cells prior to infection are markedly protected from PcP-related respiratory deficit and death, despite progressive lung infection. However, the therapeutic effectiveness of Ab-mediated disruption of T cell function in mice already displaying clinical symptoms of disease has not been determined. Therefore, a murine model of PcP-related immune reconstitution inflammatory syndrome was used to assess whether Ab to the pan-T cell molecule CD3 is effective for reducing the severity of PcP when administered after the onset of disease. Mice that received anti-CD3 Ab exhibited a rapid and dramatic halt in the PcP-associated pulmonary function decline within 1 week after treatment, and a striking enhancement of survival rate compared with mice receiving the control Ab. Physiologic improvement in anti-CD3 treated mice was associated with a significant reduction in the number of CD4(+) and CD8(+) T cells recovered in lung lavage fluid. This effectiveness of anti-CD3 was noted whether the mice also received antibiotic therapy with trimethoprim-sulfamethoxazole. These data suggest that monoclonal Ab-mediated disruption of T cell function may represent a specific and effective adjunctive therapy to rapidly reverse the ongoing pathologic immune response occurring during active PcP. Thus, the anti-human CD3 monoclonal Ab OKT3, which is already in clinical use, has the potential to be developed as an adjunctive therapy for PcP.


Asunto(s)
Anticuerpos/uso terapéutico , Complejo CD3/inmunología , Inflamación/tratamiento farmacológico , Depleción Linfocítica/métodos , Neumonía por Pneumocystis/tratamiento farmacológico , Animales , Antiinfecciosos/uso terapéutico , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/microbiología , Ratones , Neumonía por Pneumocystis/inmunología , Neumonía por Pneumocystis/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Combinación Trimetoprim y Sulfametoxazol/uso terapéutico
20.
J Immunol ; 181(2): 1409-19, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606695

RESUMEN

The opportunistic organism Pneumocystis carinii (Pc) produces a life-threatening pneumonia (PcP) in patients with low CD4(+) T cell counts. Animal models of HIV-AIDS-related PcP indicate that development of severe disease is dependent on the presence of CD8(+) T cells and the TNF receptors (TNFR) TNFRsf1a and TNFRsf1b. To distinguish roles of parenchymal and hematopoietic cell TNF signaling in PcP-related lung injury, murine bone marrow transplant chimeras of wild-type, C57BL6/J, and TNFRsf1a/1b double-null origin were generated, CD4(+) T cell depleted, and inoculated with Pc. As expected, C57 --> C57 chimeras (donor marrow --> recipient) developed significant disease as assessed by weight loss, impaired pulmonary function (lung resistance and dynamic lung compliance), and inflammatory cell infiltration. In contrast, TNFRsf1a/1b(-/-) --> TNFRsf1a/1b(-/-) mice were relatively mildly affected despite carrying the greatest organism burden. Mice solely lacking parenchymal TNFRs (C57 --> TNFRsf1a/1b(-/-)) had milder disease than did C57 --> C57 mice. Both groups of mice with TNFR-deficient parenchymal cells had low bronchoalveolar lavage fluid total cell counts and fewer lavageable CD8(+) T cells than did C57 --> C57 mice, suggesting that parenchymal TNFR signaling contributes to PcP-related immunopathology through the recruitment of damaging immune cells. Interestingly, mice with wild-type parenchymal cells but TNFRsf1a/1b(-/-) hematopoietic cells (TNFRsf1a/1b(-/-) --> C57) displayed exacerbated disease characterized by increased MCP-1 and KC production in the lung and increased macrophage and lymphocyte numbers in the lavage, indicating a dysregulated immune response. This study supports a key role of parenchymal cell TNFRs in lung injury induced by Pc and a potential protective effect of receptors on radiosensitive, bone marrow-derived cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Neumonía por Pneumocystis/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Trasplante de Médula Ósea , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/microbiología , Hidroliasas/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pneumocystis carinii , Neumonía por Pneumocystis/microbiología , Neumonía por Pneumocystis/patología , Neumonía por Pneumocystis/fisiopatología , Receptores del Factor de Necrosis Tumoral/inmunología , Quimera por Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA