Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Antimicrob Agents Chemother ; 68(1): e0099223, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38059634

RESUMEN

Vancomycin is the first-line agent to treat pulmonary infections caused by methicillin-resistant Staphylococcus aureus (MRSA) in people with cystic fibrosis (PwCF). However, there is no consensus on vancomycin initial dosing in this population among health institutions, and there is a large variability in initial dosing across the United States. In this study, we characterized the pharmacokinetics (PK) of vancomycin in PwCF using a population PK approach. The clinical PK data to develop the population PK model were obtained from vancomycin therapeutic monitoring data from PwCF undergoing treatment for infections due to MRSA. The population PK model was then used to perform comprehensive Monte Carlo simulations to evaluate the probability of target attainment (PTA) of 12 different initial dosing scenarios. The area under the curve to minimum inhibitory concentration (MIC) ratio ≥400 mg*h/L and <650 mg*h/L were used as efficacy and toxicity targets for PTA analysis. A total of 181 vancomycin plasma concentrations were included in the analysis. A one-compartment model with first-order elimination best described the data. Weight significantly influenced the vancomycin PK (P < 0.05). In the final model, clearance was estimated as 5.52 L/h/70 kg, and the volume of distribution was 31.5 L/70 kg. The PTA analysis showed that at MIC = 1 µg/mL, doses 1,500 q8h and 2,000 q12h showed the highest %PTA in achieving both efficacy and toxicity targets. The PTA results from this study may potentially inform the initial dosing regimens of vancomycin to treat pulmonary infections due to MRSA in PwCF.


Asunto(s)
Fibrosis Quística , Staphylococcus aureus Resistente a Meticilina , Adulto , Humanos , Vancomicina/farmacología , Antibacterianos/farmacología , Fibrosis Quística/tratamiento farmacológico , Pruebas de Sensibilidad Microbiana , Método de Montecarlo
2.
Pediatr Pulmonol ; 59(1): 89-94, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37792309

RESUMEN

BACKGROUND: The American Thoracic Society Guidelines recommend vancomycin as first line option for treatment of methicillin-resistant Staphylococcus aureus. Two studies have described the pharmacokinetics (PK) of intermittent intravenous (IV) vancomycin in adult people with cystic fibrosis (PwCF). Currently, there have not been any studies describing the PK of continuous infusion vancomycin in PwCF. Our study aimed to describe the PK of continuous infusion vancomycin in adult PwCF. METHODS: Included patients were adult PwCF, who were admitted to University of Utah Hospital between May 11, 2014 and August 31, 2020, and received continuous infusion vancomycin for the treatment of an pulmonary exacerbations. The primary outcome was to describe vancomycin clearance rate (CLvanco ) and total daily dose (TDD). Secondary outcomes included rates of acute kidney injury (AKI), liver injury, and infusion-related reactions. RESULTS: Twenty patients were included in this study. The mean CLvanco was 5.08 L/h on Day 3 and 4.58 L/h on Day 7 (p = .04), and the TDD increased from 2444 mg on Day 3 to 2556 on Day 7, although not statistically significant (p = 0.26). Zero patients experienced an AKI, two patients experienced liver injury, and no patients experienced infusion-related reactions. CONCLUSIONS: This study demonstrates that continuous infusion vancomycin PK, namely CLvanco , is similar to previously reported CLvanco for intermittent dosed IV vancomycin in adult PwCF. This study suggests that continuous infusion vancomycin is likely safe to use in adult PwCF.


Asunto(s)
Lesión Renal Aguda , Fibrosis Quística , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Adulto , Humanos , Vancomicina/uso terapéutico , Antibacterianos/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Lesión Renal Aguda/inducido químicamente , Infusiones Intravenosas , Infecciones Estafilocócicas/tratamiento farmacológico , Estudios Retrospectivos
3.
Pediatr Pulmonol ; 57(11): 2646-2651, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35836330

RESUMEN

BACKGROUND: Antibiotics have altered pharmacokinetics (PK) in persons with cystic fibrosis (PwCF) during treatment for an acute pulmonary exacerbation (APE). The Cystic Fibrosis Foundation Pulmonary Guidelines-Treatment of Pulmonary Exacerbations do not provide specific recommendations for treatment of methicillin-resistant Staphylococcus aureus (MRSA) lung infections. However, the American Thoracic Society Guidelines recommend vancomycin as the first-line therapy. Only one study has previously described a single dose of intravenous (IV) vancomycin PK in adult PwCF. Our study aimed to describe intermittent IV vancomycin PK at steady-state in adult PwCF. METHODS: Adult PwCF who were admitted to University of Utah Hospital between May 11, 2014 and August 31, 2020, and received intermittent IV vancomycin for the treatment of an APE were included in this study. The primary outcome was to describe the drug volume of distribution (Vd), drug clearance, elimination half-life, and total daily dose of vancomycin. Secondary outcomes were rates of acute kidney injury (AKI), liver injury, and infusion-related reactions. RESULTS: Thirteen patients were included. The mean Vd was 0.54 L/kg on Day 3 and 0.53L/kg on Day 7. CLvanco was 5.11L/h on Day 3 and 4.69 L/h on Day 7. Zero patients experienced an AKI, two patients experienced liver injury, and no patients experienced infusion-related reactions. CONCLUSIONS: Our results demonstrate that in PwCF intermittent IV vancomycin steady-state PK are similar to previously reported single-dose IV vancomycin. Additionally, CLvanco minimally changes from Day 3 to Day 7, although this study was not powered to detect a difference.


Asunto(s)
Lesión Renal Aguda , Fibrosis Quística , Hominidae , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Adulto , Animales , Antibacterianos/uso terapéutico , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Humanos , Infecciones Estafilocócicas/tratamiento farmacológico , Vancomicina/uso terapéutico
4.
J Asthma Allergy ; 14: 405-413, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33911879

RESUMEN

BACKGROUND: Data regarding the inflammatory profile of patients with asthma and chronic rhinosinusitis (CRS-A) with (CRSwNP-A) and without (CRSsNP-A) nasal polyposis remain limited. OBJECTIVE: Define and compare systemic transcriptional changes in patients with CRS-A to those with non-asthma-related CRS with (CRSwNP) and without nasal polyposis (CRSsNP). METHODS: Thirty-four patients with CRS-A (n=19) and CRS (n=15) were prospectively enrolled into an observational study. Demographic information and subjective and objective disease severity measures were recorded. Multiplex gene expression analysis of mRNA extracted from peripheral blood was performed. A total of 594 genes associated with innate/adaptive immunity were analyzed using NanoString technology. Gene expression ratios were reported for genes that were differentially expressed among these cohorts. Linear regression analysis was used to compare the mRNA transcript copy numbers for each gene with disease severity. RESULTS: There was no significant difference in age, gender, nasal polyposis, or health-related quality of life measures between the two groups (p>0.05). HLA class II histocompatibility antigen, DRB3-1 beta chain (HLA-DRB3) was significantly upregulated in the peripheral blood of patients with CRSsNP-A compared to CRSsNP, whereas chemokine (C-C motif) ligands 4 (CCL4) and zinc finger protein helios (IKZF2) were significantly upregulated in CRSwNP-A compared to CRSwNP (p<0.05). CONCLUSION: Patients with CRSsNP-A demonstrate a molecular endotype associated with a Th2-dominant inflammatory profile compared to CRSsNP. Patients with CRSwNP-A similarly demonstrate an overrepresentation of genes associated with Th2-driven inflammation compared to patients with CRSwNP.

5.
Int Forum Allergy Rhinol ; 11(8): 1197-1206, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33629540

RESUMEN

BACKGROUND: Chronic rhinosinusitis with asthma (CRS-A) has a significant impact on patient morbidity and quality of life. Nevertheless, little is known about the natural history of endoscopic sinus surgery (ESS) in this cohort. The objective of this study was to evaluate revision rates of ESS in CRS-A and identify risk factors associated with increased likelihood for revision surgery compared to those with CRS without asthma (CRS-alone). METHODS: The Utah Population Database was queried for patients age >18 years with CRS who underwent at least 1 ESS between 1996 and 2018. Demographic information and history of ESS were collected and compared between CRS-A and CRS-alone using chi-square tests for categorical variables and t tests for continuous variables. Risk factors for revision surgery were analyzed using Cox proportional hazard models. RESULTS: A total of 33,090 patients (7693 CRS-A and 25,397 CRS-alone) were included in the final analysis. Mean follow up was 9.8 years in CRS-A and 9.1 years in CRS-alone (p < 0.001). The revision rate among patients with CRS-A (21.5%) was twice that of CRS-alone (10.8%) (p < 0.001). Among patients with CRS, a history of allergy (p < 0.001), asthma (p < 0.001), and nasal polyposis (p < 0.001) was independently associated with increased risk of revision ESS. Patients with CRS-A and nasal polyposis were 6 times more likely to require revision surgery than those with CRS-alone (p < 0.010). CONCLUSION: The rate of revision ESS in CRS-A was twice that of CRS-alone; patients with CRS-A and nasal polyposis were 6 times more likely to require revision than those with CRS-alone. ©2021 ARSAAOA, LLC.


Asunto(s)
Asma , Pólipos Nasales , Rinitis , Sinusitis , Adolescente , Asma/epidemiología , Enfermedad Crónica , Endoscopía , Humanos , Pólipos Nasales/epidemiología , Pólipos Nasales/cirugía , Calidad de Vida , Rinitis/epidemiología , Rinitis/cirugía , Sinusitis/epidemiología , Sinusitis/cirugía
6.
J Interprof Care ; 32(3): 386-390, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29172761

RESUMEN

Interprofessional care teams are the backbone of intensive care units (ICUs) where severity of illness is high and care requires varied skills and experience. Despite this care model, longitudinal educational programmes for such workplace teams rarely include all professions. In this article, we report findings on the initial assessment and evaluation of an ongoing, longitudinal simulation-based curriculum for interprofessional workplace critical care teams. The study had two independent components, quantitative learner assessment and qualitative curricular evaluation. To assess curriculum effectiveness at meeting learning objectives, participant-reported key learning points identified using a self-assessment tool administered immediately following curricular participation were mapped to session learning objectives. To evaluate the curriculum, we conducted a qualitative study using a phenomenology approach involving purposeful sampling of nine curricular participants undergoing recorded semi-structured interviews. Verbatim transcripts were reviewed by two independent readers to derive themes further subdivided into successes and barriers. Learner self-assessment demonstrated that the majority of learners, across all professions, achieved at least one intended learning objective with senior learners more likely to report team-based objectives and junior learners more likely to report knowledge/practice objectives. Successes identified by curricular evaluation included authentic critical care curricular content, safe learning environment, and team comradery from shared experience. Barriers included unfamiliarity with the simulation environment and clinical coverage for curricular participation. This study suggests that a sustainable interprofessional curriculum for workplace ICU critical care teams can achieve the desired educational impact and effectively deliver authentic simulated work experiences if barriers to educational engagement and participation can be overcome.


Asunto(s)
Cuidados Críticos , Personal de Salud/educación , Relaciones Interprofesionales , Entrenamiento Simulado/organización & administración , Competencia Clínica , Curriculum , Ambiente , Humanos , Entrevistas como Asunto , Grupo de Atención al Paciente , Investigación Cualitativa , Factores de Tiempo
8.
Bioengineered ; 5(2): 143-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24637705

RESUMEN

There is increasing urgency in the battle against drug-resistant bacterial pathogens, and this public health crisis has created a desperate need for novel antimicrobial agents. Recombinant human lysozyme represents one interesting candidate for treating pulmonary infections, but the wild type enzyme is subject to electrostatic mediated inhibition by anionic biopolymers that accumulate in the infected lung. We have redesigned lysozyme's electrostatic potential field, creating a genetically engineered variant that is less susceptible to polyanion inhibition, yet retains potent bactericidal activity. A recent publication demonstrated that the engineered enzyme outperforms wild type lysozyme in a murine model of Pseudomonas aeruginosa lung infection. Here, we expand upon our initial studies and consider dual therapies that combine lysozymes with an antimicrobial peptide. Consistent with our earlier results, the charge modified lysozyme combination outperformed its wild type counterpart, yielding more than an order-of-magnitude reduction in bacterial burden following treatment with a single dose.


Asunto(s)
Antibacterianos/uso terapéutico , Muramidasa/genética , Muramidasa/uso terapéutico , Neumonía Bacteriana/tratamiento farmacológico , Ingeniería de Proteínas/métodos , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Bioingeniería/métodos , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Quimioterapia Combinada/métodos , Ratones , Ratones Endogámicos C57BL , Neumonía Bacteriana/diagnóstico , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/diagnóstico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/fisiología , Resultado del Tratamiento
9.
Am J Respir Crit Care Med ; 189(4): 463-74, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24325366

RESUMEN

RATIONALE: The death receptor Fas is critical for bacterial clearance and survival of mice after Pseudomonas aeruginosa infection. OBJECTIVES: Fas ligand (FasL)-induced apoptosis is augmented by S-glutathionylation of Fas (Fas-SSG), which can be reversed by glutaredoxin-1 (Grx1). Therefore, the objective of this study was to investigate the interplay between Grx1 and Fas in regulating the clearance of P. aeruginosa infection. METHODS: Lung samples from patients with bronchopneumonia were analyzed by immunofluorescence. Primary tracheal epithelial cells, mice lacking the gene for Grx1 (Glrx1(-/-)), Glrx1(-/-) mice treated with caspase inhibitor, or transgenic mice overexpressing Grx1 in the airway epithelium were analyzed after infection with P. aeruginosa. MEASUREMENTS AND MAIN RESULTS: Patient lung samples positive for P. aeruginosa infection demonstrated increased Fas-SSG compared with normal lung samples. Compared with wild-type primary lung epithelial cells, infection of Glrx1(-/-) cells with P. aeruginosa showed enhanced caspase 8 and 3 activities and cell death in association with increases in Fas-SSG. Infection of Glrx1(-/-) mice with P. aeruginosa resulted in enhanced caspase activity and increased Fas-SSG as compared with wild-type littermates. Absence of Glrx1 significantly enhanced bacterial clearance, and decreased mortality postinfection with P. aeruginosa. Inhibition of caspases significantly decreased bacterial clearance postinfection with P. aeruginosa, in association with decreased Fas-SSG. In contrast, transgenic mice that overexpress Grx1 in lung epithelial cells had significantly higher lung bacterial loads, enhanced mortality, decreased caspase activation, and Fas-SSG in the lung after infection with P. aeruginosa, compared with wild-type control animals. CONCLUSIONS: These results suggest that S-glutathionylation of Fas within the lung epithelium enhances epithelial apoptosis and promotes clearance of P. aeruginosa and that glutaredoxin-1 impairs bacterial clearance and increases the severity of pneumonia in association with deglutathionylation of Fas.


Asunto(s)
Bronconeumonía/metabolismo , Glutarredoxinas/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Receptor fas/metabolismo , Animales , Apoptosis , Carga Bacteriana , Biomarcadores/metabolismo , Bronconeumonía/microbiología , Caspasas/metabolismo , Citocinas/metabolismo , Glutatión/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Infecciones por Pseudomonas/microbiología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Índice de Severidad de la Enfermedad
10.
Antimicrob Agents Chemother ; 57(11): 5559-64, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979752

RESUMEN

The spread of drug-resistant bacterial pathogens is a growing global concern and has prompted an effort to explore potential adjuvant and alternative therapies derived from nature's repertoire of bactericidal proteins and peptides. In humans, the airway surface liquid layer is a rich source of antibiotics, and lysozyme represents one of the most abundant and effective antimicrobial components of airway secretions. Human lysozyme is active against both Gram-positive and Gram-negative bacteria, acting through several mechanisms, including catalytic degradation of cell wall peptidoglycan and subsequent bacterial lysis. In the infected lung, however, lysozyme's dense cationic character can result in sequestration and inhibition by polyanions associated with airway inflammation. As a result, the efficacy of the native enzyme may be compromised in the infected and inflamed lung. To address this limitation, we previously constructed a charge-engineered variant of human lysozyme that was less prone to electrostatic-mediated inhibition in vitro. Here, we employ a murine model to show that this engineered enzyme is superior to wild-type human lysozyme as a treatment for mucoid Pseudomonas aeruginosa lung infections. The engineered enzyme effectively decreases the bacterial burden and reduces markers of inflammation and lung injury. Importantly, we found no evidence of acute toxicity or allergic hypersensitivity upon repeated administration of the engineered biotherapeutic. Thus, the charge-engineered lysozyme represents an interesting therapeutic candidate for P. aeruginosa lung infections.


Asunto(s)
Antibacterianos/farmacología , Pulmón/efectos de los fármacos , Muramidasa/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Recuento de Colonia Microbiana , Citocinas/biosíntesis , Citocinas/inmunología , Glicosaminoglicanos/biosíntesis , Glicosaminoglicanos/metabolismo , Humanos , Inflamación/prevención & control , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Muramidasa/química , Muramidasa/genética , Ingeniería de Proteínas , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/crecimiento & desarrollo , Electricidad Estática
11.
J Breath Res ; 7(1): 016003, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23307645

RESUMEN

The identification of bacteria by their volatilomes is of interest to many scientists and clinicians as it holds the promise of diagnosing infections in situ, particularly lung infections via breath analysis. While there are many studies reporting various bacterial volatile biomarkers or fingerprints using in vitro experiments, it has proven difficult to translate these data to in vivo breath analyses. Therefore, we aimed to create secondary electrospray ionization-mass spectrometry (SESI-MS) pathogen fingerprints directly from the breath of mice with lung infections. In this study we demonstrated that SESI-MS is capable of differentiating infected versus uninfected mice, P. aeruginosa-infected versus S. aureus-infected mice, as well as distinguish between infections caused by P. aeruginosa strains PAO1 versus FRD1, with statistical significance (p < 0.05). In addition, we compared in vitro and in vivo volatiles and observed that only 25-34% of peaks are shared between the in vitro and in vivo SESI-MS fingerprints. To the best of our knowledge, these are the first breath volatiles measured for P. aeruginosa PAO1, FRD1, and S. aureus RN450, and the first comparison of in vivo and in vitro volatile profiles from the same strains using the murine infection model.


Asunto(s)
Pruebas Respiratorias , Pseudomonas aeruginosa/metabolismo , Infecciones del Sistema Respiratorio/microbiología , Staphylococcus aureus/metabolismo , Compuestos Orgánicos Volátiles/metabolismo , Animales , Biomarcadores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/diagnóstico , Espectrometría de Masa por Ionización de Electrospray , Infecciones Estafilocócicas/diagnóstico
12.
J Allergy Clin Immunol ; 128(3): 508-15.e1-2, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21782230

RESUMEN

BACKGROUND: Asthma in obese subjects is poorly understood, and these patients are often refractory to standard therapy. OBJECTIVES: We sought to gain insights into the pathogenesis and treatment of asthma in obese subjects by determining how obesity and bariatric surgery affect asthma control, airway hyperresponsiveness (AHR), and markers of asthmatic inflammation. METHODS: We performed a prospective study of (1) asthmatic and nonasthmatic patients undergoing bariatric surgery compared at baseline and (2) asthmatic patients followed for 12 months after bariatric surgery. RESULTS: We studied 23 asthmatic and 21 nonasthmatic patients undergoing bariatric surgery. At baseline, asthmatic patients had lower FEV(1) and forced vital capacity and lower numbers of lymphocytes in bronchoalveolar lavage fluid. After surgery, asthmatic participants experienced significant improvements in asthma control (asthma control score, 1.55 to 0.74; P < .0001) and asthma quality of life (4.87 to 5.87, P < .0001). Airways responsiveness to methacholine improved significantly (methacholine PC(20), 3.9 to 7.28, P = .03). There was a statistically significant interaction between IgE status and change in airways responsiveness (P for interaction = .01). The proportion of lymphocytes in bronchoalveolar lavage fluid and the production of cytokines from activated peripheral blood CD4(+) T cells increased significantly. CONCLUSIONS: Bariatric surgery improves AHR in obese asthmatic patients with normal serum IgE levels. Weight loss has dichotomous effects on airway physiology and T-cell function typically involved in the pathogenesis of asthma, suggesting that obesity produces a unique phenotype of asthma that will require a distinct therapeutic approach.


Asunto(s)
Asma/complicaciones , Asma/prevención & control , Cirugía Bariátrica/métodos , Hiperreactividad Bronquial , Obesidad/complicaciones , Obesidad/cirugía , Adulto , Asma/inmunología , Asma/fisiopatología , Líquido del Lavado Bronquioalveolar , Linfocitos T CD4-Positivos/inmunología , Estudios Transversales , Femenino , Humanos , Inflamación , Masculino , Cloruro de Metacolina/farmacología , Persona de Mediana Edad , Estudios Prospectivos , Pruebas de Función Respiratoria , Resultado del Tratamiento , Pérdida de Peso
13.
Stem Cells ; 29(7): 1137-48, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21544902

RESUMEN

Bone marrow-derived mesenchymal stromal cells (BMSCs) mitigate inflammation in mouse models of acute lung injury. However, specific mechanisms of BMSC actions on CD4 T lymphocyte-mediated inflammation in vivo remain poorly understood. Limited data suggests promotion of Th2 phenotype in models of Th1-mediated diseases. However, whether this might alleviate or worsen Th2-mediated diseases such as allergic asthma is unknown. To ascertain the effects of systemic administration of BMSCs in a mouse model of Th2-mediated allergic airways inflammation, ovalbumin (OVA)-induced allergic airways inflammation was induced in wild-type C57BL/6 and BALB/c mice as well as in interferon-γ (IFNγ) receptor null mice. Effects of systemic administration during antigen sensitization of either syngeneic or allogeneic BMSC on airways hyperreactivity, lung inflammation, antigen-specific CD4 T lymphocytes, and serum immunoglobulins were assessed. Both syngeneic and allogeneic BMSCs inhibited airways hyperreactivity and lung inflammation through a mechanism partly dependent on IFNγ. However, contrary to existing data, BMSCs did not affect antigen-specific CD4 T lymphocyte proliferation but rather promoted Th1 phenotype in vivo as assessed by both OVA-specific CD4 T lymphocyte cytokine production and OVA-specific circulating immunoglobulins. BMSCs treated to prevent release of soluble mediators and a control cell population of primary dermal skin fibroblasts only partly mimicked the BMSC effects and in some cases worsened inflammation. In conclusion, BMSCs inhibit Th2-mediated allergic airways inflammation by influencing antigen-specific CD4 T lymphocyte differentiation. Promotion of a Th1 phenotype in antigen-specific CD4 T lymphocytes by BMSCs is sufficient to inhibit Th2-mediated allergic airways inflammation through an IFNγ-dependent process.


Asunto(s)
Células Madre Mesenquimatosas/inmunología , Hipersensibilidad Respiratoria/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Inflamación/inmunología , Interferón gamma/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología
14.
Am J Respir Crit Care Med ; 184(3): 345-54, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21562128

RESUMEN

RATIONALE: The opportunistic pathogen Pseudomonas aeruginosa causes both acute and chronic lung infections and is particularly problematic in patients with cystic fibrosis and those undergoing mechanical ventilation. Decreased lung function contributes significantly to morbidity and mortality during P. aeruginosa infection, and damage inflicted by P. aeruginosa virulence factors contributes to lung function decline. OBJECTIVES: We sought to describe direct contribution of a bacterial phospholipase C/sphingomyelinase, PlcHR, to alteration of host lung physiology and characterize a potential therapeutic for protection of lung function. METHODS: We infected C57Bl/6 mice with P. aeruginosa wild-type or isogenic plcHR deletion strains and measured lung function using computer-controlled ventilators. For in vivo testing, miltefosine was delivered intraperitoneally 1 hour after infection. Infection and respiratory endpoints were at 24 hours after infection. MEASUREMENTS AND MAIN RESULTS: P. aeruginosa wild-type infection caused significant lung function impairment, whereas the effects of a ΔplcHR strain infection were much less severe. Surfactometry analysis of bronchoalveolar lavage fluid indicated that PlcHR decreased pulmonary surfactant function. Miltefosine has structural similarity to the PC and sphingomyelin substrates of PlcHR, and we found that it inhibits the cleavage of these choline-containing lipids in vitro. Miltefosine administration after P. aeruginosa infection limited the negative effects of PlcHR activity on lung function. CONCLUSIONS: We have directly linked production of a single virulence factor in P. aeruginosa with effects on lung function, and demonstrated that the inhibitor miltefosine protects lung function from PlcHR-dependent surfactant dysfunction.


Asunto(s)
Fibrosis Quística/microbiología , Infecciones por Pseudomonas/microbiología , Infecciones del Sistema Respiratorio/etiología , Animales , Antifúngicos/administración & dosificación , Antifúngicos/farmacología , Líquido del Lavado Bronquioalveolar/química , Fibrosis Quística/complicaciones , Modelos Animales de Enfermedad , Humanos , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/fisiología , Ratones , Ratones Endogámicos C57BL , Infecciones Oportunistas/microbiología , Fosforilcolina/administración & dosificación , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Respiración Artificial/efectos adversos , Infecciones del Sistema Respiratorio/microbiología
15.
Am J Respir Cell Mol Biol ; 44(1): 11-23, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20118221

RESUMEN

Indoleamine 2,3-dioxygenase (IDO) suppresses the functions of CD4(+) T cells through its ability to metabolize the essential amino acid tryptophan. Although the activity of IDO is required for the immunosuppression of allergic airway disease by the Toll-Like-Receptor 9 (TLR9) agonist, oligonucleotides comprised of cytosine and guanine nucleotides linked by phosphodiester bonds (CpG) DNA, it is unclear whether IDO expression by resident lung epithelial cells is sufficient to elicit these effects. Therefore, we created a transgenic mouse inducibly overexpressing IDO within nonciliated airway epithelial cells. Upon inhalation of formalin-fixed Aspergillus fumigatus hyphal antigens, the overexpression of IDO from airway epithelial cells of these mice reduced the number of CD4(+) T cells within the inflamed lung and impaired the capacity of antigen-specific splenic CD4(+) effector T cells to secrete the cytokines IL-4, IL-5, IL-13, and IFN-γ. Despite these effects, allergic airway disease pathology was largely unaffected in mice expressing IDO in airway epithelium. In support of the concept that dendritic cells are the major cell type contributing to the IDO-inducing effects of CpG DNA, mice expressing TLR9 only in the airway epithelium did not augment IDO expression subsequent to the administration of CpG DNA. Furthermore, the systemic depletion of CD11c(+) cells rendered mice incapable of CpG DNA-induced IDO expression. Our results demonstrate that an overexpression of IDO within the airway epithelium represents a novel mechanism by which the number of CD4(+) T cells recruited to the lung and their capacity to produce cytokines can be diminished in a model of allergic airway disease, and these results also highlight the critical role of dendritic cells in the antiasthmatic effects of IDO induction by CpG DNA.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Pulmón/enzimología , Activación de Linfocitos , Aspergilosis Pulmonar/enzimología , Mucosa Respiratoria/enzimología , Animales , Antígenos Fúngicos/inmunología , Aspergillus fumigatus/inmunología , Pruebas de Provocación Bronquial , Líquido del Lavado Bronquioalveolar/inmunología , Broncoconstricción , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/microbiología , Línea Celular Transformada , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Quinurenina/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/fisiopatología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oligodesoxirribonucleótidos/farmacología , Aspergilosis Pulmonar/inmunología , Aspergilosis Pulmonar/microbiología , Aspergilosis Pulmonar/fisiopatología , Ratas , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Regulación hacia Arriba , Uteroglobina/genética , Uteroglobina/metabolismo
16.
Med Mycol ; 48 Suppl 1: S125-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21067324

RESUMEN

Cystic fibrosis (CF) is a common genetic disease whose major clinical manifestations include repeated episodes of airway infection and inflammation that ultimately result in premature death from respiratory failure. The consequences of infection by individual bacteria have been well studied and the evidence is building that fungal pathogens may be playing an important role in lung disease progression. In contrast, though many CF patients have airway infections characterized by the presence of both bacteria and fungi, our understanding of the impact of such polymicrobial infections on the host is limited. In this review, we discuss what is currently known about incidence of mixed bacterial-fungal infections, and the potential consequences of these mixed infections on the progression of CF lung disease.


Asunto(s)
Infecciones Bacterianas/complicaciones , Fibrosis Quística/complicaciones , Enfermedades Pulmonares Fúngicas/complicaciones , Enfermedades Pulmonares/complicaciones , Pulmón/microbiología , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Fibrosis Quística/microbiología , Hongos/clasificación , Hongos/aislamiento & purificación , Hongos/patogenicidad , Humanos , Incidencia , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares Fúngicas/microbiología , Adulto Joven
17.
ACS Chem Biol ; 5(9): 809-18, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20604527

RESUMEN

Lysozymes contain a disproportionately large fraction of cationic residues, and are thereby attracted toward the negatively charged surface of bacterial targets. Importantly, this conserved biophysical property may inhibit lysozyme antibacterial function during acute and chronic infections. A mouse model of acute pulmonary Pseudomonas aeruginosa infection demonstrated that anionic biopolymers accumulate to high concentrations in the infected lung, and the presence of these species correlates with decreased endogenous lysozyme activity. To develop antibacterial enzymes designed specifically to be used as antimicrobial agents in the infected airway, the electrostatic potential of human lysozyme (hLYS) was remodeled by protein engineering. A novel, high-throughput screen was implemented to functionally interrogate combinatorial libraries of charge-engineered hLYS proteins, and variants with improved bactericidal activity were isolated and characterized in detail. These studies illustrate a general mechanism by which polyanions inhibit lysozyme function, and they are the first direct demonstration that decreasing hLYS's net cationic character improves its antibacterial activity in the presence of disease-associated biopolymers. In addition to avoiding electrostatic sequestration, at least one charge-engineered variant also kills bacteria more rapidly in the absence of inhibitory biopolymers; this observation supports a novel hypothesis that tuning the cellular affinity of peptidoglycan hydrolases may be a general strategy for improving kinetics of bacterial killing.


Asunto(s)
Antibacterianos/química , Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Muramidasa/química , Muramidasa/uso terapéutico , Ingeniería de Proteínas , Infecciones por Pseudomonas/tratamiento farmacológico , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Ratones , Micrococcus luteus/efectos de los fármacos , Modelos Moleculares , Muramidasa/genética , Muramidasa/farmacología , Mutación , Pseudomonas aeruginosa/efectos de los fármacos , Electricidad Estática
18.
Respir Res ; 11: 102, 2010 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-20659336

RESUMEN

BACKGROUND: Nitrogen dioxide (NO2) is an air pollutant associated with poor respiratory health, asthma exacerbation, and an increased likelihood of inhalational allergies. NO2 is also produced endogenously in the lung during acute inflammatory responses. NO2 can function as an adjuvant, allowing for allergic sensitization to an innocuous inhaled antigen and the generation of an antigen-specific Th2 immune response manifesting in an allergic asthma phenotype. As CD11c+ antigen presenting cells are considered critical for naïve T cell activation, we investigated the role of CD11c+ cells in NO2-promoted allergic sensitization. METHODS: We systemically depleted CD11c+ cells from transgenic mice expressing a simian diphtheria toxin (DT) receptor under of control of the CD11c promoter by administration of DT. Mice were then exposed to 15 ppm NO2 followed by aerosolized ovalbumin to promote allergic sensitization to ovalbumin and were studied after subsequent inhaled ovalbumin challenges for manifestation of allergic airway disease. In addition, pulmonary CD11c+ cells from wildtype mice were studied after exposure to NO2 and ovalbumin for cellular phenotype by flow cytometry and in vitro cytokine production. RESULTS: Transient depletion of CD11c+ cells during sensitization attenuated airway eosinophilia during allergen challenge and reduced Th2 and Th17 cytokine production. Lung CD11c+ cells from wildtype mice exhibited a significant increase in MHCII, CD40, and OX40L expression 2 hours following NO2 exposure. By 48 hours, CD11c+MHCII+ DCs within the mediastinal lymph node (MLN) expressed maturation markers, including CD80, CD86, and OX40L. CD11c+CD11b- and CD11c+CD11b+ pulmonary cells exposed to NO2 in vivo increased uptake of antigen 2 hours post exposure, with increased ova-Alexa 647+ CD11c+MHCII+ DCs present in MLN from NO2-exposed mice by 48 hours. Co-cultures of ova-specific CD4+ T cells from naïve mice and CD11c+ pulmonary cells from NO2-exposed mice produced IL-1, IL-12p70, and IL-6 in vitro and augmented antigen-induced IL-5 production. CONCLUSIONS: CD11c+ cells are critical for NO2-promoted allergic sensitization. NO2 exposure causes pulmonary CD11c+ cells to acquire a phenotype capable of increased antigen uptake, migration to the draining lymph node, expression of MHCII and co-stimulatory molecules required to activate naïve T cells, and secretion of polarizing cytokines to shape a Th2/Th17 response.


Asunto(s)
Alérgenos , Asma/inmunología , Antígeno CD11c/metabolismo , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Pulmón/inmunología , Óxido Nítrico/inmunología , Administración por Inhalación , Animales , Asma/prevención & control , Antígeno CD11b/metabolismo , Antígeno CD11c/genética , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Genes Codificadores de los Receptores de Linfocitos T , Factor de Crecimiento Similar a EGF de Unión a Heparina , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ganglios Linfáticos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/administración & dosificación , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Fenotipo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA