Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 291(42): 22118-22135, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27587397

RESUMO

The biochemical mechanisms through which eosinophils contribute to asthma pathogenesis are unclear. Here we show eosinophil peroxidase (EPO), an abundant granule protein released by activated eosinophils, contributes to characteristic asthma-related phenotypes through oxidative posttranslational modification (PTM) of proteins in asthmatic airways through a process called carbamylation. Using a combination of studies we now show EPO uses plasma levels of the pseudohalide thiocyanate (SCN-) as substrate to catalyze protein carbamylation, as monitored by PTM of protein lysine residues into Nϵ-carbamyllysine (homocitrulline), and contributes to the pathophysiological sequelae of eosinophil activation. Studies using EPO-deficient mice confirm EPO serves as a major enzymatic source for protein carbamylation during eosinophilic inflammatory models, including aeroallergen challenge. Clinical studies similarly revealed significant enrichment in carbamylation of airway proteins recovered from atopic asthmatics versus healthy controls in response to segmental allergen challenge. Protein-bound homocitrulline is shown to be co-localized with EPO within human asthmatic airways. Moreover, pathophysiologically relevant levels of carbamylated protein either incubated with cultured human airway epithelial cells in vitro, or provided as an aerosolized exposure in non-sensitized mice, induced multiple asthma-associated phenotypes including induction of mucin, Th2 cytokines, IFNγ, TGFß, and epithelial cell apoptosis. Studies with scavenger receptor-A1 null mice reveal reduced IL-13 generation following exposure to aerosolized carbamylated protein, but no changes in other asthma-related phenotypes. In summary, EPO-mediated protein carbamylation is promoted during allergen-induced asthma exacerbation, and can both modulate immune responses and trigger a cascade of many of the inflammatory signals present in asthma.


Assuntos
Asma/imunologia , Citrulina/análogos & derivados , Peroxidase de Eosinófilo/imunologia , Eosinófilos/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Células A549 , Animais , Asma/patologia , Citrulina/imunologia , Eosinófilos/patologia , Humanos , Interferon gama/imunologia , Interleucina-13/imunologia , Camundongos , Células Th2/imunologia , Células Th2/patologia , Fator de Crescimento Transformador beta/imunologia
2.
Crit Care Med ; 45(2): e132-e137, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27632677

RESUMO

OBJECTIVE: To compare usage patterns and outcomes of a nurse practitioner-staffed medical ICU and a resident-staffed physician medical ICU. DESIGN: Retrospective chart review of 1,157 medical ICU admissions from March 2012 to February 2013. SETTING: Large urban academic university hospital. SUBJECTS: One thousand one hundred fifty-seven consecutive medical ICU admissions including 221 nurse practitioner-staffed medical ICU admissions (19.1%) and 936 resident-staffed medical ICU admissions (80.9%). INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Data obtained included age, gender, race, medical ICU admitting diagnosis, location at time of ICU transfer, code status at ICU admission, and severity of illness using both Acute Physiology and Chronic Health Evaluation II scores and a model for relative expected mortality. Primary outcomes compared included ICU mortality, in-hospital mortality, medical ICU length of stay, and post-ICU discharge hospital length of stay. Patients admitted to the nurse practitioner-staffed medical ICU were older (63 ± 16.5 vs 59.2 ± 16.9 yr for resident-staffed medical ICU; p = 0.019), more likely to be transferred from an inpatient unit (52.0% vs 40.0% for the resident-staffed medical ICU; p = 0.002), and had a higher severity of illness by relative expected mortality (21.3 % vs 17.2 % for the resident-staffed medical ICU; p = 0.001). There were no differences among primary outcomes except for medical ICU length of stay (nurse practitioner-resident-staffed 7.9 ± 7.5 d vs resident-staffed medical ICU 5.6 ± 6.5 d; p = 0.0001). Post-hospital discharge to nonhome location was also significantly higher in the nurse practitioner-ICU (31.7% in nurse practitioner-staffed medical ICU vs 23.9% in resident-staffed medical ICU; p = 0.24). CONCLUSIONS: We found no difference in mortality between an nurse practitioner-staffed medical ICU and a resident-staffed physician medical ICU. Our study adds further evidence that advanced practice providers can render safe and effective ICU care.


Assuntos
Unidades de Terapia Intensiva/estatística & dados numéricos , Internato e Residência/estatística & dados numéricos , Profissionais de Enfermagem/estatística & dados numéricos , Centros Médicos Acadêmicos/estatística & dados numéricos , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Mortalidade Hospitalar , Humanos , Tempo de Internação/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Índice de Gravidade de Doença , Recursos Humanos
3.
Respir Res ; 17(1): 93, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27460362

RESUMO

BACKGROUND: Sarcoidosis is a multisystem immuno-inflammatory disorder of unknown etiology that most commonly involves the lungs. We hypothesized that an unbiased approach to identify pathways activated in bronchoalveolar lavage (BAL) cells can shed light on the pathogenesis of this complex disease. METHODS: We recruited 15 patients with various stages of sarcoidosis and 12 healthy controls. All subjects underwent bronchoscopy with lavage. For each subject, total RNA was extracted from BAL cells and hybridized to an Affymetrix U133A microarray. Rigorous statistical methods were applied to identify differential gene expression between subjects with sarcoidosis vs. CONTROLS: To better elucidate pathways differentially activated between these groups, we integrated network and gene set enrichment analyses of BAL cell transcriptional profiles. RESULTS: Sarcoidosis patients were either non-smokers or former smokers, all had lung involvement and only two were on systemic prednisone. Healthy controls were all non-smokers. Comparison of BAL cell gene expression between sarcoidosis and healthy subjects revealed over 1500 differentially expressed genes. Several previously described immune mediators, such as interferon gamma, were upregulated in the sarcoidosis subjects. Using an integrative computational approach we constructed a modular network of over 80 gene sets that were highly enriched in patients with sarcoidosis. Many of these pathways mapped to inflammatory and immune-related processes including adaptive immunity, T-cell signaling, graft vs. host disease, interleukin 12, 23 and 17 signaling. Additionally, we uncovered a close association between the proteasome machinery and adaptive immunity, highlighting a potentially important and targetable relationship in the pathobiology of sarcoidosis. CONCLUSIONS: BAL cells in sarcoidosis are characterized by enrichment of distinct transcriptional programs involved in immunity and proteasomal processes. Our findings add to the growing evidence implicating alveolar resident immune effector cells in the pathogenesis of sarcoidosis and identify specific pathways whose activation may modulate disease progression.


Assuntos
Líquido da Lavagem Broncoalveolar/citologia , Regulação da Expressão Gênica/genética , Sarcoidose Pulmonar/genética , Sarcoidose Pulmonar/metabolismo , Adulto , Idoso , Broncoscopia , Contagem de Células , Citocinas/metabolismo , Feminino , Redes Reguladoras de Genes/genética , Humanos , Imunidade/genética , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Complexo de Endopeptidases do Proteassoma/genética , RNA/biossíntese , RNA/isolamento & purificação , Sarcoidose Pulmonar/imunologia , Fumar/genética
4.
Am J Respir Cell Mol Biol ; 53(1): 74-86, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25409201

RESUMO

Lipid-laden macrophages, or "foam cells," are observed in the lungs of patients with fibrotic lung disease, but their contribution to disease pathogenesis remains unexplored. Here, we demonstrate that fibrosis induced by bleomycin, silica dust, or thoracic radiation promotes early and sustained accumulation of foam cells in the lung. In the bleomycin model, we show that foam cells arise from neighboring alveolar epithelial type II cells, which respond to injury by dumping lipids into the distal airspaces of the lungs. We demonstrate that oxidized phospholipids accumulate within alveolar macrophages (AMs) after bleomycin injury and that murine and human AMs treated with oxidized phosphatidylcholine (oxPc) become polarized along an M2 phenotype and display enhanced production of transforming growth factor-ß1. The direct instillation of oxPc into the mouse lung induces foam cell formation and triggers a severe fibrotic reaction. Further, we show that reducing pulmonary lipid clearance by targeted deletion of the lipid efflux transporter ATP-binding cassette subfamily G member 1 increases foam cell formation and worsens lung fibrosis after bleomycin. Conversely, we found that treatment with granulocyte-macrophage colony-stimulating factor attenuates fibrotic responses, at least in part through its ability to decrease AM lipid accumulation. In summary, this work describes a novel mechanism leading to foam cell formation in the mouse lung and suggests that strategies aimed at blocking foam cell formation might be effective for treating fibrotic lung disorders.


Assuntos
Células Epiteliais Alveolares/metabolismo , Células Espumosas/metabolismo , Metabolismo dos Lipídeos , Macrófagos Alveolares/metabolismo , Fibrose Pulmonar/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/farmacologia , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Células Espumosas/patologia , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Macrófagos Alveolares/patologia , Camundongos , Camundongos Knockout , Fosfatidilcolinas/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia
6.
Lung ; 192(5): 639-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25108403

RESUMO

Lung cancer is the most common and lethal malignancy in the world. The landmark National lung screening trial (NLST) showed a 20% relative reduction in mortality in high-risk individuals with screening low-dose computed tomography. However, the poor specificity and low prevalence of lung cancer in the NLST provide major limitations to its widespread use. Furthermore, a lung nodule on CT scan requires a nuanced and individualized approach towards management. In this regard, advances in high through-put technology (molecular diagnostics, multi-gene chips, proteomics, and bronchoscopic techniques) have led to discovery of lung cancer biomarkers that have shown potential to complement the current screening standards. Early detection of lung cancer can be achieved by analysis of biomarkers from tissue samples within the respiratory tract such as sputum, saliva, nasal/bronchial airway epithelial cells and exhaled breath condensate or through peripheral biofluids such as blood, serum and urine. Autofluorescence bronchoscopy has been employed in research setting to identify pre-invasive lesions not identified on CT scan. Although these modalities are not yet commercially available in clinic setting, they will be available in the near future and clinicians who care for patients with lung cancer should be aware. In this review, we present up-to-date state of biomarker development, discuss their clinical relevance and predict their future role in lung cancer management.


Assuntos
Biomarcadores Tumorais/metabolismo , Detecção Precoce de Câncer/métodos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/metabolismo , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Doses de Radiação , Tomografia Computadorizada por Raios X , Biomarcadores Tumorais/genética , Humanos , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Valor Preditivo dos Testes , Prognóstico
7.
BMC Immunol ; 14: 41, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24044676

RESUMO

BACKGROUND: Activin A is a pleiotrophic regulatory cytokine, the ablation of which is neonatal lethal. Healthy human alveolar macrophages (AMs) constitutively express activin A, but AMs of patients with pulmonary alveolar proteinosis (PAP) are deficient in activin A. PAP is an autoimmune lung disease characterized by neutralizing autoantibodies to Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF). Activin A can be stimulated, however, by GM-CSF treatment of AMs in vitro. To further explore pulmonary activin A regulation, we examined AMs in bronchoalveolar lavage (BAL) from wild-type C57BL/6 compared to GM-CSF knockout mice which exhibit a PAP-like histopathology. Both human PAP and mouse GM-CSF knockout AMs are deficient in the transcription factor, peroxisome proliferator activated receptor gamma (PPARγ). RESULTS: In sharp contrast to human PAP, activin A mRNA was elevated in mouse GM-CSF knockout AMs, and activin A protein was increased in BAL fluid. Investigation of potential causative factors for activin A upregulation revealed intrinsic overexpression of IFNγ, a potent inducer of the M1 macrophage phenotype, in GM-CSF knockout BAL cells. IFNγ mRNA was not elevated in PAP BAL cells. In vitro studies confirmed that IFNγ stimulated activin A in wild-type AMs while antibody to IFNγ reduced activin A in GM-CSF knockout AMs. Both IFNγ and Activin A were also reduced in GM-CSF knockout mice in vivo after intratracheal instillation of lentivirus-PPARγ compared to control lentivirus vector. Examination of other M1 markers in GM-CSF knockout mice indicated intrinsic elevation of the IFNγ-regulated gene, inducible Nitrogen Oxide Synthetase (iNOS), CCL5, and interleukin (IL)-6 compared to wild-type. The M2 markers, IL-10 and CCL2 were also intrinsically elevated. CONCLUSIONS: Data point to IFNγ as the primary upregulator of activin A in GM-CSF knockout mice which in addition, exhibit a unique mix of M1-M2 macrophage phenotypes.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Macrófagos Alveolares/metabolismo , Proteinose Alveolar Pulmonar/metabolismo , Ativinas/genética , Ativinas/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/citologia , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imuno-Histoquímica , Interferon gama/genética , Interferon gama/metabolismo , Interferon gama/farmacologia , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos Alveolares/classificação , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Proteinose Alveolar Pulmonar/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Int J Mol Sci ; 14(12): 23858-71, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24322444

RESUMO

Sarcoidosis, a chronic granulomatous disease of unknown cause, has been linked to several environmental risk factors, among which are some that may favor carbon nanotube formation. Using gene array data, we initially observed that bronchoalveolar lavage (BAL) cells from sarcoidosis patients displayed elevated mRNA of the transcription factor, Twist1, among many M1-associated genes compared to healthy controls. Based on this observation we hypothesized that Twist1 mRNA and protein expression might become elevated in alveolar macrophages from animals bearing granulomas induced by carbon nanotube instillation. To address this hypothesis, wild-type and macrophage-specific peroxisome proliferator-activated receptor gamma (PPARγ) knock out mice were given oropharyngeal instillation of multiwall carbon nanotubes (MWCNT). BAL cells obtained 60 days later exhibited significantly elevated Twist1 mRNA expression in granuloma-bearing wild-type or PPARγ knock out alveolar macrophages compared to sham controls. Overall, Twist1 expression levels in PPARγ knock out mice were higher than those of wild-type. Concurrently, BAL cells obtained from sarcoidosis patients and healthy controls validated gene array data: qPCR and protein analysis showed significantly elevated Twist1 in sarcoidosis compared to healthy controls. In vitro studies of alveolar macrophages from healthy controls indicated that Twist1 was inducible by classical (M1) macrophage activation stimuli (LPS, TNFα) but not by IL-4, an inducer of alternative (M2) macrophage activation. Findings suggest that Twist1 represents a PPARγ-sensitive alveolar macrophage M1 biomarker which is induced by inflammatory granulomatous disease in the MWCNT model and in human sarcoidosis.


Assuntos
Macrófagos Alveolares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Adulto , Animais , Líquido da Lavagem Broncoalveolar/citologia , Feminino , Humanos , Ativação de Macrófagos , Macrófagos Alveolares/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Nanotubos de Carbono/química , Nanotubos de Carbono/toxicidade , PPAR gama/deficiência , PPAR gama/genética , PPAR gama/metabolismo , RNA Mensageiro/metabolismo , Sarcoidose Pulmonar/induzido quimicamente , Sarcoidose Pulmonar/metabolismo , Sarcoidose Pulmonar/patologia , Proteína 1 Relacionada a Twist/genética , Regulação para Cima
9.
Respir Res ; 13: 46, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22697800

RESUMO

RATIONALE: Pulmonary Alveolar Proteinosis (PAP) patients exhibit an acquired deficiency of biologically active granulocyte-macrophage colony stimulating factor (GM-CSF) attributable to GM-CSF specific autoantibodies. PAP alveolar macrophages are foamy, lipid-filled cells with impaired surfactant clearance and markedly reduced expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) and the PPARγ-regulated ATP binding cassette (ABC) lipid transporter, ABCG1. An open label proof of concept Phase II clinical trial was conducted in PAP patients using rituximab, a chimeric murine-human monoclonal antibody directed against B lymphocyte specific antigen CD20. Rituximab treatment decreased anti-GM-CSF antibody levels in bronchoalveolar lavage (BAL) fluid, and 7/9 patients completing the trial demonstrated clinical improvement as measured by arterial blood oxygenation. OBJECTIVES: This study sought to determine whether rituximab therapy would restore lipid metabolism in PAP alveolar macrophages. METHODS: BAL samples were collected from patients pre- and 6-months post-rituximab infusion for evaluation of mRNA and lipid changes. RESULTS: Mean PPARγ and ABCG1 mRNA expression increased 2.8 and 5.3-fold respectively (p ≤ 0.05) after treatment. Lysosomal phospholipase A2 (LPLA2) (a key enzyme in surfactant degradation) mRNA expression was severely deficient in PAP patients pre-treatment but increased 2.8-fold post-treatment. In supplemental animal studies, LPLA2 deficiency was verified in GM-CSF KO mice but was not present in macrophage-specific PPARγ KO mice compared to wild-type controls. Oil Red O intensity of PAP alveolar macrophages decreased after treatment, indicating reduced intracellular lipid while extracellular free cholesterol increased in BAL fluid. Furthermore, total protein and Surfactant protein A were significantly decreased in the BAL fluid post therapy. CONCLUSIONS: Reduction in GM-CSF autoantibodies by rituximab therapy improves alveolar macrophage lipid metabolism by increasing lipid transport and surfactant catabolism. Mechanisms may involve GM-CSF stimulation of alveolar macrophage ABCG1 and LPLA2 activities by distinct pathways.


Assuntos
Anticorpos Monoclonais Murinos/uso terapêutico , Homeostase , Macrófagos Alveolares/efeitos dos fármacos , Lipídeos de Membrana/fisiologia , Proteinose Alveolar Pulmonar/tratamento farmacológico , Alvéolos Pulmonares/efeitos dos fármacos , Adulto , Animais , Feminino , Homeostase/efeitos dos fármacos , Homeostase/imunologia , Humanos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estudos Prospectivos , Proteinose Alveolar Pulmonar/imunologia , Proteinose Alveolar Pulmonar/patologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Rituximab
10.
Am J Respir Cell Mol Biol ; 45(4): 858-66, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21398620

RESUMO

Lung granulomas are associated with numerous conditions, including inflammatory disorders, exposure to environmental pollutants, and infection. Osteopontin is a chemotactic cytokine produced by macrophages, and is implicated in extracellular matrix remodeling. Furthermore, osteopontin is up-regulated in granulomatous disease, and osteopontin null mice exhibit reduced granuloma formation. Animal models currently used to investigate chronic lung granulomatous inflammation bear a pathological resemblance, but lack the chronic nature of human granulomatous disease. Carbon nanoparticles are generated as byproducts of combustion. Interestingly, experimental exposures to carbon nanoparticles induce pulmonary granuloma-like lesions. However, the recruited cellular populations and extracellular matrix gene expression profiles within these lesions have not been explored. Because of the rapid resolution of granulomas in current animal models, the mechanisms responsible for persistence have been elusive. To overcome the limitations of previous models, we investigated whether a model using multiwall carbon nanoparticles would resemble chronic human lung granulomatous inflammation. We hypothesized that pulmonary exposure to multiwall carbon nanoparticles would induce granulomas, elicit a macrophage and T-cell response, and mimic other granulomatous disorders with an up-regulation of osteopontin. This model demonstrates: (1) granulomatous inflammation, with macrophage and T-cell infiltration; (2) resemblance to the chronicity of human granulomas, with persistence up to 90 days; and (3) a marked elevation of osteopontin, metalloproteinases, and cell adhesion molecules in granulomatous foci isolated by laser-capture microdissection and in alveolar macrophages from bronchoalveolar lavage. The establishment of such a model provides an important platform for mechanistic studies on the persistence of granuloma.


Assuntos
Granuloma/induzido quimicamente , Pulmão/imunologia , Nanotubos de Carbono , Pneumonia/induzido quimicamente , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Moléculas de Adesão Celular/genética , Citocinas/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Granuloma/genética , Granuloma/imunologia , Granuloma/metabolismo , Granuloma/patologia , Mediadores da Inflamação/metabolismo , Integrinas/genética , Lasers , Pulmão/metabolismo , Pulmão/patologia , Macrófagos Alveolares/imunologia , Metaloproteases/genética , Camundongos , Camundongos Endogâmicos C57BL , Microdissecção/instrumentação , Osteopontina/genética , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Fatores de Tempo
11.
Am J Physiol Lung Cell Mol Physiol ; 300(1): L73-80, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21036914

RESUMO

Pulmonary alveolar proteinosis (PAP) is a lung disease characterized by a deficiency of functional granulocyte macrophage colony-stimulating factor (GM-CSF) resulting in surfactant accumulation and lipid-engorged alveolar macrophages. GM-CSF is a positive regulator of PPARγ that is constitutively expressed in healthy alveolar macrophages. We previously reported decreased PPARγ and ATP-binding cassette transporter G1 (ABCG1) levels in alveolar macrophages from PAP patients and GM-CSF knockout (KO) mice, suggesting PPARγ and ABCG1 involvement in surfactant catabolism. Because ABCG1 represents a PPARγ target, we hypothesized that PPARγ restoration would increase ABCG1 and reduce macrophage lipid accumulation. Upregulation of PPARγ was achieved using a lentivirus expression system in vivo. GM-CSF KO mice received intratracheal instillation of lentivirus (lenti)-PPARγ or control lenti-eGFP. Ten days postinstillation, 79% of harvested alveolar macrophages expressed eGFP, demonstrating transduction. Alveolar macrophages showed increased PPARγ and ABCG1 expression after lenti-PPARγ instillation, whereas PPARγ and ABCG1 levels remained unchanged in lenti-eGFP controls. Alveolar macrophages from lenti-PPARγ-treated mice also exhibited reduced intracellular phospholipids and increased cholesterol efflux to HDL, an ABCG1-mediated pathway. In vivo instillation of lenti-PPARγ results in: 1) upregulating ABCG1 and PPARγ expression of GM-CSF KO alveolar macrophages, 2) reducing intracellular lipid accumulation, and 3) increasing cholesterol efflux activity.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , PPAR gama/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Colesterol/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Humanos , Lipídeos/fisiologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , PPAR gama/uso terapêutico , Proteinose Alveolar Pulmonar/tratamento farmacológico , Proteinose Alveolar Pulmonar/genética , Proteinose Alveolar Pulmonar/metabolismo , Surfactantes Pulmonares/metabolismo
12.
J Immunol ; 182(9): 5816-22, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380830

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) is constitutively expressed at high levels in healthy alveolar macrophages, in contrast to other tissue macrophages and blood monocytes. PPARgamma ligands have been shown to down-regulate IFN-gamma-stimulated inducible NO synthase (iNOS) in macrophages. Because NO is an important inflammatory mediator in the lung, we hypothesized that deletion of alveolar macrophage PPARgamma in vivo would result in up-regulation of iNOS and other inflammatory mediators. The loss of PPARgamma in macrophages was achieved by crossing floxed (+/+) PPARgamma mice and a transgenic mouse containing the CRE recombinase gene under the control of the murine M lysozyme promoter (PPARgammaKO). Alveolar macrophages were harvested by bronchoalveolar lavage (BAL). Lymphocytes (CD8:CD4 ratio = 2.8) were increased in BAL of PPARgammaKO vs wild-type C57BL6; p < or = 0.0001. Both iNOS and IFN-gamma expression were significantly elevated (p < or = 0.05) in BAL cells. Th-1 associated cytokines including IL-12 (p40), MIP-1alpha (CCL3), and IFN inducible protein-10 (IP-10, CXCL10) were also elevated. IL-4 and IL-17A were not detected. To test whether these alterations were due to the lack of PPARgamma, PPARgamma KO mice were intratracheally inoculated with a PPARgamma lentivirus construct. PPARgamma transduction resulted in significantly decreased iNOS and IFN-gamma mRNA expression, as well as reduced BAL lymphocytes. These results suggest that lack of PPARgamma in alveolar macrophages disrupts lung homeostasis and results in a Th1-like inflammatory response.


Assuntos
Deleção de Genes , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/patologia , Macrófagos Alveolares/imunologia , PPAR gama/deficiência , Células Th1/imunologia , Células Th1/patologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia , Células Cultivadas , Homeostase/genética , Homeostase/imunologia , Humanos , Mediadores da Inflamação/fisiologia , Lentivirus/genética , Lentivirus/imunologia , Pulmão/virologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/fisiologia , PPAR gama/genética , Células Th1/virologia , Transdução Genética , Regulação para Cima/genética , Regulação para Cima/imunologia
13.
J Lipid Res ; 51(6): 1325-31, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20064973

RESUMO

Surfactant accumulates in alveolar macrophages of granulocyte-macrophage colony-stimulating factor (GM-CSF) knockout (KO) mice and pulmonary alveolar proteinosis (PAP) patients with a functional loss of GM-CSF resulting from neutralizing anti-GM-CSF antibody. Alveolar macrophages from PAP patients and GM-CSF KO mice are de-ficient in peroxisome proliferator-activated receptor-gamma (PPARgamma) and ATP-binding cassette (ABC) lipid transporter ABCG1. Previous studies have demonstrated that GM-CSF induces PPARgamma. We therefore hypothesized that PPARgamma promotes surfactant catabolism through regulation of ABCG1. To address this hypothesis, macrophage-specific PPARgamma (MacPPARgamma) knockout mice were utilized. MacPPARgamma KO mice develop foamy, lipid-engorged Oil Red O positive alveolar macrophages. Lipid analyses revealed significant increases in the cholesterol and phospholipid contents of MacPPARgamma KO alveolar macrophages and extracellular bronchoalveolar lavage (BAL)-derived fluids. MacPPARgamma KO alveolar macrophages showed decreased expression of ABCG1 and a deficiency in ABCG1-mediated cholesterol efflux to HDL. Lipid metabolism may also be regulated by liver X receptor (LXR)-ABCA1 pathways. Interestingly, ABCA1 and LXRbeta expression were elevated, indicating that this pathway is not sufficient to prevent surfactant accumulation in alveolar macrophages. These results suggest that PPARgamma mediates a critical role in surfactant homeostasis through the regulation of ABCG1.


Assuntos
Macrófagos Alveolares/metabolismo , PPAR gama/deficiência , PPAR gama/genética , Tensoativos/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Colesterol/metabolismo , Técnicas de Inativação de Genes , Lipoproteínas/metabolismo , Lipoproteínas HDL/metabolismo , Receptores X do Fígado , Pulmão/metabolismo , Camundongos , Especificidade de Órgãos , Receptores Nucleares Órfãos/metabolismo
14.
Biochem Biophys Res Commun ; 393(4): 682-7, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20170635

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a nuclear transcription factor involved in lipid metabolism that is constitutively expressed in the alveolar macrophages of healthy individuals. PPARgamma has recently been implicated in the catabolism of surfactant by alveolar macrophages, specifically the cholesterol component of surfactant while the mechanism remains unclear. Studies from other tissue macrophages have shown that PPARgamma regulates cholesterol influx, efflux, and metabolism. PPARgamma promotes cholesterol efflux through the liver X receptor-alpha (LXRalpha) and ATP-binding cassette G1 (ABCG1). We have recently shown that macrophage-specific PPARgamma knockout (PPARgamma KO) mice accumulate cholesterol-laden alveolar macrophages that exhibit decreased expression of LXRalpha and ABCG1 and reduced cholesterol efflux. We hypothesized that in addition to the dysregulation of these cholesterol efflux genes, the expression of genes involved in cholesterol synthesis and influx was also dysregulated and that replacement of PPARgamma would restore regulation of these genes. To investigate this hypothesis, we have utilized a Lentivirus expression system (Lenti-PPARgamma) to restore PPARgamma expression in the alveolar macrophages of PPARgamma KO mice. Our results show that the alveolar macrophages of PPARgamma KO mice have decreased expression of key cholesterol synthesis genes and increased expression of cholesterol receptors CD36 and scavenger receptor A-I (SRA-I). The replacement of PPARgamma (1) induced transcription of LXRalpha and ABCG1; (2) corrected suppressed expression of cholesterol synthesis genes; and (3) enhanced the expression of scavenger receptors CD36. These results suggest that PPARgamma regulates cholesterol metabolism in alveolar macrophages.


Assuntos
Colesterol/metabolismo , Regulação da Expressão Gênica , Metabolismo dos Lipídeos/genética , Macrófagos Alveolares/metabolismo , PPAR gama/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Antígenos CD36/genética , Colesterol/genética , Lipoproteínas/genética , Receptores X do Fígado , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos/genética , PPAR gama/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Regulação para Cima
15.
Curr Opin Pulm Med ; 15(5): 491-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19561506

RESUMO

PURPOSE OF REVIEW: This review discusses the most recent clinical and basic research literature on pulmonary alveolar proteinosis (PAP) as it relates to pathogenesis, diagnosis, and management. RECENT FINDINGS: The discovery of Granulocyte macrophage-colony stimulating factor (GM-CSF) and the alveolar macrophage as critical regulators of surfactant protein and lipid homeostasis has led to significant advances in PAP. Adults affected by PAP have circulating neutralizing anti-GM-CSF antibodies. Reduced localized GM-CSF activity in the lung (from neutralizing anti-GM-CSF antibodies), decreases alveolar macrophage surfactant degradation with surfactant excess and accumulation. Cause, source of antibodies or downstream effects of GM-CSF deficiency is speculative. GM-CSF antibodies above a threshold level have proved to be a useful diagnostic test. Research towards therapy has focused on improving the technique for therapeutic whole lung lavage as well as overcoming effects of neutralizing anti-GM-CSF, which include GM-CSF therapy (systemic and inhaled) and anecdotal reports of anti-B cell therapy. Whereas this approach has been somewhat successful for primary PAP, other causes of PAP (i.e. alveolar macrophage dysfunction, surfactant protein alterations) are still without therapy. SUMMARY: Understanding of the pathogenesis of PAP has greatly increased in the last decade; study has brought better comprehension of lung biology and recognition of the critical role for GM-CSF and alveolar macrophage in surfactant clearance. Balance between resident immune cell population and normal lung function still needs further study. Resident alveolar macrophages have an essential role in surfactant homeostasis. With this knowledge more effective diagnostic tests (e.g. anti-GM-CSF antibody) and therapies for PAP are under investigation.


Assuntos
Linfócitos B/imunologia , Lavagem Broncoalveolar/métodos , Imunidade Celular/imunologia , Imunossupressores/uso terapêutico , Proteinose Alveolar Pulmonar , Biópsia , Líquido da Lavagem Broncoalveolar/química , Broncoscopia , Diagnóstico Diferencial , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Macrófagos Alveolares/metabolismo , Proteinose Alveolar Pulmonar/diagnóstico , Proteinose Alveolar Pulmonar/imunologia , Proteinose Alveolar Pulmonar/terapia , Tomografia Computadorizada por Raios X
16.
Clin Dermatol ; 25(3): 267-75, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17560304

RESUMO

Susceptibility to most human diseases is polygenic, with complex interactions between functional polymorphisms of single genes governing disease incidence, phenotype, or both. In this context, the contribution of any discrete gene is generally modest for a single individual, but may confer substantial attributable risk on a population level. Environmental exposure can modify the effects of a polymorphism, either by providing a necessary substrate for development of human disease or because the effects of a given exposure modulate the effects of the gene. In several diseases, genetic polymorphisms have been shown to be context dependent, ie, the effects of a genetic variant are realized only in the setting of a relevant exposure. Because sarcoidosis susceptibility is dependent on both genetic and environmental modifiers, the study of gene-environment interactions may yield important pathogenetic information and will likely be crucial for uncovering the range of genetic susceptibility loci. The complexity of these relationships implies, however, that investigations of gene-environment interactions will require the study of large cohorts with carefully defined exposures and similar clinical phenotypes. A general principle is that the study of gene-environment interactions requires a sample size at least severalfold greater than for either factor alone. To date, the presence of environmental modifiers has been demonstrated for one sarcoidosis susceptibility locus, HLA-DQB1, in African-American families. This article reviews general considerations obtaining for the study of gene-environment interactions in sarcoidosis. It also describes the limited current understanding of the role of environmental influences on sarcoidosis susceptibility genes.


Assuntos
Exposição Ambiental , Sarcoidose/epidemiologia , Sarcoidose/genética , Análise por Conglomerados , Predisposição Genética para Doença , Genótipo , Antígenos HLA/imunologia , Humanos , Risco , Sarcoidose/imunologia
17.
J Leukoc Biol ; 79(1): 133-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16275889

RESUMO

Pulmonary alveolar proteinosis (PAP) is an anti-granulocyte macrophage-colony stimulating factor (GM-CSF) autoimmune disease resulting in the accumulation of phospholipids in the alveoli. GM-CSF knockout (KO) mice exhibit a strikingly similar lung pathology to patients with PAP. The lack of functionally active GM-CSF correlates with highly elevated concentrations of M-CSF in the lungs of PAP patients and GM-CSF KO mice. M-CSF has been associated with alternative macrophage activation, and in models of pulmonary fibrosis, M-CSF also contributes to tissue resorption and fibrosis. Matrix metalloproteinase-2 (MMP-2) and MMP-9 have been implicated in extracellular matrix degradation in animal models of fibrosis and asthma. We show for the first time that the lungs of PAP patients contain highly elevated levels of MMP-2 and MMP-9. PAP broncholaveolar lavage (BAL) cells but not bronchial epithelial cells expressed increased MMP-2 and MMP-9 mRNA relative to healthy controls. Both MMPs were detectable as pro and active proteins by gelatin zymography; and by fluorometric global assay, PAP-MMP activity was elevated. BAL cells/fluids from GM-CSF KO mice also demonstrated significantly elevated MMP-2 and MMP-9 gene expression, protein, and activity. Finally, PAP patients undergoing GM-CSF therapy exhibited significantly reduced MMPs and M-CSF. These data suggest that in the absence of GM-CSF, excess M-CSF in PAP may redirect alveolar macrophage activation, thus potentially contributing to elevated MMP expression in the lung.


Assuntos
Regulação Enzimológica da Expressão Gênica/imunologia , Pulmão/enzimologia , Ativação de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos Alveolares/enzimologia , Proteinose Alveolar Pulmonar/enzimologia , Animais , Lavagem Broncoalveolar , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Gelatinases , Regulação Enzimológica da Expressão Gênica/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Pulmão/imunologia , Pulmão/patologia , Ativação de Macrófagos/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Metaloproteinases da Matriz , Camundongos , Camundongos Knockout , Proteinose Alveolar Pulmonar/tratamento farmacológico , Proteinose Alveolar Pulmonar/patologia
18.
Chest ; 151(4): 930-935, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28089817

RESUMO

Health care is at a crossroads and under pressure to add value by improving patient experience and health outcomes and reducing costs to the system. Efforts to improve the care model in primary care, such as the patient-centered medical home, have enjoyed some success. However, primary care accounts for only a small portion of total health-care spending, and there is a need for policies and frameworks to support high-quality, cost-efficient care in specialty practices of the medical neighborhood. The Patient-Centered Specialty Practice (PCSP) model offers ambulatory-based specialty practices one such framework, supported by a formal recognition program through the National Committee for Quality Assurance. The key elements of the PCSP model include processes to support timely access to referral requests, improved communication and coordination with patients and referring clinicians, reduced unnecessary and duplicative testing, and an emphasis on continuous measurement of quality, safety, and performance improvement for a population of patients. Evidence to support the model remains limited, and estimates of net costs and value to practices are not fully understood. The PCSP model holds promise for promoting value-based health care in specialty practices. The continued development of appropriate incentives is required to ensure widespread adoption.


Assuntos
Assistência Centrada no Paciente/organização & administração , Papel do Médico , Administração da Prática Médica/organização & administração , Especialização , Aquisição Baseada em Valor , Assistência Ambulatorial/organização & administração , Humanos , Qualidade da Assistência à Saúde , Estados Unidos
19.
Chest ; 130(1): 227-37, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16840407

RESUMO

Pulmonary alveolar proteinosis (PAP) is a rare idiopathic autoimmune lung disease in adults characterized by the accumulation of lipoproteinaceous material within the alveoli of the lung. The natural history of this disease is poorly defined. Current therapy of bilateral whole-lung lavage (WLL) under general anesthesia is invasive and has its limitations. Data suggest that relative granulocyte macrophage colony stimulating factor (GM-CSF) deficiency may be involved in the pathogenesis of this disease. There have been several case series that have described clinical improvement with exogenous GM-CSF therapy in a subset of patients with PAP. We describe the results of a prospective, open-label clinical trial of daily subcutaneous GM-CSF therapy in a group of adult patients with idiopathic PAP. In this series of 25 patients, the largest reported to date, administration of GM-CSF improved oxygenation as assessed by a 10 mm Hg decrease in alveolar-arterial oxygen gradient, as well as improvement in other clinical and quality of life parameters in 12 of 25 patients (48%) with moderate symptomatic disease who completed the trial. In addition, the serum anti-GM-CSF antibody titer correlated with lung disease activity and was a predictor for responsiveness to therapy. These data indicate that subcutaneous GM-CSF therapy is a promising alternative to WLL for symptomatic patients with PAP.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Proteinose Alveolar Pulmonar/tratamento farmacológico , Adulto , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Proteinose Alveolar Pulmonar/fisiopatologia , Qualidade de Vida , Resultado do Tratamento
20.
Chest ; 130(3): 855-62, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16963686

RESUMO

STUDY OBJECTIVE: To test the hypothesis that sibling pairs, who share genes and environmental exposures, might have similar phenotypic expressions of sarcoidosis beyond what would be expected by chance alone. DESIGN: Multicenter family study with study subjects recruited from 11 clinical centers. SUBJECTS: Subjects were African-American sibling pairs with sarcoidosis. Sarcoidosis and organ pattern involvement were defined according to specific criteria. Fifteen different organ systems were evaluated. RESULTS: For full-sibling pairs, ocular involvement was found in both siblings more often than expected by chance alone (p < 0.05), but the concordance was weak (kappa = 0.18). When analyzing full-sibling and half-sibling pairs, ocular and liver involvement showed a significant concordance between sibling pairs (p < 0.05), but again the agreement was poor (kappa = 0.16 for both). Concordance in pulmonary function change over time was also weak. Clinical outcomes of sibling pairs were not significantly correlated except for whether treatment was prescribed, and this level of agreement was poor (kappa = 0.14 for full-sibling and half-sibling pairs; kappa = 0.15 for full-sibling pairs only). Modeling phenotypic expression in sibling pairs using logistic regression did show that the presence of ocular and liver sarcoidosis in the first affected sibling conferred a statistically significant increased risk to the second affected sibling for having those organs involved (odds ratio [OR], 3; 95% confidence interval [CI], 1.7 to 5.4 for ocular; OR, 3.3; 95% CI, 1.5 to 7.4 for liver). CONCLUSIONS: The phenotypic features and clinical outcomes of sarcoidosis in sibling pairs show minimal concordance, with the possible exception that the presence of ocular or liver involvement in the first sibling with a diagnosis of sarcoidosis makes involvement of these organs more likely in other affected siblings.


Assuntos
Negro ou Afro-Americano/genética , Fenótipo , Sarcoidose/genética , Adulto , Idoso , Exposição Ambiental , Olho/patologia , Olho/fisiopatologia , Feminino , Humanos , Fígado/patologia , Fígado/fisiopatologia , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Sarcoidose/patologia , Sarcoidose/fisiopatologia , Irmãos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA