Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
N Engl J Med ; 377(21): 2024-2035, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29099333

RESUMEN

BACKGROUND: Cystic fibrosis is an autosomal recessive disease caused by mutations in the CFTR gene that lead to progressive respiratory decline. Some mutant CFTR proteins show residual function and respond to the CFTR potentiator ivacaftor in vitro, whereas ivacaftor alone does not restore activity to Phe508del mutant CFTR. METHODS: We conducted a randomized, double-blind, placebo-controlled, phase 3, crossover trial to evaluate the efficacy and safety of ivacaftor alone or in combination with tezacaftor, a CFTR corrector, in 248 patients 12 years of age or older who had cystic fibrosis and were heterozygous for the Phe508del mutation and a CFTR mutation associated with residual CFTR function. Patients were randomly assigned to one of six sequences, each involving two 8-week intervention periods separated by an 8-week washout period. They received tezacaftor-ivacaftor, ivacaftor monotherapy, or placebo. The primary end point was the absolute change in the percentage of predicted forced expiratory volume in 1 second (FEV1) from the baseline value to the average of the week 4 and week 8 measurements in each intervention period. RESULTS: The number of analyzed intervention periods was 162 for tezacaftor-ivacaftor, 157 for ivacaftor alone, and 162 for placebo. The least-squares mean difference versus placebo with respect to the absolute change in the percentage of predicted FEV1 was 6.8 percentage points for tezacaftor-ivacaftor and 4.7 percentage points for ivacaftor alone (P<0.001 for both comparisons). Scores on the respiratory domain of the Cystic Fibrosis Questionnaire-Revised, a quality-of-life measure, also significantly favored the active-treatment groups. The incidence of adverse events was similar across intervention groups; most events were mild or moderate in severity, with no discontinuations of the trial regimen due to adverse events for tezacaftor-ivacaftor and few for ivacaftor alone (1% of patients) and placebo (<1%). CONCLUSIONS: CFTR modulator therapy with tezacaftor-ivacaftor or ivacaftor alone was efficacious in patients with cystic fibrosis who were heterozygous for the Phe508del deletion and a CFTR residual-function mutation. (Funded by Vertex Pharmaceuticals and others; EXPAND ClinicalTrials.gov number, NCT02392234 .).


Asunto(s)
Aminofenoles/uso terapéutico , Benzodioxoles/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Indoles/uso terapéutico , Quinolonas/uso terapéutico , Adolescente , Adulto , Aminofenoles/efectos adversos , Aminofenoles/farmacología , Benzodioxoles/efectos adversos , Benzodioxoles/farmacología , Niño , Estudios Cruzados , Fibrosis Quística/genética , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos adversos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/farmacología , Método Doble Ciego , Combinación de Medicamentos , Femenino , Volumen Espiratorio Forzado/efectos de los fármacos , Heterocigoto , Humanos , Indoles/efectos adversos , Indoles/farmacología , Masculino , Mutación , Calidad de Vida , Quinolonas/efectos adversos , Quinolonas/farmacología , Adulto Joven
2.
N Engl J Med ; 377(21): 2013-2023, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29099344

RESUMEN

BACKGROUND: Combination treatment with the cystic fibrosis transmembrane conductance regulator (CFTR) modulators tezacaftor (VX-661) and ivacaftor (VX-770) was designed to target the underlying cause of disease in patients with cystic fibrosis. METHODS: In this phase 3, randomized, double-blind, multicenter, placebo-controlled, parallel-group trial, we evaluated combination therapy with tezacaftor and ivacaftor in patients 12 years of age or older who had cystic fibrosis and were homozygous for the CFTR Phe508del mutation. Patients were randomly assigned in a 1:1 ratio to receive either 100 mg of tezacaftor once daily and 150 mg of ivacaftor twice daily or matched placebo for 24 weeks. The primary end point was the absolute change in the percentage of the predicted forced expiratory volume in 1 second (FEV1) through week 24 (calculated in percentage points); relative change in the percentage of the predicted FEV1 through week 24 (calculated as a percentage) was a key secondary end point. RESULTS: Of the 510 patients who underwent randomization, 509 received tezacaftor-ivacaftor or placebo, and 475 completed 24 weeks of the trial regimen. The mean FEV1 at baseline was 60.0% of the predicted value. The effects on the absolute and relative changes in the percentage of the predicted FEV1 in favor of tezacaftor-ivacaftor over placebo were 4.0 percentage points and 6.8%, respectively (P<0.001 for both comparisons). The rate of pulmonary exacerbation was 35% lower in the tezacaftor-ivacaftor group than in the placebo group (P=0.005). The incidence of adverse events was similar in the two groups. Most adverse events were of mild severity (in 41.8% of patients overall) or moderate severity (in 40.9% overall), and serious adverse events were less frequent with tezacaftor-ivacaftor (12.4%) than with placebo (18.2%). A total of 2.9% of patients discontinued the assigned regimen owing to adverse events. Fewer patients in the tezacaftor-ivacaftor group than in the placebo group had respiratory adverse events, none of which led to discontinuation. CONCLUSIONS: The combination of tezacaftor and ivacaftor was efficacious and safe in patients 12 years of age or older who had cystic fibrosis and were homozygous for the CFTR Phe508del mutation. (Funded by Vertex Pharmaceuticals; EVOLVE ClinicalTrials.gov number, NCT02347657 .).


Asunto(s)
Aminofenoles/uso terapéutico , Benzodioxoles/uso terapéutico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Indoles/uso terapéutico , Quinolonas/uso terapéutico , Adolescente , Adulto , Aminofenoles/efectos adversos , Aminofenoles/farmacología , Benzodioxoles/efectos adversos , Benzodioxoles/farmacología , Índice de Masa Corporal , Niño , Fibrosis Quística/genética , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos adversos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/farmacología , Método Doble Ciego , Combinación de Medicamentos , Femenino , Volumen Espiratorio Forzado/efectos de los fármacos , Homocigoto , Humanos , Indoles/efectos adversos , Indoles/farmacología , Masculino , Mutación , Calidad de Vida , Quinolonas/efectos adversos , Quinolonas/farmacología , Adulto Joven
3.
Chest ; 148(6): 1489-1496, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26226215

RESUMEN

BACKGROUND: High bronchodilator reversibility in adult asthma is associated with distinct clinical characteristics. This analysis compares lung function, biomarker profiles, and disease control in patients with high reversibility (HR) and low reversibility (LR) asthma. METHODS: A retrospective analysis was performed with data from two completed clinical trials of similar design. Patients were divided into HR and LR subgroups based on their response to bronchodilators (HR = ΔFEV1 postbronchodilator ≥ 20%). Blood eosinophil count, serum IgE level, and fraction of exhaled nitric oxide concentration, biomarkers commonly used to stratify patients into T-helper (Th)-2-high vs Th2-low phenotypes, were measured in patients with not well controlled (1.5 ≤ Asthma Control Questionnaire [ACQ] ≤ 2.143) and very poorly controlled (ACQ > 2.143) disease. RESULTS: The majority of patients in the HR and LR subgroups displayed Th2-low biomarker profiles and very poor disease control. HR was more frequently associated with Th2-high biomarker profiles (40.1% vs 29.4%, P = .006), lower lung function (FEV1, 63.5 ± 7.7% predicted vs 67.9 ± 8.4% predicted; P < .001), and atopy (93.7% vs 86.5%, P = .005). CONCLUSIONS: HR is a physiologic indicator of reduced lung function and is more often associated with elevations in Th2 biomarkers than LR in moderate to severe asthma. However, the majority of patients with HR and LR asthma in this analysis had a Th2-low biomarker profile. Moreover, a Th2-high biomarker profile was not associated with worse disease control.


Asunto(s)
Asma , Broncodilatadores/farmacología , Eosinófilos/inmunología , Glucocorticoides/farmacología , Inmunoglobulina E/sangre , Óxido Nítrico/análisis , Sistema Respiratorio , Células Th2/inmunología , Adulto , Asma/sangre , Asma/diagnóstico , Asma/tratamiento farmacológico , Asma/fisiopatología , Biomarcadores/análisis , Biomarcadores/sangre , Pruebas Respiratorias/métodos , Monitoreo de Drogas/métodos , Femenino , Humanos , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Prostaglandina D2/antagonistas & inhibidores , Pruebas de Función Respiratoria/métodos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/inmunología , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Estadística como Asunto , Resultado del Tratamiento
4.
Transl Res ; 163(4): 363-76, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24316173

RESUMEN

Lung regeneration research is yielding data with increasing translational value. The classical models of lung development, postnatal alveolarization, and postpneumonectomy alveolarization have contributed to a broader understanding of the cellular participants including stem-progenitor cells, cell-cell signaling pathways, and the roles of mechanical deformation and other physiologic factors that have the potential to be modulated in human and animal patients. Although recent information is available describing the lineage fate of lung fibroblasts, genetic fate mapping, and clonal studies are lacking in the study of lung regeneration and deserve further examination. In addition to increasing knowledge concerning classical alveolarization (postnatal, postpneumonectomy), there is increasing evidence for remodeling of the adult lung after partial pneumonectomy. Though limited in scope, compelling data have emerged describing restoration of lung tissue mass in the adult human and in large animal models. The basis for this long-term adaptation to pneumonectomy is poorly understood, but investigations into mechanisms of lung regeneration in older animals that have lost their capacity for rapid re-alveolarization are warranted, as there would be great translational value in modulating these mechanisms. In addition, quantitative morphometric analysis has progressed in conjunction with developments in advanced imaging, which allow for longitudinal and nonterminal evaluation of pulmonary regenerative responses in animals and humans. This review focuses on the cellular and molecular events that have been observed in animals and humans after pneumonectomy because this model is closest to classical regeneration in other mammalian systems and has revealed several new fronts of translational research that deserve consideration.


Asunto(s)
Pulmón/fisiología , Regeneración/fisiología , Investigación Biomédica Traslacional/tendencias , Animales , Humanos , Neumonectomía
5.
Stem Cells Dev ; 22(24): 3214-25, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23895415

RESUMEN

Aging is a critical determinant of regenerative capacity in many organ systems, but it remains unresolved in the lung. This study examines murine lung cell dynamics during age-dependent lung regeneration. Proliferation of lung progenitor cells (EpCAM(neg)/Sca-1(high) lung mesenchymal stromal cells - LMSCs, EpCAM(pos)/Sca-1(low) epithelial progenitor cells, proSP-C(pos) alveolar type II epithelial cells - AECII, and CD31(pos) - endothelial cells) was tracked to day 3 or 7 after pneumonectomy (PNX) or SHAM surgery in 3, 9, and 17 month mice. In 3 month mice, post-PNX LMSC proliferation peaked early (3 days), with 50%-80% more BrdU-positive cells than the other cell types, which peaked later (4-7 days). In older mice (9 and 17 month), abundance and post-PNX proliferation of LMSCs at day 3 were reduced (40%-80%). In both young and old mice, LMSCs were isolated and compared phenotypically with whole lung non-LMSCs. Donor age had no qualitative effect on the phenotype (LMSC vs. non-LMSC), with increased expression of CD90/Thy1, CD105/Eng, CD106/Vcam, CD146/Mcam, and Pdgfrα, and up-regulation of mRNA encoding Fap, Eln, Col1a1, Col3a1, Aldh1a3, Arhgef25, Dner, Fgfr1, and Midkine. However, compared with LMSCs isolated from young mice, LMSCs from older mice exhibited reduced mRNA expression of retinoic acid (Aldh1a3, Rbp4), Fgf/Wnt (Fgfr1, Sfrp1, Wnt2, and Ctnnb1), and elastogenesis (Col1a1, Eln, Fbn1, and Sdc2) pathway genes. Isolated LMSCs from older mice also demonstrated lower colony-forming units (-67%), growth potential (-60% by day 7), ALDH activity (-49%), and telomerase activity (-47%). Therefore, age is associated with declining proliferative potential and regenerative functions of LMSCs in the lung.


Asunto(s)
Envejecimiento/genética , Diferenciación Celular , Pulmón/citología , Células Madre Mesenquimatosas/citología , Animales , Proliferación Celular , Células Madre Mesenquimatosas/metabolismo , Ratones , Neumonectomía , Regeneración/genética , Células Madre/citología
6.
J Tissue Eng Regen Med ; 7(7): 572-83, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22585451

RESUMEN

Compensatory growth is mediated by multiple cell types that interact during organ repair. To elucidate the relationship between stem/progenitor cells that proliferate or differentiate and somatic cells of the lung, we used a novel organotypic ex vivo pneumoexplant system. Applying this technique, we identified a sustained culture of repopulating adult progenitors in the form of free-floating anchorage-independent cells (AICs). AICs did not express integrin proteins α5, ß3 and ß7, and constituted 37% of the total culture at day 14, yielding a mixed yet conservative population that recapitulated RNA expression patterns of the healthy lung. AICs exhibited rapid proliferation manifested by a marked 60-fold increase in cell numbers by day 21. More than 50% of the AIC population was c-KIT(+) or double-positive for CD45(+) and CD11b(+) antigenic determinants, consistent with cells of hematopoietic origin. The latter subset was found to be enriched with prosurfactant protein-C and SCGB1A1 expressing putative stem cells and with aquaporin-5 producing cells, characteristic of terminally differentiated alveolar epithelial type-1 pneumocytes. At the air/gel interface, AICs undergo remodeling to form a cellular lining, whereas TGF(ß)1 treatment modifies protein expression properties to further imply a robust effect of the microenvironment on AIC phenotypic changes. These data confirm the active participation of clonogenic hematopoietic stem cells in a mammalian model of lung repair and validate mixed stem/somatic cell cultures, which license sustained cell viability, proliferation and differentiation, for use in studies of compensatory pulmonary growth.


Asunto(s)
Células Madre Adultas/metabolismo , Células Epiteliales Alveolares/metabolismo , Antígeno CD11b , Células Epiteliales/metabolismo , Antígenos Comunes de Leucocito , Pulmón/metabolismo , Adulto , Células Madre Adultas/citología , Células Epiteliales Alveolares/citología , Animales , Proliferación Celular , Células Cultivadas , Células Epiteliales/citología , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Integrinas/biosíntesis , Pulmón/citología , Masculino
7.
Front Physiol ; 3: 387, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23060811

RESUMEN

RATIONALE: In clinical trials, homogeneous emphysema patients have responded well to upper lobe volume reduction but not lower lobe volume reduction. MATERIALS/METHODS: To understand the physiological basis for this observation, a computer model was developed to simulate the effects of upper and lower lobe lung volume reduction on RV/TLC and lung recoil in homogeneous emphysema. RESULTS: Patients with homogeneous emphysema received either upper or lower lobe volume reduction therapy based on findings of radionucleotide scintigraphy scanning. CT analysis of lobar volumes showed that patients undergoing upper (n = 18; -265 mL/site) and lower lobe treatment (LLT; n = 11; -217 mL/site) experienced similar reductions in lung volume. However, only upper lobe treatment (ULT) improved FEV(1) (+11.1 ± 14.7 versus -4.4 ± 15.8%) and RV/TLC (-5.4 ± 8.1 versus -2.4 ± 8.6%). Model simulations provided an unexpected explanation for this response. Increases in transpulmonary pressure subsequent to volume reduction increased RV/TLC in upper lobe alveoli, while caudal shifts in airway closure decreased RV/TLC in lower lobe alveoli. ULT, which eliminates apical alveoli with high RV/TLC values, lowers the average RV/TLC of the lung. Conversely, LLT, which eliminates caudal alveoli with low RV/TLC values, has less effect. CONCLUSION: LLT in homogeneous emphysema is uniformly less effective than ULT.

8.
Cell Transplant ; 21(1): 175-89, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21294955

RESUMEN

Autologous lung-derived mesenchymal stem cells (LMSCs) were transplanted endoscopically into sheep with experimental emphysema to assess their capacity to regenerate functional tissue. LMSC lines were derived from transbronchial biopsies, cloned at passage 2, expanded in culture, and labeled. A delivery scaffold containing 1% fibrinogen, 20 µg/ml of fibronectin, and 20 µg/ml of poly-L-lysine was used to promote cell attachment and spreading. Treatment animals received scaffold containing 5-10 × 10(6) cells/site; control animals received scaffold alone. Phenotypic markers, differentiation capacity, extracellular matrix protein expression, and paracrine function of LMSCs were characterized in vitro. Responses to LMSC transplantation in vivo were assessed in terms of clinical toxicity, lung physiology, change in tissue mass (measured by CT scanning) and perfusion (measured by scintigraphy scanning), and tissue histology. At 4-week follow-up, transplants were well tolerated and associated with increased tissue mass and lung perfusion compared to control treatment. Histology confirmed cell retention, increased cellularity, and increased extracellular matrix content following LMSC treatment. Labeled cells were distributed in the alveolar septum and peribronchiolar interstitium. Some label was also present within phagocytes, indicating that a fraction of autologous LMSCs do not survive transplantation. These results suggest that endobronchial delivery of autologous LMSCs has potential therapeutic utility for regenerating functional lung in emphysema.


Asunto(s)
Enfisema/terapia , Pulmón/citología , Pulmón/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Enfisema/fisiopatología , Células Madre Mesenquimatosas/metabolismo , Regeneración , Ovinos
9.
PLoS One ; 6(8): e23232, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21912590

RESUMEN

While aging leads to a reduction in the capacity for regeneration after pneumonectomy (PNX) in most mammals, this biological phenomenon has not been characterized over the lifetime of mice. We measured the age-specific (3, 9, 24 month) effects of PNX on physiology, morphometry, cell proliferation and apoptosis, global gene expression, and lung fibroblast phenotype and clonogenicity in female C57BL6 mice. The data show that only 3 month old mice were fully capable of restoring lung volumes by day 7 and total alveolar surface area by 21 days. By 9 months, the rate of regeneration was slower (with incomplete regeneration by 21 days), and by 24 months there was no regrowth 21 days post-PNX. The early decline in regeneration rate was not associated with changes in alveolar epithelial cell type II (AECII) proliferation or apoptosis rate. However, significant apoptosis and lack of cell proliferation was evident after PNX in both total cells and AECII cells in 24 mo mice. Analysis of gene expression at several time points (1, 3 and 7 days) post-PNX in 9 versus 3 month mice was consistent with a myofibroblast signature (increased Tnc, Lox1, Col3A1, Eln and Tnfrsf12a) and more alpha smooth muscle actin (αSMA) positive myofibroblasts were present after PNX in 9 month than 3 month mice. Isolated lung fibroblasts showed a significant age-dependent loss of clonogenicity. Moreover, lung fibroblasts isolated from 9 and 17 month mice exhibited higher αSMA, Col3A1, Fn1 and S100A expression, and lower expression of the survival gene Mdk consistent with terminal differentiation. These data show that concomitant loss of clonogenicity and progressive myofibroblastic differentiation contributes to the age-dependent decline in the rate of lung regeneration.


Asunto(s)
Envejecimiento/fisiología , Diferenciación Celular , Pulmón/citología , Pulmón/fisiología , Miofibroblastos/citología , Regeneración/fisiología , Actinas/metabolismo , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Apoptosis/genética , Diferenciación Celular/genética , Proliferación Celular , Colágeno/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Femenino , Homeostasis/genética , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Neumonectomía , Alveolos Pulmonares/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/genética , Transcriptoma
10.
Expert Rev Med Devices ; 8(3): 307-12, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21542703

RESUMEN

AeriSeal Emphysematous Lung Sealant is a novel endoscopic lung-volume reduction therapy designed to reduce hyperinflation and improve pulmonary function and quality of life in patients with advanced emphysema. The device is administered to the subsegmental bronchus via a catheter as a 20 ml volume of liquid-foam. It flows into the peripheral airways and alveoli where it polymerizes and functions as a tissue glue, forming a film of material on the lung surface that seals the target region to cause durable absorption atelectasis. The AeriSeal System received CE mark approval for the treatment of patients with advanced upper lobe predominant and homogeneous emphysema based upon favorable results from clinical studies, and is commercially available in Europe. Patient and treatment site selection algorithms have been developed to simplify product use and optimize outcomes. This manuscript summarizes how the device is used, its mechanism of action and clinical trial results supporting its safety and efficacy.


Asunto(s)
Endoscopía/métodos , Neumonectomía/instrumentación , Neumonectomía/métodos , Enfisema Pulmonar/terapia , Ensayos Clínicos como Asunto , Humanos , Mercadotecnía , Neumonectomía/economía
11.
Stem Cells Dev ; 20(10): 1779-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21585237

RESUMEN

While multipotent mesenchymal stromal cells have been recently isolated from adult lung (L-MSCs), there is very limited data on their biological properties and therapeutic potential in vivo. How L-MSCs compare with bone marrow-derived MSCs (BM-MSCs) is also unclear. In this study, we characterized L-MSC phenotype, clonogenicity, and differentiation potential, and compared L-MSCs to BM-MSCs in vivo survival, retention, paracrine gene expression, and repair or elastase injury after transplantation. L-MSCs were highly clonogenic, frequently expressed aldehyde dehydrogenase activity, and differentiated into osteocytes, chondrocytes, adipocytes, myofibroblasts, and smooth muscle cells. After intravenous injection (2 h), L-MSCs showed greater survival than BM-MSCs; similarly, L-MSCs were significantly more resistant than BM-MSCs to anchorage independent culture (4 h) in vitro. Long after transplantation (4 or 32 days), a significantly higher number of CD45(neg) L-MSCs were retained than BM-MSCs. By flow cytometry, L-MSCs expressed more intercellular adhesion molecule-1 (ICAM-1), platelet derived growth factor receptor alpha (PDGFRα), and integrin α2 than BM-MSCs; these proteins were found to modulate endothelial adherence, directional migration, and migration across Matrigel in L-MSCs. Further, L-MSCs with low ICAM-1 showed poorer lung retention and higher phagocytosis in vivo. Compared with BM-MSCs, L-MSCs expressed higher levels of several transcripts (e.g., Ccl2, Cxcl2, Cxcl10, IL-6, IL-11, Hgf, and Igf2) in vitro, although gene expression in vivo was increased by L-MSCs and BM-MSCs equivalently. Accordingly, both L-MSCs and BM-MSCs reduced elastase injury to the same extent. This study demonstrates that tissue-specific L-MSCs possess mechanisms that enhance their lung retention after intravenous transplantation, and produce substantial healing of elastase injury comparable to BM-MSCs.


Asunto(s)
Lesión Pulmonar/patología , Lesión Pulmonar/terapia , Pulmón/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Comunicación Paracrina , Cicatrización de Heridas , Animales , Células de la Médula Ósea/citología , Adhesión Celular , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Pulmón/metabolismo , Lesión Pulmonar/genética , Masculino , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Elastasa Pancreática , Comunicación Paracrina/genética , Análisis de Supervivencia , Cicatrización de Heridas/genética
12.
Respir Med ; 105(4): 602-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21115237

RESUMEN

BACKGROUND: Patients undergoing bronchoscopy are usually monitored only by pulse oximetry, hence hypoventilation cannot be assessed. Transcutaneous carbon dioxide tension (TcPCO(2)) monitoring is a non-invasive technique to assess hypoventilation. Patients with severe chronic obstructive pulmonary disease (COPD) undergoing bronchoscopy are at increased risk for sedation-induced hypoventilation. The aim of the study was to measure TcPCO(2) using a digital sensor to examine the occurrence of hypoventilation during bronchoscopic lung volume reduction (BLVR). METHODS: Combined TcPCO(2) and SpO(2) saturation and arterial blood gases (ABG) were prospectively measured in 15 patients with severe COPD (Mean FEV(1) 29%) undergoing BLVR under conscious sedation with IV midazolam and IV alfentanil. RESULTS: A highly significant correlation was noted between simultaneous ABG PCO(2) samplings and TcPCO(2) measured (R = 0.85, p < 0.001). Mean baseline TcPCO(2) level was 41.7 ± 10.3 mm Hg (±SD) (range 35-66 mmHg)], and peak measurement during the procedure was 61 ± 17.1 mm Hg (range 41-111 mmHg). The mean increase in TcPCO(2) during bronchoscopy was 19.2 (range 3.7-45 mmHg) [p < 0.0001]. Mean duration of significant hypercapnea (TcPCO(2) > 55 mmHg), observed in 7 (46%) patients, was 9 min (range 0-53). CONCLUSIONS: Bronchoscopy performed under conscious sedation in patients with severe COPD is frequently associated with significant hypoventilation that can only be detected by TcPCO(2) monitoring. Combined measurement of SpO(2) and TcPCO(2) during bronchoscopy enhances patient safety, helps guide administration of sedation, and can alert physicians to the need for anesthesia reversal following completion of bronchoscopic interventions.


Asunto(s)
Broncoscopía/métodos , Dióxido de Carbono/sangre , Hipoventilación/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/cirugía , Adulto , Anciano , Monitoreo de Gas Sanguíneo Transcutáneo , Broncoscopía/efectos adversos , Sedación Consciente/efectos adversos , Sedación Consciente/métodos , Femenino , Humanos , Hipoventilación/etiología , Hipoventilación/fisiopatología , Masculino , Persona de Mediana Edad , Monitoreo Intraoperatorio , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
13.
Respir Res ; 11: 69, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20525205

RESUMEN

BACKGROUND: Peroxisome proliferator-activated receptor (PPAR)-gamma is a nuclear hormone receptor that regulates gene expression, cell proliferation and differentiation. We previously described airway epithelial cell PPARgamma deficient mice that develop airspace enlargement with decreased tissue resistance and increased lung volumes. We sought to understand the impact of airspace enlargement in conditionally targeted mice upon the physio-mechanical properties of the lung. METHODS: We measured elastic recoil and its determinants, including tissue structure and surface forces. We measured alveolar number using radial alveolar counts, and airspace sizes and their distribution using computer-assisted morphometry. RESULTS: Air vs. saline-filled pressure volume profiles demonstrated loss of lung elastic recoil in targeted mice that was contributed by both tissue components and surface tension, but was proportional to lung volume. There were no significant differences in surfactant quantity/function nor in elastin and collagen content between targeted animals and littermate controls. Importantly, radial alveolar counts were significantly reduced in the targeted animals and at 8 weeks of age there were 18% fewer alveoli with 32% more alveolar ducts. Additionally, the alveolar ducts were 19% larger in the targeted animals. CONCLUSIONS: Our data suggest that the functional abnormalities, including loss of recoil are secondary to altered force transmission due to differences in the structure of alveolar ducts, rather than changes in surfactant function or elastin or collagen content. These data further define the nature of abnormal lung maturation in the absence of airway epithelial cell PPARgamma and identify a putative genetic determinant of dysanapsis, which may serve as a precursor to chronic lung disease.


Asunto(s)
Pulmón/anomalías , PPAR gamma/deficiencia , Alveolos Pulmonares/anomalías , Factores de Edad , Animales , Fenómenos Biomecánicos , Colágeno/metabolismo , Elasticidad , Elastina/metabolismo , Pulmón/metabolismo , Mediciones del Volumen Pulmonar , Mecanotransducción Celular , Ratones , Ratones Noqueados , PPAR gamma/genética , Alveolos Pulmonares/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Mucosa Respiratoria/anomalías , Mucosa Respiratoria/metabolismo , Tensión Superficial
14.
J Tissue Eng Regen Med ; 4(4): 259-72, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20020503

RESUMEN

This study summarizes the development and testing of a scaffold to promote engraftment of cells in the distal lung. A fibrinogen-fibronectin-vitronectin hydrogel (FFVH) was developed and optimized with respect to its mechanical and biological properties for this application. In vitro, FFVH scaffolds promoted attachment, histiotypic growth and expression of basement membrane proteins by primary ovine lung mesenchymal cells derived from lung biopsies. In vivo testing was then performed to assess the ability of FFVHs to promote cell engraftment in the sheep lung. Treatment with autologous cells delivered using FFVH was clinically well tolerated. Cells labelled with a fluorescent dye (PKH-26) were detected at treatment sites after 1 month. Tissue mass (assessed by CT imaging) and lung perfusion (assessed by nuclear scintigraphy) were increased at emphysema test sites. Post-treatment histology demonstrated cell proliferation and increased elastin expression without scarring or collapse. No treatment-related pathology was observed at healthy control sites. FFVH scaffolds promote cell attachment, spreading and extracellular matrix expression in vitro and apparent engraftment in vivo, with evidence of trophic effects on the surrounding tissue. Scaffolds of this type may contribute to the development of cell-based therapies for patients with end-stage pulmonary diseases.


Asunto(s)
Pulmón/citología , Pulmón/patología , Ensayo de Materiales/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Bovinos , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibrinógeno/farmacología , Fibronectinas/farmacología , Citometría de Flujo , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Inmunohistoquímica , Pulmón/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Fenotipo , Ovinos , Técnicas de Cultivo de Tejidos , Trasplante Autólogo , Vitronectina/farmacología
15.
Respir Res ; 10: 92, 2009 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-19804646

RESUMEN

BACKGROUND: Adult mice have a remarkable capacity to regenerate functional alveoli following either lung resection or injury that exceeds the regenerative capacity observed in larger adult mammals. The molecular basis for this unique capability in mice is largely unknown. We examined the transcriptomic responses to single lung pneumonectomy in adult mice in order to elucidate prospective molecular signaling mechanisms used in this species during lung regeneration. METHODS: Unilateral left pneumonectomy or sham thoracotomy was performed under general anesthesia (n = 8 mice per group for each of the four time points). Total RNA was isolated from the remaining lung tissue at four time points post-surgery (6 hours, 1 day, 3 days, 7 days) and analyzed using microarray technology. RESULTS: The observed transcriptomic patterns revealed mesenchymal cell signaling, including up-regulation of genes previously associated with activated fibroblasts (Tnfrsf12a, Tnc, Eln, Col3A1), as well as modulation of Igf1-mediated signaling. The data set also revealed early down-regulation of pro-inflammatory cytokine transcripts and up-regulation of genes involved in T cell development/function, but few similarities to transcriptomic patterns observed during embryonic or post-natal lung development. Immunohistochemical analysis suggests that early fibroblast but not myofibroblast proliferation is important during lung regeneration and may explain the preponderance of mesenchymal-associated genes that are over-expressed in this model. This again appears to differ from embryonic alveologenesis. CONCLUSION: These data suggest that modulation of mesenchymal cell transcriptome patterns and proliferation of S100A4 positive mesenchymal cells, as well as modulation of pro-inflammatory transcriptome patterns, are important during post-pneumonectomy lung regeneration in adult mice.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Pulmón/cirugía , Neumonectomía , Regeneración/genética , Toracotomía , Actinas/metabolismo , Animales , Proliferación Celular , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Inmunohistoquímica , Inflamación/genética , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión al Calcio S100A4 , Proteínas S100/metabolismo , Transducción de Señal/genética , Factores de Tiempo
16.
Am J Respir Cell Mol Biol ; 41(4): 407-14, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19188661

RESUMEN

Theories of emphysema traditionally revolved around proteolytic destruction of extracellular matrix. Models have recently been developed that show airspace enlargement with the induction of pulmonary cell apoptosis. The purpose of this study was to determine the mechanism by which a model of epithelial cell apoptosis caused airspace enlargement. Mice were treated with either intratracheal microcystin (MC) to induce apoptosis, intratracheal porcine pancreatic elastase (PPE), or their respective vehicles. Mice from all groups were inflated and morphometry was measured at various time points. Physiology measurements were performed for airway resistance, tissue elastance, and lung volumes. The groups were further analyzed by air-saline quasistatic measurements, surfactant staining, and surfactant functional studies. Mice treated with MC showed evidence of reversible airspace enlargement. In contrast, PPE-treated mice showed irreversible airspace enlargement. The airspace enlargement in MC-treated mice was associated with an increase in elastic recoil due to an increase in alveolar surface tension. PPE-treated mice showed a loss of lung elastic recoil and normal alveolar surface tension, a pattern more consistent with human emphysema. Airspace enlargement that occurs with the MC model of pulmonary epithelial cell apoptosis displays physiology distinct from human emphysema. Reversibility, restrictive physiology due to changes in surface tension, and alveolar enlargement associated with heterogeneous alveolar collapse are most consistent with a mild acute lung injury. Inflation near total lung capacity gives the appearance of enlarged alveoli as neighboring collapsed alveoli exert tethering forces.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Apoptosis/fisiología , Células Epiteliales/patología , Alveolos Pulmonares/efectos de los fármacos , Enfisema Pulmonar/etiología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/diagnóstico , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Resistencia de las Vías Respiratorias , Animales , Apoptosis/efectos de los fármacos , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Elasticidad , Femenino , Inflamación/inducido químicamente , Inflamación/fisiopatología , Mediciones del Volumen Pulmonar , Ratones , Ratones Endogámicos C57BL , Microcistinas/toxicidad , Modelos Biológicos , Elastasa Pancreática/toxicidad , Alveolos Pulmonares/patología , Enfisema Pulmonar/diagnóstico , Surfactantes Pulmonares , Tensión Superficial , Capacidad Pulmonar Total
17.
Mol Carcinog ; 48(6): 488-97, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18942115

RESUMEN

Tumor colonization involves changes in cell permeability and remodeling. Paracellular permeability is regulated by claudins, integrated tight junction (TJ) proteins, located on the apicolateral portion of epithelial cells. Epidermal growth factor (EGF) was reported to modify cellular claudin levels and induce remodeling. To investigate a role for EGF receptor (EGFR) activation in tumor colonization we studied the effect of EGF and claudin-2 overexpression on permeability and cell reorganization in the human A549 non-small cell lung cancer (NSCLC) cell line. Our data demonstrated that A549 cells possess functional TJs and that EGF treatment increased levels of claudin-2 expression by 46%. Furthermore, EGFR signaling reduced monolayer permeability to choline and triggered cellular remodeling. The mitogen-activated protein kinase inhibitor PD98059 blocked the effect on A549 permeability and remodeling. EGF stimulation also exacerbated a fourfold increase in cell colonization elicited by claudin-2 upregulation. Our findings are consistent with the hypothesis that EGFR signaling plays an important role in A549 cell physiology and acts synergistically with claudin-2 to accelerate tumor colonization. Understanding the influence of EGF on A549 cell permeability and reorganization will help shed light on NSCLC tumor colonization and contribute to the development of novel anti-cancer treatments.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Permeabilidad de la Membrana Celular , Receptores ErbB/fisiología , Neoplasias Pulmonares/patología , Proteínas de la Membrana/fisiología , Secuencia de Bases , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , División Celular , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Claudinas , Cartilla de ADN , Receptores ErbB/metabolismo , Flavonoides/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
18.
Proc Am Thorac Soc ; 5(4): 416-20, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18453348

RESUMEN

It is not readily apparent how pulmonary function could be improved by resecting portions of the lung in patients with emphysema. In emphysema, elevation in residual volume relative to total lung capacity reduces forced expiratory volumes, increases inspiratory effort, and impairs inspiratory muscle mechanics. Lung volume reduction surgery (LVRS) better matches the size of the lungs to the size of the thorax containing them. This restores forced expiratory volumes and the mechanical advantage of the inspiratory muscles. In patients with heterogeneous emphysema, LVRS may also allow space occupied by cysts to be reclaimed by more normal lung. Newer, bronchoscopic methods for lung volume reduction seek to achieve similar ends by causing localized atelectasis, but may be hindered by the low collateral resistance of emphysematous lung. Understanding of the mechanisms of improved function after LVRS can help select patients more likely to benefit from this approach.


Asunto(s)
Enfisema Pulmonar/fisiopatología , Enfisema Pulmonar/cirugía , Mecánica Respiratoria/fisiología , Broncoscopía , Volumen Espiratorio Forzado/fisiología , Capacidad Residual Funcional/fisiología , Humanos , Volumen Residual/fisiología , Pruebas de Función Respiratoria , Músculos Respiratorios/fisiopatología , Resultado del Tratamiento
19.
Proc Am Thorac Soc ; 5(4): 454-60, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18453355

RESUMEN

Lung volume reduction surgery (LVRS) produces physiological, symptomatic, and survival benefits in selected patients with advanced emphysema. Because it is associated with significant morbidity, mortality, and cost, nonsurgical alternatives for achieving volume reduction have been developed. Three bronchoscopic lung volume reduction (BLVR) approaches have shown promise and reached later-stage clinical trials. These include the following: (1) placement of endobronchial one-way valves designed to promote atelectasis by blocking inspiratory flow; (2) formation of airway bypass tracts using a radiofrequency catheter designed to facilitate emptying of damaged lung regions with long expiratory times; and (3) instillation of biological adhesives designed to collapse and remodel hyperinflated lung. The limited clinical data currently available suggest that all three techniques are reasonably safe. However, efficacy signals have been substantially smaller and less durable than those observed after LVRS. Studies to optimize patient selection, refine treatment strategies, characterize procedural safety, elucidate mechanisms of action, and characterize short- and longer-term effectiveness of these approaches are ongoing. Results will be available over the next few years and will determine whether BLVR represents a safe and effective alternative to LVRS.


Asunto(s)
Broncoscopía , Neumonectomía/métodos , Enfisema Pulmonar/cirugía , Broncoscopios , Ensayos Clínicos como Asunto , Diseño de Equipo , Humanos , Selección de Paciente , Enfisema Pulmonar/fisiopatología , Pruebas de Función Respiratoria , Factores de Riesgo
20.
Semin Thorac Cardiovasc Surg ; 19(2): 142-50, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17870010

RESUMEN

Medical treatment for patients with stable chronic obstructive pulmonary disease (COPD) has evolved significantly over the past 2 decades. Current World Health Organization recommendations suggest a stepwise approach to therapy depending upon disease severity. As-needed use of short-acting bronchodilators is recommended for patients with mild disease. Scheduled dosing of bronchodilators is recommended for patients with more advanced disease. Inhaled beta-agonists and anti-cholinergic agents in combination have proved to be more effective than either agent alone. Long-acting preparations are associated with better disease control and have not been associated with tachyphylaxis. Inhaled corticosteroids are useful for reducing the frequency of exacerbations in patients who experience one or more episodes per year. Oxygen therapy is clearly beneficial in patients with advanced COPD and chronic respiratory failure, and its potential benefits in less severe disease are currently being studied. Pulmonary rehabilitation benefits patients with mild-to-severe disease, although the greatest benefits have been demonstrated in those with moderate COPD. New ultra-long-acting inhaled bronchodilators, phosphodiesterase inhibitors, protease inhibitors, and retinoids intended to promote tissue regeneration are currently being evaluated in clinical trials as future therapeutic agents.


Asunto(s)
Agonistas Adrenérgicos beta/uso terapéutico , Broncodilatadores/uso terapéutico , Antagonistas Colinérgicos/uso terapéutico , Terapia por Inhalación de Oxígeno/métodos , Inhibidores de Fosfodiesterasa/uso terapéutico , Guías de Práctica Clínica como Asunto , Enfermedad Pulmonar Obstructiva Crónica/terapia , Quimioterapia Combinada , Humanos , Resultado del Tratamiento , Organización Mundial de la Salud
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA