Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nitric Oxide ; 147: 13-25, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38588917

RESUMEN

In the developing lung, nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) signaling are essential in regulating lung formation and vascular tone. Animal studies have linked many anatomical and pathophysiological features of newborn lung disease to abnormalities in the NO/cGMP signaling system. They have demonstrated that driving this system with agonists and antagonists alleviates many of them. This research has spurred the rapid clinical development, testing, and application of several NO/cGMP-targeting therapies with the hope of treating and potentially preventing significant pediatric lung diseases. However, there are instances when the therapeutic effectiveness of these agents is limited. Studies indicate that injury-induced disruption of several critical components within the signaling system may hinder the promise of some of these therapies. Recent research has identified basic mechanisms that suppress NO/cGMP signaling in the injured newborn lung. They have also pinpointed biomarkers that offer insight into the activation of these pathogenic mechanisms and their influence on the NO/cGMP signaling system's integrity in vivo. Together, these will guide the development of new therapies to protect NO/cGMP signaling and safeguard newborn lung development and function. This review summarizes the important role of the NO/cGMP signaling system in regulating pulmonary development and function and our evolving understanding of how it is disrupted by newborn lung injury.


Asunto(s)
GMP Cíclico , Pulmón , Óxido Nítrico , Óxido Nítrico/metabolismo , Humanos , Pulmón/metabolismo , Animales , GMP Cíclico/metabolismo , Recién Nacido , Transducción de Señal , Feto/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L204-L223, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34878944

RESUMEN

During newborn lung injury, excessive activity of lysyl oxidases (LOXs) disrupts extracellular matrix (ECM) formation. Previous studies indicate that TGFß activation in the O2-injured mouse pup lung increases lysyl oxidase (LOX) expression. But how TGFß regulates this, and whether the LOXs generate excess pulmonary aldehydes are unknown. First, we determined that O2-mediated lung injury increases LOX protein expression in TGFß-stimulated pup lung interstitial fibroblasts. This regulation appeared to be direct; this is because TGFß treatment also increased LOX protein expression in isolated pup lung fibroblasts. Then using a fibroblast cell line, we determined that TGFß stimulates LOX expression at a transcriptional level via Smad2/3-dependent signaling. LOX is translated as a pro-protein that requires secretion and extracellular cleavage before assuming amine oxidase activity and, in some cells, reuptake with nuclear localization. We found that pro-LOX is processed in the newborn mouse pup lung. Also, O2-mediated injury was determined to increase pro-LOX secretion and nuclear LOX immunoreactivity particularly in areas populated with interstitial fibroblasts and exhibiting malformed ECM. Then, using molecular probes, we detected increased aldehyde levels in vivo in O2-injured pup lungs, which mapped to areas of increased pro-LOX secretion in lung sections. Increased activity of LOXs plays a critical role in the aldehyde generation; an inhibitor of LOXs prevented the elevation of aldehydes in the O2-injured pup lung. These results reveal new mechanisms of TGFß and LOX in newborn lung disease and suggest that aldehyde-reactive probes might have utility in sensing the activation of LOXs in vivo during lung injury.


Asunto(s)
Aldehídos/metabolismo , Lesión Pulmonar/metabolismo , Pulmón/enzimología , Pulmón/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Aldehídos/química , Animales , Animales Recién Nacidos , Embrión de Mamíferos/patología , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Sondas Moleculares/metabolismo , Células 3T3 NIH , Proteína-Lisina 6-Oxidasa/genética , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Transducción de Señal , Proteínas Smad/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Telemed J E Health ; 27(12): 1433-1439, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33729001

RESUMEN

Background: We investigated the ability of a novel stand-alone, smartphone-based system, the cvrPhone, in estimating the minute ventilation (MV) from body surface electrocardiographic (ECG) signals. Methods: Twelve lead ECG signals were collected from anesthetized and mechanically ventilated swine (n = 9) using standard surface electrodes and the cvrPhone. The tidal volume delivered to the animals was varied between 0, 250, 500, and 750 mL at respiration rates of 6 and 14 breaths/min. MV estimates were determined by the cvrPhone and were compared with the delivered ones. Results: The median relative estimation errors were 17%, -4%, 35%, -3%, -9%, and 1%, for true MVs of 1,500, 3,000, 3,500, 4,500, 7,000, and 10,500 breaths*mL/min, respectively. The MV estimates at each of the settings were significantly different from each other (p < 0.05). Conclusions: We have demonstrated that accurate MV estimations can be derived from standard body surface ECG signals, using a smartphone.


Asunto(s)
Electrocardiografía , Teléfono Inteligente , Animales , Porcinos
4.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L21-L34, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374672

RESUMEN

Cyclic guanosine monophosphate (cGMP) signaling is an important regulator of newborn lung function and development. Although cGMP signaling is decreased in many models of newborn lung injury, the mechanisms are poorly understood. We determined how IL-1ß regulates the expression of the α1-subunit of soluble guanylate cyclase (sGCα1), a prime effector of pulmonary cGMP signaling. Physiologic levels of IL-1ß were discovered to rapidly decrease sGCα1 mRNA expression in a human fetal lung fibroblast cell line (IMR-90 cells) and protein levels in primary mouse pup lung fibroblasts. This sGCα1 expression inhibition appeared to be at a transcriptional level; IL-1ß treatment did not alter sGCα1 mRNA stability, although it reduced sGCα1 promoter activity. Transforming growth factor-ß (TGFß)-activated kinase-1 (TAK1) was determined to be required for IL-1ß's regulation of sGCα1 expression; TAK1 knockdown protected sGCα1 mRNA expression in IL-1ß-treated IMR-90 cells. Moreover, heterologously expressed TAK1 was sufficient to decrease sGCα1 mRNA levels in those cells. Nuclear factor-κB (NF-κB) signaling played a critical role in the IL-1ß-TAK1-sGCα1 regulatory pathway; chromatin immunoprecipitation studies demonstrated enhanced activated NF-κB subunit (RelA) binding to the sGCα1 promoter after IL-1ß treatment unless treated with an IκB kinase-2 inhibitor. Also, this NF-κB signaling inhibition protected sGCα1 expression in IL-1ß-treated fibroblasts. Lastly, using transgenic mice in which active IL-1ß was conditionally expressed in lung epithelial cells, we established that IL-1ß expression is sufficient to stimulate TAK1 and decrease sGCα1 protein expression in the newborn lung. Together these results detail the role and mechanisms by which IL-1ß inhibits cGMP signaling in the newborn lung.

5.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L20-L34, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30260287

RESUMEN

TGFß activation during newborn lung injury decreases the expression of pulmonary artery smooth muscle cell (PASMC)-soluble guanylate cyclase (sGC), a critical mediator of nitric oxide signaling. Using a rat PASMC line (CS54 cells), we determined how TGFß downregulates sGC expression. We found that TGFß decreases sGC expression through stimulating its type I receptor; TGFß type I receptor (TGFßR1) inhibitors prevented TGFß-1-mediated decrease in sGCα1 subunit mRNA levels in the cells. However, TGFßR1-Smad mechanisms do not regulate sGC; effective knockdown of Smad2 and Smad3 expression and function did not protect sGCα1 mRNA levels during TGFß-1 exposure. A targeted small-molecule kinase inhibitor screen suggested that MEK signaling regulates sGC expression in TGFß-stimulated PASMC. TGFß activates PASMC MEK/ERK signaling; CS54 cell treatment with TGFß-1 increased MEK and ERK phosphorylation in a biphasic, time- and dose-dependent manner. Moreover, MEK/ERK activity appears to be required for TGFß-mediated sGC expression inhibition in PASMC; MEK and ERK inhibitors protected sGCα1 mRNA expression in TGFß-1-treated CS54 cells. Nuclear ERK activity is sufficient for sGC regulation; heterologous expression of a nucleus-retained, constitutively active ERK2-MEK1 fusion protein decreased CS54 cell sGCα1 mRNA levels. The in vivo relevance of this TGFß-MEK/ERK-sGC downregulation pathway is suggested by the detection of ERK activation and sGCα1 protein expression downregulation in TGFß-associated mouse pup hyperoxic lung injury, and the determination that ERK decreases sGCα1 protein expression in TGFß-1-treated primary PASMC obtained from mouse pups. These studies identify MEK/ERK signaling as an important pathway by which TGFß regulates sGC expression in PASMC.


Asunto(s)
Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Guanilil Ciclasa Soluble/biosíntesis , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Sistema de Señalización de MAP Quinasas , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Arteria Pulmonar/citología , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L615-L627, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28642261

RESUMEN

The intracellular signaling mechanisms through which TGF-ß regulates pulmonary development are incompletely understood. Canonical TGF-ß signaling involves Smad2/3 phosphorylation, Smad2/3·Smad4 complex formation and nuclear localization, and gene regulation. Here, we show that physiologically relevant TGF-ß1 levels also stimulate Smad1/5 phosphorylation, which is typically a mediator of bone morphogenetic protein (BMP) signaling, in mouse pup pulmonary artery smooth muscle cells (mPASMC) and lung fibroblasts and other interstitial lung cell lines. This cross-talk mechanism likely has in vivo relevance because mixed Smad1/5/8·Smad2/3 complexes, which are indicative of TGF-ß-stimulated Smad1/5 activation, were detected in the developing mouse lung using a proximity ligation assay. Although mixed Smad complexes have been shown not to transduce nuclear signaling, we determined that TGF-ß stimulates nuclear localization of phosphorylated Smad1/5 and induces the expression of prototypical BMP-regulated genes in the mPASMC. Small-molecule kinase inhibitor studies suggested that TGF-ß-regulated Smad1/5 phosphorylation in these cells is mediated by TGF-ß-type I receptors, not BMP-type I receptors, but possibly the accessory activin-like kinase (ALK1) receptor. Although work by others suggested that ALK1 is expressed exclusively in endothelial cells in the vasculature, we detected ALK1 mRNA and protein expression in mPASMC in vitro and in mouse pup lungs. Moreover, using an antimurine ALK1 antibody and mPASMC, we determined that ALK1 regulates Smad1/5 phosphorylation by TGF-ß. Together, these studies characterize an accessory TGF-ß-stimulated BMP R-Smad signaling mechanism in interstitial cells of the developing lung. They also indicate the importance of considering alternate Smad pathways in studies directed at determining how TGF-ß regulates newborn lung development.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Fibroblastos/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo II , Animales , Animales Recién Nacidos , Benzodioxoles/farmacología , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Humanos , Imidazoles/farmacología , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Ratones , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , Ratas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
7.
Am J Respir Cell Mol Biol ; 55(1): 105-16, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27082727

RESUMEN

Lysophosphatidic acid (LPA) signaling through one of its receptors, LPA1, contributes to both the development and the pathological remodeling after injury of many organs. Because we found previously that LPA-LPA1 signaling contributes to pulmonary fibrosis, here we investigated whether this pathway is also involved in lung development. Quantitative assessment of lung architecture of LPA1-deficient knock-out (KO) and wild-type (WT) mice at 3, 12, and 24 weeks of age using design-based stereology suggested the presence of an alveolarization defect in LPA1 KO mice at 3 weeks, which persisted as alveolar numbers increased in WT mice into adulthood. Across the ages examined, the lungs of LPA1 KO mice exhibited decreased alveolar numbers, septal tissue volumes, and surface areas, and increased volumes of the distal airspaces. Elastic fibers, critical to the development of alveolar septa, appeared less organized and condensed and more discontinuous in KO alveoli starting at P4. Tropoelastin messenger RNA expression was decreased in KO lungs, whereas expression of matrix metalloproteinases degrading elastic fibers was either decreased or unchanged. These results are consistent with the abnormal lung phenotype of LPA1 KO mice, being attributable to reduced alveolar septal formation during development, rather than to increased septal destruction as occurs in the emphysema of chronic obstructive pulmonary disease. Peripheral septal fibroblasts and myofibroblasts, which direct septation in late alveolarization, demonstrated reduced production of tropoelastin and matrix metalloproteinases, and diminished LPA-induced migration, when isolated from LPA1 KO mice. Taken together, our data suggest that LPA-LPA1 signaling is critically required for septation during alveolarization.


Asunto(s)
Lisofosfolípidos/metabolismo , Morfogénesis , Alveolos Pulmonares/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Animales , Recuento de Células , Movimiento Celular , Tamaño de la Célula , Elasticidad , Elastina/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Tropoelastina/metabolismo
8.
Am J Physiol Cell Physiol ; 308(11): C944-58, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25855081

RESUMEN

cGMP-dependent protein kinase I (PKGI) is an important effector of cGMP signaling that regulates vascular smooth muscle cell (SMC) phenotype and proliferation. PKGI has been detected in the perinuclear region of cells, and recent data indicate that proprotein convertases (PCs) typically resident in the Golgi apparatus (GA) can stimulate PKGI proteolysis and generate a kinase fragment that localizes to the nucleus and regulates gene expression. However, the role of the endomembrane system in PKGI compartmentation and processing is unknown. Here, we demonstrate that PKGI colocalizes with endoplasmic reticulum (ER), ER-Golgi intermediate compartment, GA cisterna, and trans-Golgi network proteins in pulmonary artery SMC and cell lines. Moreover, PKGI localizes with furin, a trans-Golgi network-resident PC known to cleave PKGI. ER protein transport influences PKGI localization because overexpression of a constitutively inactive Sar1 transgene caused PKGI retention in the ER. Additionally, PKGI appears to reside within the GA because PKGI immunoreactivity was determined to be resistant to cytosolic proteinase K treatment in live cells. The GA appears to play a role in PKGI proteolysis because overexpression of inositol 1,4,5-trisphosphate receptor-associated cGMP kinase substrate, not only tethered heterologous PKGI-ß to the ER and decreased its localization to the GA, but also diminished PKGI proteolysis and nuclear translocation. Also, inhibiting intra-GA protein transport with monensin was observed to decrease PKGI cleavage. These studies detail a role for the endomembrane system in regulating PKGI compartmentation and proteolysis. Moreover, they support the investigation of mechanisms regulating PKGI-dependent nuclear cGMP signaling in the pulmonary vasculature with Golgi dysfunction.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , GMP Cíclico/metabolismo , Aparato de Golgi/metabolismo , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/metabolismo , Animales , Línea Celular , Células Cultivadas , Cricetulus , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Furina/genética , Furina/metabolismo , Regulación de la Expresión Génica , Aparato de Golgi/efectos de los fármacos , Células HEK293 , Humanos , Monensina/farmacología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
9.
Paediatr Anaesth ; 25(2): 150-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24916144

RESUMEN

BACKGROUND: Electrical Cardiometry(™) (EC) estimates cardiac parameters by measuring changes in thoracic electrical bioimpedance during the cardiac cycle. The ICON(®), using four electrocardiogram electrodes (EKG), estimates the maximum rate of change of impedance to peak aortic blood acceleration (based on the premise that red blood cells change from random orientation during diastole (high impedance) to an aligned state during systole (low impedance)). OBJECTIVE: To determine whether continuous cardiac output (CO) data provide additional information to current anesthesia monitors that is useful to practitioners. METHODS: After IRB approval and verbal consent, 402 children were enrolled. Data were uploaded to our anesthesia record at one-minute intervals. Ten-second measurements (averaged over the previous 20 heart beats) were downloaded to separate files for later comparison with routine OR monitors. RESULTS: Data from 374 were in the final cohort (loss of signal or improper lead placement); 292,012 measurements during 58,049 min of anesthesia were made in these children (1 day to 19 years and 1 to 107 kg). Four events had a ≥25% reduction in cardiac index at least 1 min before a clinically important change in other monitored parameters; 18 events in 14 children confirmed manifestations of other hemodynamic measures; eight events may have represented artifacts because the observed measurements did not seem to fit the clinical parameters of the other monitors; three other events documented decreased stroke index with extreme tachycardia. CONCLUSIONS: Electrical cardiometry provides real-time cardiovascular information regarding developing hemodynamic events and successfully tracked the rapid response to interventions in children of all sizes. Intervention decisions must be based on the combined data from all monitors and the clinical situation. Our experience suggests that this type of monitor may be an important addition to real-time hemodynamic monitoring.


Asunto(s)
Gasto Cardíaco/fisiología , Monitoreo Intraoperatorio/instrumentación , Monitoreo Intraoperatorio/métodos , Adolescente , Adulto , Cardiografía de Impedancia , Niño , Preescolar , Electrocardiografía/instrumentación , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Reproducibilidad de los Resultados , Adulto Joven
10.
Am J Respir Cell Mol Biol ; 50(2): 459-69, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24066869

RESUMEN

Many pediatric pulmonary diseases are associated with significant morbidity and mortality due to impairment of alveolar development. The lack of an appropriate in vitro model system limits the identification of therapies aimed at improving alveolarization. Herein, we characterize an ex vivo lung culture model that facilitates investigation of signaling pathways that influence alveolar septation. Postnatal Day 4 (P4) mouse pup lungs were inflated with 0.4% agarose, sliced, and cultured within a collagen matrix in medium that was optimized to support cell proliferation and promote septation. Lung slices were grown with and without 1D11, an active transforming growth factor-ß-neutralizing antibody. After 4 days, the lung sections (designated P4 + 4) and noncultured lung sections were examined using quantitative morphometry to assess alveolar septation and immunohistochemistry to evaluate cell proliferation and differentiation. We observed that the P4 + 4 lung sections exhibited ex vivo alveolarization, as evidenced by an increase in septal density, thinning of septal walls, and a decrease in mean linear intercept comparable to P8, age-matched, uncultured lungs. Moreover, immunostaining showed ongoing cell proliferation and differentiation in cultured lungs that were similar to P8 controls. Cultured lungs exposed to 1D11 had a distinct phenotype of decreased septal density when compared with untreated P4 + 4 lungs, indicating the utility of investigating signaling in these lung slices. These results indicate that this novel lung culture system is optimized to permit the investigation of pathways involved in septation, and potentially the identification of therapeutic targets that enhance alveolarization.


Asunto(s)
Enfermedades Pulmonares/metabolismo , Pulmón/patología , Alveolos Pulmonares/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Pulmón/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos/métodos , Alveolos Pulmonares/patología , Factor de Crecimiento Transformador beta/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 306(3): L246-59, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24285264

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common and serious complication of premature birth, characterized by a pronounced arrest of alveolar development. The underlying pathophysiological mechanisms are poorly understood although perturbations to the maturation and remodeling of the extracellular matrix (ECM) are emerging as candidate disease pathomechanisms. In this study, the expression and regulation of three members of the lysyl hydroxylase family of ECM remodeling enzymes (Plod1, Plod2, and Plod3) in clinical BPD, as well as in an experimental animal model of BPD, were addressed. All three enzymes were localized to the septal walls in developing mouse lungs, with Plod1 also expressed in the vessel walls of the developing lung and Plod3 expressed uniquely at the base of developing septa. The expression of plod1, plod2, and plod3 was upregulated in the lungs of mouse pups exposed to 85% O2, an experimental animal model of BPD. Transforming growth factor (TGF)-ß increased plod2 mRNA levels and activated the plod2 promoter in vitro in lung epithelial cells and in lung fibroblasts. Using in vivo neutralization of TGF-ß signaling in the experimental animal model of BPD, TGF-ß was identified as the regulator of aberrant plod2 expression. PLOD2 mRNA expression was also elevated in human neonates who died with BPD or at risk for BPD, compared with neonates matched for gestational age at birth or chronological age at death. These data point to potential roles for lysyl hydroxylases in normal lung development, as well as in perturbed late lung development associated with BPD.


Asunto(s)
Displasia Broncopulmonar/fisiopatología , Pulmón/embriología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/enzimología , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Línea Celular , Células Epiteliales/metabolismo , Femenino , Humanos , Hiperoxia/fisiopatología , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/biosíntesis , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
12.
Eur Respir J ; 44(1): 109-21, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24603819

RESUMEN

Aberrant remodelling of the extracellular matrix in the developing lung may underlie arrested alveolarisation associated with bronchopulmonary dysplasia (BPD). Transglutaminases are regulators of extracellular matrix remodelling. Therefore, the expression and activity of transglutaminases were assessed in lungs from human neonates with BPD and in a rodent model of BPD. Transglutaminase expression and localisation were assessed by RT-PCR, immunoblotting, activity assay and immunohistochemical analyses of human and mouse lung tissues. Transglutaminase regulation by transforming growth factor (TGF)-ß was investigated in lung cells by luciferase-based reporter assay and RT-PCR. TGF-ß signalling was neutralised in vivo in an animal model of BPD, to determine whether TGF-ß mediated the hyperoxia-induced changes in transglutaminase expression. Transglutaminase 2 expression was upregulated in the lungs of preterm infants with BPD and in the lungs of hyperoxia-exposed mouse pups, where lung development was arrested. Transglutaminase 2 localised to the developing alveolar septa. TGF-ß was identified as a regulator of transglutaminase 2 expression in human and mouse lung epithelial cells. In vivo neutralisation of TGF-ß signalling partially restored normal lung structure and normalised lung transglutaminase 2 mRNA expression. Our data point to a role for perturbed transglutaminase 2 activity in the arrested alveolarisation associated with BPD.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación Enzimológica de la Expresión Génica , Transglutaminasas/metabolismo , Animales , Displasia Broncopulmonar/mortalidad , Células Epiteliales/citología , Matriz Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Hiperoxia/metabolismo , Lactante , Recién Nacido , Recien Nacido Prematuro , Pulmón/metabolismo , Masculino , Ratones , Proteína Glutamina Gamma Glutamiltransferasa 2 , Alveolos Pulmonares/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
13.
Sci Rep ; 14(1): 167, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168512

RESUMEN

Respiratory rate (RR) is a critical vital sign used to assess pulmonary function. Currently, RR estimating instrumentation is specialized and bulky, therefore unsuitable for remote health monitoring. Previously, RR was estimated using proprietary software that extract surface electrocardiogram (ECG) waveform features obtained at several thoracic locations. However, developing a non-proprietary method that uses minimal ECG leads, generally available from mobile cardiac monitors is highly desirable. Here, we introduce an open-source and well-documented Python-based algorithm that estimates RR requiring only single-stream ECG signals. The algorithm was first developed using ECGs from awake, spontaneously breathing adult human subjects. The algorithm-estimated RRs exhibited close linear correlation to the subjects' true RR values demonstrating an R2 of 0.9092 and root mean square error of 2.2 bpm. The algorithm robustness was then tested using ECGs generated by the ischemic hearts of anesthetized, mechanically ventilated sheep. Although the ECG waveforms during ischemia exhibited severe morphologic changes, the algorithm-determined RRs exhibited high fidelity with a resolution of 1 bpm, an absolute error of 0.07 ± 0.07 bpm, and a relative error of 0.67 ± 0.64%. This optimized Python-based RR estimation technique will likely be widely adapted for remote lung function assessment in patients with cardiopulmonary disease.


Asunto(s)
Respiración , Frecuencia Respiratoria , Adulto , Humanos , Animales , Ovinos , Programas Informáticos , Algoritmos , Electrocardiografía , Procesamiento de Señales Asistido por Computador
14.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L130-40, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23686857

RESUMEN

Nitric oxide and cGMP modulate vascular smooth muscle cell (SMC) phenotype by regulating cell differentiation and proliferation. Recent studies suggest that cGMP-dependent protein kinase I (PKGI) cleavage and the nuclear translocation of a constitutively active kinase fragment, PKGIγ, are required for nuclear cGMP signaling in SMC. However, the mechanisms that control PKGI proteolysis are unknown. Inspection of the amino acid sequence of a PKGI cleavage site that yields PKGIγ and a protease database revealed a putative minimum consensus sequence for proprotein convertases (PCs). Therefore we investigated the role of PCs in regulating PKGI proteolysis. We observed that overexpression of PCs, furin and PC5, but not PC7, which are all expressed in SMC, increase PKGI cleavage in a dose-dependent manner in human embryonic kidney (HEK) 293 cells. Moreover, furin-induced proteolysis of mutant PKGI, in which alanines were substituted into the putative PC consensus sequence, was decreased in these cells. In addition, overexpression of furin increased PKGI proteolysis in LoVo cells, which is an adenocarcinoma cell line expressing defective furin without PC activity. Also, expression of α1-PDX, an engineered serpin-like PC inhibitor, reduced PC activity and decreased PKGI proteolysis in HEK293 cells. Last, treatment of low-passage rat aortic SMC with membrane-permeable PC inhibitor peptides decreased cGMP-stimulated nuclear PKGIγ translocation. These data indicate for the first time that PCs have a role in regulating PKGI proteolysis and the nuclear localization of its active cleavage product, which are important for cGMP-mediated SMC phenotype.


Asunto(s)
Núcleo Celular/enzimología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Furina/metabolismo , Proproteína Convertasa 5/metabolismo , Proteolisis , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Núcleo Celular/genética , GMP Cíclico/genética , GMP Cíclico/metabolismo , Furina/genética , Células HEK293 , Humanos , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Proproteína Convertasa 5/genética , Ratas , Subtilisinas/genética , Subtilisinas/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 305(8): L569-81, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23934926

RESUMEN

Nitric oxide (NO) regulates lung development through incompletely understood mechanisms. NO controls pulmonary vascular smooth muscle cell (SMC) differentiation largely through stimulating soluble guanylate cyclase (sGC) to produce cGMP and increase cGMP-mediated signaling. To examine the role of sGC in regulating pulmonary development, we tested whether decreased sGC activity reduces alveolarization in the normal and injured newborn lung. For these studies, mouse pups with gene-targeted sGC-α1 subunit truncation were used because we determined that they have decreased pulmonary sGC enzyme activity. sGC-α1 knockout (KO) mouse pups were observed to have decreased numbers of small airway structures and lung volume compared with wild-type (WT) mice although lung septation and body weights were not different. However, following mild lung injury caused by breathing 70% O2, the sGC-α1 KO mouse pups had pronounced inhibition of alveolarization, as evidenced by an increase in airway mean linear intercept, reduction in terminal airway units, and decrease in lung septation and alveolar openings, as well as reduced somatic growth. Because cGMP regulates SMC phenotype, we also tested whether decreased sGC activity reduces lung myofibroblast differentiation. Cellular markers revealed that vascular SMC differentiation decreased, whereas myofibroblast activation increased in the hyperoxic sGC-α1 KO pup lung. These results indicate that lung development, particularly during hyperoxic injury, is impaired in mouse pups with diminished sGC activity. These studies support the investigation of sGC-targeting agents as therapies directed at improving development in the newborn lung exposed to injury.


Asunto(s)
Diferenciación Celular/fisiología , Guanilato Ciclasa/metabolismo , Alveolos Pulmonares/enzimología , Alveolos Pulmonares/crecimiento & desarrollo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Animales Recién Nacidos , GMP Cíclico/genética , GMP Cíclico/metabolismo , Guanilato Ciclasa/genética , Hiperoxia/tratamiento farmacológico , Hiperoxia/enzimología , Hiperoxia/genética , Hiperoxia/patología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/enzimología , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Miofibroblastos/enzimología , Receptores Citoplasmáticos y Nucleares/genética , Guanilil Ciclasa Soluble
16.
Circ Res ; 109(6): 616-28, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21778429

RESUMEN

RATIONALE: Integrin-linked kinase (ILK) is located at focal adhesions and links the extracellular matrix (ECM) to the actin cytoskeleton via ß1- and ß3-integrins. ILK plays a role in the activation of kinases including protein kinase B/Akt and glycogen synthase kinase 3ß and regulates cell proliferation, motility, and survival. OBJECTIVE: To determine the function of ILK in vascular smooth muscle cells (SMCs) in vivo. METHODS AND RESULTS: SM22Cre(+)Ilk(Fl/Fl) conditional mutant mice were generated in which the Ilk gene was selectively ablated in SMCs. SM22Cre(+)Ilk(Fl/Fl) conditional mutant mice survive to birth but die in the perinatal period exhibiting multiple vascular pathologies including aneurysmal dilatation of the aorta and patent ductus arteriosus (PDA). Defects in morphogenetic development of the aorta were observed as early as E12.5 in SM22Cre(+)Ilk(Fl/Fl) mutant embryos. By late gestation (E16.5 to 18.5), striking expansion of the thoracic aorta was observed in ILK mutant embryos. Histological analyses revealed that the structural organization of the arterial tunica media is severely disrupted with profound derangements in SMC morphology, cell-cell, and cell-matrix relationships, including disruption of the elastic lamellae. ILK deletion in primary aortic SMCs results in alterations of RhoA/cytoskeletal signaling transduced through aberrant localization of myocardin-related transcription factor (MRTF)-A repressing the transcription and expression of SMC genes, which are required for the maintenance of the contractile SMC phenotype. CONCLUSIONS: These data identify a molecular pathway linking ILK signaling to the contractile SMC gene program. Activation of this pathway is required for morphogenetic development of the aorta and ductus arteriosus during embryonic and postnatal survival.


Asunto(s)
Aneurisma de la Aorta/enzimología , Eliminación de Gen , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Animales , Aneurisma de la Aorta/patología , Células Cultivadas , Femenino , Marcación de Gen/métodos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Miocitos del Músculo Liso/citología , Embarazo
17.
Sci Rep ; 13(1): 11246, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37438462

RESUMEN

The study of mouse lung mechanics provides essential insights into the physiological mechanisms of pulmonary disease. Consequently, investigators assemble custom systems comprising infusion-withdrawal syringe pumps and analog pressure sensors to investigate the lung function of these animals. But these systems are expensive and require ongoing regulation, making them challenging to use. Here I introduce LungElast, an open-source, inexpensive, and self-contained instrument that can experimentally determine lung elasticity and volumes even in immature mice. It is assembled using custom 3D printed parts and readily available or easily constructed components. In this device, a microprocessor-controlled stepper motor automatically regulates lung volume by precisely driving a syringe piston whose position is determined using time-of-flight LIDAR technology. The airway pressures associated with the lung volumes are determined using compact sensor-on-chip technology, retrieved in a digital format, and stored by the microcontroller. The instrument software is modular, which eases device testing, calibration, and use. Data are also provided here that specify the accuracy and precision of the elastometer's sensors and volume delivery and demonstrate its use with lung models and mouse pups. This instrument has excellent potential for research and educational work.


Asunto(s)
Trastorno de Personalidad Antisocial , Cultura , Animales , Ratones , Calibración , Escolaridad , Microcomputadores
18.
Mol Imaging Biol ; 25(5): 944-953, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37610609

RESUMEN

PURPOSE: Idiopathic pulmonary fibrosis (IPF) is a destructive lung disease with a poor prognosis, an unpredictable clinical course, and inadequate therapies. There are currently no measures of disease activity to guide clinicians making treatment decisions. The aim of this study was to develop a PET probe to identify lung fibrogenesis using a pre-clinical model of pulmonary fibrosis, with potential for translation into clinical use to predict disease progression and inform treatment decisions. METHODS: Eight novel allysine-targeting chelators, PIF-1, PIF-2, …, PIF-8, with different aldehyde-reactive moieties were designed, synthesized, and radiolabeled with gallium-68 or copper-64. PET probe performance was assessed in C57BL/6J male mice 2 weeks after intratracheal bleomycin challenge and in naïve mice by dynamic PET/MR imaging and with biodistribution at 90 min post injection. Lung hydroxyproline and allysine were quantified ex vivo and histological staining for fibrosis and aldehyde was performed. RESULTS: In vivo screening of probes identified 68GaPIF-3 and 68GaPIF-7 as probes with high uptake in injured lung, high uptake in injured lung versus normal lung, and high uptake in injured lung versus adjacent liver and heart tissue. A crossover, intra-animal PET/MR imaging study of 68GaPIF-3 and 68GaPIF-7 confirmed 68GaPIF-7 as the superior probe. Specificity for fibrogenesis was confirmed in a crossover, intra-animal PET/MR imaging study with 68GaPIF-7 and a non-binding control compound, 68GaPIF-Ctrl. Substituting copper-64 for gallium-68 did not affect lung uptake or specificity indicating that either isotope could be used. CONCLUSION: A series of allysine-reactive PET probes with variations in the aldehyde-reactive moiety were evaluated in a pre-clinical model of lung fibrosis. The hydrazine-bearing probe, 68GaPIF-7, exhibited the highest uptake in fibrogenic lung, low uptake in surrounding liver or heart tissue, and low lung uptake in healthy mice and should be considered for further clinical translation.

19.
HardwareX ; 12: e00335, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35873736

RESUMEN

Conscious respiratory pattern and rate control is desired by patients with some forms of pulmonary disease that are undergoing respiratory muscle conditioning and rehabilitation, by practitioners of meditation hoping to improve mindfulness and wellbeing, by athletes striving to obtain breathing control in order to increase competitiveness, and by engineers and scientists that wish to use the data from breathing subjects to test hypotheses and develop physiological monitoring systems. Although prerecorded audio sources and computer applications are available that guide breathing exercises, they often suffer from being inflexible and allow only limited customization of the breathing cues. Here we describe a small, lightweight, battery-powered, microprocessor-based respiratory coaching device (RespiCo), which through wireless or wired connections, can be easily customized to precisely guide subjects to breathe at desired respiratory rates using specific breathing patterns through visual, auditory, or haptic cues. Digital signals can also be captured from the device to document the breathing cues provided by the device for research purposes. It is anticipated that this device will have important utility for those who wish to be guided to breathe in a precise manner or in research and development of physiologic monitoring systems.

20.
Sci Transl Med ; 14(663): eabq6297, 2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36130015

RESUMEN

Liver fibrosis plays a critical role in the evolution of most chronic liver diseases and is characterized by a buildup of extracellular matrix, which can progress to cirrhosis, hepatocellular carcinoma, liver failure, or death. Now, there are no noninvasive methods available to accurately assess disease activity (fibrogenesis) to sensitively detect early onset of fibrosis or to detect early response to treatment. Here, we hypothesized that extracellular allysine aldehyde (LysAld) pairs formed by collagen oxidation during active fibrosis could be a target for assessing fibrogenesis with a molecular probe. We showed that molecular magnetic resonance imaging (MRI) using an extracellular probe targeting these LysAld pairs acts as a noninvasive biomarker of fibrogenesis and demonstrated its high sensitivity and specificity in detecting fibrogenesis in toxin- and dietary-induced mouse models, a cholestasis rat model of liver fibrogenesis, and in human fibrotic liver tissues. Quantitative molecular MRI was highly correlated with fibrogenesis markers and enabled noninvasive detection of early onset fibrosis and response to antifibrotic treatment, showing high potential for clinical translation.


Asunto(s)
Aldehídos , Hígado , Animales , Biomarcadores , Colágeno , Fibrosis , Humanos , Hígado/diagnóstico por imagen , Hígado/patología , Cirrosis Hepática/patología , Imagen por Resonancia Magnética , Ratones , Sondas Moleculares , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA