Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nitric Oxide ; 147: 13-25, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38588917

RESUMO

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.


Assuntos
GMP Cíclico , Pulmão , Óxido Nítrico , Óxido Nítrico/metabolismo , Humanos , Pulmão/metabolismo , Animais , GMP Cíclico/metabolismo , Recém-Nascido , Transdução de Sinais , Feto/metabolismo
2.
Sci Rep ; 14(1): 167, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38168512

RESUMO

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.


Assuntos
Respiração , Taxa Respiratória , Adulto , Humanos , Animais , Ovinos , Software , Algoritmos , Eletrocardiografia , Processamento de Sinais Assistido por Computador
4.
Mol Imaging Biol ; 25(5): 944-953, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37610609

RESUMO

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.

5.
Sci Rep ; 13(1): 11246, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37438462

RESUMO

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.


Assuntos
Transtorno da Personalidade Antissocial , Cultura , Animais , Camundongos , Calibragem , Escolaridade , Microcomputadores
6.
Sci Transl Med ; 14(663): eabq6297, 2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36130015

RESUMO

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.


Assuntos
Aldeídos , Fígado , Animais , Biomarcadores , Colágeno , Fibrose , Humanos , Fígado/diagnóstico por imagem , Fígado/patologia , Cirrose Hepática/patologia , Imageamento por Ressonância Magnética , Camundongos , Sondas Moleculares , Ratos
7.
HardwareX ; 12: e00335, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35873736

RESUMO

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.

9.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L204-L223, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34878944

RESUMO

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.


Assuntos
Aldeídos/metabolismo , Lesão Pulmonar/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Proteína-Lisina 6-Oxidase/metabolismo , Aldeídos/química , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/patologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Sondas Moleculares/metabolismo , Células NIH 3T3 , Proteína-Lisina 6-Oxidase/genética , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Transdução de Sinais , Proteínas Smad/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
10.
Eur Heart J Digit Health ; 2(3): 494-510, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34604759

RESUMO

The pandemic has brought to everybody's attention the apparent need of remote monitoring, highlighting hitherto unseen challenges in healthcare. Today, mobile monitoring and real-time data collection, processing and decision-making, can drastically improve the cardiorespiratory-haemodynamic health diagnosis and care, not only in the rural communities, but urban ones with limited healthcare access as well. Disparities in socioeconomic status and geographic variances resulting in regional inequity in access to healthcare delivery, and significant differences in mortality rates between rural and urban communities have been a growing concern. Evolution of wireless devices and smartphones has initiated a new era in medicine. Mobile health technologies have a promising role in equitable delivery of personalized medicine and are becoming essential components in the delivery of healthcare to patients with limited access to in-hospital services. Yet, the utility of portable health monitoring devices has been suboptimal due to the lack of user-friendly and computationally efficient physiological data collection and analysis platforms. We present a comprehensive review of the current cardiac, pulmonary, and haemodynamic telemonitoring technologies. We also propose a novel low-cost smartphone-based system capable of providing complete cardiorespiratory assessment using a single platform for arrhythmia prediction along with detection of underlying ischaemia and sleep apnoea; we believe this system holds significant potential in aiding the diagnosis and treatment of cardiorespiratory diseases, particularly in underserved populations.

11.
Telemed J E Health ; 27(12): 1433-1439, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33729001

RESUMO

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.


Assuntos
Eletrocardiografia , Smartphone , Animais , Suínos
12.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L21-L34, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32374672

RESUMO

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.

13.
J Cereb Blood Flow Metab ; 39(12): 2379-2391, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31423931

RESUMO

Recent works highlight the therapeutic potential of targeting cyclic guanosine monophosphate (cGMP)-dependent pathways in the context of brain ischemia/reperfusion injury (IRI). Although cGMP-dependent protein kinase I (cGKI) has emerged as a key mediator of the protective effects of nitric oxide (NO) and cGMP, the mechanisms by which cGKI attenuates IRI remain poorly understood. We used a novel, conditional cGKI knockout mouse model to study its role in cerebral IRI. We assessed neurological deficit, infarct volume, and cerebral perfusion in tamoxifen-inducible vascular smooth muscle cell-specific cGKI knockout mice and control animals. Stroke experiments revealed greater cerebral infarct volume in smooth muscle cell specific cGKI knockout mice (males: 96 ± 16 mm3; females: 93 ± 12 mm3, mean±SD) than in all control groups: wild type (males: 66 ± 19; females: 64 ± 14), cGKI control (males: 65 ± 18; females: 62 ± 14), cGKI control with tamoxifen (males: 70 ± 8; females: 68 ± 10). Our results identify, for the first time, a protective role of cGKI in vascular smooth muscle cells during ischemic stroke injury. Moreover, this protective effect of cGKI was found to be independent of gender and was mediated via improved reperfusion. These results suggest that cGKI in vascular smooth muscle cells should be targeted by therapies designed to protect brain tissue against ischemic stroke.


Assuntos
Infarto Cerebral/enzimologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Traumatismo por Reperfusão/enzimologia , Acidente Vascular Cerebral/enzimologia , Animais , Infarto Cerebral/genética , Infarto Cerebral/patologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia
14.
PLoS One ; 14(6): e0217217, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31206522

RESUMO

BACKGROUND: Sleep disordered breathing manifested as sleep apnea (SA) is prevalent in the general population, and while it is associated with increased morbidity and mortality risk in some patient populations, it remains under-diagnosed. The objective of this study was to assess the accuracy of respiration-rate (RR) and tidal-volume (TV) estimation algorithms, from body-surface ECG signals, using a smartphone based ambulatory respiration monitoring system (cvrPhone). METHODS: Twelve lead ECG signals were collected using the cvrPhone from anesthetized and mechanically ventilated swine (n = 9). During ECG data acquisition, the mechanical ventilator tidal-volume (TV) was varied from 250 to 0 to 750 to 0 to 500 to 0 to 750 ml at respiratory rates (RR) of 6 and 14 breaths/min, respectively, and the RR and TV values were estimated from the ECG signals using custom algorithms. RESULTS: TV estimations from any two different TV settings showed statistically significant difference (p < 0.01) regardless of the RR. RRs were estimated to be 6.1±1.1 and 14.0±0.2 breaths/min at 6 and 14 breaths/min, respectively (when 250, 500 and 750 ml TV settings were combined). During apnea, the estimated TV and RR values were 11.7±54.9 ml and 0.0±3.5 breaths/min, which were significantly different (p<0.05) than TV and RR values during non-apnea breathing. In addition, the time delay from the apnea onset to the first apnea detection was 8.6±6.7 and 7.0±3.2 seconds for TV and RR respectively. CONCLUSIONS: We have demonstrated that apnea can reliably be detected using ECG-derived RR and TV algorithms. These results support the concept that our algorithms can be utilized to detect SA in conjunction with ECG monitoring.


Assuntos
Eletrocardiografia , Monitorização Fisiológica/instrumentação , Síndromes da Apneia do Sono/diagnóstico , Síndromes da Apneia do Sono/fisiopatologia , Smartphone , Animais , Masculino , Taxa Respiratória , Processamento de Sinais Assistido por Computador , Suínos , Volume de Ventilação Pulmonar
15.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L20-L34, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30260287

RESUMO

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.


Assuntos
Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Guanilil Ciclase Solúvel/biossíntese , Fator de Crescimento Transformador beta1/metabolismo , Animais , Sistema de Sinalização das MAP Quinases , Camundongos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Artéria Pulmonar/citologia , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L615-L627, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28642261

RESUMO

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.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Fibroblastos/metabolismo , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/citologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Receptores de Activinas Tipo II , Animais , Animais Recém-Nascidos , Benzodioxóis/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Humanos , Imidazóis/farmacologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
17.
Am J Respir Cell Mol Biol ; 55(1): 105-16, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27082727

RESUMO

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.


Assuntos
Lisofosfolipídeos/metabolismo , Morfogênese , Alvéolos Pulmonares/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Movimento Celular , Tamanho Celular , Elasticidade , Elastina/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores Teciduais de Metaloproteinases/metabolismo , Tropoelastina/metabolismo
18.
Am J Physiol Cell Physiol ; 308(11): C944-58, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25855081

RESUMO

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.


Assuntos
Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , GMP Cíclico/metabolismo , Complexo de Golgi/metabolismo , Músculo Liso Vascular/metabolismo , Artéria Pulmonar/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Cricetulus , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Furina/genética , Furina/metabolismo , Regulação da Expressão Gênica , Complexo de Golgi/efeitos dos fármacos , Células HEK293 , Humanos , Monensin/farmacologia , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais
19.
Paediatr Anaesth ; 25(2): 150-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24916144

RESUMO

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.


Assuntos
Débito Cardíaco/fisiologia , Monitorização Intraoperatória/instrumentação , Monitorização Intraoperatória/métodos , Adolescente , Adulto , Cardiografia de Impedância , Criança , Pré-Escolar , Eletrocardiografia/instrumentação , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Reprodutibilidade dos Testes , Adulto Jovem
20.
Cell Signal ; 26(12): 2633-44, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25172423

RESUMO

cGMP-dependent protein kinase I (PKGI) plays an important role in regulating how cGMP specifies vascular smooth muscle cell (SMC) phenotype. Although studies indicate that PKGI nuclear localization controls how cGMP regulates gene expression in SMC, information about the mechanisms that regulate PKGI nuclear compartmentation and its role in directly regulating cell phenotype is limited. Here we characterize a nuclear localization signal sequence (NLS) in PKGIγ, a proteolytically cleaved PKGI kinase fragment that translocates to the nucleus of SMC. Immuno-localization studies using cells expressing native and NLS-mutant PKGIγ, and treated with a small molecule nuclear transport inhibitor, indicated that PKGIγ encodes a constitutively active NLS that requires importin α and ß for regulation of its compartmentation. Moreover, studies utilizing a genetically encoded nuclear phospho-CREB biosensor probe and fluorescence lifetime imaging microscopy demonstrated that this NLS controls PKGIγ nuclear function. In addition, although cytosolic PKGIγ-activity was observed to stimulate MAPK/ERK-mediated nuclear CREB signaling in SMC, NLS-mediated PKGIγ nuclear activity alone was determined to increase the expression of differentiation marker proteins in these cells. These results indicate that NLS-mediated nuclear PKGIγ localization plays an important role in how PKGI regulates vascular SMC phenotype.


Assuntos
Núcleo Celular/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Sinais de Localização Nuclear/metabolismo , Animais , Transporte Biológico/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA