Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 155(5): 1178-87, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267896

RESUMEN

There are few substantive methods to measure the health of the immune system, and the connection between immune strength and the viral component of the microbiome is poorly understood. Organ transplant recipients are treated with posttransplant therapies that combine immunosuppressive and antiviral drugs, offering a window into the effects of immune modulation on the virome. We used sequencing of cell-free DNA in plasma to investigate drug-virome interactions in a cohort of organ transplant recipients (656 samples, 96 patients) and find that antivirals and immunosuppressants strongly affect the structure of the virome in plasma. We observe marked virome compositional dynamics at the onset of the therapy and find that the total viral load increases with immunosuppression, whereas the bacterial component of the microbiome remains largely unaffected. The data provide insight into the relationship between the human virome, the state of the immune system, and the effects of pharmacological treatment and offer a potential application of the virome state to predict immunocompetence.


Asunto(s)
Antivirales/uso terapéutico , Sangre/virología , Trasplante de Corazón , Inmunosupresores/uso terapéutico , Trasplante de Pulmón , Virus/aislamiento & purificación , Adulto , Profilaxis Antibiótica , Sangre/microbiología , Niño , ADN/sangre , ADN/genética , Humanos , Virus/clasificación
2.
Am J Respir Cell Mol Biol ; 69(4): 470-483, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37290124

RESUMEN

Worldwide, the incidence of both preterm births and chronic lung disease of infancy, or bronchopulmonary dysplasia, remains high. Infants with bronchopulmonary dysplasia have larger and fewer alveoli, a lung pathology that can persist into adulthood. Although recent data point to a role for hypoxia-inducible factor-1α (HIF-1α) in mediating pulmonary angiogenesis and alveolarization, the cell-specific role of HIF-1α remains incompletely understood. Thus, we hypothesized that HIF-1α, in a distinct subset of mesenchymal cells, mediates postnatal alveolarization. To test the hypothesis, we generated mice with a cell-specific deletion of HIF-1α by crossing SM22α promoter-driven Cre mice with HIF-1αflox/flox mice (SM22α-HIF-1α-/-), determined SM-22α-expressing cell identity using single-cell RNA sequencing, and interrogated samples from preterm infants. Deletion of HIF-1α in SM22α-expressing cells had no effect on lung structure at day 3 of life. However, at 8 days, there were fewer and larger alveoli, a difference that persisted into adulthood. Microvascular density, elastin organization, and peripheral branching of the lung vasculature were decreased in SM22α-HIF-1α-/- mice, compared with control mice. Single-cell RNA sequencing demonstrated that three mesenchymal cell subtypes express SM22α: myofibroblasts, airway smooth muscle cells, and vascular smooth muscle cells. Pulmonary vascular smooth muscle cells from SM22α-HIF-1α-/- mice had decreased angiopoietin-2 expression and, in coculture experiments, a diminished capacity to promote angiogenesis that was rescued by angiopoietin-2. Angiopoietin-2 expression in tracheal aspirates of preterm infants was inversely correlated with overall mechanical ventilation time, a marker of disease severity. We conclude that SM22α-specific HIF-1α expression drives peripheral angiogenesis and alveolarization in the lung, perhaps by promoting angiopoietin-2 expression.


Asunto(s)
Angiopoyetina 2 , Displasia Broncopulmonar , Subunidad alfa del Factor 1 Inducible por Hipoxia , Animales , Humanos , Recién Nacido , Ratones , Angiopoyetina 2/metabolismo , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Recien Nacido Prematuro , Pulmón/patología
3.
Am J Physiol Lung Cell Mol Physiol ; 325(6): L741-L755, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37847687

RESUMEN

Pulmonary arterial hypertension (PAH) is a disease characterized by increased vasoconstriction and vascular remodeling. Pulmonary artery smooth muscle cells (PASMCs) highly express the transcription factor hypoxia-inducible factor-1α (HIF-1α), yet the role of PASMC HIF-1α in the development of PAH remains controversial. To study the role of SMC HIF-1α in the pulmonary vascular response to acute and chronic hypoxia, we used a gain-of-function strategy to stabilize HIF-1α in PASMC by generating mice lacking prolyl hydroxylase domain (PHD) 1 and 2 in SM22α-expressing cells. This strategy increased HIF-1α expression and transcriptional activity under conditions of normoxia and hypoxia. Acute hypoxia increased right ventricular systolic pressure (RVSP) in control, but not in SM22α-PHD1/2-/- mice. Chronic hypoxia increased RVSP and vascular remodeling more in control SM22α-PHD1/2+/+ than in SM22α-PHD1/2-/- mice. In vitro studies demonstrated increased contractility and myosin light chain phosphorylation in isolated PHD1/2+/+ compared with PHD1/2-/- PASMC under both normoxic and hypoxic conditions. After chronic hypoxia, there was more p27 and less vascular remodeling in SM22α-PHD1/2-/- compared with SM22α-PHD1/2+/+ mice. Hypoxia increased p27 in PASMC isolated from control patients, but not in cells from patients with idiopathic pulmonary arterial hypertension (IPAH). These findings highlight an SM22α-expressing cell-specific role for HIF-1α in the inhibition of pulmonary vasoconstriction and vascular remodeling. Modulating HIF-1α expression in PASMC may represent a promising preventative and therapeutic strategy for patients with PAH.NEW & NOTEWORTHY In a mouse model wherein hypoxia-inducible factor 1 alpha (HIF-1α) is stabilized in vascular smooth muscle cells, we found that HIF-1α regulates vasoconstriction by limiting phosphorylation of myosin light chain and regulates vascular remodeling through p27 induction. These findings highlight a cell-specific role for HIF-1α in the inhibition of pulmonary vasoconstriction and vascular remodeling.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Animales , Humanos , Ratones , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Prolil Hidroxilasas/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar/metabolismo , Remodelación Vascular
4.
Eur Respir J ; 61(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37024132

RESUMEN

INTRODUCTION: Pulmonary arterial hypertension (PAH) is characterised by loss of microvessels. The Wnt pathways control pulmonary angiogenesis but their role in PAH is incompletely understood. We hypothesised that Wnt activation in pulmonary microvascular endothelial cells (PMVECs) is required for pulmonary angiogenesis, and its loss contributes to PAH. METHODS: Lung tissue and PMVECs from healthy and PAH patients were screened for Wnt production. Global and endothelial-specific Wnt7a -/- mice were generated and exposed to chronic hypoxia and Sugen-hypoxia (SuHx). RESULTS: Healthy PMVECs demonstrated >6-fold Wnt7a expression during angiogenesis that was absent in PAH PMVECs and lungs. Wnt7a expression correlated with the formation of tip cells, a migratory endothelial phenotype critical for angiogenesis. PAH PMVECs demonstrated reduced vascular endothelial growth factor (VEGF)-induced tip cell formation as evidenced by reduced filopodia formation and motility, which was partially rescued by recombinant Wnt7a. We discovered that Wnt7a promotes VEGF signalling by facilitating Y1175 tyrosine phosphorylation in vascular endothelial growth factor receptor 2 (VEGFR2) through receptor tyrosine kinase-like orphan receptor 2 (ROR2), a Wnt-specific receptor. We found that ROR2 knockdown mimics Wnt7a insufficiency and prevents recovery of tip cell formation with Wnt7a stimulation. While there was no difference between wild-type and endothelial-specific Wnt7a -/- mice under either chronic hypoxia or SuHx, global Wnt7a +/- mice in hypoxia demonstrated higher pulmonary pressures and severe right ventricular and lung vascular remodelling. Similar to PAH, Wnt7a +/- PMVECs exhibited an insufficient angiogenic response to VEGF-A that improved with Wnt7a. CONCLUSIONS: Wnt7a promotes VEGF signalling in lung PMVECs and its loss is associated with an insufficient VEGF-A angiogenic response. We propose that Wnt7a deficiency contributes to progressive small vessel loss in PAH.


Asunto(s)
Hipertensión Arterial Pulmonar , Ratones , Animales , Hipertensión Arterial Pulmonar/complicaciones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Endoteliales/metabolismo , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipoxia/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 323(2): L129-L141, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35762602

RESUMEN

Though survival rates for preterm infants are improving, the incidence of chronic lung disease of infancy, or bronchopulmonary dysplasia (BPD), remains high. Histologically, BPD is characterized by larger and fewer alveoli. Hypoxia-inducible factors (HIFs) may be protective in the context of hyperoxia-induced lung injury, but the cell-specific effects of HIF expression in neonatal lung injury remain unknown. Thus, we sought to determine whether HIF stabilization in SM22α-expressing cells can limit hyperoxia-induced neonatal lung injury. We generated SM22α-specific HIF-1α-stabilized mice (SM22α-PHD1/2-/- mice) by cross-breeding SM22α-promotor-driven Cre recombinase mice with prolyl hydroxylase PHD1flox/flox and PHD2flox/flox mice. Neonatal mice were randomized to 21% O2 (normoxia) or 80% O2 (hyperoxia) exposure for 14 days. For the hyperoxia recovery studies, neonatal mice were recovered from normoxia for an additional 10 wk. SM22α-specific HIF-1α stabilization mitigated hyperoxia-induced lung injury and preserved microvessel density compared with control mice for both neonates and adults. In SM22α-PHD1/2-/- mice, pulmonary artery endothelial cells (PAECs) were more proliferative and pulmonary arteries expressed more collagen IV compared with control mice, even under hyperoxic conditions. Angiopoietin-2 (Ang2) mRNA expression in pulmonary artery smooth muscle cells (PASMC) was greater in SM22α-PHD1/2-/- compared with control mice in both normoxia and hyperoxia. Pulmonary endothelial cells (PECs) cocultured with PASMC isolated from SM22α-PHD1/2-/- mice formed more tubes and branches with greater tube length compared with PEC cocultured with PASMC isolated from SM22α-PHD1/2+/+ mice. Addition of Ang2 recombinant protein further augmented tube formation for both PHD1/2+/+ and PHD1/2-/- PASMC. Cell-specific deletion of PHD1 and 2 selectively increases HIF-1α expression in SM22α-expressing cells and protects neonatal lung development despite prolonged hyperoxia exposure. HIF stabilization in SM22α-expressing cells preserved endothelial cell proliferation, microvascular density, increased angiopoietin-2 expression, and lung structure, suggesting a role for cell-specific HIF-1α stabilization to prevent neonatal lung injury.


Asunto(s)
Hiperoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Lesión Pulmonar , Angiopoyetina 2/metabolismo , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Células Endoteliales/metabolismo , Humanos , Hiperoxia/metabolismo , Hiperoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Recién Nacido , Recien Nacido Prematuro , Pulmón/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/prevención & control , Ratones
6.
J Pediatr ; 231: 278-283.e2, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33359301

RESUMEN

ABCA3 deficiency is a rare cause of neonatal respiratory failure. Biallelic complete loss of function variants lead to neonatal demise without lung transplantation, but children with partial function variants have variable outcomes. The favorable clinical course of 3 such infants presenting with respiratory distress at birth is described.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/deficiencia , Transportadoras de Casetes de Unión a ATP/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Humanos , Recién Nacido , Masculino , Mutación , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Síndrome de Dificultad Respiratoria del Recién Nacido/terapia
7.
Curr Opin Pediatr ; 33(3): 302-310, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33938476

RESUMEN

PURPOSE OF REVIEW: Pediatric coronavirus disease 2019 (COVID-19) respiratory disease is a distinct entity from adult illness, most notable in its milder phenotype. This review summarizes the current knowledge of the clinical patterns, cellular pathophysiology, and epidemiology of COVID-19 respiratory disease in children with specific attention toward factors that account for the maturation-related differences in disease severity. RECENT FINDINGS: Over the past 14 months, knowledge of the clinical presentation and pathophysiology of COVID-19 pneumonia has rapidly expanded. The decreased disease severity of COVID-19 pneumonia in children was an early observation. Differences in the efficiency of viral cell entry and timing of immune recognition and response between children and adults remain at the center of ongoing research. SUMMARY: The clinical spectrum of COVID-19 respiratory disease in children is well defined. The age-related differences protecting children from severe disease and death remain incompletely understood.


Asunto(s)
COVID-19 , Trastornos Respiratorios , Enfermedades Respiratorias , Adulto , Niño , Humanos , SARS-CoV-2 , Índice de Severidad de la Enfermedad
11.
Am J Respir Crit Care Med ; 198(3): 320-328, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29688023

RESUMEN

RATIONALE: Asthma management depends on prompt identification of symptoms, which challenges both patients and providers. In asthma, a misapprehension of health between exacerbations can compromise compliance. Thus, there is a need for a tool that permits objective longitudinal monitoring without increasing the burden of patient compliance. OBJECTIVES: We sought to determine whether changes in nocturnal physiology are associated with asthma symptoms in pediatric patients. METHODS: Using a contactless bed sensor, nocturnal heart rate (HR), respiratory rate, relative stroke volume, and movement in children with asthma 5-18 years of age (n = 16) were recorded. Asthma symptoms and asthma control test (ACT) score were reported every 2 weeks. Random forest model was used to identify physiologic parameters associated with asthma symptoms. Elastic net regression was used to identify variables associated with ACT score. MEASUREMENTS AND MAIN RESULTS: The model on the full cohort performed with sensitivity of 47.2%, specificity of 96.3%, and accuracy of 87.4%; HR and respiratory parameters were the most important variables in this model. The model predicted asthma symptoms 35% of the time on the day before perception of symptoms, and 100% of the time for a select subject for which the model performed with greater sensitivity. Multivariable and bivariable analyses demonstrated significant association between HR and respiratory rate parameters and ACT score. CONCLUSIONS: Nocturnal physiologic changes correlate with asthma symptoms, supporting the notion that nocturnal physiologic monitoring represents an objective diagnostic tool capable of longitudinally assessing disease control and predicting asthma exacerbations in children with asthma at home.


Asunto(s)
Asma/diagnóstico , Asma/fisiopatología , Monitoreo Fisiológico/métodos , Adolescente , Niño , Preescolar , Ritmo Circadiano , Estudios de Cohortes , Progresión de la Enfermedad , Diagnóstico Precoz , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Estudios Longitudinales , Masculino , Estudios Prospectivos , Reproducibilidad de los Resultados , Frecuencia Respiratoria/fisiología , Sensibilidad y Especificidad , Volumen Sistólico/fisiología
12.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L66-L77, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29597831

RESUMEN

Compromised pulmonary endothelial cell (PEC) barrier function characterizes acute respiratory distress syndrome (ARDS), a cause of substantial morbidity and mortality. Survival from ARDS is greater in children compared with adults. Whether developmental differences intrinsic to PEC barrier function contribute to this survival advantage remains unknown. To test the hypothesis that PEC barrier function is more well-preserved in neonatal lungs compared with adult lungs in response to inflammation, we induced lung injury in neonatal and adult mice with systemic lipopolysaccharide (LPS). We assessed PEC barrier function in vivo and in vitro, evaluated changes in the expression of focal adhesion kinase 1 (FAK1) and phosphorylation in response to LPS, and determined the effect of FAK silencing and overexpression on PEC barrier function. We found that LPS induced a greater increase in lung permeability and PEC barrier disruption in the adult mice, despite similar degrees of inflammation and apoptosis. Although baseline expression was similar, LPS increased FAK1 expression in neonatal PEC but increased FAK1 phosphorylation and decreased FAK1 expression in adult PEC. Pharmacologic inhibition of FAK1 accentuated LPS-induced barrier disruption most in adult PEC. Finally, in response to LPS, FAK silencing markedly impaired neonatal PEC barrier function, whereas FAK overexpression preserved adult PEC barrier function. Thus, developmental differences in FAK expression during inflammatory injury serve to preserve neonatal pulmonary endothelial barrier function compared with that of adults and suggest that intrinsic differences in the immature versus pulmonary endothelium, especially relative to FAK1 phosphorylation, may contribute to the improved outcomes of children with ARDS.


Asunto(s)
Apoptosis , Barrera Alveolocapilar/enzimología , Células Endoteliales/enzimología , Endotelio/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , Transducción de Señal , Animales , Barrera Alveolocapilar/crecimiento & desarrollo , Barrera Alveolocapilar/patología , Células Endoteliales/patología , Endotelio/patología , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Inflamación/inducido químicamente , Inflamación/enzimología , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones
13.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L265-L275, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29644895

RESUMEN

Accessory subunits associated with the calcium-sensitive potassium channel (BKCa), a major determinant of vascular tone, confer functional and anatomical diversity. The ß1 subunit increases Ca2+ and voltagesensitivity of the BKCa channel and is expressed exclusively in smooth muscle cells. Evidence supporting the physiological significance of the ß1 subunit includes the observations that murine models with deletion of the ß1 subunit are hypertensive and that humans with a gain-of-function ß1 mutation are at a decreased risk of diastolic hypertension. However, whether the ß1 subunit of the BKCa channel contributes to the low tone that characterizes the normal pulmonary circulation or modulates the pulmonary vascular response to hypoxia remains unknown. To determine the role of the BKCa channel ß1 subunit in the regulation of pulmonary vascular tone and the response to acute and chronic hypoxia, mice with deletion of the Kcnmb1 gene that encodes for the ß1 subunit ( Kcnmb1-/-) were placed in chronic hypoxia (10% O2) for 21-24 days. In normoxia, right ventricular systolic pressure (RVSP) did not differ between Kcnmb1+/+ (controls) and Kcnmb1-/- mice. After exposure to either acute or chronic hypoxia, RVSP was higher in Kcnmb1-/- mice compared with Kcnmb1+/+ mice, without increased vascular remodeling. ß1 subunit expression was predominantly confined to pulmonary artery smooth muscle cells (PASMCs) from vessels ≤ 150 µm. Peripheral PASMCs contracted collagen gels irrespective of ß1 expression. Focal adhesion expression and Rho kinase activity were greater in Kcnmb1-/- compared with Kcnmb1+/+ PASMCs. Compromised PASMC ß1 function may contribute to the heightened microvascular vasoconstriction that characterizes pulmonary hypertension.


Asunto(s)
Hipoxia/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Pulmón/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Enfermedad Aguda , Animales , Enfermedad Crónica , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Adhesiones Focales/patología , Eliminación de Gen , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipoxia/genética , Hipoxia/patología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Vasoconstricción
14.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L422-L431, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29745253

RESUMEN

Pulmonary artery smooth muscle cells (PASMCs) express endothelin (ET-1), which modulates the pulmonary vascular response to hypoxia. Although cross-talk between hypoxia-inducible factor-1α (HIF-1α), an O2-sensitive transcription factor, and ET-1 is established, the cell-specific relationship between HIF-1α and ET-1 expression remains incompletely understood. We tested the hypotheses that in PASMCs 1) HIF-1α expression constrains ET-1 expression, and 2) a specific microRNA (miRNA) links HIF-1α and ET-1 expression. In human (h)PASMCs, depletion of HIF-1α with siRNA increased ET-1 expression at both the mRNA and protein levels ( P < 0.01). In HIF-1α-/- murine PASMCs, ET-1 gene and protein expression was increased ( P < 0.0001) compared with HIF-1α+/+ cells. miRNA profiles were screened in hPASMCs transfected with siRNA-HIF-1α, and RNA hybridization was performed on the Agilent (Santa Clara, CA) human miRNA microarray. With HIF-1α depletion, miRNA-543 increased 2.4-fold ( P < 0.01). In hPASMCs, miRNA-543 overexpression increased ET-1 gene ( P < 0.01) and protein ( P < 0.01) expression, decreased TWIST gene expression ( P < 0.05), and increased ET-1 gene and protein expression, compared with nontargeting controls ( P < 0.01). Moreover, we evaluated low passage hPASMCs from control and patients with idiopathic pulmonary arterial hypertension (IPAH). Compared with controls, protein expression of HIF-1α and Twist-related protein-1 (TWIST1) was decreased ( P < 0.05), and miRNA-543 and ET-1 expression increased ( P < 0.001) in hPASMCs from patients with IPAH. Thus, in PASMCs, loss of HIF-1α increases miRNA-543, which decreases Twist expression, leading to an increase in PASMC ET-1 expression. This previously undescribed link between HIF-1α and ET-1 via miRNA-543 mediated Twist suppression represents another layer of molecular regulation that might determine pulmonary vascular tone.


Asunto(s)
Endotelina-1/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/metabolismo , Animales , Células Cultivadas , Endotelina-1/genética , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Noqueados , MicroARNs/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
15.
FASEB J ; 31(2): 650-662, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27811062

RESUMEN

Pulmonary arterial hypertension (PAH) is an often fatal disease with limited treatment options. Whereas current data support the notion that, in pulmonary artery endothelial cells (PAECs), expression of transcription factor hypoxia inducible factor-1α (HIF-1α) is increased, the role of HIF-1α in pulmonary artery smooth muscle cells (PASMCs) remains controversial. This study investigates the hypothesis that, in PASMCs from patients with PAH, decreases in HIF-1α expression and activity underlie augmented pulmonary vascular contractility. PASMCs and tissues were isolated from nonhypertensive control patients and patients with PAH. Compared with controls, HIF-1α and Kv1.5 protein expression were decreased in PAH smooth muscle cells (primary culture). Myosin light chain (MLC) phosphorylation and MLC kinase (MLCK) activity-major determinants of vascular tone-were increased in patients with PAH. Cofactors involved in prolyl hydroxylase domain activity were increased in PAH smooth muscle cells. Functionally, PASMC contractility was inversely correlated with HIF-1α activity. In PASMCs derived from patients with PAH, HIF-1α expression is decreased, and MLCK activity, MLC phosphorylation, and cell contraction are increased. We conclude that compromised PASMC HIF-1α expression may contribute to the increased tone that characterizes pulmonary hypertension.-Barnes, E. A., Chen, C.-H., Sedan, O., Cornfield, D. N. Loss of smooth muscle cell hypoxia inducible factor-1α underlies increased vascular contractility in pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/fisiología , Vasoconstricción/fisiología , Aminoácidos Dicarboxílicos/farmacología , Dimetilsulfóxido/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Prolil Hidroxilasas/genética , Prolil Hidroxilasas/metabolismo
17.
Curr Opin Pediatr ; 29(3): 320-325, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28338487

RESUMEN

PURPOSE OF REVIEW: Bronchopulmonary dysplasia (BPD) or chronic lung disease of infancy BPD was originally described 50 years ago, in 1967 by Northway et al. This article possesses two fundamental objectives to provide: a brief historical perspective on BPD; and an update relative to current notions of epidemiology, pathophysiology, evaluation, and clinical management of BPD complicated by vascular disease. The review highlights areas of consensus and ongoing uncertainty. RECENT FINDINGS: The clinical cause and presentation of infants with BPD has evolved over the past several decades. Considerable improvements in neonatal care, including surfactant replacement therapies, antenatal steroids, nutritional support, ventilator management, and attention to the potential of oxygen toxicity, underlie the evolution of BPD. Most children with BPD improve over time. However, in the presence of vascular disease, the morbidity and mortality associated with BPD increases considerably. Though recent recommendations include procuring an echocardiogram to screen for pulmonary hypertension in infants with established BPD, there is less agreement surrounding the additional diagnostic and putative treatment modalities for infants with BPD and pulmonary hypertension. The indications, rationale, potential benefits, and risks of vasodilator therapy in BPD are discussed. SUMMARY: The pediatric community has 50 years of experience with BPD. Past experience should be used to inform present and future diagnostic and treatment strategies. This review seeks to arm the clinician with evidence that motivates a physiology-based approach to the management of infants with BPD and pulmonary hypertension.


Asunto(s)
Displasia Broncopulmonar/complicaciones , Hipertensión Pulmonar/etiología , Displasia Broncopulmonar/fisiopatología , Displasia Broncopulmonar/terapia , Enfermedad Crónica , Progresión de la Enfermedad , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/terapia , Lactante , Recién Nacido , Recien Nacido Prematuro
18.
Kidney Int ; 90(3): 598-609, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27470678

RESUMEN

Medial vascular calcification is a common complication of chronic kidney disease (CKD). Although elevated inorganic phosphate stimulates vascular smooth muscle cell (VSMC) osteogenic transdifferentiation and calcification, the mechanisms involved in their calcification during CKD are not fully defined. Because hypoxic gene activation is linked to CKD and stimulates bone cell osteogenic differentiation, we used in vivo and in vitro rodent models to define the role of hypoxic signaling during elevated inorganic phosphate-induced VSMC calcification. Cell mineralization studies showed that elevated inorganic phosphate rapidly induced VSMC calcification. Hypoxia strongly enhanced elevated inorganic phosphate-induced VSMC calcification and osteogenic transdifferentiation, as seen by osteogenic marker expression. Hypoxia-inducible factor-1 (HIF-1), the key hypoxic transcription factor, was essential for enhanced VSMC calcification. Targeting HIF-1 expression in murine VSMC blocked calcification in hypoxia with elevated inorganic phosphate while HIF-1 activators, including clinically used FG-4592/Roxadustat, recreated a procalcifying environment. Elevated inorganic phosphate rapidly activated HIF-1, even in normal oxygenation; an effect mediated by HIF-1α subunit stabilization. Thus, hypoxia synergizes with elevated inorganic phosphate to enhance VSMC osteogenic transdifferentiation. Our work identifies HIF-1 as an early CKD-related pathological event, prospective marker, and potential target against vascular calcification in CKD-relevant conditions.


Asunto(s)
Transdiferenciación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Músculo Liso Vascular/patología , Fosfatos/metabolismo , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Glicina/análogos & derivados , Glicina/farmacología , Humanos , Hipoxia/metabolismo , Inmunohistoquímica , Isoquinolinas/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Calcificación Vascular/etiología , Rigidez Vascular
20.
Curr Opin Pediatr ; 28(3): 324-30, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27043088

RESUMEN

PURPOSE OF REVIEW: This review is written from the perspective of the pediatric clinician involved in the care of premature infants at risk for pulmonary hypertension. The main objective is to better inform the clinician in the diagnosis and treatment of pulmonary hypertension in premature infants by reviewing the available relevant literature and focusing on the areas for which there is the greatest need for continued research. RECENT FINDINGS: Continued knowledge regarding the epidemiology of pulmonary hypertension in the premature infant population has aided better diagnostic screening algorithms. Included in this knowledge, is the association of pulmonary hypertension in infants with bronchopulmonary dysplasia (BPD). However, it is also known that beyond BPD, low birth weight and other conditions that result in increased systemic inflammation are associated with pulmonary hypertension. This information has led to the recent recommendation that all infants with BPD should have an echocardiogram to evaluate for evidence of pulmonary hypertension prior to discharge from the neonatal ICU. SUMMARY: Pulmonary hypertension can be a significant comorbidity for premature infants. This review aims to focus the clinician on the available literature to improve recognition of the condition to allow for more timely interventions.


Asunto(s)
Displasia Broncopulmonar , Ecocardiografía , Hipertensión Pulmonar , Enfermedades del Prematuro/fisiopatología , Displasia Broncopulmonar/diagnóstico , Displasia Broncopulmonar/fisiopatología , Displasia Broncopulmonar/terapia , Comorbilidad , Investigación sobre Servicios de Salud , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/terapia , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Unidades de Cuidado Intensivo Neonatal , Guías de Práctica Clínica como Asunto , Poblaciones Vulnerables
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA