Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 204(3): 326-338, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33705684

RESUMEN

Rationale: Although pulmonary endothelial progenitor cells (EPCs) hold promise for cell-based therapies for neonatal pulmonary disorders, whether EPCs can be derived from pluripotent embryonic stem cells (ESCs) or induced pluripotent stem cells remains unknown.Objectives: To investigate the heterogeneity of pulmonary EPCs and derive functional EPCs from pluripotent ESCs.Methods: Single-cell RNA sequencing of neonatal human and mouse lung was used to identify the heterogeneity of pulmonary EPCs. CRISPR/Cas9 gene editing was used to genetically label and purify mouse pulmonary EPCs. Functional properties of the EPCs were assessed after cell transplantation into neonatal mice with S52F Foxf1 mutation, a mouse model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Interspecies mouse-rat chimeras were produced through blastocyst complementation to generate EPCs from pluripotent ESCs for cell therapy in ACDMPV mice.Measurements and Main Results: We identified a unique population of EPCs, FOXF1+cKIT+ EPCs, as a subset of recently described general capillary cells (gCAPs) expressing SMAD7, ZBTB20, NFIA, and DLL4 but lacking mature arterial, venous, and lymphatic markers. FOXF1+cKIT+ gCAPs are reduced in ACDMPV, and their transcriptomic signature is conserved in mouse and human lungs. After cell transplantation into the neonatal circulation of ACDMPV mice, FOXF1+cKIT+ gCAPs engraft into the pulmonary vasculature, stimulate angiogenesis, improve oxygenation, and prevent alveolar simplification. FOXF1+cKIT+ gCAPs, produced from ESCs in interspecies chimeras, are fully competent to stimulate neonatal lung angiogenesis and alveolarization in ACDMPV mice.Conclusions: Cell-based therapy using donor or ESC/induced pluripotent stem cell-derived FOXF1+cKIT+ endothelial progenitors may be considered for treatment of human ACDMPV.


Asunto(s)
Células Madre Embrionarias/citología , Células Progenitoras Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Síndrome de Circulación Fetal Persistente/terapia , Trasplante de Células Madre , Animales , Animales Recién Nacidos , Sistemas CRISPR-Cas , Quimera , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/trasplante , Factores de Transcripción Forkhead/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido , Ratones , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/patología , Células Madre Pluripotentes , RNA-Seq , Ratas , Análisis de la Célula Individual
2.
Hum Mutat ; 42(6): 694-698, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33739555

RESUMEN

The FOXF1 gene, causative for a neonatal lethal lung developmental disorder alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), maps 1.7 kb away from the long noncoding RNA gene FENDRR on the opposite strand, suggesting they may be coregulated. Using RNA sequencing in lung tissue from ACDMPV patients with heterozygous deletions of the FOXF1 distant enhancer located 286 kb upstream, leaving FOXF1 and FENDRR intact, we have found that the FENDRR and FOXF1 expressions were reduced by approximately 75% and 50%, respectively, and were monoallelic from the intact chromosome 16q24.1. In contrast, ACDMPV patients with FOXF1 SNVs had biallelic FENDRR expression reduced by 66%-82%. Corroboratively, depletion of FOXF1 by small interfering RNA in lung fibroblasts resulted in a 50% decrease of FENDRR expression. These data indicate that FENDRR expression in the lungs is regulated both in cis by the FOXF1 distant enhancer and in trans by FOXF1. Our findings are compatible with the involvement of FENDRR in FOXF1-related disorders, including ACDMPV.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Factores de Transcripción Forkhead/genética , Pulmón/metabolismo , Síndrome de Circulación Fetal Persistente/genética , ARN Largo no Codificante/genética , Estudios de Casos y Controles , Femenino , Factores de Transcripción Forkhead/metabolismo , Mutación del Sistema de Lectura , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Recién Nacido , Masculino , Mutación Missense , Especificidad de Órganos/genética , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/patología , Polimorfismo de Nucleótido Simple , Embarazo , ARN Largo no Codificante/metabolismo
3.
Respir Res ; 22(1): 212, 2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315444

RESUMEN

BACKGROUND: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare lethal congenital lung disorder in neonates characterized by severe progressive respiratory failure and refractory pulmonary hypertension, resulting from underdevelopment of the peripheral pulmonary tree. Causative heterozygous single nucleotide variants (SNVs) or copy-number variant (CNV) deletions involving FOXF1 or its distant lung-specific enhancer on chromosome 16q24.1 have been identified in 80-90% of ACDMPV patients. FOXF1 maps closely to and regulates the oppositely oriented FENDRR, with which it also shares regulatory elements. METHODS: To better understand the transcriptional networks downstream of FOXF1 that are relevant for lung organogenesis, using RNA-seq, we have examined lung transcriptomes in 12 histopathologically verified ACDMPV patients with or without pathogenic variants in the FOXF1 locus and analyzed gene expression profile in FENDRR-depleted fetal lung fibroblasts, IMR-90. RESULTS: RNA-seq analyses in ACDMPV neonates revealed changes in the expression of several genes, including semaphorins (SEMAs), neuropilin 1 (NRP1), and plexins (PLXNs), essential for both epithelial branching and vascular patterning. In addition, we have found deregulation of the vascular endothelial growth factor (VEGF) signaling that also controls pulmonary vasculogenesis and a lung-specific endothelial gene TMEM100 known to be essential in vascular morphogenesis. Interestingly, we have observed a substantial difference in gene expression profiles between the ACDMPV samples with different types of FOXF1 defect. Moreover, partial overlap between transcriptome profiles of ACDMPV lungs with FOXF1 SNVs and FENDRR-depleted IMR-90 cells suggests contribution of FENDRR to ACDMPV etiology. CONCLUSIONS: Our transcriptomic data imply potential crosstalk between several lung developmental pathways, including interactions between FOXF1-SHH and SEMA-NRP or VEGF/VEGFR2 signaling, and provide further insight into complexity of lung organogenesis in humans.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Pulmón/metabolismo , Síndrome de Circulación Fetal Persistente/metabolismo , Semaforinas/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen/métodos , Humanos , Recién Nacido , Pulmón/patología , Masculino , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/patología , Semaforinas/genética , Factor A de Crecimiento Endotelial Vascular/genética
4.
Am J Respir Cell Mol Biol ; 63(4): 510-518, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32609558

RESUMEN

Optimal oxygen saturation as measured by pulse oximetry (SpO2) in neonatal lung injury, such as meconium aspiration syndrome (MAS) and persistent pulmonary hypertension of newborn (PPHN), is not known. Our goal was to determine the SpO2 range in lambs with MAS and PPHN that results in the highest brain oxygen delivery (bDO2) and pulmonary blood flow (Qp) and the lowest pulmonary vascular resistance and oxidative stress. Meconium was instilled into endotracheal tubes in 25 near-term gestation lambs, and the umbilical cord was occluded to induce asphyxia and gasping, causing MAS and PPHN. Lambs were randomized into four groups and ventilated for 6 hours with fixed fraction of inspired oxygen (FiO2) = 1.0 irrespective of SpO2, and three groups had FiO2 titrated to keep preductal SpO2 between 85% and 89%, 90% and 94%, and 95% and 99%, respectively. Tissues were collected to measure nitric oxide synthase activity, 3-nitrotyrosine, and 8-isoprostanes. Throughout the 6-hour exposure period, lambs in the 95-99% SpO2 target group had the highest Qp, lowest pulmonary vascular resistance, and highest bDO2 but were exposed to higher FiO2 (0.5 ± 0.21 vs. 0.29 ± 0.17) with higher lung 3-nitrotyrosine (0.67 [interquartile range (IQR), 0.43-0.73] ng/mcg protein vs. 0.1 [IQR, 0.09-0.2] ng/mcg protein) and lower lung nitric oxide synthase activity (196 [IQR, 192-201] mMol nitrite/mg protein vs. 270 [IQR, 227-280] mMol nitrite/mg protein) compared with the 90-94% target group. Brain 3-nitrotyrosine was lower in the 85-89% target group, and brain/lung 8-isoprostane levels were not significantly different. In term lambs with MAS and PPHN, Qp and bDO2 through the first 6 hours are higher with target SpO2 in the 95-99% range. However, the 90-94% target range is associated with significantly lower FiO2 and lung oxidative stress. Clinical trials comparing the 90-94% versus the 95-99% SpO2 target range in term infants with PPHN are warranted.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Pulmón/metabolismo , Síndrome de Aspiración de Meconio/metabolismo , Oxígeno/metabolismo , Animales , Animales Recién Nacidos , Dinoprost/análogos & derivados , Dinoprost/farmacología , Femenino , Hipertensión Pulmonar/tratamiento farmacológico , Pulmón/efectos de los fármacos , Masculino , Síndrome de Aspiración de Meconio/tratamiento farmacológico , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oximetría/métodos , Síndrome de Circulación Fetal Persistente/tratamiento farmacológico , Síndrome de Circulación Fetal Persistente/metabolismo , Embarazo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ovinos/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacología , Resistencia Vascular/efectos de los fármacos
5.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L933-L944, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30234376

RESUMEN

Persistent pulmonary hypertension of the newborn (PPHN) features hypoxemia, pulmonary vasoconstriction, and impaired cardiac inotropy. We previously reported low basal and stimulated cAMP in hypoxic pulmonary artery smooth muscle cells (PASMCs). We now examine pulmonary arterial adenylyl cyclase (AC) activity and regulation in hypoxic PPHN. PPHN was induced in newborn swine by normobaric hypoxia (fraction of inspired oxygen 0.10) for 72 h and compared with age-matched normoxic controls. We studied relaxation of pulmonary arterial (PA) rings to AC activator forskolin and cGMP activator sodium nitroprusside (SNP) by isometric myography, ATP content, phosphodiesterase activity, AC content, isoform expression, and catalytic activity in presence or absence of Gαs-coupled receptor agonists, forskolin, or transnitrosylating agents in human and neonatal porcine PASMCs and HEK293T stably expressing AC isoform 6, after 72 h hypoxia (10% O2) or normoxia (21% O2). Relaxation to forskolin and SNP were equally impaired in PPHN PA. AC-specific activity decreased in hypoxia. PASMC from PPHN swine had reduced AC activity despite exposure to normoxia in culture; transient hypoxia in vitro further decreased AC activity. Prostacyclin receptor ligand affinity decreased, but its association with Gαs increased in hypoxia. Total AC content was unchanged by hypoxia, but AC6 increased in hypoxic cells and PPHN pulmonary arteries. Impairment of AC6 activity in hypoxia was associated with nitrosylation. PPHN PA relaxation is impaired because of loss of AC activity. Hypoxic AC is inhibited because of S-nitrosylation; inhibition persists after removal from hypoxia. Downregulation of AC-mediated relaxation in hypoxic PA has implications for utility of Gαs-coupled receptor agonists in PPHN treatment.


Asunto(s)
Adenilil Ciclasas/metabolismo , Animales Recién Nacidos/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Síndrome de Circulación Fetal Persistente/metabolismo , Animales , Animales Recién Nacidos/fisiología , Línea Celular , GMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Células HEK293 , Humanos , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Relajación Muscular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Nitroprusiato/farmacología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Porcinos
6.
BMC Med Genet ; 19(1): 202, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30458719

RESUMEN

BACKGROUND: Genetic defects in the mitochondrial aminoacyl-tRNA synthetase are important causes of mitochondrial disorders. VARS2 is one of the genes encoding aminoacyl-tRNA synthetases. Recently, an increasing number of pathogenic variants of VARS2 have been reported. CASE PRESENTATION: We report the novel compound heterozygous pathogenic VARS2 mutations c.643 C > T (p. His215Tyr) and c.1354 A > G (p. Met452Val) in a female infant who presented with poor sucking at birth, poor activity, hyporeflexia, hypertonia, persistent pulmonary hypertension of newborn (PPHN), metabolic acidosis, severe lactic acidosis, expansion and hypertrophic cardiomyopathy. These heterozygous mutations were carried individually by the proband's parents and elder sister; the two mutations segregated in the family and were the cause of the disease in the proband.The c.643 C > T (p. His215Tyr) mutation was not described in the ExaC, GNomAD and 1000 Genomes Project databases, and the frequency of c.1354 A > G (p. Met452Val) was < 0.001 in these gene databases.The two mutated amino acids were located in a highly conserved region of the VARS2 protein that is important for its interaction with the cognate tRNA. The two missense mutations were predicted by online tools to be damaging and deleterious. CONCLUSIONS: Our report expands the spectrum of known pathogenicVARS2 variants associated with mitochondrial disorders in humans.VARS2 deficiency may cause a severe neonatal presentation with structural cardiac abnormalities.


Asunto(s)
Acidosis Láctica/genética , Cardiomiopatía Hipertrófica/genética , Antígenos HLA/genética , Paro Cardíaco/genética , Enfermedades Mitocondriales/genética , Mutación Missense , Síndrome de Circulación Fetal Persistente/genética , Valina-ARNt Ligasa/genética , Acidosis Láctica/diagnóstico , Acidosis Láctica/metabolismo , Acidosis Láctica/fisiopatología , Adulto , Alelos , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Preescolar , Resultado Fatal , Femenino , Expresión Génica , Frecuencia de los Genes , Paro Cardíaco/diagnóstico , Paro Cardíaco/metabolismo , Paro Cardíaco/fisiopatología , Heterocigoto , Humanos , Recién Nacido , Masculino , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Linaje , Síndrome de Circulación Fetal Persistente/diagnóstico , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/fisiopatología
7.
Histochem Cell Biol ; 147(6): 707-719, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28247028

RESUMEN

Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.


Asunto(s)
Cadherinas/metabolismo , Síndrome de Circulación Fetal Persistente/metabolismo , Infecciones Neumocócicas/metabolismo , Alveolos Pulmonares/anomalías , Humanos , Síndrome de Circulación Fetal Persistente/microbiología , Infecciones Neumocócicas/microbiología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/microbiología , Streptococcus pneumoniae
8.
Am J Perinatol ; 34(8): 749-758, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28099979

RESUMEN

Objective To describe the pharmacokinetics and pharmacodynamics of milrinone in infants with persistent pulmonary hypertension of the newborn (PPHN) and to explore the impact of age on milrinone disposition. Design Randomized, open label pilot study. Setting Multicenter; level 3 and level 4 neonatal intensive care units. Patients Six infants ≥34 weeks' gestational age and <10 days of life with persistent hypoxemia receiving inhaled nitric oxide. Intervention Intravenous milrinone lactate in one of two dosing regimens: (1) low dose, 20 mcg/kg bolus followed by 0.2 mcg/kg/minute, and (2) standard dose, 50 mcg/kg bolus followed by 0.5 mcg/kg/minute. Measurements and Main Results The final structural model was a two-compartment disposition model with interindividual variability estimated on clearance (CL). The estimated value of CL is 7.65 mL/minute/3.4 kg (3.05 mL/minute/kg). The addition of age improved the precision of the CL estimate, and CL increased with chronological age in days. The oxygenation index was highly variable within each participant and improved with time. There were no observed safety concerns in either dosing group. Conclusion The CL of milrinone in newborns with PPHN is reduced and increases with age. In this pilot study, we did not see significant pharmacodynamic or safety effects associated with drug exposure.


Asunto(s)
Milrinona , Óxido Nítrico/administración & dosificación , Síndrome de Circulación Fetal Persistente , Administración Intravenosa , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacocinética , Relación Dosis-Respuesta a Droga , Factores Relajantes Endotelio-Dependientes/administración & dosificación , Femenino , Edad Gestacional , Humanos , Recién Nacido , Unidades de Cuidado Intensivo Neonatal/estadística & datos numéricos , Masculino , Tasa de Depuración Metabólica , Milrinona/administración & dosificación , Milrinona/farmacocinética , Consumo de Oxígeno/efectos de los fármacos , Síndrome de Circulación Fetal Persistente/diagnóstico , Síndrome de Circulación Fetal Persistente/tratamiento farmacológico , Síndrome de Circulación Fetal Persistente/metabolismo , Proyectos Piloto , Resultado del Tratamiento
9.
Am J Physiol Lung Cell Mol Physiol ; 310(1): L40-9, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519208

RESUMEN

Impaired vasodilation in persistent pulmonary hypertension of the newborn (PPHN) is characterized by mitochondrial dysfunction. We investigated the hypothesis that a decreased endothelial nitric oxide synthase level leads to impaired mitochondrial biogenesis and function in a lamb model of PPHN induced by prenatal ductus arteriosus constriction. We ventilated PPHN lambs with 100% O2 alone or with inhaled nitric oxide (iNO). We treated pulmonary artery endothelial cells (PAECs) from normal and PPHN lambs with detaNONOate, an NO donor. We observed decreased mitochondrial (mt) DNA copy number, electron transport chain (ETC) complex subunit levels, and ATP levels in PAECs and lung tissue of PPHN fetal lambs at baseline compared with gestation matched controls. Phosphorylation of AMP-activated kinase (AMPK) and levels of peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC-1α) and sirtuin-1, which facilitate mitochondrial biogenesis, were decreased in PPHN. Ventilation with 100% O2 was associated with larger decreases in ETC subunits in the lungs of PPHN lambs compared with unventilated PPHN lambs. iNO administration, which facilitated weaning of FiO2 , partly restored mtDNA copy number, ETC subunit levels, and ATP levels. DetaNONOate increased eNOS phosphorylation and its interaction with heat shock protein 90 (HSP90); increased levels of superoxide dismutase 2 (SOD2) mRNA, protein, and activity; and decreased the mitochondrial superoxide levels in PPHN-PAECs. Knockdown of eNOS decreased ETC protein levels in control PAECs. We conclude that ventilation with 100% O2 amplifies oxidative stress and mitochondrial dysfunction in PPHN, which are partly improved by iNO and weaning of oxygen.


Asunto(s)
Células Endoteliales/metabolismo , Hipertensión Pulmonar/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Síndrome de Circulación Fetal Persistente/metabolismo , Animales , Animales Recién Nacidos , Femenino , Feto/inmunología , Feto/metabolismo , Mitocondrias/inmunología , Óxido Nítrico/metabolismo , Receptores Activados del Proliferador del Peroxisoma/inmunología , Embarazo , Ovinos
10.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L196-203, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26024892

RESUMEN

This study was designed to determine whether cyclic stretch induces a persistent pulmonary hypertension of the newborn (PPHN) phenotype of increased NADPH oxidase (Nox) 4 signaling in control pulmonary artery smooth muscle cells (PASMC), and to identify the signal transduction molecules involved. To achieve this, PPHN was induced in lambs by antenatal ligation of the ductus arteriosus at 128 days gestation. After 9 days, lungs and PASMC were isolated from control (twin) and PPHN lambs. Control PASMC were exposed to cyclic stretch at 1 Hz and 15% elongation for 24 h. Stretch-induced Nox4 expression was attenuated by inhibition of mitochondrial complex III and NF-κB, and stretch-induced protein thiol oxidation was attenuated by Nox4 small interfering RNA and complex III inhibition. NF-κB activity was increased by stretch in a complex III-dependent fashion, and stretch-induced cyclin D1 expression was attenuated by complex III inhibition and Nox4 small interfering RNA. This is the first study to show that cyclic stretch increases Nox4 expression via mitochondrial complex III-induced activation of NF-κB in fetal PASMC, resulting in ROS signaling and increased cyclin D1 expression. Targeting these signaling molecules may attenuate pulmonary vascular remodeling associated with PPHN.


Asunto(s)
Hipertensión Pulmonar/etiología , Mitocondrias/metabolismo , Miocitos del Músculo Liso/patología , NADPH Oxidasas/metabolismo , Síndrome de Circulación Fetal Persistente/etiología , Arteria Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Estrés Mecánico , Animales , Western Blotting , Células Cultivadas , Femenino , Feto/metabolismo , Feto/patología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Mitocondrias/patología , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/patología , Arteria Pulmonar/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ovinos , Transducción de Señal
11.
Am J Physiol Lung Cell Mol Physiol ; 306(3): L299-308, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24375792

RESUMEN

Peroxisome proliferator-activated receptor-γ (PPARγ) and Rho-kinase (ROCK) regulate smooth muscle cell (SMC) proliferation and contribute to vascular remodeling in adult pulmonary hypertension. Whether these pathways interact to contribute to the development of vascular remodeling in persistent pulmonary hypertension of the newborn (PPHN) remains unknown. We hypothesized that ROCK-PPARγ interactions increase SMC proliferation resulting in vascular remodeling in experimental PPHN. Pulmonary artery SMCs (PASMCs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Cell counts were performed daily for 5 days with or without PPARγ agonists and ROCK inhibition. PPARγ and ROCK protein expression/activity were measured by Western blot in normal and PPHN PASMCs. We assessed PPARγ-ROCK interactions by studying the effect of ROCK activation on PPARγ activity and PPARγ inhibition (siRNA) on ROCK activity and PASMC proliferation. At baseline, PPHN PASMC cell number was increased by 38% above controls on day 5. ROCK protein expression/activity were increased by 25 and 34% and PPARγ protein/activity decreased by 40 and 50% in PPHN PASMC. ROCK inhibition and PPARγ activation restored PPHN PASMC growth to normal values. ROCK inhibition increased PPARγ activity by 50% in PPHN PASMC, restoring PPARγ activity to normal. In normal PASMCs, ROCK activation decreased PPARγ activity and PPARγ inhibition increased ROCK activity and cell proliferation, resulting in a PPHN hyperproliferative PASMC phenotype. PPARγ-ROCK interactions regulate SMC proliferation and contribute to increased PPHN PASMC proliferation and vascular remodeling in PPHN. Restoring normal PPARγ-ROCK signaling may prevent vascular remodeling and improve outcomes in PPHN.


Asunto(s)
PPAR gamma/metabolismo , Síndrome de Circulación Fetal Persistente/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , PPAR gamma/antagonistas & inhibidores , PPAR gamma/biosíntesis , Ovinos , Transducción de Señal , Quinasas Asociadas a rho/biosíntesis
12.
Am J Physiol Lung Cell Mol Physiol ; 306(4): L361-71, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337925

RESUMEN

Increased endothelin-1 (ET-1) disrupts angiogenesis in persistent pulmonary hypertension of the newborn (PPHN), but pathogenic mechanisms are unclear. Peroxisome proliferator activated receptor γ (PPARγ) is decreased in adult pulmonary hypertension, but whether ET-1-PPARγ interactions impair endothelial cell function and angiogenesis in PPHN remains unknown. We hypothesized that increased PPHN pulmonary artery endothelial cell (PAEC) ET-1 production decreases PPARγ signaling and impairs tube formation in vitro. Proximal PAECs were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. PPARγ and phospho-PPARγ protein were compared between normal and PPHN PAECs ± ET-1 and bosentan (ETA/ETB receptor blocker). Tube formation was assessed in response to PPARγ agonists ± ET-1, N-nitro-l-arginine (LNA) (NOS inhibitor), and PPARγ siRNA. Endothelial NO synthase (eNOS), phospho-eNOS, and NO production were measured after exposure to PPARγ agonists and PPARγ siRNA. At baseline, PPHN PAECs demonstrate decreased tube formation and PPARγ protein expression and activity. PPARγ agonists restored PPHN tube formation to normal. ET-1 decreased normal and PPHN PAEC tube formation, which was rescued by PPARγ agonists. ET-1 decreased PPARγ protein and activity, which was prevented by bosentan. PPARγ agonists increased eNOS protein and activity and NO production in normal and PPHN PAECs. LNA inhibited the effect of PPARγ agonists on tube formation. PPARγ siRNA decreased eNOS protein and tube formation in normal PAECs. We conclude that ET-1 decreases PPARγ signaling and contributes to PAEC dysfunction and impaired angiogenesis in PPHN. We speculate that therapies aimed at decreasing ET-1 production will restore PPARγ signaling, preserve endothelial function, and improve angiogenesis in PPHN.


Asunto(s)
Células Endoteliales/metabolismo , Endotelina-1/fisiología , Neovascularización Fisiológica , PPAR gamma/metabolismo , Animales , Bosentán , Células Cultivadas , Antagonistas de los Receptores de Endotelina , Humanos , Recién Nacido , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR gamma/agonistas , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/fisiopatología , Arteria Pulmonar/patología , Receptores de Endotelina/metabolismo , Ovinos , Transducción de Señal , Sulfonamidas/farmacología
13.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L877-87, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25281640

RESUMEN

In hypoxic pulmonary arterial (PA) myocytes, challenge with thromboxane mimetic U46619 induces marked actin polymerization and contraction, phenotypic features of persistent pulmonary hypertension of the newborn (PPHN). Rho GTPases regulate the actin cytoskeleton. We previously reported that U46619-induced actin polymerization in hypoxic PA myocytes occurs independently of the RhoA pathway and hypothesized involvement of the Cdc42 pathway. PA myocytes grown in normoxia or hypoxia for 72 h were stimulated with U46619, then analyzed for Rac/Cdc42 activation by affinity precipitation, phosphatidylinositide-3-kinase (PI3K) activity by phospho-Akt, phospho-p21-activated kinase (PAK) by immunoblot, and association of Cdc42 with neuronal Wiskott Aldrich Syndrome protein (N-WASp) by immunoprecipitation. The effect of Rac or PAK inhibition on filamentous actin was quantified by laser-scanning cytometry and by cytoskeletal fractionation; effects of actin-modifying agents were measured by isometric myography. Basal Cdc42 activity increased in hypoxia, whereas Rac activity decreased. U46619 challenge increased Cdc42 and Rac activity in hypoxic cells, independently of PI3K. Hypoxia increased phospho-PAK, unaltered by U46619. Association of Cdc42 with N-WASp decreased in hypoxia but increased after U46619 exposure. Hypoxia doubled filamentous-to-globular ratios of α- and γ-actin isoforms. Jasplakinolide stabilized γ-filaments, increasing force; cytochalasin D depolymerized all actin isoforms, decreasing force. Rac and PAK inhibition decreased filamentous actin in tissues although without decrease in force. Rho inhibition decreased myosin phosphorylation and force. Hypoxia induces actin polymerization in PA myocytes, particularly increasing filamentous α- and γ-actin, contributing to U46619-induced contraction. Hypoxic PA myocytes challenged with a thromboxane mimetic polymerize actin via the Cdc42 pathway, reflecting increased Cdc42 association with N-WASp. Mechanisms regulating thromboxane-mediated actin polymerization are potential targets for future PPHN pharmacotherapy.


Asunto(s)
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Actinas/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Tromboxanos/farmacología , Proteína de Unión al GTP cdc42/metabolismo , Actinas/biosíntesis , Aminoquinolinas/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Citocalasina D/farmacología , Contracción Muscular/fisiología , Miocitos del Músculo Liso/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Síndrome de Circulación Fetal Persistente/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Isoformas de Proteínas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/metabolismo , Pirimidinas/farmacología , Transducción de Señal , Porcinos , Vasoconstrictores/farmacología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
14.
Nat Commun ; 15(1): 5233, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898031

RESUMEN

Mutations in the FOXF1 gene, a key transcriptional regulator of pulmonary vascular development, cause Alveolar Capillary Dysplasia with Misalignment of Pulmonary Veins, a lethal lung disease affecting newborns and infants. Identification of new FOXF1 upstream regulatory elements is critical to explain why frequent non-coding FOXF1 deletions are linked to the disease. Herein, we use multiome single-nuclei RNA and ATAC sequencing of mouse and human patient lungs to identify four conserved endothelial and mesenchymal FOXF1 enhancers. We demonstrate that endothelial FOXF1 enhancers are autoactivated, whereas mesenchymal FOXF1 enhancers are regulated by EBF1 and GLI1. The cell-specificity of FOXF1 enhancers is validated by disrupting these enhancers in mouse embryonic stem cells using CRISPR/Cpf1 genome editing followed by lineage-tracing of mutant embryonic stem cells in mouse embryos using blastocyst complementation. This study resolves an important clinical question why frequent non-coding FOXF1 deletions that interfere with endothelial and mesenchymal enhancers can lead to the disease.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead , Mesodermo , Síndrome de Circulación Fetal Persistente , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Animales , Humanos , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/patología , Síndrome de Circulación Fetal Persistente/metabolismo , Ratones , Elementos de Facilitación Genéticos/genética , Mesodermo/metabolismo , Mesodermo/embriología , Pulmón/patología , Células Endoteliales/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Células Madre Embrionarias/metabolismo , Alveolos Pulmonares/anomalías
15.
Hum Mutat ; 34(6): 801-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23505205

RESUMEN

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACD/MPV) is a rare and lethal developmental disorder of the lung defined by a constellation of characteristic histopathological features. Nonpulmonary anomalies involving organs of gastrointestinal, cardiovascular, and genitourinary systems have been identified in approximately 80% of patients with ACD/MPV. We have collected DNA and pathological samples from more than 90 infants with ACD/MPV and their family members. Since the publication of our initial report of four point mutations and 10 deletions, we have identified an additional 38 novel nonsynonymous mutations of FOXF1 (nine nonsense, seven frameshift, one inframe deletion, 20 missense, and one no stop). This report represents an up to date list of all known FOXF1 mutations to the best of our knowledge. Majority of the cases are sporadic. We report four familial cases of which three show maternal inheritance, consistent with paternal imprinting of the gene. Twenty five mutations (60%) are located within the putative DNA-binding domain, indicating its plausible role in FOXF1 function. Five mutations map to the second exon. We identified two additional genic and eight genomic deletions upstream to FOXF1. These results corroborate and extend our previous observations and further establish involvement of FOXF1 in ACD/MPV and lung organogenesis.


Asunto(s)
Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Mutación , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Secuencia de Aminoácidos , Mapeo Cromosómico , Bases de Datos Genéticas , Femenino , Factores de Transcripción Forkhead/química , Dosificación de Gen , Orden Génico , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Síndrome de Circulación Fetal Persistente/mortalidad , Síndrome de Circulación Fetal Persistente/patología , Alineación de Secuencia
16.
Pediatr Res ; 73(3): 252-62, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23202724

RESUMEN

BACKGROUND: Endothelin-1 (ET-1) and Rho-kinase (ROCK) increase vascular tone in experimental persistent pulmonary hypertension of the newborn (PPHN). Whether ET-1 activates ROCK to decrease angiogenesis in the developing lung remains unknown. METHODS: Proximal pulmonary artery endothelial cells (PAECs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Growth and tube formation were assessed after ET-1 treatment. The effect of ET-1 antagonism on tube formation was studied using ET-1 small interfering RNA (siRNA), ET-1 monoclonal antibodies (ET-1mAbs), BQ-123 (an ET(A) blocker), and bosentan (an ET(A)/ET(B) blocker). ET-1 gene and protein and ET(A)/ET(B) receptor protein expression were measured in normal and PPHN PAECs. ET-1-ROCK interactions were assessed by measuring ROCK activity after ET-1, ET-1 siRNA, and bosentan treatments, and tube formation with ET-1 and Y-27632 (ROCK inhibitor). RESULTS: ET-1 did not affect growth but decreased tube formation in normal and PPHN PAECs. ET-1 protein and gene expression were increased and ET(B) receptor protein decreased in PPHN PAECs. ET-1 siRNA, ET-1mAbs, and bosentan, but not BQ-123, increased tube formation. ROCK activity was increased in PPHN PAECs and decreased with ET-1 siRNA and bosentan treatments. Y-27632 prevented the decrease in tube formation with ET-1. CONCLUSION: ET-1 activation of ROCK impairs angiogenesis of fetal PAECs. Disruption of ET-1-ROCK interactions may increase vascular growth in PPHN.


Asunto(s)
Endotelina-1/farmacología , Activación Enzimática/efectos de los fármacos , Pulmón/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Síndrome de Circulación Fetal Persistente/metabolismo , Quinasas Asociadas a rho/metabolismo , Amidas , Análisis de Varianza , Animales , Western Blotting , Cartilla de ADN/genética , Conducto Arterial/cirugía , Células Endoteliales/efectos de los fármacos , Endotelina-1/metabolismo , Ensayo de Inmunoadsorción Enzimática , Feto/fisiopatología , Humanos , Técnicas In Vitro , Recién Nacido , Ligadura , Piridinas , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Ovinos
17.
Am J Respir Cell Mol Biol ; 47(5): 669-78, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22842492

RESUMEN

Increased pulmonary artery endothelial cell (PAEC) endothelium-dependent nitric oxide synthase (eNOS) activity mediates perinatal pulmonary vasodilation. Compromised eNOS activity is central to the pathogenesis of persistent pulmonary hypertension of the newborn (PPHN). Voltage-derived anion channel (VDAC)-1 was recently demonstrated to bind eNOS in the systemic circulation. We hypothesized that VDAC isoforms modulate eNOS activity in the pulmonary circulation, and that decreased VDAC expression contributes to PPHN. In PAECs derived from an ovine model of PPHN: (1) there is eNOS activity, but not expression; and (2) VDAC1 and -2 proteins are decreased. Immunocytochemistry, coimmunoprecipitation, and in situ proximity ligation assays in human PAECs (hPAECs) demonstrate binding between eNOS and both VDAC1 and -2, which increased upon stimulation with NO agonists. The ability of agonists to increase the eNOS/VDAC interaction was significantly blunted in hypertensive, compared with normotensive, ovine PAECs. Depletion of VDAC2, but not VDAC1, blocked the agonist-induced increase in eNOS activity in hPAECs. Overexpression of VDAC2 in hypertensive PAECs increased eNOS activity. Binding of VDAC2 enhances eNOS activity in the pulmonary circulation, and diminished VDAC2 constrains eNOS in PAECs derived from fetal lambs with chronic intrauterine pulmonary hypertension. We speculate that decreases in VDAC2 may contribute to the limited eNOS activity that characterizes pulmonary hypertension.


Asunto(s)
Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Arteria Pulmonar/patología , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Animales , Calcimicina/farmacología , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/patología , Expresión Génica , Histamina/fisiología , Humanos , Recién Nacido , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Síndrome de Circulación Fetal Persistente/enzimología , Síndrome de Circulación Fetal Persistente/metabolismo , Síndrome de Circulación Fetal Persistente/patología , Unión Proteica , Mapeo de Interacción de Proteínas , Ovinos , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/genética
18.
Am J Physiol Lung Cell Mol Physiol ; 302(6): L595-603, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22198909

RESUMEN

In the pulmonary vasculature, cGMP levels are regulated by soluble guanylate cyclase (sGC) and phosphodiesterase 5 (PDE5). We previously reported that lambs with persistent pulmonary hypertension of the newborn (PPHN) demonstrate increased reactive oxygen species (ROS) and altered sGC and PDE5 activity, with resultant decreased cGMP. The objective of this study was to evaluate the effects of hydrocortisone on pulmonary vascular function, ROS, and cGMP in the ovine ductal ligation model of PPHN. PPHN lambs were ventilated with 100% O(2) for 24 h. Six lambs received 5 mg/kg hydrocortisone every 8 h times three doses (PPHN-hiHC), five lambs received 3 mg/kg hydrocortisone followed by 1 mg·kg(-1)·dose(-1) times two doses (PPHN-loHC), and six lambs were ventilated with O(2) alone (PPHN). All groups were compared with healthy 1-day spontaneously breathing lambs (1DSB). O(2) ventilation of PPHN lambs decreased sGC activity, increased PDE5 activity, and increased ROS vs. 1DSB lambs. Both hydrocortisone doses significantly improved arterial-to-alveolar ratios relative to PPHN lambs, decreased PDE5 activity, and increased cGMP relative to PPHN lambs. High-dose hydrocortisone also increased sGC activity, decreased PDE5 expression, decreased ROS, and increased total vascular SOD activity vs. PPHN lambs. These data suggest that hydrocortisone treatment in clinically relevant doses improves oxygenation and decreases hyperoxia-induced changes in sGC and PDE5 activity, increasing cGMP levels. Hydrocortisone reduces ROS levels in part by increasing SOD activity in PPHN lambs ventilated with 100% O(2.) We speculate that hydrocortisone increases cGMP by direct effects on sGC and PDE5 expression and by attenuating abnormalities induced by oxidant stress.


Asunto(s)
GMP Cíclico/metabolismo , Hidrocortisona/farmacología , Oxígeno/metabolismo , Síndrome de Circulación Fetal Persistente/tratamiento farmacológico , Síndrome de Circulación Fetal Persistente/metabolismo , Arteria Pulmonar/efectos de los fármacos , Animales , Animales Recién Nacidos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Hiperoxia/tratamiento farmacológico , Hiperoxia/genética , Hiperoxia/metabolismo , Hiperoxia/fisiopatología , Recién Nacido , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/fisiopatología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Ovinos , Guanilil Ciclasa Soluble , Superóxido Dismutasa/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L13-26, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21926266

RESUMEN

Actin polymerization (APM), regulated by Rho GTPases, promotes myocyte force generation. Hypoxia is known to impede postnatal disassembly of the actin cytoskeleton in pulmonary arterial (PA) myocytes. We compared basal and agonist-induced APM in myocytes from PA and descending aorta (Ao), under hypoxic and normoxic conditions. We also examined effects of thromboxane challenge on force generation and cytoskeletal assembly in resistance PA and renal arteries from neonatal swine with persistent pulmonary hypertension (PPHN) induced by 72-h normobaric hypoxia, compared with age-matched controls. Synthetic and contractile phenotype myocytes from neonatal porcine PA or Ao were grown in hypoxia (10% O(2)) or normoxia (21% O(2)) for 7 days, then challenged with 10(-6) M thromboxane mimetic U46619. F/G actin ratio was quantified by laser-scanning cytometry and by cytoskeletal fractionation. Thromboxane receptor (TP) G protein coupling was measured by immunoprecipitation and probing for Gαq, G12, or G13, RhoA activation by Rhotekin-RBD affinity precipitation, and LIM kinase (LIMK) and cofilin phosphorylation by Western blot. Isometric force to serial concentrations of U46619 was measured in muscular pulmonary and renal arteries from PPHN and control swine; APM was quantified in fixed contracted vessels. Contractile PA myocytes exhibit marked Rho-dependent APM in hypoxia, with increased active RhoA and LIMK phosphorylation. Their additional APM response to U46619 challenge is independent of RhoA, reflecting decreased TP association with G12/13 in favor of Gαq. In contrast, hypoxic contractile Ao myocytes polymerize actin modestly and depolymerize to U46619. Both basal APM and the APM response to U46619 are increased in PPHN PA. APM corresponds with increased force generation to U46619 challenge in PPHN PA but not renal arteries.


Asunto(s)
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Citoesqueleto de Actina/metabolismo , Hipoxia de la Célula/fisiología , Células Musculares , Síndrome de Circulación Fetal Persistente/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Animales Recién Nacidos , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Humanos , Hipertensión Renovascular , Hipoxia/metabolismo , Hipoxia/fisiopatología , Recién Nacido , Citometría de Barrido por Láser , Quinasas Lim/metabolismo , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Fosforilación , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , Receptores de Tromboxanos/metabolismo , Arteria Renal/citología , Arteria Renal/efectos de los fármacos , Arteria Renal/fisiología , Porcinos , Tromboxanos/metabolismo , Tromboxanos/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
20.
Nat Commun ; 13(1): 2080, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440116

RESUMEN

Pulmonary endothelial progenitor cells (EPCs) are critical for neonatal lung angiogenesis and represent a subset of general capillary cells (gCAPs). Molecular mechanisms through which EPCs stimulate lung angiogenesis are unknown. Herein, we used single-cell RNA sequencing to identify the BMP9/ACVRL1/SMAD1 pathway signature in pulmonary EPCs. BMP9 receptor, ACVRL1, and its downstream target genes were inhibited in EPCs from Foxf1WT/S52F mutant mice, a model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Expression of ACVRL1 and its targets were reduced in lungs of ACDMPV subjects. Inhibition of FOXF1 transcription factor reduced BMP9/ACVRL1 signaling and decreased angiogenesis in vitro. FOXF1 synergized with ETS transcription factor FLI1 to activate ACVRL1 promoter. Nanoparticle-mediated silencing of ACVRL1 in newborn mice decreased neonatal lung angiogenesis and alveolarization. Treatment with BMP9 restored lung angiogenesis and alveolarization in ACVRL1-deficient and Foxf1WT/S52F mice. Altogether, EPCs promote neonatal lung angiogenesis and alveolarization through FOXF1-mediated activation of BMP9/ACVRL1 signaling.


Asunto(s)
Células Progenitoras Endoteliales , Síndrome de Circulación Fetal Persistente , Neumonía , Animales , Ratones , Receptores de Activinas Tipo II/metabolismo , Células Progenitoras Endoteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Pulmón/metabolismo , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/metabolismo , Neumonía/metabolismo , Alveolos Pulmonares/anomalías
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA