Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell Neurosci ; 129: 103933, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663691

RESUMEN

Astrocytes are in constant communication with neurons during the establishment and maturation of functional networks in the developing brain. Astrocytes release extracellular vesicles (EVs) containing microRNA (miRNA) cargo that regulates transcript stability in recipient cells. Astrocyte released factors are thought to be involved in neurodevelopmental disorders. Healthy astrocytes partially rescue Rett Syndrome (RTT) neuron function. EVs isolated from stem cell progeny also correct aspects of RTT. EVs cross the blood-brain barrier (BBB) and their cargo is found in peripheral blood which may allow non-invasive detection of EV cargo as biomarkers produced by healthy astrocytes. Here we characterize miRNA cargo and sequence motifs in healthy human astrocyte derived EVs (ADEVs). First, human induced Pluripotent Stem Cells (iPSC) were differentiated into Neural Progenitor Cells (NPCs) and subsequently into astrocytes using a rapid differentiation protocol. iPSC derived astrocytes expressed specific markers, displayed intracellular calcium transients and secreted ADEVs. miRNAs were identified by RNA-Seq on astrocytes and ADEVs and target gene pathway analysis detected brain and immune related terms. The miRNA profile was consistent with astrocyte identity, and included approximately 80 miRNAs found in astrocytes that were relatively depleted in ADEVs suggestive of passive loading. About 120 miRNAs were relatively enriched in ADEVs and motif analysis discovered binding sites for RNA binding proteins FUS, SRSF7 and CELF5. miR-483-5p was the most significantly enriched in ADEVs. This miRNA regulates MECP2 expression in neurons and has been found differentially expressed in blood samples from RTT patients. Our results identify potential miRNA biomarkers selectively sorted into ADEVs and implicate RNA binding protein sequence dependent mechanisms for miRNA cargo loading.


Asunto(s)
Astrocitos , Vesículas Extracelulares , Células Madre Pluripotentes Inducidas , MicroARNs , Neuronas , Humanos , Vesículas Extracelulares/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Astrocitos/metabolismo , Neuronas/metabolismo , Diferenciación Celular , Células Cultivadas , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología
2.
Am J Respir Crit Care Med ; 206(4): 476-487, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35671495

RESUMEN

Rationale: Pulmonary hypoplasia secondary to congenital diaphragmatic hernia is characterized by reduced branching morphogenesis, which is responsible for poor clinical outcomes. Administration of amniotic fluid stem cell extracellular vesicles (AFSC-EVs) rescues branching morphogenesis in rodent fetal models of pulmonary hypoplasia. Herein, we hypothesized that AFSC-EVs exert their regenerative potential by affecting autophagy, a process required for normal lung development. Objectives: To evaluate autophagy in hypoplastic lungs throughout gestation and establish whether AFSC-EV administration improves branching morphogenesis through autophagy-mediated mechanisms. Methods: EVs were isolated from c-kit+ AFSC-conditioned medium by ultracentrifugation and characterized for size, morphology, and EV markers. Branching morphogenesis was inhibited in rat fetuses by nitrofen administration to dams and in human fetal lung explants by blocking RAC1 activity with NSC23766. The expression of autophagy activators (BECN1 and ATG5) and adaptor (SQSTM1/p62) was analyzed in vitro (rat and human fetal lung explants) and in vivo (rat fetal lungs). Mechanistic studies on rat fetal primary lung epithelial cells were conducted using inhibitors for microRNA-17 and -20a contained in the AFSC-EV cargo and known to regulate autophagy. Measurements and Main Results: Rat and human models of fetal pulmonary hypoplasia showed reduced autophagy mainly at pseudoglandular and canalicular stages. AFSC-EV administration restored autophagy in both pulmonary hypoplasia models by transferring miR-17∼92 cluster members contained in the EV cargo. Conclusions: AFSC-EV treatment rescues branching morphogenesis partly by restoring autophagy through microRNA cargo transfer. This study enhances our understanding of pulmonary hypoplasia pathogenesis and creates new opportunities for fetal therapeutic intervention in congenital diaphragmatic hernia babies.


Asunto(s)
Vesículas Extracelulares , Hernias Diafragmáticas Congénitas , MicroARNs , Anomalías del Sistema Respiratorio , Líquido Amniótico/metabolismo , Animales , Autofagia , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , MicroARNs/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo
3.
Pediatr Surg Int ; 39(1): 194, 2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37160490

RESUMEN

Congenital diaphragmatic hernia (CDH) is a birth defect characterized by the incomplete closure of the diaphragm and herniation of abdominal organs into the chest during gestation. This invariably leads to an impairment in fetal lung development (pulmonary hypoplasia) that involves the pulmonary vessels (vascular remodeling) leading to postnatal pulmonary hypertension. Moreover, approximately 60% of CDH survivors have long-term comorbidities, including critical cardiac anomalies, neurodevelopmental impairment, gastroesophageal reflux, and musculoskeletal malformations. While the pathophysiology of the diaphragmatic defect and pulmonary hypoplasia have been studied in detail over the decades, less is known about the other organs affected in CDH. In this review, we searched the literature for reports on other organs beyond the lung and diaphragm in human and experimental models of CDH. We found studies reporting gross morphometric changes and alterations to biological pathways in the heart, brain, liver, kidney, gastrointestinal tract, and musculoskeletal system. Given the paucity of literature and the importance that these comorbidities play in the life of patients with CDH, further studies are needed to comprehensively uncover the pathophysiology of the changes observed in these other organs.


Asunto(s)
Hernias Diafragmáticas Congénitas , Hipertensión Pulmonar , Humanos , Diafragma , Hígado , Pulmón
4.
Pediatr Surg Int ; 39(1): 291, 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37955723

RESUMEN

PURPOSE: Congenital diaphragmatic hernia (CDH) survivors may experience neurodevelopmental impairment, whose etiology remains elusive. Preclinical evidence indicates that amniotic fluid stem cell extracellular vesicle (AFSC-EV) administration promotes lung development but their effects on other organs are unknown. Herein, we investigated the brain of rat fetuses with CDH for signs of inflammation and response to AFSC-EVs. METHODS: CDH was induced by maternal nitrofen administration at E9.5. At E18.5, fetuses were injected intra-amniotically with saline or AFSC-EVs (isolated by ultracentrifugation, characterized as per MISEV guidelines). Fetuses from vehicle-gavaged dams served as controls. Groups were compared for: lung hypoplasia, TNFa and IL-1B brain expression, and activated microglia (Iba1) density in the subgranular zone (SGZ). RESULTS: CDH lungs had fewer airspaces compared to controls, whereas AFSC-EV-treated lungs had rescued branching morphogenesis. Fluorescently labeled AFSC-EVs injected intra-amniotically into CDH fetuses had fluorescent signal in the brain. Compared to controls, the brain of CDH fetuses had higher TNFa and IL-1B levels, and increased activated microglia density. Conversely, the brain of AFSC-EV treated fetuses had inflammatory marker expression levels and microglia density similar to controls. CONCLUSION: This study shows that the brain of rat fetuses with CDH has signs of inflammation that are abated by the intra-amniotic administration of AFSC-EVs.


Asunto(s)
Vesículas Extracelulares , Hernias Diafragmáticas Congénitas , Femenino , Embarazo , Animales , Ratas , Encéfalo , Líquido Amniótico , Inflamación , Antiinflamatorios
5.
Pediatr Surg Int ; 39(1): 296, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37981587

RESUMEN

PURPOSE: Lineage tracing is key to study the fate of individual cells and their progeny especially in developmental biology. To conduct these studies, we aimed to establish a reproducible model of CDH in the most commonly used genetic background strain that is C57BL/6J mice. METHODS: CDH was induced in C57BL/6J dams by maternal administration of nitrofen + bisdiamine at E8.5. Fetuses from olive oil-gavaged mothers served as controls. Lungs from CDH and control fetuses were compared for (1) growth via radial airspace count (RAC), mean linear intercept (MLI) and gene expression for Fgf10, Nrp1, and Ctnnb1; (2) maturation (Pdpn, Spc, Ager, Abca3, Eln, Acta2, Pdgfra) via gene and protein expression; (3) vascularization via gene and protein expression (CD31, Vegfa, Vegfr1/2, Epas1, Enos). STATISTICS: unpaired t-test or Mann-Whitney test. RESULTS: Nitrofen + bisdiamine administration resulted in 36% left-sided CDH (31% mortality). CDH fetuses had hypoplastic lungs and impaired growth (lower RAC, higher MLI, lower Fgf10, Nrp1, Ctnnb1), maturation (decreased Pdpn, Ager, Eln gene expression), and vascularization (decreased Cd31, Vegfr1/2; Epas1 and Enos). Lower protein expression was confirmed for PDPN, ELN and CD31. CONCLUSION: Modeling CDH in C57BL/6J mouse fetuses is effective in reproducing the classical CDH hallmarks. This model will be critical for lineage tracing experiments.


Asunto(s)
Hernias Diafragmáticas Congénitas , Ratones , Animales , Humanos , Femenino , Embarazo , Ratones Endogámicos C57BL , Hernias Diafragmáticas Congénitas/genética , Feto , Atención Prenatal , Factores de Transcripción
6.
Blood ; 136(23): 2679-2690, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-32663239

RESUMEN

Quebec platelet disorder (QPD) is an autosomal dominant bleeding disorder with a unique, platelet-dependent, gain-of-function defect in fibrinolysis, without systemic fibrinolysis. The hallmark feature of QPD is a >100-fold overexpression of PLAU, specifically in megakaryocytes. This overexpression leads to a >100-fold increase in platelet stores of urokinase plasminogen activator (PLAU/uPA); subsequent plasmin-mediated degradation of diverse α-granule proteins; and platelet-dependent, accelerated fibrinolysis. The causative mutation is a 78-kb tandem duplication of PLAU. How this duplication causes megakaryocyte-specific PLAU overexpression is unknown. To investigate the mechanism that causes QPD, we used epigenomic profiling, comparative genomics, and chromatin conformation capture approaches to study PLAU regulation in cultured megakaryocytes from participants with QPD and unaffected controls. QPD duplication led to ectopic interactions between PLAU and a conserved megakaryocyte enhancer found within the same topologically associating domain (TAD). Our results support a unique disease mechanism whereby the reorganization of sub-TAD genome architecture results in a dramatic, cell-type-specific blood disorder phenotype.


Asunto(s)
Elementos de Facilitación Genéticos , Deficiencia del Factor V , Duplicación de Gen , Regulación de la Expresión Génica , Megacariocitos/metabolismo , Proteínas de la Membrana , Animales , Deficiencia del Factor V/genética , Deficiencia del Factor V/metabolismo , Deficiencia del Factor V/patología , Femenino , Humanos , Megacariocitos/patología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Pez Cebra
7.
Prenat Diagn ; 42(3): 364-372, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35191057

RESUMEN

The poor outcomes of babies with congenital diaphragmatic hernia (CDH) are directly related to pulmonary hypoplasia, a condition characterized by impaired lung development. Although the pathogenesis of pulmonary hypoplasia is not fully elucidated, there is now evidence that CDH patients have missing or dysregulated microRNAs (miRNAs) that regulate lung development. A prenatal therapy that supplements these missing/dysregulated miRNAs could be a strategy to rescue normal lung development. Extracellular vesicles (EVs), also known as exosomes when of small dimensions, are lipid-bound nanoparticles that can transfer their heterogeneous cargo (proteins, lipids, small RNAs) to target cells to induce biological responses. Herein, we review all studies that show evidence for stem cell-derived EVs as a regenerative therapy to rescue normal development in CDH fetal lungs. Particularly, we report studies showing that administration of EVs derived from amniotic fluid stem cells (AFSC-EVs) to models of pulmonary hypoplasia promotes fetal lung growth and maturation via transfer of miRNAs that are known to regulate lung developmental processes. We also describe that stem cell-derived EVs exert effects on vascular remodeling, thus possibly preventing postnatal pulmonary hypertension. Finally, we discuss future perspectives and challenges to translate this promising stem cell EV-based therapy to clinical practice.


Asunto(s)
Vesículas Extracelulares , Hernias Diafragmáticas Congénitas , MicroARNs , Anomalías del Sistema Respiratorio , Femenino , Hernias Diafragmáticas Congénitas/complicaciones , Hernias Diafragmáticas Congénitas/patología , Hernias Diafragmáticas Congénitas/terapia , Humanos , Pulmón , Embarazo , Regeneración , Anomalías del Sistema Respiratorio/complicaciones , Células Madre
8.
Pediatr Surg Int ; 37(3): 301-309, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33566163

RESUMEN

PURPOSE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease. Amniotic fluid stem cells (AFSC) improve NEC injury but human translation remains difficult. We aimed to evaluate the use of extracellular vesicles (EV) derived from human AFSC. METHODS: Human AFSC (hAFSC) were cultured according to the protocol (Celprogen Inc., California, U.S.A.). Conditioned medium was obtained, ultra-centrifuged, and EV were suspended in phosphate-buffered saline (PBS). C57BL/6 pups were grouped into: (1) breast-fed (Control, n = 11); (2) NEC + placebo (NEC + PBS; n = 10); and (3) NEC + treatment (NEC + EV; n = 11). NEC was induced post-natal days P5-9 by (A) gavage feeding hyperosmolar formula; (B) hypoxia for 10 min; and (C) lipopolysaccharide. Intra-peritoneal injections of PBS or hAFSC-EV were given on P6-7. All animals were sacrificed on P9 and terminal ileum harvested. RESULTS: hAFSC-EV administration reduced intestinal injury (p = 0.0048), NEC incidence (score ≥ 2), and intestinal inflammation (IL-6 p < 0.0001; TNF-α p < 0.0001). Intestinal stem cell expression (Lgr5 +) and cellular proliferation (Ki67) were enhanced above control levels following hAFSC-EV administration (Lgr5 p = 0.0003; Ki67 p < 0.0001). CONCLUSION: hAFSC-EV administration reduced intestinal NEC injury and inflammation while increasing stem cell expression and cellular proliferation. hAFSC-EV administration may induce similar beneficial effects to exogenous stem cells.


Asunto(s)
Líquido Amniótico/citología , Enterocolitis Necrotizante/metabolismo , Vesículas Extracelulares/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/terapia , Femenino , Humanos , Íleon/metabolismo , Recién Nacido , Enfermedades del Recién Nacido , Inflamación/metabolismo , Intestinos , Ratones , Ratones Endogámicos C57BL , Células Madre/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Development ; 144(13): 2428-2444, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28536097

RESUMEN

The transcriptional pathways activated downstream of vascular endothelial growth factor (VEGF) signaling during angiogenesis remain incompletely characterized. By assessing the signals responsible for induction of the Notch ligand delta-like 4 (DLL4) in endothelial cells, we find that activation of the MAPK/ERK pathway mirrors the rapid and dynamic induction of DLL4 transcription and that this pathway is required for DLL4 expression. Furthermore, VEGF/ERK signaling induces phosphorylation and activation of the ETS transcription factor ERG, a prerequisite for DLL4 induction. Transcription of DLL4 coincides with dynamic ERG-dependent recruitment of the transcriptional co-activator p300. Genome-wide gene expression profiling identified a network of VEGF-responsive and ERG-dependent genes, and ERG chromatin immunoprecipitation (ChIP)-seq revealed the presence of conserved ERG-bound putative enhancer elements near these target genes. Functional experiments performed in vitro and in vivo confirm that this network of genes requires ERK, ERG and p300 activity. Finally, genome-editing and transgenic approaches demonstrate that a highly conserved ERG-bound enhancer located upstream of HLX (which encodes a transcription factor implicated in sprouting angiogenesis) is required for its VEGF-mediated induction. Collectively, these findings elucidate a novel transcriptional pathway contributing to VEGF-dependent angiogenesis.


Asunto(s)
Proteína p300 Asociada a E1A/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Bovinos , Elementos de Facilitación Genéticos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Intrones/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Neovascularización Fisiológica/genética , Regulador Transcripcional ERG/metabolismo , Pez Cebra/embriología
10.
J Neuroinflammation ; 16(1): 97, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31077225

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal disease primarily affecting preterm neonates. Neonates with NEC suffer from a degree of neurodevelopmental delay that is not explained by prematurity alone. There is a need to understand the pathogenesis of neurodevelopmental delay in NEC. In this study, we assessed the macroscopic and microscopic changes that occur to brain cell populations in specific brain regions in a neonatal mouse model of NEC. Moreover, we investigated the role of intestinal inflammation as part of the mechanism responsible for the changes observed in the brain of pups with NEC. METHODS: Brains of mice were assessed for gross morphology and cerebral cortex thickness (using histology). Markers for mature neurons, oligodendrocytes, neural progenitor cells, microglia, and astrocytes were used to quantify their cell populations in different regions of the brain. Levels of cell apoptosis in the brain were measured by Western blotting and immunohistochemistry. Endoplasmic reticulum (ER) stress markers and levels of pro-inflammatory cytokines (in the ileum and brain) were measured by RT-qPCR and Western blotting. A Pearson test was used to correlate the levels of cytokines (ELISA) in the brain and ileum and to correlate activated microglia and astrocyte populations to the severity of NEC. RESULTS: NEC pups had smaller brain weights, higher brain-to-body weight ratios, and thinner cortices compared to control pups. NEC pups had increased levels of apoptosis and ER stress. In addition, NEC was associated with a reduction in the number of neurons, oligodendrocytes, and neural progenitors in specific regions of the brain. Levels of pro-inflammatory cytokines and the density of activated microglia and astrocytes were increased in the brain and positively correlated with the increase in the levels pro-inflammatory cytokines in the gut and the severity of NEC damage respectively. CONCLUSIONS: NEC is associated with severe changes in brain morphology, a pro-inflammatory response in the brain that alters cell homeostasis and density of brain cell populations in specific cerebral regions. We show that the severity of neuroinflammation is associated with the severity of NEC. Our findings suggest that early intervention during NEC may reduce the chance of acute neuroinflammation and cerebral damage.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Mediadores de Inflamación/metabolismo , Animales , Animales Recién Nacidos , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL
11.
Pediatr Res ; 85(6): 754-768, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30780153

RESUMEN

Congenital diaphragmatic hernia (CDH) is a severe birth defect that is characterized by pulmonary hypoplasia and pulmonary hypertension (PHTN). PHTN secondary to CDH is a result of vascular remodeling, a structural alteration in the pulmonary vessel wall that occurs in the fetus. Factors involved in vascular remodeling have been reported in several studies, but their interactions remain unclear. To help understand PHTN pathophysiology and design novel preventative and treatment strategies, we have conducted a systematic review of the literature and comprehensively analyzed all factors and pathways involved in the pathogenesis of pulmonary vascular remodeling secondary to CDH in the nitrofen model. Moreover, we have linked the dysregulated factors with pathways involved in human CDH. Of the 358 full-text articles screened, 75 studies reported factors that play a critical role in vascular remodeling secondary to CDH. Overall, the impairment of epithelial homeostasis present in pulmonary hypoplasia results in altered signaling to endothelial cells, leading to endothelial dysfunction. This causes an impairment of the crosstalk between endothelial cells and pulmonary artery smooth muscle cells, resulting in increased smooth muscle cell proliferation, resistance to apoptosis, and vasoconstriction, which clinically translate into PHTN.


Asunto(s)
Hernias Diafragmáticas Congénitas/complicaciones , Hipertensión Pulmonar/etiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Células Endoteliales/fisiología , Femenino , Hernias Diafragmáticas Congénitas/patología , Hernias Diafragmáticas Congénitas/fisiopatología , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Recién Nacido , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Éteres Fenílicos/toxicidad , Embarazo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Factores de Riesgo , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/fisiología
12.
Pediatr Surg Int ; 35(1): 63-69, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30386898

RESUMEN

PURPOSE: Pulmonary hypoplasia secondary to congenital diaphragmatic hernia (CDH) is characterized by impaired epithelial homeostasis. Recently, amniotic fluid stem cells (AFSCs) have been shown to promote growth in hypoplastic lungs of rat fetuses with CDH. Herein, we investigated whether CDH hypoplastic lungs mount an endoplasmic reticulum (ER) stress response and whether AFSCs could re-establish pulmonary epithelial homeostasis. METHODS: Primary epithelial cells were isolated from fetal rat lungs at E14.5 from control and nitrofen-exposed dams at E9.5. Nitrofen-exposed epithelial cells were grown in medium alone or co-cultured with AFSCs. Epithelial cell cultures were compared for apoptosis (TUNEL), cytotoxicity (LIVE/DEAD assay), proliferation (5'EdU), and ER stress (CHOP, Bcl-2) using one-way ANOVA (Dunn's post-test). RESULTS: Compared to control, nitrofen-exposed epithelial cells had increased cytotoxicity and apoptosis, reduced proliferation, and activated ER stress. AFSCs restored apoptosis, proliferation, and ER stress back to control levels, and significantly reduced cytotoxicity. CONCLUSIONS: This study shows for the first time that ER stress-induced apoptosis is activated in the pulmonary epithelium of hypoplastic lungs from fetuses with CDH. AFSC treatment restores epithelial cellular homeostasis by attenuating the ER stress response and apoptosis, by increasing proliferation and migration ability, and by reducing cytotoxicity.


Asunto(s)
Anomalías Múltiples/metabolismo , Líquido Amniótico/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Estrés del Retículo Endoplásmico , Hernias Diafragmáticas Congénitas/metabolismo , Enfermedades Pulmonares/metabolismo , Pulmón/anomalías , Preñez , Células Madre/citología , Anomalías Múltiples/embriología , Anomalías Múltiples/terapia , Animales , Apoptosis , Modelos Animales de Enfermedad , Femenino , Hernias Diafragmáticas Congénitas/embriología , Hernias Diafragmáticas Congénitas/terapia , Pulmón/embriología , Pulmón/metabolismo , Enfermedades Pulmonares/embriología , Enfermedades Pulmonares/terapia , Éteres Fenílicos/toxicidad , Embarazo , Ratas , Ratas Sprague-Dawley
13.
Genet Epidemiol ; 41(5): 455-466, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28421636

RESUMEN

Tissue factor pathway inhibitor (TFPI) regulates the formation of intravascular blood clots, which manifest clinically as ischemic heart disease, ischemic stroke, and venous thromboembolism (VTE). TFPI plasma levels are heritable, but the genetics underlying TFPI plasma level variability are poorly understood. Herein we report the first genome-wide association scan (GWAS) of TFPI plasma levels, conducted in 251 individuals from five extended French-Canadian Families ascertained on VTE. To improve discovery, we also applied a hypothesis-driven (HD) GWAS approach that prioritized single nucleotide polymorphisms (SNPs) in (1) hemostasis pathway genes, and (2) vascular endothelial cell (EC) regulatory regions, which are among the highest expressers of TFPI. Our GWAS identified 131 SNPs with suggestive evidence of association (P-value < 5 × 10-8 ), but no SNPs reached the genome-wide threshold for statistical significance. Hemostasis pathway genes were not enriched for TFPI plasma level associated SNPs (global hypothesis test P-value = 0.147), but EC regulatory regions contained more TFPI plasma level associated SNPs than expected by chance (global hypothesis test P-value = 0.046). We therefore stratified our genome-wide SNPs, prioritizing those in EC regulatory regions via stratified false discovery rate (sFDR) control, and reranked the SNPs by q-value. The minimum q-value was 0.27, and the top-ranked SNPs did not show association evidence in the MARTHA replication sample of 1,033 unrelated VTE cases. Although this study did not result in new loci for TFPI, our work lays out a strategy to utilize epigenomic data in prioritization schemes for future GWAS studies.


Asunto(s)
Biomarcadores/sangre , Lipoproteínas/sangre , Lipoproteínas/genética , Polimorfismo de Nucleótido Simple/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Tromboembolia Venosa/sangre , Tromboembolia Venosa/genética , Adulto , Canadá , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Epigenómica , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Tromboembolia Venosa/diagnóstico
14.
Sci Rep ; 14(1): 13680, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871804

RESUMEN

Congenital diaphragmatic hernia (CDH) is a birth defect characterized by incomplete closure of the diaphragm, herniation of abdominal organs into the chest, and compression of the lungs and the heart. Besides complications related to pulmonary hypoplasia, 1 in 4 survivors develop neurodevelopmental impairment, whose etiology remains unclear. Using a fetal rat model of CDH, we demonstrated that the compression exerted by herniated organs on the mediastinal structures results in decreased brain perfusion on ultrafast ultrasound, cerebral hypoxia with compensatory angiogenesis, mature neuron and oligodendrocyte loss, and activated microglia. In CDH fetuses, apoptosis was prominent in the subventricular and subgranular zones, areas that are key for neurogenesis. We validated these findings in the autopsy samples of four human fetuses with CDH compared to age- and sex-matched controls. This study reveals the molecular mechanisms and cellular changes that occur in the brain of fetuses with CDH and creates opportunities for therapeutic targets.


Asunto(s)
Encéfalo , Hernias Diafragmáticas Congénitas , Neuronas , Oligodendroglía , Animales , Hernias Diafragmáticas Congénitas/patología , Hernias Diafragmáticas Congénitas/diagnóstico por imagen , Neuronas/patología , Neuronas/metabolismo , Oligodendroglía/patología , Oligodendroglía/metabolismo , Ratas , Humanos , Encéfalo/patología , Encéfalo/diagnóstico por imagen , Encéfalo/embriología , Femenino , Células Madre/patología , Feto/patología , Modelos Animales de Enfermedad , Embarazo , Masculino
15.
J Pediatr Surg ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38519389

RESUMEN

BACKGROUND: The severity of pulmonary hypoplasia is a main determinant of outcome for babies with congenital diaphragmatic hernia (CDH). Antenatal administration of extracellular vesicles derived from amniotic fluid stem cells (AFSC-EVs) has been shown to rescue morphological features of lung development in the rat nitrofen model of CDH. Herein, we evaluated whether AFSC-EV administration to fetal rats with CDH is associated with neonatal improvement in lung function. METHODS: AFSC-EVs were isolated by ultracentrifugation and characterized by size, morphology, and canonical marker expression. At embryonic (E) day 9.5, dams were gavaged with olive oil (control) or nitrofen to induce CDH. At E18.5, fetuses received an intra-amniotic injection of either saline or AFSC-EVs. At E21.5, rats were delivered and subjected to a tracheostomy for mechanical ventilation (flexiVent system). Groups were compared for lung compliance, resistance, Newtonian resistance, tissue damping and elastance. Lungs were evaluated for branching morphogenesis and collagen quantification. RESULTS: Compared to healthy control, saline-treated pups with CDH had fewer airspaces, more collagen deposition, and functionally exhibited reduced compliance and increased airway resistance, elastance, and tissue damping. Conversely, AFSC-EV administration resulted in improvement of lung mechanics (compliance, resistance, tissue damping, elastance) as well as lung branching morphogenesis and collagen deposition. CONCLUSIONS: Our studies show that the rat nitrofen model reproduces lung function impairment similar to that of human babies with CDH. Antenatal administration of AFSC-EVs improves lung morphology and function in neonatal rats with CDH. LEVEL OF EVIDENCE: N/A (animal and laboratory study).

16.
Eur J Pediatr Surg ; 33(1): 11-16, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35858641

RESUMEN

INTRODUCTION: The etiology of congenital diaphragmatic hernia (CDH) remains unknown and only 10 to 30% of patients have a genetic cause. Seasonal variation is known to contribute to the development of some congenital anomalies. Our aim was to investigate whether CDH births have seasonal variation. MATERIALS AND METHODS: A literature review was conducted for CDH and seasonality. Moreover, data from the CDH International Patient Registry Database were collected for infants with due dates between 2008 and 2014. Due dates were used to determine seasonal distribution of births. Birth rates per month in the United States and Canada were extracted from publicly available databases. Data were analyzed using analysis of variance and contingency tables. RESULTS: First, the literature review revealed 11 articles, of which 3 were eligible for inclusion. These studies reported conflicting results on seasonality of CDH. Second, we extracted due dates from the CDH International Patient Registry Database (1,259 patients) and found that there were fewer due dates in winter months (12.1 ± 4 patients/month) than in summer (16.7 ± 6 patients/month; p = 0.011) and fall months (16.3 ± 5 patients/month; p = 0.022). Although this trend was similar to that of all births in the United States and Canada, a lower incidence was observed in winter for CDH infants (20.2%) than for the general population (24.1%, p = 0.0012). CDH survival rate did not vary by season. CONCLUSION: This study provides evidence for a seasonal variation of CDH births. No causative link was established between CDH development and seasonality. Population-based studies with a focus on exposome data are needed to explain seasonal variation in CDH.


Asunto(s)
Anomalías Múltiples , Hernias Diafragmáticas Congénitas , Lactante , Humanos , Estados Unidos/epidemiología , Hernias Diafragmáticas Congénitas/epidemiología , Hernias Diafragmáticas Congénitas/etiología , Estaciones del Año , Anomalías Múltiples/epidemiología , Incidencia , Canadá/epidemiología
17.
Semin Pediatr Surg ; 31(6): 151228, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36442455

RESUMEN

Congenital lung malformations comprise a diverse group of anomalies including congenital pulmonary airway malformation (CPAM, previously known as congenital cystic adenomatoid malformation or CCAM), bronchopulmonary sequestration (BPS), congenital lobar emphysema (CLE), bronchogenic cysts, and hybrid lesions. Little is known about the signaling pathways that underlie the pathophysiology of these lesions and the processes that may promote their malignant transformation. In the last decade, the use of transgenic/knockout animal models and the implementation of next generation sequencing on surgical lung specimens have increased our knowledge on the pathophysiology of these lesions. Herein, we provide an overview of normal lung development in humans and rodents, and we discuss the current state of knowledge on the pathophysiology and molecular pathways that are altered in each congenital lung malformation.


Asunto(s)
Secuestro Broncopulmonar , Malformación Adenomatoide Quística Congénita del Pulmón , Enfermedades Pulmonares , Anomalías del Sistema Respiratorio , Animales , Humanos , Malformación Adenomatoide Quística Congénita del Pulmón/diagnóstico , Pulmón/anomalías , Anomalías del Sistema Respiratorio/diagnóstico , Anomalías del Sistema Respiratorio/genética , Transducción de Señal
18.
Stem Cells Transl Med ; 11(10): 1089-1102, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36103370

RESUMEN

Pulmonary hypoplasia secondary to congenital diaphragmatic hernia (CDH) is characterized by impaired branching morphogenesis and differentiation. We have previously demonstrated that administration of extracellular vesicles derived from rat amniotic fluid stem cells (AFSC-EVs) rescues development of hypoplastic lungs at the pseudoglandular and alveolar stages in rodent models of CDH. Herein, we tested whether AFSC-EVs exert their regenerative effects at the canalicular and saccular stages, as these are translationally relevant for clinical intervention. To induce fetal pulmonary hypoplasia, we gavaged rat dams with nitrofen at embryonic day 9.5 and demonstrated that nitrofen-exposed lungs had impaired branching morphogenesis, dysregulated signaling pathways relevant to lung development (FGF10/FGFR2, ROBO/SLIT, Ephrin, Neuropilin 1, ß-catenin) and impaired epithelial and mesenchymal cell marker expression at both stages. AFSC-EVs administered to nitrofen-exposed lung explants rescued airspace density and increased the expression levels of key factors responsible for branching morphogenesis. Moreover, AFSC-EVs rescued the expression of alveolar type 1 and 2 cell markers at both canalicular and saccular stages and restored markers of club, ciliated epithelial, and pulmonary neuroendocrine cells at the saccular stage. AFSC-EV-treated lungs also had restored markers of lipofibroblasts and PDGFRA+ cells to control levels at both stages. EV tracking showed uptake of AFSC-EV RNA cargo throughout the fetal lung and an mRNA-miRNA network analysis identified that several miRNAs responsible for regulating lung development processes were contained in the AFSC-EV cargo. These findings suggest that AFSC-EV-based therapies hold potential for restoring fetal lung growth and maturation in babies with pulmonary hypoplasia secondary to CDH.


Asunto(s)
Vesículas Extracelulares , Hernias Diafragmáticas Congénitas , MicroARNs , Ratas , Animales , Hernias Diafragmáticas Congénitas/metabolismo , beta Catenina/metabolismo , Líquido Amniótico/metabolismo , Neuropilina-1/metabolismo , Ratas Sprague-Dawley , Pulmón/metabolismo , Células Madre/metabolismo , Diferenciación Celular , ARN Mensajero/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Efrinas/metabolismo , Modelos Animales de Enfermedad
19.
Nat Rev Dis Primers ; 8(1): 37, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650272

RESUMEN

Congenital diaphragmatic hernia (CDH) is a rare birth defect characterized by incomplete closure of the diaphragm and herniation of fetal abdominal organs into the chest that results in pulmonary hypoplasia, postnatal pulmonary hypertension owing to vascular remodelling and cardiac dysfunction. The high mortality and morbidity rates associated with CDH are directly related to the severity of cardiopulmonary pathophysiology. Although the aetiology remains unknown, CDH has a polygenic origin in approximately one-third of cases. CDH is typically diagnosed with antenatal ultrasonography, which also aids in risk stratification, alongside fetal MRI and echocardiography. At specialized centres, prenatal management includes fetal endoscopic tracheal occlusion, which is a surgical intervention aimed at promoting lung growth in utero. Postnatal management focuses on cardiopulmonary stabilization and, in severe cases, can involve extracorporeal life support. Clinical practice guidelines continue to evolve owing to the rapidly changing landscape of therapeutic options, which include pulmonary hypertension management, ventilation strategies and surgical approaches. Survivors often have long-term, multisystem morbidities, including pulmonary dysfunction, gastroesophageal reflux, musculoskeletal deformities and neurodevelopmental impairment. Emerging research focuses on small RNA species as biomarkers of severity and regenerative medicine approaches to improve fetal lung development.


Asunto(s)
Hernias Diafragmáticas Congénitas , Hipertensión Pulmonar , Endoscopía , Femenino , Hernias Diafragmáticas Congénitas/diagnóstico , Hernias Diafragmáticas Congénitas/cirugía , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/terapia , Pulmón/anomalías , Pulmón/diagnóstico por imagen , Embarazo , Ultrasonografía Prenatal/efectos adversos
20.
Eur J Pediatr Surg ; 31(4): 326-334, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34161984

RESUMEN

The novel coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), continues to be a major health concern. In search for novel treatment strategies against COVID-19, exosomes have attracted the attention of scientists and pharmaceutical companies worldwide. Exosomes are small extracellular vesicles, secreted by all types of cells, and considered as key mediators of intercellular communication and stem-cell paracrine signaling. Herein, we reviewed the most recent literature about the role of exosomes as potential agents for treatment, prevention, diagnosis, and pathogenesis of COVID-19. Several studies and ongoing clinical trials have been investigating the anti-inflammatory, immunomodulatory, and reparative effects of exosomes derived from mesenchymal stem/stromal cells for COVID-19-related acute lung injury. Other studies reported that exosomes play a key role in convalescent plasma therapy for COVID-19, and that they could be of use for the treatment of COVID-19 Kawasaki's-like multisystem inflammatory syndrome and as drug delivery nanocarriers for antiviral therapy. Harnessing some advantageous aspects of exosome biology, such as their endogenous origin, capability of crossing biological barriers, high stability in circulation, and low toxicity and immunogenicity, several companies have been testing exosome-based vaccines against SARS-CoV-2. As they carry cargos that mimic the status of parent cells, exosomes can be isolated from a variety of sources, including plasma, and employed as biomarkers of COVID-19. Lastly, there is growing evidence supporting the role of exosomes in COVID-19 infection, spread, reactivation, and reinfection. The lessons learned using exosomes for COVID-19 will help determine their efficacy and applicability in other clinical conditions.


Asunto(s)
COVID-19/terapia , Exosomas/inmunología , COVID-19/diagnóstico , COVID-19/prevención & control , Vacunas contra la COVID-19 , Portadores de Fármacos , Humanos , Inmunización Pasiva , Células Madre Mesenquimatosas/citología , Sueroterapia para COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA