Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Pediatr Surg Int ; 40(1): 78, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38472353

ABSTRACT

BACKGROUND: The RNA-binding protein Quaking (QKI) increases during epithelial-to-mesenchymal transition and its expression is controlled by microRNA-200 family members. Here, we aimed to describe the expression of QKI in the developing lungs of control and nitrofen-induced congenital diaphragmatic hernia lungs (CDH). METHODS: To investigate the expression of QKI, we dissected lungs from control and nitrofen-induced CDH rats on embryonic day 15, 18, 21 (E15, E18, E21). We performed immunofluorescence (IF) and quantitative reverse transcription PCR (RT-qPCR) for QKI expression. Additionally, we assessed Interleukin-6 (IL-6) abundance using IF. RESULTS: On E21, IF showed that the abundance of all three QKI isoforms and IL-6 protein was higher in CDH lungs compared to control lungs (QKI5: p = 0.023, QKI6: p = 0.006, QKI7: p = 0.014, IL-6: p = 0.045, respectively). Furthermore, RT-qPCR data showed increased expression of QKI5, QKI6, and QKI7 mRNA in E21 nitrofen lungs by 1.63 fold (p = 0.001), 1.63 fold (p = 0.010), and 1.48 fold (p = 0.018), respectively. CONCLUSIONS: Our data show an increase in the abundance and expression of QKI at the end of gestation in nitrofen-induced CDH lungs. Therefore, a disruption in the regulation of QKI during the late stage of pregnancy could be associated with the pathogenesis of abnormal lung development in CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital , Pregnancy , Female , Rats , Animals , Hernias, Diaphragmatic, Congenital/metabolism , Interleukin-6/metabolism , Rats, Sprague-Dawley , Lung/abnormalities , Phenyl Ethers , Disease Models, Animal , Gene Expression Regulation, Developmental
2.
Nat Med ; 30(3): 875-887, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38438734

ABSTRACT

Isolation of tissue-specific fetal stem cells and derivation of primary organoids is limited to samples obtained from termination of pregnancies, hampering prenatal investigation of fetal development and congenital diseases. Therefore, new patient-specific in vitro models are needed. To this aim, isolation and expansion of fetal stem cells during pregnancy, without the need for tissue samples or reprogramming, would be advantageous. Amniotic fluid (AF) is a source of cells from multiple developing organs. Using single-cell analysis, we characterized the cellular identities present in human AF. We identified and isolated viable epithelial stem/progenitor cells of fetal gastrointestinal, renal and pulmonary origin. Upon culture, these cells formed clonal epithelial organoids, manifesting small intestine, kidney tubule and lung identity. AF organoids exhibit transcriptomic, protein expression and functional features of their tissue of origin. With relevance for prenatal disease modeling, we derived lung organoids from AF and tracheal fluid cells of congenital diaphragmatic hernia fetuses, recapitulating some features of the disease. AF organoids are derived in a timeline compatible with prenatal intervention, potentially allowing investigation of therapeutic tools and regenerative medicine strategies personalized to the fetus at clinically relevant developmental stages.


Subject(s)
Hernias, Diaphragmatic, Congenital , Pregnancy , Female , Humans , Hernias, Diaphragmatic, Congenital/metabolism , Amniotic Fluid/metabolism , Prenatal Care , Lung/metabolism , Organoids/metabolism
3.
J Pediatr Surg ; 59(5): 847-853, 2024 May.
Article in English | MEDLINE | ID: mdl-38413261

ABSTRACT

BACKGROUND: Fetoscopic endoluminal tracheal occlusion (FETO) improves the survival rate in fetuses with severe congenital diaphragmatic hernia (CDH). We hypothesize that prenatal therapies into the trachea during FETO can further improve outcomes. Here, we present an ex vivo microinjection technique with rat lung explants to study prenatal therapy with nanoparticles. METHODS: We used microsurgery to isolate lungs from rats on embryonic day 18. We injected chitosan nanoparticles loaded with fluorescein (FITC) into the trachea of the lung explants. We compared the difference in biodistribution of two types of nanoparticles, functionalized IgG-conjugated nanoparticles (IgG-nanoparticles) and bare nanoparticles after 24 h culture with immunofluorescence (IF). We used IF to mark lung epithelial cells with E-cadherin and to investigate an apoptosis (Active-caspase 3) and inflammatory marker (Interleukin, IL-6) and compared its abundance between the two experimental groups and control lung explants. RESULTS: We detected the presence of nanoparticles in the lung explants, and the relative number of nanoparticles to cells was 2.49 fold higher in IgG-nanoparticles than bare nanoparticles (p < 0.001). Active caspase-3 protein abundance was similar in the control, bare nanoparticles (1.20 fold higher), and IgG-nanoparticles (1.34 fold higher) groups (p = 0.34). Similarly, IL-6 protein abundance was not different in the control, bare nanoparticles (1.13 fold higher), and IgG-nanoparticles (1.12 fold higher) groups (p = 0.33). CONCLUSIONS: Functionalized nanoparticles had a higher presence in lung cells and this did not result in more apoptosis or inflammation. Our proof-of-principle study will guide future research with therapies to improve lung development prenatally. LEVELS OF EVIDENCE: N/A TYPE OF STUDY: Animal and laboratory study.


Subject(s)
Hernias, Diaphragmatic, Congenital , Pregnancy , Female , Animals , Rats , Hernias, Diaphragmatic, Congenital/surgery , Hernias, Diaphragmatic, Congenital/metabolism , Pilot Projects , Interleukin-6/metabolism , Microinjections , Tissue Distribution , Lung/abnormalities , Fetoscopy/methods , Trachea/surgery , Immunoglobulin G/metabolism
4.
Pediatr Surg Int ; 40(1): 43, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38291157

ABSTRACT

PURPOSE: CITED2 both modulates lung, heart and diaphragm development. The role of CITED2 in the pathogenesis of congenital diaphragmatic hernia (CDH) is unknown. We aimed to study CITED2 during abnormal lung development in the nitrofen model. METHODS: Timed-pregnant rats were given nitrofen on embryonic day (E) 9 to induce CDH. Fetal lungs were harvested on E15, 18 and 21. We performed RT-qPCR, RNAscope™ in situ hybridization and immunofluorescence staining for CITED2. RESULTS: We observed no difference in RT-qPCR (control: 1.09 ± 0.22 and nitrofen: 0.95 ± 0.18, p = 0.64) and in situ hybridization (1.03 ± 0.03; 1.04 ± 0.03, p = 0.97) for CITED2 expression in E15 nitrofen and control pups. At E18, CITED2 expression was reduced in in situ hybridization of nitrofen lungs (1.47 ± 0.05; 1.14 ± 0.07, p = 0.0006), but not altered in RT-qPCR (1.04 ± 0.16; 0.81 ± 0.13, p = 0.33). In E21 nitrofen lungs, CITED2 RNA expression was increased in RT-qPCR (1.04 ± 0.11; 1.52 ± 0.17, p = 0.03) and in situ hybridization (1.08 ± 0.07, 1.29 ± 0.04, p = 0.02). CITED2 protein abundance was higher in immunofluorescence staining of E21 nitrofen lungs (2.96 × 109 ± 0.13 × 109; 4.82 × 109 ± 0.25 × 109, p < 0.0001). CONCLUSION: Our data suggest that dysregulation of CITED2 contributes to abnormal lung development of CDH, as demonstrated by the distinct spatial-temporal distribution in nitrofen-induced lungs.


Subject(s)
Hernias, Diaphragmatic, Congenital , Lung Diseases , Respiratory System Abnormalities , Animals , Female , Pregnancy , Rats , 2,4-Dinitrophenol , Disease Models, Animal , Gene Expression Regulation, Developmental , Hernias, Diaphragmatic, Congenital/chemically induced , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Lung/abnormalities , Lung Diseases/metabolism , Phenyl Ethers/toxicity , Rats, Sprague-Dawley
5.
Am J Physiol Lung Cell Mol Physiol ; 325(4): L477-L486, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37605849

ABSTRACT

Abnormal pulmonary vascular development and function in congenital diaphragmatic hernia (CDH) is a significant factor leading to pulmonary hypertension. The lung is a very heterogenous organ and has marked cellular diversity that is differentially responsive to injury and therapeutic agents. Spatial transcriptomics provides the unmatched capability of discerning the differences in the transcriptional signature of these distinct cell subpopulations in the lung with regional specificity. We hypothesized that the distal lung parenchyma (selected as a region of interest) would show a distinct transcriptomic profile in the CDH lung compared with control (normal lung). We subjected lung sections obtained from male and female CDH and control neonates to spatial transcriptomics using the Nanostring GeoMx platform. Spatial transcriptomic analysis of the human CDH and control lung revealed key differences in the gene expression signature. Increased expression of alveolar epithelial-related genes (SFTPA1 and SFTPC) and angiogenesis-related genes (EPAS1 and FHL1) was seen in control lungs compared with CDH lungs. Response to vitamin A was enriched in the control lungs as opposed to abnormality of the coagulation cascade and TNF-alpha signaling via NF-kappa B in the CDH lung parenchyma. In male patients with CDH, higher expression of COL1A1 (ECM remodeling) and CD163 was seen. Increased type 2 alveolar epithelial cells (AT-2) and arterial and lung capillary endothelial cells were seen in control lung samples compared with CDH lung samples. To the best of our knowledge, this is the first use of spatial transcriptomics in patients with CDH that identifies the contribution of different lung cellular subpopulations in CDH pathophysiology and highlights sex-specific differences.NEW & NOTEWORTHY This is the first use of spatial transcriptomics in patients with congenital diaphragmatic hernia (CDH) that identifies the contribution of different lung cellular subpopulations in CDH pathophysiology and highlights sex-specific differences.


Subject(s)
Hernias, Diaphragmatic, Congenital , Hypertension, Pulmonary , Infant, Newborn , Humans , Male , Female , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Transcriptome/genetics , Endothelial Cells/metabolism , Lung/metabolism , Hypertension, Pulmonary/metabolism , Phenyl Ethers/metabolism , Muscle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism
6.
Am J Respir Cell Mol Biol ; 69(5): 545-555, 2023 11.
Article in English | MEDLINE | ID: mdl-37552822

ABSTRACT

Abnormal lung development is the main cause of morbidity and mortality in neonates with congenital diaphragmatic hernia (CDH), a common birth defect (1:2,500) of largely unknown pathobiology. Recent studies discovered that inflammatory processes, and specifically NF-κB-associated pathways, are enriched in human and experimental CDH. However, the molecular signaling of NF-κB in abnormal CDH lung development and its potential as a therapeutic target require further investigation. Using sections and hypoplastic lung explant cultures from the nitrofen rat model of CDH and human fetal CDH lungs, we demonstrate that NF-κB and its downstream transcriptional targets are hyperactive during abnormal lung formation in CDH. NF-κB activity was especially elevated in the airway epithelium of nitrofen and human CDH lungs at different developmental stages. Fetal rat lung explants had impaired pseudoglandular airway branching after exposure to nitrofen, together with increased phosphorylation and transcriptional activity of NF-κB. Dexamethasone, the broad and clinically applicable antiinflammatory NF-κB antagonist, rescued lung branching and normalized NF-κB signaling in hypoplastic lung explants. Moreover, specific NF-κB inhibition with curcumenol similarly rescued ex vivo lung hypoplasia and restored NF-κB signaling. Last, we showed that prenatal intraperitoneal dexamethasone administration to pregnant rat dams carrying fetuses with hypoplastic lungs significantly improves lung branching and normalizes NF-κB in vivo. Our results indicate that NF-κB is aberrantly activated in human and nitrofen CDH lungs. Antiinflammatory treatment with dexamethasone and/or specific NF-κB inhibition should be investigated further as a therapeutic avenue to target lung hypoplasia in CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital , Lung Diseases , Pregnancy , Female , Humans , Rats , Animals , Hernias, Diaphragmatic, Congenital/metabolism , NF-kappa B/metabolism , Rats, Sprague-Dawley , Lung/metabolism , Lung Diseases/metabolism , Dexamethasone/metabolism , Disease Models, Animal
7.
Pharmacol Res ; 191: 106749, 2023 05.
Article in English | MEDLINE | ID: mdl-37004830

ABSTRACT

Left congenital diaphragmatic hernia (CDH) can lead to pulmonary arteries abnormalities in the contralateral and ipsilateral sides of the diaphragm. Nitric oxide (NO) is the main therapy used to attenuate the vascular effects of CDH, but it is not always effective. We hypothesized that the left and right pulmonary arteries do not respond similarly to NO donors during CDH. Therefore, vasorelaxant responses of the left and right pulmonary arteries to sodium nitroprusside (SNP, a NO donor) were determined in a rabbit experimental model of left CDH. CDH was surgically induced in the fetuses of rabbits on the 25th day of pregnancy. On the 30th day of pregnancy, a midline laparotomy was performed to access the fetuses. The fetuses' left and right pulmonary arteries were isolated and mounted in myograph chambers. Vasodilation was evaluated by cumulative concentration-effect curves to SNP. Protein expression of guanylate cyclase isoforms (GCα, GCß) and the α isoform of cGMP-dependent protein kinase 1 (PKG1α), and the concentration of NO and cGMP were determined in the pulmonary arteries. The left and right pulmonary arteries of newborns with CDH exhibited increased vasorelaxant responses to SNP (i.e. the potency of SNP was increased) compared to the control group. GCα, GCß, and PKG1α expression were decreased, while NO and cGMP concentrations were increased in the pulmonary arteries of newborns with CDH compared to the control group. The increased cGMP mobilization may be responsible for the increased vasorelaxant responses to the SNP in the pulmonary arteries during left CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital , Animals , Pregnancy , Female , Rabbits , Hernias, Diaphragmatic, Congenital/metabolism , Pulmonary Artery , Nitric Oxide/metabolism , Lung , Vasodilator Agents/pharmacology
8.
Pediatr Surg Int ; 39(1): 180, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37055635

ABSTRACT

PURPOSE: Congenital diaphragmatic hernia (CDH) pathogenesis is poorly understood. We hypothesize that fetal CDH lungs are chronically hypoxic because of lung hypoplasia and tissue compression, affecting the cell bioenergetics as a possible explanation for abnormal lung development. METHODS: To investigate this theory, we conducted a study using the rat nitrofen model of CDH. We evaluated the bioenergetics status using H1 Nuclear magnetic resonance and studied the expression of enzymes involved in energy production, the hypoxia-inducible factor 1α, and the glucose transporter 1. RESULTS: The nitrofen-exposed lungs have increased levels of hypoxia-inducible factor 1α and the main fetal glucose transporter, more evident in the CDH lungs. We also found imbalanced AMP:ATP and ADP:ATP ratios, and a depleted energy cellular charge. Subsequent transcription levels and protein expression of the enzymes involved in bioenergetics confirm the attempt to prevent the energy collapse with the increase in lactate dehydrogenase C, pyruvate dehydrogenase kinase 1 and 2, adenosine monophosphate deaminase, AMP-activated protein kinase, calcium/calmodulin-dependent protein kinase 2, and liver kinase B1, while decreasing ATP synthase. CONCLUSION: Our study suggests that changes in energy production could play a role in CDH pathogenesis. If confirmed in other animal models and humans, this could lead to the development of novel therapies targeting the mitochondria to improve outcomes.


Subject(s)
Hernias, Diaphragmatic, Congenital , Lung Diseases , Humans , Rats , Animals , Hernias, Diaphragmatic, Congenital/metabolism , Rats, Sprague-Dawley , Lung/abnormalities , Phenyl Ethers/toxicity , Lung Diseases/metabolism , Hypoxia/metabolism , Adenosine Triphosphate/adverse effects , Adenosine Triphosphate/metabolism , Disease Models, Animal
9.
Pediatr Surg Int ; 39(1): 61, 2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36564649

ABSTRACT

PURPOSE: Here, we establish a tracheal occlusion (TO) model with rat lung explants in nitrofen-induced pulmonary hypoplasia in the congenital diaphragmatic hernia (CDH). METHODS: We extracted lungs from rats on an embryonic day 18. We mimicked TO in the lung explants by tying the trachea. We assessed lung weight, morphometry, and abundance of Ki-67, Active caspase-3, and Prosurfactant Protein C (proSP-C) with immunofluorescence. RESULTS: Lung weight was higher in TO + than TO - on day 1. Abundance of Ki-67 was higher in TO + than TO - (0.15 vs. 0.32, p = 0.009 for day 1, 0.07 vs. 0.17, p = 0.004 for day 2, 0.07 vs. 0.12, p = 0.044 for day 3), and Active caspase-3 was higher in TO + than TO - on day 2 and day 3 (0.04 vs. 0.03 p = 0.669 for day 1, 0.03 vs. 0.13 p < 0.001 for day 2, 0.04 vs. 0.17 p = 0.008 for day3). However, proSP-C protein abundance was lower in TO + than TO - (67.9 vs. 59.1 p = 0.033 for day 1, 73.5 vs. 51.6 p = 0.038 for day 2, 83.1 vs. 56.4 p = 0.009 for day 3). CONCLUSIONS: The TO model in lung explants mimics the outcomes of current surgical models of TO and further studies can reveal the cellular and molecular effects of TO in CDH lungs.


Subject(s)
Airway Obstruction , Hernias, Diaphragmatic, Congenital , Rats , Animals , Hernias, Diaphragmatic, Congenital/surgery , Hernias, Diaphragmatic, Congenital/metabolism , Caspase 3/metabolism , Ki-67 Antigen/metabolism , Rats, Sprague-Dawley , Lung , Phenyl Ethers/toxicity , Disease Models, Animal
10.
Stem Cells Transl Med ; 11(10): 1089-1102, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36103370

ABSTRACT

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.


Subject(s)
Extracellular Vesicles , Hernias, Diaphragmatic, Congenital , MicroRNAs , Rats , Animals , Hernias, Diaphragmatic, Congenital/metabolism , beta Catenin/metabolism , Amniotic Fluid/metabolism , Neuropilin-1/metabolism , Rats, Sprague-Dawley , Lung/metabolism , Stem Cells/metabolism , Cell Differentiation , RNA, Messenger/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Ephrins/metabolism , Disease Models, Animal
11.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563656

ABSTRACT

The Hedgehog (HH) signaling pathway plays an essential role in mouse lung development. We hypothesize that the HH pathway is necessary for branching during human lung development and is impaired in pulmonary hypoplasia. Single-cell, bulk RNA-sequencing data, and human fetal lung tissues were analyzed to determine the spatiotemporal localization of HH pathway actors. Distal human lung segments were cultured in an air-liquid interface and treated with an SHH inhibitor (5E1) to determine the effect of HH inhibition on human lung branching, epithelial-mesenchymal markers, and associated signaling pathways in vitro. Our results showed an early and regulated expression of HH pathway components during human lung development. Inhibiting HH signaling caused a reduction in branching during development and dysregulated epithelial (SOX2, SOX9) and mesenchymal (ACTA2) progenitor markers. FGF and Wnt pathways were also disrupted upon HH inhibition. Finally, we demonstrated that HH signaling elements were downregulated in lung tissues of patients with a congenital diaphragmatic hernia (CDH). In this study, we show for the first time that HH signaling inhibition alters important genes and proteins required for proper branching of the human developing lung. Understanding the role of the HH pathway on human lung development could lead to the identification of novel therapeutic targets for childhood pulmonary diseases.


Subject(s)
Hedgehog Proteins , Lung , Signal Transduction , Animals , Child , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hernias, Diaphragmatic, Congenital/metabolism , Humans , Lung/growth & development , Lung/metabolism , Mice , Morphogenesis , Organogenesis , Wnt Signaling Pathway
12.
Pediatr Surg Int ; 38(5): 695-700, 2022 May.
Article in English | MEDLINE | ID: mdl-35235015

ABSTRACT

PURPOSE: We previously demonstrated that absence of miR-200b results in abnormal lung development in congenital diaphragmatic hernia due to imbalance between epithelial and mesenchymal cells. Tenascin C is a highly conserved extracellular matrix protein involved in epithelial to mesenchymal transition, tissue regeneration and lung development. Considering the involvement of Tenascin C and miR-200b and their potential interaction, we aimed to study Tenascin C during lung development in the absence of miR-200b. METHODS: We collected lungs of miR-200b-/- mice (male, 8 weeks). We performed Western blot (WB) analysis (N = 6) and immunofluorescence (N = 5) for Tenascin C and alpha smooth muscle actin and RT-qPCR for Tenascin C gene expression (N = 4). RESULTS: Using WB analysis, we observed a decreased total protein abundance of Tenascin C in miR-200b-/- lungs (miR-200b+/+: 3.8 × 107 ± 1 × 107; miR-200b-/-: 1.9 × 107 ± 5 × 106; p = 0.002). Immunofluorescence confirmed decreased total Tenascin C in miR-200b-/- lungs. Tenascin C was significantly decreased in the mesenchyme but relatively increased in the airways of mutant lungs. Total lung RNA expression of Tenascin C was higher in miR-200b-/- lungs. CONCLUSION: We report dysregulation of Tenascin C in lungs of miR-200b-/- mice. This suggests that absence of miR-200b results in abnormal Tenascin C abundance contributing to the lung hypoplasia observed in miR-200b-/- mice.


Subject(s)
Hernias, Diaphragmatic, Congenital , MicroRNAs , Tenascin , Animals , Epithelial-Mesenchymal Transition , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Lung/abnormalities , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Tenascin/genetics , Tenascin/metabolism
13.
Pediatr Surg Int ; 38(5): 713-719, 2022 May.
Article in English | MEDLINE | ID: mdl-35226175

ABSTRACT

BACKGROUND: Congenital diaphragmatic hernia (CDH) is a birth defect associated with abnormal lung development. Yes-associated protein (YAP) is a core kinase of the Hippo pathway, which controls organ size during development. The absence of YAP protein during lung development results in hypoplastic lungs comparable to the lung phenotype in CDH (Mahoney, Dev Cell 30(2):137-150, 2014). We aimed to describe the expression of YAP during normal and nitrofen-induced abnormal lung development. METHODS: Intra-gastric administration of dams with 100 mg of nitrofen was used to induce CDH and abnormal lung development in the embryos. Immunofluorescence was performed to visualize the localization of YAP and p-YAP during lung development (E15, E18, E21). Western Blotting was used to determine the abundance of YAP and p-YAP in E21 control and nitrofen-induced hypoplastic CDH lungs. RESULTS: Immunofluorescence demonstrated cytoplasmic localization of YAP protein in airway epithelial and mesenchymal cells of nitrofen-induced hypoplastic lungs compared to nuclear localization in control lungs. Western Blotting showed a decrease (p = 0.0188) in abundance of YAP (active form) and increase in p-YAP (inactive form) in hypoplastic lungs compared to control lungs. CONCLUSION: Our results demonstrate that YAP protein is mostly phosphorylated, inactive, and expressed in the cytoplasm at the later stages of nitrofen-induced hypoplastic lung development indicating that the alteration in regulation of YAP can be associated with the pathogenesis of abnormal lung development in experimental CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital , Animals , Disease Models, Animal , Gene Expression Regulation, Developmental , Hernias, Diaphragmatic, Congenital/metabolism , Humans , Lung/abnormalities , Phenyl Ethers/toxicity , Rats , Rats, Sprague-Dawley , YAP-Signaling Proteins
14.
Am J Physiol Renal Physiol ; 322(1): F14-F26, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34747197

ABSTRACT

The multiligand receptors megalin (Lrp2) and cubilin (Cubn) and their endocytic adaptor protein Dab2 (Dab2) play essential roles in maintaining the integrity of the apical endocytic pathway of proximal tubule (PT) cells and have complex and poorly understood roles in the development of chronic kidney disease. Here, we used RNA-sequencing and CRISPR/Cas9 knockout (KO) technology in a well-differentiated cell culture model to identify PT-specific transcriptional changes that are directly consequent to the loss of megalin, cubilin, or Dab2 expression. KO of Lrp2 had the greatest transcriptional effect, and nearly all genes whose expression was affected in Cubn KO and Dab2 KO cells were also changed in Lrp2 KO cells. Pathway analysis and more granular inspection of the altered gene profiles suggested changes in pathways with immunomodulatory functions that might trigger the pathological changes observed in KO mice and patients with Donnai-Barrow syndrome. In addition, differences in transcription patterns between Lrp2 and Dab2 KO cells suggested the possibility that altered spatial signaling by aberrantly localized receptors contributes to transcriptional changes upon the disruption of PT endocytic function. A reduction in transcripts encoding sodium-glucose cotransporter isoform 2 was confirmed in Lrp2 KO mouse kidney lysates by quantitative PCR analysis. Our results highlight the role of megalin as a master regulator and coordinator of ion transport, metabolism, and endocytosis in the PT. Compared with the studies in animal models, this approach provides a means to identify PT-specific transcriptional changes that are directly consequent to the loss of these target genes.NEW & NOTEWORTHY Megalin and cubilin receptors together with their adaptor protein Dab2 represent major components of the endocytic machinery responsible for efficient uptake of filtered proteins by the proximal tubule (PT). Dab2 and megalin expression have been implicated as both positive and negative modulators of kidney disease. We used RNA sequencing to knock out CRISPR/Cas9 cubilin, megalin, and Dab2 in highly differentiated PT cells to identify PT-specific changes that are directly consequent to knockout of each component.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Knockout Techniques , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Receptors, Cell Surface/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Databases, Genetic , Gene Regulatory Networks , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Humans , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice, Knockout , Monodelphis , Myopia/genetics , Myopia/metabolism , Myopia/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , Receptors, Cell Surface/genetics , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/pathology
15.
Pediatr Res ; 92(1): 118-124, 2022 07.
Article in English | MEDLINE | ID: mdl-34465875

ABSTRACT

BACKGROUND: Teratogen-induced congenital diaphragmatic hernia (CDH) rat models are commonly used to study the pathophysiology. We have created a new and reliable surgically induced diaphragmatic hernia (DH) model to obtain a purely mechanical DH rat model, and avoid the confounding teratogen-induced effects on the lung development. METHODS: Fetal DH was surgically created on fetuses at E18.5 and harvested at E21.5 in rats. Four groups were evaluated (n = 16): control (CONT), control exposed to Nitrofen (CONT NIT), DH surgically created (DH SURG), and CDH Nitrofen (CDH NIT). Body weight, total lung weights, and their ratio (BW, TLW, and TLBR) were compared. Air space (AS), parenchyma (PA), total protein, and DNA contents were measured to verify lung hypoplasia. Medial wall thickness (MWT) of pulmonary arterioles was also analyzed. RESULTS: DH SURG showed significant hypoplasia (decreased in total protein and DNA) vs CONT (p < 0.05); DH SURG vs CDH NIT were similar in TLW and TLBR. DH SURG has less AS than CONT (p < 0.05) and similar PA compared to CONT NIT and CDH NIT, MWT were similarly increased in CONT NIT, DH SURG, and CDH NIT. CONCLUSIONS: This novel surgical model generates fetal lung hypoplasia contributing to the study of the mechanical compression effect on fetal lung development in DH. IMPACT: There is a critical need to develop a surgical model in rat to complement the findings of the well-known Nitrofen-induced CDH model. This experimental study is pioneer and can help to understand better the CDH pathophysiological changes caused by herniated abdominal viscera compression against the lung during the final stage of gestation in CDH fetuses, and also to develop more efficient treatments in near future.


Subject(s)
Hernias, Diaphragmatic, Congenital , Animals , DNA/metabolism , Disease Models, Animal , Fetus , Hernias, Diaphragmatic, Congenital/metabolism , Lung , Models, Anatomic , Phenyl Ethers/toxicity , Rats , Rats, Sprague-Dawley , Teratogens/metabolism , Teratogens/pharmacology
16.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502408

ABSTRACT

Oxidative stress plays a pathological role in pulmonary hypoplasia and pulmonary hypertension in congenital diaphragmatic hernia (CDH). This study investigated the effect of molecular hydrogen (H2), an antioxidant, on CDH pathology induced by nitrofen. Sprague-Dawley rats were divided into three groups: control, CDH, and CDH + hydrogen-rich water (HW). Pregnant dams of CDH + HW pups were orally administered HW from embryonic day 10 until parturition. Gasometric evaluation and histological, immunohistochemical, and real-time polymerase chain reaction analyses were performed. Gasometric results (pH, pO2, and pCO2 levels) were better in the CDH + HW group than in the CDH group. The CDH + HW group showed amelioration of alveolarization and pulmonary artery remodeling compared with the CDH group. Oxidative stress (8-hydroxy-2'-deoxyguanosine-positive-cell score) in the pulmonary arteries and mRNA levels of protein-containing pulmonary surfactant that protects against pulmonary collapse (surfactant protein A) were significantly attenuated in the CDH + HW group compared with the CDH group. Overall, prenatal H2 administration improved respiratory function by attenuating lung morphology and pulmonary artery thickening in CDH rat models. Thus, H2 administration in pregnant women with diagnosed fetal CDH might be a novel antenatal intervention strategy to reduce newborn mortality due to CDH.


Subject(s)
Hernias, Diaphragmatic, Congenital/drug therapy , Hydrogen/pharmacology , Animals , Animals, Newborn , Antioxidants/pharmacology , Deuterium Oxide/pharmacology , Disease Models, Animal , Female , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Hydrogen/metabolism , Hypertension, Pulmonary/metabolism , Lung/pathology , Male , Organogenesis/drug effects , Phenyl Ethers/adverse effects , Phenyl Ethers/pharmacology , Pregnancy , Pulmonary Artery , Pulmonary Surfactants/metabolism , Rats , Rats, Sprague-Dawley , Vascular Remodeling/drug effects
17.
Cells ; 10(6)2021 06 14.
Article in English | MEDLINE | ID: mdl-34198576

ABSTRACT

Infants with congenital diaphragmatic hernia (CDH) are at high risk of postnatal mortality due to lung hypoplasia and arterial pulmonary hypertension. In severe cases, prenatal intervention by fetal endoscopic tracheal occlusion (FETO) can improve survival by accelerating lung growth. However, postnatal mortality remains in the range of about 50% despite fetal treatment, and there is currently no clear explanation for this different clinical response to FETO. We evaluated the concentration of extracellular vesicles (EVs) and associated microRNA expression in amniotic and tracheal fluids of fetuses with CDH undergoing FETO, and we examined the association between molecular findings and postnatal survival. We observed a higher count of EVs in the amniotic fluid of non-survivors and in the tracheal fluid sampled in utero at the time of reversal of tracheal occlusion, suggesting a pro-inflammatory lung reactivity that is already established in utero and that could be associated with a worse postnatal clinical course. In addition, we observed differential regulation of four EV-enclosed miRNAs (miR-379-5p, miR-889-3p; miR-223-3p; miR-503-5p) in relation to postnatal survival, with target genes possibly involved in altered lung development. Future research should investigate molecular therapeutic agents targeting differentially regulated miRNAs to normalize their expression and potentially improve clinical outcomes.


Subject(s)
Amniotic Fluid/metabolism , Extracellular Vesicles/metabolism , Fetal Diseases/metabolism , Fetus/metabolism , Hernias, Diaphragmatic, Congenital/metabolism , MicroRNAs/metabolism , Trachea/embryology , Extracellular Vesicles/pathology , Female , Fetal Diseases/surgery , Fetus/surgery , Hernias, Diaphragmatic, Congenital/surgery , Humans , Severity of Illness Index , Trachea/surgery
18.
Molecules ; 26(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530644

ABSTRACT

The aim of this study was to analyze the exhaled volatile organic compounds (VOCs) profile, airway microbiome, lung function and exercise performance in congenital diaphragmatic hernia (CDH) patients compared to healthy age and sex-matched controls. A total of nine patients (median age 9 years, range 6-13 years) treated for CDH were included. Exhaled VOCs were measured by GC-MS. Airway microbiome was determined from deep induced sputum by 16S rRNA gene sequencing. Patients underwent conventional spirometry and exhausting bicycle spiroergometry. The exhaled VOC profile showed significantly higher levels of cyclohexane and significantly lower levels of acetone and 2-methylbutane in CDH patients. Microbiome analysis revealed no significant differences for alpha-diversity, beta-diversity and LefSe analysis. CDH patients had significantly lower relative abundances of Pasteurellales and Pasteurellaceae. CDH patients exhibited a significantly reduced Tiffeneau Index. Spiroergometry showed no significant differences. This is the first study to report the VOCs profile and airway microbiome in patients with CDH. Elevations of cyclohexane observed in the CDH group have also been reported in cases of lung cancer and pneumonia. CDH patients had no signs of impaired physical performance capacity, fueling controversial reports in the literature.


Subject(s)
Bacteria/classification , Hernias, Diaphragmatic, Congenital/surgery , Herniorrhaphy/methods , RNA, Ribosomal, 16S/genetics , Volatile Organic Compounds/analysis , Acetone/analysis , Adolescent , Bacteria/genetics , Bacteria/isolation & purification , Child , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Exercise , Female , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/physiopathology , Humans , Male , Microbiota , Pentanes/analysis , Phylogeny , Spirometry , Vital Capacity
19.
Arch Biochem Biophys ; 700: 108769, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33484710

ABSTRACT

A congenital diaphragmatic hernia (CDH) is an anomaly caused by defects in the diaphragm; the resulting limited thorax cavity in turn restricts lung growth (pulmonary hypoplasia). This condition is related to pulmonary hypertension. Despite advances in neonatal CDH therapy, the mortality for severe pulmonary hypoplasia remains high. Therefore, it is essential to establish prenatal therapeutic interventions. Vitamin D was reported to have beneficial effects on adult pulmonary hypertension. This study aims to evaluate the efficacy of prenatal vitamin D administration for CDH. First, serum 25-hydroxyvitamin D [25(OH)D] levels in umbilical cord blood were evaluated among CDH newborns. Second, Sprague Dawley rat CDH models were exposed to nitrofen on embryo day 9 (E9). Randomly selected rats in the nitrofen-treated group were infused with calcitriol from E9 to E21. Samples from CDH pups diagnosed after birth were used for lung weight measurements, blood gas analysis, and immunohistochemical analysis. Third, microarray analysis was performed to examine the effect of vitamin D on gene expression profiles in CDH pulmonary arterial tissues. Serum 25(OH)D levels in the umbilical cord blood of newborns who did not survive were significantly lower than those who were successfully discharged. Prenatal vitamin D showed no significant effect on CDH incidence or lung weight but attenuated alveolarization and pulmonary artery remodeling accompanied the improved blood gas parameters. Vitamin D inhibited several gene expression pathways in the pulmonary arteries of CDH rats. Our results suggest that prenatal vitamin D administration attenuates pulmonary vascular remodeling by influencing several gene pathways in CDH.


Subject(s)
Gene Expression Regulation/drug effects , Hernias, Diaphragmatic, Congenital , Phenyl Ethers/toxicity , Vitamin D/analogs & derivatives , Animals , Disease Models, Animal , Hernias, Diaphragmatic, Congenital/chemically induced , Hernias, Diaphragmatic, Congenital/drug therapy , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Humans , Rats , Rats, Sprague-Dawley , Vitamin D/pharmacokinetics , Vitamin D/pharmacology
20.
Pediatr Surg Int ; 37(1): 49-57, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33161446

ABSTRACT

BACKGROUND: Congenital diaphragmatic hernia (CDH) and congenital pulmonary airway malformation (CPAM) are two inborn pathologies of the lung of unknown origin. Alterations of gene expression in airway epithelial cells are involved in the pathobiology of both diseases. We previously found decreased expression of the epithelial cell adhesion protein cadherin 26 (CDH26) in hypoplastic mice lungs. Here, our objective was to describe the expression and localization of CDH26 in hypoplastic CDH lungs and hyperproliferative CPAM tissues. METHODS: After ethical approval, we used human lung tissues from CDH and CPAM cases and age-matched control samples from a previously established biobank. Furthermore, lungs from the nitrofen rat model of CDH were included in the study. We performed immunohistochemistry and western blot analysis with antibodies against CDH26 to examine protein localization and abundance. Statistical analysis was performed using Mann-Whitney U test with significance set at p < 0.05. RESULTS: We observed an overexpression of CDH26 within the epithelium of cystic CPAM lesions compared to normal airways within the same lung and compared to control lungs. Western blot demonstrated a downregulation of CDH26 in the nitrofen rat model of CDH compared to healthy controls. Immunohistochemistry could not show consistent differences between CDH and control in human and rat lungs. In the studied human lung samples, CDH26 was localized to the apical part of the airway epithelial cells. CONCLUSION: CDH26 is differentially expressed in human CPAM lung tissues and may be downregulated in nitrofen-induced hypoplastic rat lungs compared to control lungs. Disruption of CDH26 associated pathways in lung development may be involved in the pathogenesis of lung hypoplasia or cystic lung disease.


Subject(s)
Cadherins/metabolism , Hernias, Diaphragmatic, Congenital/metabolism , Lung Diseases/metabolism , Lung/abnormalities , Animals , Cadherins/genetics , Disease Models, Animal , Down-Regulation , Epithelial Cells/metabolism , Hernias, Diaphragmatic, Congenital/genetics , Humans , Infant , Infant, Newborn , Lung/metabolism , Lung Diseases/genetics , Mice , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...