Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Cell Res ; 437(1): 113997, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38508328

ABSTRACT

Bronchopulmonary dysplasia (BPD) is characterized by shortened secondary septa and fewer, larger alveoli. Elastin deposition to the distal tips of the secondary septa is critical for elongation of the secondary septa. Alveolar myofibroblasts, which are thought to migrate to the septal tips during alveolarization, are mainly responsible for elastin production and deposition. Antenatal exposure to inflammation induces abnormal elastin deposition, thereby increasing the risk of developing BPD. Here, we found that lipopolysaccharide (LPS) significantly increased the expression of transforming growth factor-α (TGF-α) in an LPS-induced rat model of BPD and in LPS-treated human pulmonary epithelial cells (BEAS-2B). In addition, in vitro experiments suggested that LPS upregulated TGF-α expression via toll-like receptor 4 (TLR4)/tumor necrosis factor α-converting enzyme (TACE) signaling. Increased TGF-α levels via its receptor epidermal growth factor receptor (EGFR)-induced lysyl oxidase (LOX) overactivation and cell division cycle 42 (Cdc42) activity inhibition of myofibroblasts. Similarly, in vivo LOX overactivation and inhibition of Cdc42 activity were observed in the lungs of LPS-exposed pups. LOX overactivation led to abnormal elastin deposition, and inhibition of Cdc42 activity disturbed the directional migration of myofibroblasts and disrupted elastin localization. Most importantly, the EGFR inhibitor erlotinib partially rescued LOX overactivation and Cdc42 activity inhibition, and improved elastin deposition and alveolar development in antenatal LPS-treated rats. Taken together, our data suggest that TGF-α/EGFR signaling is critically involved in the regulation of elastin deposition and represents a novel therapeutic target.


Subject(s)
Bronchopulmonary Dysplasia , Lipopolysaccharides , Animals , Female , Humans , Infant, Newborn , Pregnancy , Rats , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/metabolism , Elastin , ErbB Receptors/metabolism , Lipopolysaccharides/pharmacology , Lung/metabolism , Transforming Growth Factor alpha
2.
Am J Pathol ; 191(12): 2080-2090, 2021 12.
Article in English | MEDLINE | ID: mdl-34508690

ABSTRACT

Lung inflammation interrupts alveolarization and causes bronchopulmonary dysplasia (BPD). Besides mechanical ventilation and hyperoxia, sepsis contributes to BPD pathogenesis. Adrenomedullin (Adm) is a multifunctional peptide that exerts anti-inflammatory effects in the lungs of adult rodents. Whether Adm mitigates sepsis-induced neonatal lung injury is unknown. The lung phenotype of mice exposed to early postnatal lipopolysaccharide (LPS) was recently shown to be similar to that in human BPD. This model was used to test the hypothesis that Adm-deficient neonatal mice will display increased LPS-induced lung injury than their wild-type (WT) littermates. Adm-deficient mice or their WT littermates were intraperitoneally administered 6 mg/kg of LPS or vehicle daily on postnatal days (PNDs) 3 to 5. The lungs were harvested at several time points to quantify inflammation, alveolarization, and vascularization. The extent of LPS-induced lung inflammation in Adm-deficient mice was 1.6-fold to 10-fold higher than their WT littermates. Strikingly, Adm deficiency induced STAT1 activation and potentiated STAT3 activation in LPS-exposed lungs. The severity of LPS-induced interruption of lung development was also greater in Adm-deficient mice at PND7. At PND14, LPS-exposed WT littermates displayed substantial improvement in lung development, whereas LPS-exposed Adm-deficient mice continued to have decreased lung development. These data indicate that Adm is necessary to decrease lung inflammation and injury and promote repair of the injured lungs in LPS-exposed neonatal mice.


Subject(s)
Adrenomedullin/physiology , Bronchopulmonary Dysplasia/genetics , Adrenomedullin/genetics , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Female , Gene Dosage/physiology , Lipopolysaccharides , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
3.
Pediatr Res ; 92(2): 436-444, 2022 08.
Article in English | MEDLINE | ID: mdl-34725500

ABSTRACT

BACKGROUND: We aimed to compare the effect of dexamethasone (Dex), hydrocortisone (Hc), and methylprednisolone (Mpz) at equivalent doses on somatic growth, lung healing, and neurotoxicity in a hyperoxic rat model. We hypothesized that Mpz and Hc would be superior to Dex with less neurotoxicity by exerting similar therapeutic efficacy on the injured lung. METHODS: Neonatal rats were randomized to control, bronchopulmonary dysplasia (BPD), Dex, Hc, and Mpz groups. All drugs were administered daily following day 15 over 7 days. Histopathological and immunohistochemical analyses of the lung and brain were performed on day 22. RESULTS: All types had much the same impact on lung repair. Oxidative markers in the lung were similar in the steroid groups. While nuclear factor erythroid 2-related factor and heat-shock protein 70 dropped following steroid treatment, no difference was noted in other biochemical markers in the brain between the study groups. Apoptotic activity and neuron loss in the parietal cortex and hippocampus were noted utmost in Dex, but alike in other BPD groups. CONCLUSIONS: Mpz does not appear to be superior to Dex or Hc in terms of pulmonary outcomes and oxidative damage in the brain, but safer than Dex regarding apoptotic neuron loss. IMPACT: This is the first study that compared the pulmonary efficacy and neurotoxic effects of Dex, Hc, and Mpz simultaneously in an established BPD model. This study adds to the literature on the importance of possible antioxidant and protective effects of glucocorticoid therapy in an oxidative stress-exposed brain. Mpz ended up with no more additional neuron loss or apoptosis risk by having interchangeable effects with others for the treatment of established BPD. Mpz and Hc seem safe as a rescue therapy in terms of adverse outcomes for established BPD in which lung and brain tissue is already impaired.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Lung Injury , Neurotoxicity Syndromes , Animals , Humans , Infant, Newborn , Rats , Animals, Newborn , Antioxidants , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/drug therapy , Dexamethasone , Glucocorticoids/therapeutic use , HSP70 Heat-Shock Proteins , Hydrocortisone , Hyperoxia/complications , Hyperoxia/drug therapy , Lung , Lung Injury/drug therapy , Methylprednisolone/therapeutic use
4.
Am J Physiol Lung Cell Mol Physiol ; 321(3): L507-L517, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34189953

ABSTRACT

Premature infants often require mechanical ventilation and oxygen therapy, which can result in bronchopulmonary dysplasia (BPD), characterized by developmental arrest and impaired lung function. Conventional clinical methods for assessing the prenatal lung are not adequate for the detection and assessment of long-term health risks in infants with BPD, highlighting the need for a noninvasive tool for the characterization of lung microstructure and function. Theoretical diffusion models, like the model of xenon exchange (MOXE), interrogate alveolar gas exchange by predicting the uptake of inert hyperpolarized (HP) 129Xe gas measured with HP 129Xe magnetic resonance spectroscopy (MRS). To investigate HP 129Xe MRS as a tool for noninvasive characterization of pulmonary microstructural and functional changes in vivo, HP 129Xe gas exchange data were acquired in an oxygen exposure rat model of BPD that recapitulates the fewer and larger distal airways and pulmonary vascular stunting characteristics of BPD. Gas exchange parameters from MOXE, including airspace mean chord length (Lm), apparent hematocrit in the pulmonary capillaries (HCT), and pulmonary capillary transit time (tx), were compared with airspace mean axis length and area density (MAL and ρA) and percentage area of tissue and air (PTA and PAA) from histology. Lm was significantly larger in the exposed rats (P = 0.003) and correlated with MAL, ρA, PTA, and PAA (0.59<|ρ|<0.66 and P < 0.05). Observed increase in HCT (P = 0.012) and changes in tx are also discussed. These findings support the use of HP 129Xe MRS for detecting fewer, enlarged distal airways in this rat model of BPD, and potentially in humans.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Capillaries/metabolism , Lung/metabolism , Magnetic Resonance Spectroscopy , Pulmonary Gas Exchange , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/pathology , Capillaries/pathology , Disease Models, Animal , Female , Humans , Lung/blood supply , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Xenon Isotopes
5.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L497-L512, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32697651

ABSTRACT

Hyperoxia (HO)-induced lung injury contributes to bronchopulmonary dysplasia (BPD) in preterm newborns. Intractable wheezing seen in BPD survivors is associated with airway remodeling (AWRM). Sphingosine kinase 1 (SPHK1)/sphingosine-1-phosphate (S1P) signaling promotes HO-mediated neonatal BPD; however, its role in the sequela of AWRM is not known. We noted an increased concentration of S1P in tracheal aspirates of neonatal infants with severe BPD, and earlier, demonstrated that Sphk1-/- mice showed protection against HO-induced BPD. The role of SPHK1/S1P in promoting AWRM following exposure of neonates to HO was investigated in a murine model. Therapy using PF543, the specific SPHK1 inhibitor, during neonatal HO reduced alveolar simplification followed by reduced AWRM in adult mice. This was associated with reduced airway hyperreactivity to intravenous methacholine. Neonatal HO exposure was associated with increased expression of SPHK1 in lung tissue of adult mice, which was reduced with PF543 therapy in the neonatal stage. This was accompanied by amelioration of HO-induced reduction of E-cadherin in airway epithelium. This may be suggestive of arrested partial epithelial mesenchymal transition (EMT) induced by HO. In vitro studies using human primary airway epithelial cells (HAEpCs) showed that SPHK1 inhibition or deletion restored HO-induced reduction in E-cadherin and reduced formation of mitochondrial reactive oxygen species (mtROS). Blocking mtROS with MitoTempo attenuated HO-induced partial EMT of HAEpCs. These results collectively support a therapeutic role for PF543 in preventing HO-induced BPD in neonates and the long-term sequela of AWRM, thus conferring a long-term protection resulting in improved lung development and function.


Subject(s)
Airway Remodeling/drug effects , Bronchopulmonary Dysplasia/drug therapy , Hyperoxia/drug therapy , Methanol/analogs & derivatives , Pyrrolidines/pharmacology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Disease Models, Animal , Hyperoxia/chemically induced , Lung/drug effects , Lung/metabolism , Methanol/pharmacology , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sulfones
6.
BMC Pulm Med ; 19(1): 145, 2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31409326

ABSTRACT

BACKGROUND: Aerosol administration is increasingly being used as a therapeutic intervention for mechanically ventilated preterm infants. However, the effects of inhalation therapy on retinopathy of prematurity (ROP) have not yet been explored. METHODS: A retrospective cohort study was conducted in a tertiary level neonatal intensive care unit (NICU) from 2011 to 2013. All preterm infants with a gestational age (GA) of 24~29 weeks receiving invasive intubation for more than 1 week in the NICU were included. Infants with severe congenital anomalies were excluded. ROP was defined as stage II or greater according to medical records by ophthalmologists. A multivariate logistic regression model was used to estimate the risk of ROP in relation to inhalation therapy after adjusting for confounders. RESULTS: In total, 205 infants were enrolled in this study, including 154 with inhalation therapy and 51 without inhalation therapy. Univariate analyses showed an association of inhalation with the following characteristics: sex (p = 0.047), GA (p = 0.029), sepsis (p = 0.047), bronchopulmonary dysplasia (BPD) (p < 0.001), and ROP (p = 0.001). Furthermore, logistic regression analysis indicated that inhalation therapy was an independent risk factor for ROP (odds ratio (OR) = 2.639; 95% confidence interval (CI) = 1.050~6.615). In addition, infants with a GA of 24~25 weeks (OR = 6.063; 95% CI = 2.482~14.81) and 26~27 weeks (OR = 3.825; 95% CI = 1.694~8.638) were at higher risk of ROP than those with a GA of 28~29 weeks. Other factors - including sex, sepsis, BPD, and delivery mode - did not carry significant risk. CONCLUSION: Aerosol therapy with pure oxygen delivery is associated with ROP. Clinicians should exercise great caution when conducting aerosol therapy with excess oxygen in mechanically ventilated preterm infants.


Subject(s)
Bronchopulmonary Dysplasia/chemically induced , Infant, Premature , Oxygen Inhalation Therapy/adverse effects , Oxygen Inhalation Therapy/methods , Retinopathy of Prematurity/chemically induced , Birth Weight , Female , Gestational Age , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Logistic Models , Male , Multivariate Analysis , Respiration, Artificial , Retrospective Studies , Risk Factors , Taiwan
7.
J Cell Physiol ; 233(10): 7101-7112, 2018 10.
Article in English | MEDLINE | ID: mdl-29741761

ABSTRACT

The aim of this study was to identify and compare the peptidomic profiles of lung tissues from neonatal mice with and without bronchopulmonary dysplasia (BPD). Hyperoxia was used to establish the BPD mouse model. Lung tissues obtained on postnatal day (PND) 9 were processed for analysis via histological staining and label-free liquid chromatography-mass spectrometry (LC-MS/MS). Histological analysis of the lung sections from the BPD group showed significant alveolar simplification and aberrant pulmonary vascularization. We identified 3,704 total peptides, of which 63 were differentially expressed in the lung tissues from the BPD group compared with those from the control group. Within this subset, 31 peptides were downregulated, and 32 peptides were upregulated. Bioinformatics analysis suggested several potential roles of the differentially expressed peptides in the pathophysiological process of BPD. In summary, this study highlights novel peptide candidates, and provides new insights for further understanding the molecular mechanism of BPD development.


Subject(s)
Bronchopulmonary Dysplasia/physiopathology , Hyperoxia/physiopathology , Lung/physiopathology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Chromatography, Liquid/methods , Computational Biology/methods , Disease Models, Animal , Female , Humans , Infant, Newborn , Lung/pathology , Mice, Inbred C57BL , Tandem Mass Spectrometry/methods , Up-Regulation
8.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L137-L143, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28912380

ABSTRACT

The halogen bromine (Br2) is used extensively in industry and stored and transported in large quantities. Its accidental or malicious release into the atmosphere has resulted in significant casualties. The pathophysiology of Br2-induced lung injury has been studied in adult animals, but the consequences of Br2 exposure to the developing lung are completely unknown. We exposed neonatal mouse littermates on postnatal day 3 (P3) to either Br2 at 400 ppm for 30 min (400/30), to Br2 at 600 ppm for 30 min (600/30), or to room air, then returned them to their dams and observed until P14. Mice exposed to Br2 had decreased survival (S) and had decreased weight (W) at P14 in the 400/30 group (S = 63.5%, W = 6.67 ± 0.08) and in the 600/30 group (S = 36.1%, W = 5.13 ± 0.67) as compared with air breathing mice (S = 100%, W = 7.96 ± 0.30). Alveolar development was impaired, as evidenced by increased mean linear intercept at P14. At P14, Br2 exposed mice also exhibited a decrease of arterial partial pressure of oxygen, decreased quasi-static lung compliance, as well as increased alpha smooth muscle actin mRNA and protein and increased mRNA for IL-1ß, IL-6, CXCL1, and TNFα. Global gene expression, evaluated by RNA sequencing and Ingenuity Pathway Analysis, revealed persistent abnormalities in gene expression profiles at P14 involving pathways of "formation of lung" and "pulmonary development." The data indicate that Br2 inhalation injury early in life results in severe lung developmental consequences, wherein persistent inflammation and global altered developmental gene expression are likely mechanistic contributors.


Subject(s)
Bromine/toxicity , Bronchopulmonary Dysplasia/pathology , Lung Injury/pathology , Prenatal Exposure Delayed Effects/pathology , Pulmonary Alveoli/pathology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/metabolism , Female , Gene Expression Regulation , Lung Injury/chemically induced , Lung Injury/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L871-L881, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29345193

ABSTRACT

Pulmonary hypertension (PH) complicating bronchopulmonary dysplasia (BPD) worsens clinical outcomes in former preterm infants. Increased serotonin (5-hydroxytryptamine, 5-HT) signaling plays a prominent role in PH pathogenesis and progression in adults. We hypothesized that increased 5-HT signaling contributes to the pathogenesis of neonatal PH, complicating BPD and neonatal lung injury. Thus, we investigated 5-HT signaling in neonatal mice exposed to bleomycin, previously demonstrated to induce PH and alveolar simplification. Newborn wild-type mice received intraperitoneal PBS, ketanserin (1 mg/kg), bleomycin (3 U/kg) or bleomycin (3 U/kg) plus ketanserin (1 mg/kg) three times weekly for 3 wk. Following treatment with bleomycin, pulmonary expression of the rate-limiting enzyme of 5-HT synthesis, tryptophan hydroxylase-1 (Tph1), was significantly increased. Bleomycin did not affect pulmonary 5-HT 2A receptor (R) expression, but did increase pulmonary gene expression of the 5-HT 2BR and serotonin transporter. Treatment with ketanserin attenuated bleomycin-induced PH (increased RVSP and RVH) and pulmonary vascular remodeling (decreased vessel density and increased muscularization of small vessels). In addition, we found that treatment with ketanserin activated pulmonary MAPK and Akt signaling in mice exposed to bleomycin. We conclude that 5-HT signaling is increased in a murine model of neonatal PH and pharmacological inhibition of the 5-HT 2AR protects against the development of PH in neonatal lung injury. We speculate this occurs through restoration of MAPK signaling and increased Akt signaling.


Subject(s)
Bronchopulmonary Dysplasia/prevention & control , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Protective Agents/pharmacology , Receptor, Serotonin, 5-HT2A/chemistry , Vascular Remodeling/drug effects , Animals , Animals, Newborn , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Ketanserin/pharmacology , Mice , Mice, Inbred C57BL , Serotonin Antagonists/pharmacology
10.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L348-L359, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29722560

ABSTRACT

Intrauterine growth restriction (IUGR) in premature newborns increases the risk for bronchopulmonary dysplasia, a chronic lung disease characterized by disrupted pulmonary angiogenesis and alveolarization. We previously showed that experimental IUGR impairs angiogenesis; however, mechanisms that impair pulmonary artery endothelial cell (PAEC) function are uncertain. The NF-κB pathway promotes vascular growth in the developing mouse lung, and we hypothesized that IUGR disrupts NF-κB-regulated proangiogenic targets in fetal PAEC. PAECs were isolated from the lungs of control fetal sheep and sheep with experimental IUGR from an established model of chronic placental insufficiency. Microarray analysis identified suppression of NF-κB signaling and significant alterations in extracellular matrix (ECM) pathways in IUGR PAEC, including decreases in collagen 4α1 and laminin α4, components of the basement membrane and putative NF-κB targets. In comparison with controls, immunostaining of active NF-κB complexes, NF-κB-DNA binding, baseline expression of NF-κB subunits p65 and p50, and LPS-mediated inducible activation of NF-κB signaling were decreased in IUGR PAEC. Although pharmacological NF-κB inhibition did not affect angiogenic function in IUGR PAEC, angiogenic function of control PAEC was reduced to a similar degree as that observed in IUGR PAEC. These data identify reductions in endothelial NF-κB signaling as central to the disrupted angiogenesis observed in IUGR, likely by impairing both intrinsic PAEC angiogenic function and NF-κB-mediated regulation of ECM components necessary for vascular development. These data further suggest that strategies that preserve endothelial NF-κB activation may be useful in lung diseases marked by disrupted angiogenesis such as IUGR.


Subject(s)
Bronchopulmonary Dysplasia , Endothelial Cells , Fetal Growth Retardation , NF-kappa B p50 Subunit/metabolism , Pulmonary Artery , Signal Transduction , Transcription Factor RelA/metabolism , Animals , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Fetal Growth Retardation/physiopathology , Lipopolysaccharides/toxicity , Pregnancy , Pulmonary Artery/embryology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Sheep
11.
Am J Physiol Heart Circ Physiol ; 315(5): H1287-H1292, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30095998

ABSTRACT

Abnormal pulmonary vascular development is a critical factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Despite the well-established sex-specific differences in the incidence of BPD, the molecular mechanism(s) behind these are not completely understood. Exposure to a high concentration of oxygen (hyperoxia) contributes to BPD and creates a profibrotic environment in the lung. Our objective was to elucidate the sex-specific differences in neonatal human pulmonary microvascular endothelial cells (HPMECs) in normoxic and hyperoxic conditions, including the propensity for endothelial-to-mesenchymal transition. HPMECs (18- to 24-wk gestation donors, 6 male donors and 5 female donors) were subjected to hyperoxia (95% O2 and 5% CO2) or normoxia (air and 5% CO2) up to 72 h. We assessed cell migration and angiogenesis at baseline. Cell proliferation, viability, and expression of endothelial (CD31) and fibroblast markers (α-smooth muscle actin) were measured upon exposure to hyperoxia. Female HPMECs had significantly higher cell migration when assessed by the wound healing assay (40.99 ± 4.4%) compared with male HPMECs (14.76 ± 3.7%) and showed greater sprouting (1710 ± 962 µm in female cells vs. 789 ± 324 in male cells) compared with male endothelial cells in normoxia. Hyperoxia exposure decreased cell viability (by 9.8% at 48 h and 11.7% at 72 h) and proliferation (by 26.7% at 72 h) markedly in male HPMECs, whereas viability was sustained in female endothelial cells. There was greater expression of α-smooth muscle actin (2.5-fold) and decreased expression (5-fold) of CD31 in male HPMECs upon exposure to hyperoxia. The results indicate that cellular sex affects response in HPMECs in normoxia and hyperoxia. NEW & NOTEWORTHY Cellular sex affects response in human neonatal pulmonary microvascular endothelial cells in normoxia and hyperoxia. Under normoxic conditions, female human neonatal pulmonary microvascular endothelial cells display greater migration and angiogenic sprouting compared with male endothelial cells. Compared with female endothelial cells, hyperoxia exposure decreased cell viability and proliferation and increased α-smooth muscle actin and decreased CD31 expression in male endothelial cells, indicating an increased endothelial-mesenchymal transition.


Subject(s)
Bronchopulmonary Dysplasia/chemically induced , Endothelial Cells/drug effects , Oxygen/toxicity , Pulmonary Artery/drug effects , Actins/metabolism , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Child, Preschool , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant , Male , Neovascularization, Physiologic/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Sex Characteristics , Sex Factors
12.
Transgenic Res ; 26(1): 165-170, 2017 02.
Article in English | MEDLINE | ID: mdl-27730498

ABSTRACT

Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth characterized by blunted post-natal lung development. BPD can be modelled in mice by exposure of newborn mouse pups to elevated oxygen levels. Little is known about the mechanisms of perturbed lung development associated with BPD. The advent of transgenic mice, where genetic rearrangements can be induced in particular cell-types at particular time-points during organogenesis, have great potential to explore the pathogenic mechanisms at play during arrested lung development. Many inducible, conditional transgenic technologies available rely on the application of the estrogen-receptor modulator, tamoxifen. While tamoxifen is well-tolerated and has been widely employed in adult mice, or in healthy developing mice; tamoxifen is not well-tolerated in combination with hyperoxia, in the most widely-used mouse model of BPD. To address this, we set out to establish a safe and effective tamoxifen dosing regimen that can be used in newborn mouse pups subjected to injurious stimuli, such as exposure to elevated levels of environmental oxygen. Our data reveal that a single intraperitoneal dose of tamoxifen of 0.2 mg applied to newborn mouse pups in 10 µl Miglyol vehicle was adequate to successfully drive Cre recombinase-mediated genome rearrangements by the fifth day of life, in a murine model of BPD. The number of recombined cells was comparable to that observed in regular tamoxifen administration protocols. These findings will be useful to investigators where tamoxifen dosing is problematic in the background of injurious stimuli and mouse models of human and veterinary disease.


Subject(s)
Bronchopulmonary Dysplasia/genetics , Integrases/genetics , Recombination, Genetic , Tamoxifen/pharmacology , Animals , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Humans , Hyperoxia/genetics , Hyperoxia/pathology , Lung/growth & development , Lung/pathology , Mice, Transgenic , Oxygen Consumption/genetics , Premature Birth/genetics , Premature Birth/pathology
13.
Pediatr Res ; 81(4): 565-571, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27842056

ABSTRACT

BACKGROUND: Premature infants are at increased risk for wheezing disorders. Clinically, these neonates experience recurrent episodes of apnea and desaturation often treated by increasing the fraction of inspired oxygen (FIO2). We developed a novel paradigm of neonatal intermittent hypoxia with subsequent hyperoxia overshoots (CIHO/E) and hypothesized that CIHO/E elicits long-term changes on pulmonary mechanics in mice. METHODS: Neonatal C57BL/6 mice received CIHO/E, which consisted of 10% O2 (1 min) followed by a transient exposure to 50% FIO2, on 10-min repeating cycles 24 h/d from birth to P7. Baseline respiratory mechanics, methacholine challenge, RT-PCR for pro and antioxidants, radial alveolar counts, and airway smooth muscle actin were assessed at P21 after 2-wk room air recovery. Control groups were mice exposed to normoxia, chronic intermittent hyperoxia (CIHE), and chronic intermittent hypoxia (CIHO). RESULTS: CIHO/E and CIHE increased airway resistance at higher doses of methacholine and decreased baseline compliance compared with normoxia mice. Lung mRNA for NOX2 was increased by CIHO/E. Radial alveolar counts and airway smooth muscle actin was not different between groups. CONCLUSION: Neonatal intermittent hypoxia/hyperoxia exposure results in long-term changes in respiratory mechanics. We speculate that recurrent desaturation with hyperoxia overshoot may increase oxidative stress and contribute to wheezing in former preterm infants.


Subject(s)
Hyperoxia/pathology , Hypoxia/pathology , Respiratory Mechanics , Respiratory System/physiopathology , Animals , Animals, Newborn , Antioxidants/chemistry , Body Weight , Bronchopulmonary Dysplasia/chemically induced , Disease Models, Animal , Female , Methacholine Chloride/chemistry , Mice , Mice, Inbred C57BL , Oxidants/chemistry , Oxidative Stress , Oxygen/chemistry , Phenotype , Pulmonary Alveoli/metabolism , Recurrence , Respiration , Time Factors
14.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L292-302, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317685

ABSTRACT

Systemically-administered bleomycin causes inflammation, arrested lung growth, and pulmonary hypertension (PHT) in the neonatal rat, similar to human infants with severe bronchopulmonary dysplasia (BPD). Leukotrienes (LTs) are inflammatory lipid mediators produced by multiple cell types in the lung. The major LTs, LTB4 and cysteinyl LTs, are suggested to contribute to BPD, but their specific roles remain largely unexplored in experimental models. We hypothesized that LTs are increased in bleomycin-induced BPD-like injury, and that inhibition of LT production would prevent inflammatory cell influx and thereby ameliorate lung injury. Rat pups were exposed to bleomycin (1 mg·kg(-1)·day(-1) ip) or vehicle (control) from postnatal days 1-14 and were treated with either zileuton (5-lipoxygenase inhibitor), montelukast (cysteinyl LT1 receptor antagonist), or SC57461A (LTA4 hydrolase inhibitor) 10 mg·kg(-1)·day(-1) ip. Bleomycin led to increased lung content of LTB4, but not cysteinyl LTs. Bleomycin-induced increases in tissue neutrophils and macrophages and lung contents of LTB4 and tumor necrosis factor-α were all prevented by treatment with zileuton. Treatment with zileuton or SC57461A also prevented the hemodynamic and structural markers of chronic PHT, including raised pulmonary vascular resistance, increased Fulton index, and arterial wall remodeling. However, neither treatment prevented impaired alveolarization or vascular hypoplasia secondary to bleomycin. Treatment with montelukast had no effect on macrophage influx, PHT, or on abnormal lung structure. We conclude that LTB4 plays a crucial role in lung inflammation and PHT in experimental BPD. Agents targeting LTB4 or LTB4-mediated signaling may have utility in infants at risk of developing BPD-associated PHT.


Subject(s)
Bronchopulmonary Dysplasia/immunology , Hypertension, Pulmonary/immunology , Leukotriene B4/physiology , Macrophages/immunology , Animals , Animals, Newborn , Bleomycin , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/metabolism , Cell Movement/immunology , Female , Gene Expression , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Rats, Sprague-Dawley
15.
Am J Physiol Lung Cell Mol Physiol ; 309(9): L1018-26, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26342089

ABSTRACT

Vitamin D [vit D; 1,25-(OH)2D] treatment improves survival and lung alveolar and vascular growth in an experimental model of bronchopulmonary dysplasia (BPD) after antenatal exposure to endotoxin (ETX). However, little is known about lung-specific 1,25-(OH)2D3 regulation during development, especially regarding maturational changes in lung-specific expression of the vitamin D receptor (VDR), 1α-hydroxylase (1α-OHase), and CYP24A1 during late gestation and the effects of antenatal ETX exposure on 1,25-(OH)2D3 metabolism in the lung. We hypothesized that vit D regulatory proteins undergo maturation regulation in the late fetal and early neonatal lung and that prenatal exposure to ETX impairs lung growth partly through abnormal endogenous vit D metabolism. Normal fetal rat lungs were harvested between embryonic day 15 and postnatal day 14. Lung homogenates were assayed for VDR, 1α-OHase, and CYP24A1 protein contents by Western blot analysis. Fetal rats were injected on embryonic day 20 with intra-amniotic ETX, ETX + 1,25-(OH)2D3, or saline and delivered 2 days later. Pulmonary artery endothelial cells (PAECs) from fetal sheep were assessed for VDR, 1α-OHase, and CYP24A1 expression after treatment with 25-(OH)D3, 1,25-(OH)2D3, ETX, ETX + 25-(OH)D3, or ETX + 1,25-(OH)2D3. We found that lung VDR, 1α-OHase, and CYP2741 protein expression dramatically increase immediately before birth (P < 0.01 vs. early fetal values). Antenatal ETX increases CYP24A1 expression (P < 0.05) and decreases VDR and 1α-OHase expression at birth (P < 0.001), but these changes are prevented with concurrent vit D treatment (P < 0.001). ETX-induced reduction of fetal PAEC growth and tube formation and lung 1α-OHase expression are prevented by vit D treatment (P < 0.001). We conclude that lung VDR, 1α-OHase, and CYP24A1 protein content markedly increase before birth and that antenatal ETX disrupts lung vit D metabolism through downregulation of VDR and increased vit D catabolic enzyme expression, including changes in developing endothelium. We speculate that endogenous vitamin D metabolism modulates normal fetal lung development and that prenatal disruption of vit D signaling may contribute to impaired postnatal lung growth at least partly through altered angiogenic signaling.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/biosynthesis , Endothelial Cells/metabolism , Endotoxins/toxicity , Gene Expression Regulation, Developmental/drug effects , Lung/embryology , Receptors, Calcitriol/biosynthesis , Animals , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/pathology , Calcifediol/metabolism , Endothelial Cells/pathology , Lung/pathology , Pulmonary Artery/embryology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Sheep , Vitamin D3 24-Hydroxylase/biosynthesis
16.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L503-10, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25595650

ABSTRACT

Arginase is an enzyme that limits substrate L-arginine bioavailability for the production of nitric oxide by the nitric oxide synthases and produces L-ornithine, which is a precursor for collagen formation and tissue remodeling. We studied the pulmonary vascular effects of arginase inhibition in an established model of repeated systemic bleomycin sulfate administration in neonatal rats that results in pulmonary hypertension and lung injury mimicking the characteristics typical of bronchopulmonary dysplasia. We report that arginase expression is increased in the lungs of bleomycin-exposed neonatal rats and that treatment with the arginase inhibitor amino-2-borono-6-hexanoic acid prevented the bleomycin-induced development of pulmonary hypertension and deposition of collagen. Arginase inhibition resulted in increased L-arginine and L-arginine bioavailability and increased pulmonary nitric oxide production. Arginase inhibition also normalized the expression of inducible nitric oxide synthase, and reduced bleomycin-induced nitrative stress while having no effect on bleomycin-induced inflammation. Our data suggest that arginase is a promising target for therapeutic interventions in neonates aimed at preventing lung vascular remodeling and pulmonary hypertension.


Subject(s)
Aminocaproates/pharmacology , Antibiotics, Antineoplastic/adverse effects , Arginase/antagonists & inhibitors , Bleomycin/adverse effects , Boron Compounds/pharmacology , Collagen/metabolism , Hypertension, Pulmonary , Lung/enzymology , Vascular Remodeling/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Arginase/metabolism , Arginine/metabolism , Bleomycin/pharmacology , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/prevention & control , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Lung/pathology , Lung Injury/chemically induced , Lung Injury/enzymology , Lung Injury/pathology , Lung Injury/prevention & control , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley
17.
Pediatr Res ; 78(6): 634-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26322414

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) worsens clinical outcomes in former preterm infants with bronchopulmonary dysplasia (BPD). Oxidant stress disrupts alveolar and vascular development in models of BPD. Bleomycin causes oxidative stress and induces BPD and PAH in neonatal rats. Disruption in the vascular endothelial growth factor (VEGF) and nitric oxide signaling pathways contributes to BPD. We hypothesized that loss of EC-SOD would worsen PAH associated with BPD in a neonatal mouse model of bleomycin-induced BPD by disrupting the VEGF/NO signaling pathway. METHODS: Neonatal wild-type mice (WT), and mice lacking EC-SOD (EC-SOD KO) received intraperitoneal bleomycin (2 units/kg) or phosphate-buffered saline (PBS) three times weekly and were evaluated at weeks 3 or 4. RESULTS: Lack of EC-SOD impaired alveolar development and resulted in PH (elevated right ventricular systolic pressures, right ventricular hypertrophy (RVH)), decreased vessel density, and increased small vessel muscularization. Exposure to bleomycin further impaired alveolar development, worsened RVH and vascular remodeling. Lack of EC-SOD and bleomycin treatment decreased lung total and phosphorylated VEGFR2 and eNOS protein expression. CONCLUSION: EC-SOD is critical in preserving normal lung development and loss of EC-SOD results in disrupted alveolar development, PAH and vascular remodeling at baseline, which is further worsened with bleomycin and associated with decreased activation of VEGFR2.


Subject(s)
Bleomycin , Bronchopulmonary Dysplasia/enzymology , Endothelial Cells/enzymology , Hypertension, Pulmonary/enzymology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/enzymology , Pulmonary Artery/enzymology , Superoxide Dismutase/deficiency , Vascular Remodeling , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/physiopathology , Endothelial Cells/pathology , Genetic Predisposition to Disease , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Phenotype , Phosphorylation , Pulmonary Alveoli/pathology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Signal Transduction , Superoxide Dismutase/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/enzymology , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right , Ventricular Pressure
18.
Am J Physiol Lung Cell Mol Physiol ; 306(5): L420-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24414254

ABSTRACT

Vitamin D (vit D) has anti-inflammatory properties and modulates lung growth, but whether vit D can prevent lung injury after exposure to antenatal inflammation is unknown. We hypothesized that early and sustained vit D treatment could improve survival and preserve lung growth in an experimental model of bronchopulmonary dysplasia induced by antenatal exposure to endotoxin (ETX). Fetal rats (E20) were exposed to ETX (10 µg), ETX + Vit D (1 ng/ml), or saline (control) via intra-amniotic (IA) injections and delivered 2 days later. Newborn pups exposed to IA ETX received daily intraperitoneal injections of vit D (1 ng/g) or saline for 14 days. Vit D treatment improved oxygen saturations (78 vs. 87%; P < 0.001) and postnatal survival (84% vs. 57%; P < 0.001) after exposure to IA ETX compared with IA ETX alone. Postnatal vit D treatment improved alveolar and vascular growth at 14 days by 45% and 25%, respectively (P < 0.05). Vit D increased fetal sheep pulmonary artery endothelial cell (PAEC) growth and tube formation by 64% and 44%, respectively (P < 0.001), and prevented ETX-induced reductions of PAEC growth and tube formation. Vit D directly increased fetal alveolar type II cell (ATIIC) growth by 26% (P < 0.001) and enhanced ATIIC growth in the presence of ETX-induced growth suppression by 73% (P < 0.001). We conclude that antenatal vit D therapy improved oxygenation and survival in newborn rat pups and enhanced late lung structure after exposure to IA ETX in vivo, which may partly be due to direct effects on vascular and alveolar growth.


Subject(s)
Bronchopulmonary Dysplasia/prevention & control , Endotoxins/toxicity , Pulmonary Alveoli/drug effects , Vitamin D/pharmacology , Vitamins/pharmacology , Amniotic Fluid/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/mortality , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , Male , Oxygen/blood , Pregnancy , Pulmonary Alveoli/cytology , Pulmonary Alveoli/growth & development , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/growth & development , Rats , Rats, Sprague-Dawley , Sheep , Survival Rate
19.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(2): 225-9, 244, 2014 Mar.
Article in Zh | MEDLINE | ID: mdl-24749345

ABSTRACT

OBJECTIVE: To investigate the protective effects and potential mechanism of curcumin on bronchopulmonary dysplasis (BPD) induced by 600 mL/L oxygen in newborn rats. METHODS: 108 Sprague-Dawley (SD) specific pathogen-free newborn rats within 6 h after birth were randomly divided into room air group (RA group), 600 mL/L oxygen group (O2 group) and 600 mL/L oxygen + Curcumin group (O2 + Cu group). Eight rats were randomly taken from each group and killed at 4, 7 and 14 d, respectively, after the treatment, and their lung tissues were incised for HE staining. The expressions of IL-6, IL-10 in serum and lung tissue were detected by ELISA; and the protein expression of IGF-I was measured by immunohistochemical method. RESULTS Comparing with the RA group, we found that newborn rats exposed to 600 mL/L oxygen develop a heterogeneous parenchymal lung injury with areas of arrested alveolarization and growth mixed with areas of interstitial thinning, meanwhile, both the expression of IL-6 and IL-10 in serum and lung tissue increased significantly (P < 0.05). However, in O2 + Cu group, IL-6 expression was decreased in different degrees while IL-10 increased significantly (P < 0.05). The highest expression level of IGF-I in RA group were much higher from 4 d to 7 d (alveolar development period) but significantly lower in 14 d. There was a positive correlation between IGF-I and alveolar development. In comparison with RA group, the expression levels of IGF-I in O2 group were significantly lower in 4 d and 7 d but were significantly higher in 14 d (P < 0.05); In comparison with O2 group, the expression levels of IGF-I in O2 group significantly increased in 4 d and 7 d but significantly reduced in 14 d (P < 0.05). CONCLUSION: Curcumin may partly prevent the lung injury induced by prolonged hyperoxia exposure in neonatal rats probably via modulating the expressions of IL-6, IL-10 and IGF-I in serum and lung tissue.


Subject(s)
Bronchopulmonary Dysplasia/drug therapy , Curcumin/pharmacology , Hyperoxia/pathology , Oxygen/adverse effects , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/chemically induced , Insulin-Like Growth Factor I/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/drug therapy , Rats , Rats, Sprague-Dawley
20.
Am J Physiol Lung Cell Mol Physiol ; 305(1): L73-81, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23666751

ABSTRACT

Late-outgrowth endothelial colony-forming cells (ECFCs), a type of circulating endothelial progenitor cell (EPC), may contribute to pulmonary angiogenesis during development. Cord blood ECFCs from preterm newborns proliferate more rapidly than term ECFCs but are more susceptible to the adverse effects of hyperoxia. Recent studies suggest that bone marrow-derived EPCs protect against experimental lung injury via paracrine mechanisms independent of vascular engraftment. To determine whether human umbilical cord blood ECFCs from preterm and term newborns have therapeutic benefit in experimental neonatal lung injury, we isolated cord blood ECFCs from full-term and preterm newborns and prepared ECFC-conditioned medium (CM) to test its therapeutic benefit on fetal pulmonary artery endothelial cell (PAEC) proliferation and function as well as alveolar type 2 (AT2) cell growth. PAECs and AT2 cells were isolated from late-gestation fetal sheep. Additionally, we administered both ECFCs and ECFC-CM to bleomycin-exposed newborn rats, an experimental model of bronchopulmonary dysplasia (BPD). Both term ECFC-CM and preterm ECFC-CM promoted cell growth and angiogenesis in vitro. However, when ECFC-CM was collected during exposure to mild hyperoxia, the benefit of preterm ECFC-CM was no longer observed. In the bleomycin model of BPD, treatment with ECFC-CM (or CM from mature EC) effectively decreased right ventricular hypertrophy but had no effect on alveolar septation. We conclude that term ECFC-CM is beneficial both in vitro and in experimental BPD. During oxidative stress, preterm ECFC-CM, but not term ECFC-CM, loses its benefit. The inability of term ECFC-CM to promote alveolarization may limit its therapeutic potential.


Subject(s)
Bleomycin/toxicity , Bronchopulmonary Dysplasia/complications , Culture Media, Conditioned/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Hypertension, Pulmonary/prevention & control , Neovascularization, Physiologic , Animals , Animals, Newborn , Antibiotics, Antineoplastic/toxicity , Blotting, Western , Bronchopulmonary Dysplasia/chemically induced , Cell Proliferation , Cells, Cultured , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Hyperoxia , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Immunoenzyme Techniques , Infant, Newborn , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL