1
|
Roeder F, Knudsen L, Schmiedl A. The expression of the surfactant proteins SP-A and SP-B during postnatal alveolarization of the rat lung. PLoS One 2024; 19:e0297889. [PMID: 38483982 PMCID: PMC10939297 DOI: 10.1371/journal.pone.0297889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/03/2024] [Indexed: 03/17/2024] Open
Abstract
OBJECTIVE Surfactant-specific proteins (SP) are responsible for the functional and structural integrity as well as for the stabilization of the intra-alveolar surfactant. Morphological lung maturation starts in rat lungs after birth. The aim of this study was to investigate whether the expression of the hydrophilic SP-A and the hydrophobic SP-B is associated with characteristic postnatal changes characterizing morphological lung maturation. METHODS Stereological methods were performed on the light microscope. Using immunohistochemical and molecular biological methods (Western Blot, RT-qPCR), the SP-A and SP-B of adult rat lungs and of those with different postnatal developmental stages (3, 7, 14 and 21 days after birth) were characterized. RESULTS As signs of alveolarization the total septal surface and volume increased and the septal thickness decreased. The significantly highest relative surface fraction of SP-A labeled alveolar epithelial cells type II (AEII) was found together with the highest relative SP-A gene expression before the alveolarization (3th postnatal day). With the downregulation of SP-A gene expression during and after alveolarization (between postnatal days 7 and 14), the surface fraction of the SP-A labeled AEII also decreased, so they are lowest in adult animals. The surface fraction of SP-B labeled AEII and the SP-B gene expression showed the significantly highest levels in adults, the protein expression increased also significantly at the end of morphological lung maturation. There were no alterations in the SP-B expression before and during alveolarization until postnatal day 14. The protein expression as well as the gene expression of SP-A and SP-B correlated very well with the total surface of alveolar septa independent of the postnatal age. CONCLUSION The expression of SP-A and SP-B is differentially associated with morphological lung maturation and correlates with increased septation of alveoli as indirect clue for alveolarization.
Collapse
Affiliation(s)
- Franziska Roeder
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Andreas Schmiedl
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
2
|
Kumar P, Yadav A, Anand P, Debata P. Predictors of CPAP failure with RAM cannula interface for primary respiratory support in preterm neonates. Med J Armed Forces India 2024; 80:60-67. [PMID: 38261886 PMCID: PMC10793224 DOI: 10.1016/j.mjafi.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
Background RAM cannula is used as interface for delivering nasal continuous positive airway pressure (nCPAP) in many neonatal care units though the bench to bedside evidence for its use in clinical settings in lacking. Methods In this prospective cohort study from tertiary care neonatal care unit, the primary objective was to determine the rate of CPAP failure using RAM interface, within 72 h of starting of therapy and secondary objective was to look for the incidence and severity of nasal trauma with the use of RAM interface for CPAP delivery. All inborn preterm neonates between 28 and 32 weeks of gestation and weight more than 1000 g who required non-invasive respiratory support within 6 h of life were included in the study. Results The enroled cohort of 250 neonates had mean gestational age of 30.73 ± 1.32 weeks and mean birth weight of 1410 ± 210 g. CPAP failure rate with the use of RAM interface was 31.2% (78/250) in the overall cohort. The failure rate was higher in subgroup of gestation between 28 and 30 week (52.1%) in comparison with the 31-32-week gestation (18.2%; p-value = 0.0001). Nasal trauma was present in 36 (14%) neonates; of whom 33 (91%) had mild and 2 (6%) and 1 (3%) had moderate and severe, respectively. On multivariate analysis, birth weight less than 1250 g, incomplete antenatal steroids, need for PEEP more than 5 cm & FiO2 > 30% at onset of distress and administration of surfactant were found be statistically significantly associated with CPAP failure with RAM interface. Conclusion The clinical outcomes with the use of RAM interface for CPAP administration are comparable to those with other interfaces with lesser incidence of nasal injury. The results of this study warrant future randomized trials to compare different CPAP interfaces for clinical outcomes and nasal injuries from the developing countries.
Collapse
Affiliation(s)
- Pradeep Kumar
- Senior Resident (Pediatrics), Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Anita Yadav
- Associate Professor (Pediatrics), Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Pratima Anand
- Chief Medical Officer (Pediatrics), Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Pradeep Debata
- Professor (Pediatrics), Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
3
|
Bartman CM, Schiliro M, Nesbitt L, Lee KK, Prakash YS, Pabelick CM. Exogenous hydrogen sulfide attenuates hyperoxia effects on neonatal mouse airways. Am J Physiol Lung Cell Mol Physiol 2024; 326:L52-L64. [PMID: 37987780 PMCID: PMC11279744 DOI: 10.1152/ajplung.00196.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Supplemental O2 remains a necessary intervention for many premature infants (<34 wk gestation). Even moderate hyperoxia (<60% O2) poses a risk for subsequent airway disease, thereby predisposing premature infants to pediatric asthma involving chronic inflammation, airway hyperresponsiveness (AHR), airway remodeling, and airflow obstruction. Moderate hyperoxia promotes AHR via effects on airway smooth muscle (ASM), a cell type that also contributes to impaired bronchodilation and remodeling (proliferation, altered extracellular matrix). Understanding mechanisms by which O2 initiates long-term airway changes in prematurity is critical for therapeutic advancements for wheezing disorders and asthma in babies and children. Immature or dysfunctional antioxidant systems in the underdeveloped lungs of premature infants thereby heightens susceptibility to oxidative stress from O2. The novel gasotransmitter hydrogen sulfide (H2S) is involved in antioxidant defense and has vasodilatory effects with oxidative stress. We previously showed that exogenous H2S exhibits bronchodilatory effects in human developing airway in the context of hyperoxia exposure. Here, we proposed that exogenous H2S would attenuate effects of O2 on airway contractility, thickness, and remodeling in mice exposed to hyperoxia during the neonatal period. Using functional [flexiVent; precision-cut lung slices (PCLS)] and structural (histology; immunofluorescence) analyses, we show that H2S donors mitigate the effects of O2 on developing airway structure and function, with moderate O2 and H2S effects on developing mouse airways showing a sex difference. Our study demonstrates the potential applicability of low-dose H2S toward alleviating the detrimental effects of hyperoxia on the premature lung.NEW & NOTEWORTHY Chronic airway disease is a short- and long-term consequence of premature birth. Understanding effects of O2 exposure during the perinatal period is key to identify targetable mechanisms that initiate and sustain adverse airway changes. Our findings show a beneficial effect of exogenous H2S on developing mouse airway structure and function with notable sex differences. H2S donors alleviate effects of O2 on airway hyperreactivity, contractility, airway smooth muscle thickness, and extracellular matrix deposition.
Collapse
Affiliation(s)
- Colleen M Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Marta Schiliro
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Lisa Nesbitt
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Kenge K Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
4
|
Zhang EY, Bartman CM, Prakash YS, Pabelick CM, Vogel ER. Oxygen and mechanical stretch in the developing lung: risk factors for neonatal and pediatric lung disease. Front Med (Lausanne) 2023; 10:1214108. [PMID: 37404808 PMCID: PMC10315587 DOI: 10.3389/fmed.2023.1214108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Chronic airway diseases, such as wheezing and asthma, remain significant sources of morbidity and mortality in the pediatric population. This is especially true for preterm infants who are impacted both by immature pulmonary development as well as disproportionate exposure to perinatal insults that may increase the risk of developing airway disease. Chronic pediatric airway disease is characterized by alterations in airway structure (remodeling) and function (increased airway hyperresponsiveness), similar to adult asthma. One of the most common perinatal risk factors for development of airway disease is respiratory support in the form of supplemental oxygen, mechanical ventilation, and/or CPAP. While clinical practice currently seeks to minimize oxygen exposure to decrease the risk of bronchopulmonary dysplasia (BPD), there is mounting evidence that lower levels of oxygen may carry risk for development of chronic airway, rather than alveolar disease. In addition, stretch exposure due to mechanical ventilation or CPAP may also play a role in development of chronic airway disease. Here, we summarize the current knowledge of the impact of perinatal oxygen and mechanical respiratory support on the development of chronic pediatric lung disease, with particular focus on pediatric airway disease. We further highlight mechanisms that could be explored as potential targets for novel therapies in the pediatric population.
Collapse
Affiliation(s)
- Emily Y. Zhang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Colleen M. Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth R. Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Huang P, Qin X, Fan C, Wang M, Chen F, Liao M, Zhong H, Wang H, Ma L. Comparison of Biological Characteristics of Human Umbilical Cord Wharton's Jelly-Derived Mesenchymal Stem Cells from Extremely Preterm and Term Infants. Tissue Eng Regen Med 2023:10.1007/s13770-023-00538-9. [PMID: 37249837 DOI: 10.1007/s13770-023-00538-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/21/2023] [Accepted: 03/14/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Despite the progress in perinatal-neonatal medicine, complications of extremely preterm infants continue to constitute the major adverse outcomes in neonatal intensive care unit. Human umbilical cord Wharton's Jelly-derived mesenchymal stem cells (HUMSCs) may offer new hope for the treatment of intractable neonatal disorders. This study will explore the functional differences of HUMSCs between extremely preterm and term infants. METHODS UMSCs from 5 extremely preterm infants(weeks of gestation: 22+5 w,24+4 w,25+3 w,26 w,28 w) and 2 term infants(39 w,39+2 w) were isolated, and mesenchymal markers, pluripotent genes, proliferation rate were analyzed. HUVECs were injured by treated with LPS and repaired by co-cultured with HUMSCs of different gestational ages. RESULTS All HUMSCs showed fibroblast-like adherence to plastic and positively expressed surface marker of CD105,CD73 and CD90, but did not expressed CD45,CD34,CD14,CD79a and HLA-DR; HUMSCs in extremely preterm exhibited significant increase in proliferation as evidenced by CCK8, pluripotency markers OCT-4 tested by RT-PCR also showed increase. Above all, in LPS induced co-cultured inflame systerm, HUMSCs in extremely preterm were more capable to promote wound healing and tube formation in HUVEC cultures, they promoted TGFβ1 expression and inhibited IL6 expression. CONCLUSIONS Our results suggest that HUMSCs from extremely preterm infants may be more suitable as candidates in cell therapy for the preterm infants.
Collapse
Affiliation(s)
- Peng Huang
- Shenzhen Children's Hospital of China Medical University, Shenzhen, 518038, China
- Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Xiaofei Qin
- Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Chuiqin Fan
- Shenzhen Children's Hospital of China Medical University, Shenzhen, 518038, China
| | - Manna Wang
- Department of Pediatrics, The Women and Children's Medical Hospital of Guangzhou Medical University, The Third Affifiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Fuyi Chen
- Department of Pediatrics, The Women and Children's Medical Hospital of Guangzhou Medical University, The Third Affifiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Maochuan Liao
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Huifeng Zhong
- Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Hongwu Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Lian Ma
- Shenzhen Children's Hospital of China Medical University, Shenzhen, 518038, China.
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Department of Pediatrics, The Women and Children's Medical Hospital of Guangzhou Medical University, The Third Affifiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
6
|
Huang LT, Chou HC, Chen CM. Roxadustat attenuates hyperoxia-induced lung injury by upregulating proangiogenic factors in newborn mice. Pediatr Neonatol 2021; 62:369-378. [PMID: 33865748 DOI: 10.1016/j.pedneo.2021.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/28/2021] [Accepted: 03/17/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Premature infants who require oxygen therapy for respiratory distress syndrome often develop bronchopulmonary dysplasia, a chronic lung disease characterized by interrupted alveologenesis. Disrupted angiogenesis inhibits alveologenesis; however, the mechanisms through which disrupted angiogenesis affects lung development are poorly understood. Hypoxia-inducible factors (HIFs) are transcription factors that activate multiple oxygen-sensitive genes, including those encoding for vascular endothelial growth factor (VEGF). However, the HIF modulation of angiogenesis in hyperoxia-induced lung injury is not fully understood. Therefore, we explored the effects of roxadustat, an HIF stabilizer that has been shown to promote angiogenesis, in regulating pulmonary angiogenesis on hyperoxia exposure. METHODS C57BL6 mice pups reared in room air and 85% O2 were injected with phosphate-buffered saline or 5 mg/kg or 10 mg/kg roxadustat. Their daily body weight and survival rate were recorded. Their lungs were excised for histology and angiogenic factor expression analyses on postnatal Day 7. RESULTS Exposure to neonatal hyperoxia reduced body weight; survival rate; and expressions of von Willebrand factor, HIF-1α, phosphor mammalian target of rapamycin, VEGF, and endothelial nitric oxide synthase and increased the mean linear intercept values in the pups. Roxadustat administration reversed these effects. CONCLUSION Hyperoxia suppressed pulmonary vascular development and the expression of proangiogenic factors. Roxadustat promoted pulmonary angiogenesis on hyperoxia exposure by stabilizing HIF-1α and upregulating the expression of proangiogenic factors, indicating its potential in clinical and therapeutic applications.
Collapse
Affiliation(s)
- Liang-Ti Huang
- Department of Pediatrics, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
7
|
Wickramasinghe LC, van Wijngaarden P, Johnson C, Tsantikos E, Hibbs ML. An Experimental Model of Bronchopulmonary Dysplasia Features Long-Term Retinal and Pulmonary Defects but Not Sustained Lung Inflammation. Front Pediatr 2021; 9:689699. [PMID: 34527643 PMCID: PMC8435611 DOI: 10.3389/fped.2021.689699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/06/2021] [Indexed: 11/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe lung disease that affects preterm infants receiving oxygen therapy. No standardized, clinically-relevant BPD model exists, hampering efforts to understand and treat this disease. This study aimed to evaluate and confirm a candidate model of acute and chronic BPD, based on exposure of neonatal mice to a high oxygen environment during key lung developmental stages affected in preterm infants with BPD. Neonatal C57BL/6 mouse pups were exposed to 75% oxygen from postnatal day (PN)-1 for 5, 8, or 14 days, and their lungs were examined at PN14 and PN40. While all mice showed some degree of lung damage, mice exposed to hyperoxia for 8 or 14 days exhibited the greatest septal wall thickening and airspace enlargement. Furthermore, when assessed at PN40, mice exposed for 8 or 14 days to supplemental oxygen exhibited augmented septal wall thickness and emphysema, with the severity increased with the longer exposure, which translated into a decline in respiratory function at PN80 in the 14-day model. In addition to this, mice exposed to hyperoxia for 8 days showed significant expansion of alveolar epithelial type II cells as well as the greatest fibrosis when assessed at PN40 suggesting a healing response, which was not seen in mice exposed to high oxygen for a longer period. While evidence of lung inflammation was apparent at PN14, chronic inflammation was absent from all three models. Finally, exposure to high oxygen for 14 days also induced concurrent outer retinal degeneration. This study shows that early postnatal exposure to high oxygen generates hallmark acute and chronic pathologies in mice that highlights its use as a translational model of BPD.
Collapse
Affiliation(s)
- Lakshanie C Wickramasinghe
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Peter van Wijngaarden
- Department of Surgery - Ophthalmology, University of Melbourne, Melbourne, VIC, Australia.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Chad Johnson
- Monash Micro Imaging, Alfred Research Alliance, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Lung and Eye Disease Develop Concurrently in Supplemental Oxygen-Exposed Neonatal Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1801-1812. [PMID: 32526165 DOI: 10.1016/j.ajpath.2020.05.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 11/23/2022]
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are two debilitating disorders that develop in preterm infants exposed to supplemental oxygen to prevent respiratory failure. Both can lead to lifelong disabilities, such as chronic obstructive pulmonary disease and vision loss. Due to the lack of a standard experimental model of coincident disease, the underlying associations between BPD and ROP are not well characterized. To address this gap, we used the robust mouse model of oxygen-induced retinopathy exposing C57BL/6 mice to 75% oxygen from postnatal day 7 to 12. The cardinal features of ROP were replicated by this strategy, and the lungs of the same mice were simultaneously examined for evidence of BPD-like lung injury, investigating both the short- and long-term effects of early-life supplemental oxygen exposure. At postnatal days 12 and 18, mild lung disease was evident by histopathologic analysis together with the expected vasculopathy in the inner retina. At later time points, the lung lesion had progressed to severe airspace enlargement and alveolar simplification, with concurrent thinning in the outer layer of the retina. In addition, critical angiogenic oxidative stress and inflammatory factors reported to be dysregulated in ROP were similarly impaired in the lungs. These data shed new light on the interconnectedness of these two neonatal disorders, holding potential for the discovery of novel targets to treat BPD and ROP.
Collapse
|
9
|
Cheah FC, Lai CH, Tan GC, Swaminathan A, Wong KK, Wong YP, Tan TL. Intrauterine Gardnerella vaginalis Infection Results in Fetal Growth Restriction and Alveolar Septal Hypertrophy in a Rabbit Model. Front Pediatr 2020; 8:593802. [PMID: 33553066 PMCID: PMC7862757 DOI: 10.3389/fped.2020.593802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/16/2020] [Indexed: 11/15/2022] Open
Abstract
Background: Gardnerella vaginalis (GV) is most frequently associated with bacterial vaginosis and is the second most common etiology causing intrauterine infection after Ureaplasma urealyticum. Intrauterine GV infection adversely affects pregnancy outcomes, resulting in preterm birth, fetal growth restriction, and neonatal pneumonia. The knowledge of how GV exerts its effects is limited. We developed an in vivo animal model to study its effects on fetal development. Materials and Methods: A survival mini-laparotomy was conducted on New Zealand rabbits on gestational day 21 (28 weeks of human pregnancy). In each dam, fetuses in the right uterine horn received intra-amniotic 0.5 × 102 colony-forming units of GV injections each, while their littermate controls in the left horn received sterile saline injections. A second laparotomy was performed seven days later. Assessment of the fetal pups, histopathology of the placenta and histomorphometric examination of the fetal lung tissues was done. Results: Three dams with a combined total of 12 fetuses were exposed to intra-amniotic GV, and 9 fetuses were unexposed. The weights of fetuses, placenta, and fetal lung were significantly lower in the GV group than the saline-inoculated control group [mean gross weight, GV (19.8 ± 3.8 g) vs. control (27.9 ± 1.7 g), p < 0.001; mean placenta weight, GV (5.5 ± 1.0 g) vs. control (6.5 ± 0.7 g), p = 0.027; mean fetal lung weight, GV (0.59 ± 0.11 g) vs. control (0.91 ± 0.08 g), p = 0.002. There was a two-fold increase in the multinucleated syncytiotrophoblasts in the placenta of the GV group than their littermate controls (82.9 ± 14.9 vs. 41.6 ± 13.4, p < 0.001). The mean alveolar septae of GV fetuses was significantly thicker than the control (14.8 ± 2.8 μm vs. 12.4 ± 3.8 μm, p = 0.007). Correspondingly, the proliferative index in the interalveolar septum was 1.8-fold higher in the GV group than controls (24.9 ± 6.6% vs. 14.2 ± 2.9%, p = 0.011). The number of alveoli and alveolar surface area did not vary between groups. Discussion: Low-dose intra-amniotic GV injection induces fetal growth restriction, increased placental multinucleated syncytiotrophoblasts and fetal lung re-modeling characterized by alveolar septal hypertrophy with cellular proliferative changes. Conclusion: This intra-amniotic model could be utilized in future studies to elucidate the acute and chronic effects of GV intrauterine infections.
Collapse
Affiliation(s)
- Fook-Choe Cheah
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Chee Hoe Lai
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Anushia Swaminathan
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Kon Ken Wong
- Department of Microbiology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Tian-Lee Tan
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Pasha AB, Chen XQ, Zhou GP. Bronchopulmonary dysplasia: Pathogenesis and treatment. Exp Ther Med 2018; 16:4315-4321. [PMID: 30542380 PMCID: PMC6257511 DOI: 10.3892/etm.2018.6780] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/06/2018] [Indexed: 11/06/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a form of chronic lung disease of infancy, which mostly affects premature infants with significant morbidity and mortality. Premature infants who require to be treated for conditions including respiratory distress syndrome have a higher risk of developing BPD. In spite of the improvement in clinical methods, the incidence of BPD has not reduced. In the present review, the pathogenesis of BPD is described along with the treatments available at present and the role of nursing in the management of BPD. Emerging preventive therapies for BPD are also discussed, including the use of recombinant human superoxide dismutase, which has been proven effective in reducing respiratory injury and its long-term effects.
Collapse
Affiliation(s)
- Asfia Banu Pasha
- Department of Pediatrics, The First Affiliated Hospital, School of International Education, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiao-Qing Chen
- Department of Pediatrics, The First Affiliated Hospital, School of International Education, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital, School of International Education, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
11
|
Abstract
Over 50 years after its first description, Bronchopulmonary Dysplasia (BPD) remains a devastating pulmonary complication in preterm infants with respiratory failure and develops in 30-50% of infants less than 1000-gram birth weight. It is thought to involve ventilator- and oxygen-induced damage to an immature lung that results in an inflammatory response and ends in aberrant lung development with dysregulated angiogenesis and alveolarization. Significant morbidity and mortality are associated with this most common chronic lung disease of childhood. Thus, any therapies that decrease the incidence or severity of this condition would have significant impact on morbidity, mortality, human costs, and healthcare expenditure. It is clear that an inflammatory response and the elaboration of growth factors and cytokines are associated with the development of BPD. Numerous approaches to control the inflammatory process leading to the development of BPD have been attempted. This review will examine the anti-inflammatory approaches that are established or hold promise for the prevention or treatment of BPD.
Collapse
Affiliation(s)
- Rashmin C Savani
- Center for Pulmonary & Vascular Biology, Division of Neonatal-Perinatal Medicine, The Department of Pediatrics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063, USA.
| |
Collapse
|
12
|
Preeclampsia predicts higher incidence of bronchopulmonary dysplasia. J Perinatol 2018; 38:1165-1173. [PMID: 29808003 DOI: 10.1038/s41372-018-0133-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/05/2018] [Accepted: 04/16/2018] [Indexed: 11/09/2022]
Abstract
INTRODUCTION It is not known whether very preterm infants born to preeclamptic women have worse outcomes than those delivered preterm for other causes. OBJECTIVE We assessed the association between preeclampsia (PE) and the neonatal morbidity and mortality of very preterm infants. METHODS Over 2015 and 2016, 11 collaborating Portuguese level III NICUs prospectively enrolled a cohort of mothers with or without PE who delivered liveborn premature infants between 24 and 30 completed weeks of gestation. Data on neonatal morbidities were collected and their association to PE was assessed. RESULTS The final cohort consisted of 410 mothers who delivered 494 preterm infants. Infants from PE mothers weighed less than those of non-PE mothers (819 ± 207 g vs. 989 ± 256 g, p < 0.0001). Incidences of respiratory distress syndrome, patent ductus arteriosus, early and nosocomial sepsis, necrotizing enterocolitis, pneumonia, meningitis, retinopathy of prematurity, intraventricular hemorrhage, periventricular infarction, periventricular leukomalacia, and mortality did not differ significantly between infants of PE or non-PE mothers. Incidence of bronchopulmonary dysplasia (BPD-defined as oxygen dependency at 36 weeks) was higher in PE infants compared with non-PE infants by both univariate and multivariate logistic regression (p = 0.007). CONCLUSION We conclude that, when controlling for gestational age, maternal PE results in higher incidence of only BPD among preterm Portuguese infants.
Collapse
|
13
|
Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Exp Mol Med 2018; 50:1-12. [PMID: 29650962 PMCID: PMC5938045 DOI: 10.1038/s12276-018-0055-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
We previously reported the role of vascular endothelial growth factor (VEGF) secreted by mesenchymal stem cells (MSCs) in protecting against neonatal hyperoxic lung injuries. Recently, the paracrine protective effect of MSCs was reported to be primarily mediated by extracellular vesicle (EV) secretion. However, the therapeutic efficacy of MSC-derived EVs and the role of the VEGF contained within EVs in neonatal hyperoxic lung injury have not been elucidated. The aim of the study was to determine whether MSC-derived EVs attenuate neonatal hyperoxic lung injury and, if so, whether this protection is mediated via the transfer of VEGF. We compared the therapeutic efficacy of MSCs, MSC-derived EVs with or without VEGF knockdown, and fibroblast-derived EVs in vitro with a rat lung epithelial cell line challenged with H2O2 and in vivo with newborn Sprague-Dawley rats exposed to hyperoxia (90%) for 14 days. MSCs (1 × 105 cells) or EVs (20 µg) were administered intratracheally on postnatal day 5. The MSCs and MSC-derived EVs, but not the EVs derived from VEGF-knockdown MSCs or fibroblasts, attenuated the in vitro H2O2-induced L2 cell death and the in vivo hyperoxic lung injuries, such as impaired alveolarization and angiogenesis, increased cell death, and activated macrophages and proinflammatory cytokines. PKH67-stained EVs were internalized into vascular pericytes (22.7%), macrophages (21.3%), type 2 epithelial cells (19.5%), and fibroblasts (4.4%) but not into vascular endothelial cells. MSC-derived EVs are as effective as parental MSCs for attenuating neonatal hyperoxic lung injuries, and this protection was mediated primarily by the transfer of VEGF.
Collapse
|
14
|
Pabelick CM, Thompson MA, Britt RD. Effects of Hyperoxia on the Developing Airway and Pulmonary Vasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:179-194. [PMID: 29047087 DOI: 10.1007/978-3-319-63245-2_11] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although it is necessary and part of standard practice, supplemental oxygen (40-90% O2) or hyperoxia is a significant contributing factor to development of bronchopulmonary dysplasia, persistent pulmonary hypertension, recurrent wheezing, and asthma in preterm infants. This chapter discusses hyperoxia and the role of redox signaling in the context of neonatal lung growth and disease. Here, we discuss how hyperoxia promotes dysfunction in the airway and the known redox-mediated mechanisms that are important for postnatal vascular and alveolar development. Whether in the airway or alveoli, redox pathways are important and greatly influence the neonatal lung.
Collapse
Affiliation(s)
- Christina M Pabelick
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA. .,Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA.
| | - Michael A Thompson
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| | - Rodney D Britt
- Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| |
Collapse
|
15
|
Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatr Res 2018; 83:214-222. [PMID: 28972960 DOI: 10.1038/pr.2017.249] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation represents the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies, including bronchopulmonary dysplasia, hypoxic ischemic encephalopathy, and intraventricular hemorrhage. Absent engraftment and direct differentiation of transplanted MSCs, and the "hit-and-run" therapeutic effects of these MSCs suggest that their pleiotropic protection might be attributable to paracrine activity via the secretion of various biologic factors rather than to regenerative activity. The transplanted MSCs, therefore, exert their therapeutic effects not by acting as "stem cells," but rather by acting as "paracrine factors factory." The MSCs sense the microenvironment of the injury site and secrete various paracrine factors that serve several reparative functions, including antiapoptotic, anti-inflammatory, antioxidative, antifibrotic, and/or antibacterial effects in response to environmental cues to enhance regeneration of the damaged tissue. Therefore, the therapeutic efficacy of MSCs might be dependent on their paracrine potency. In this review, we focus on recent investigations that elucidate the specifically regulated paracrine mechanisms of MSCs by injury type and discuss potential strategies to enhance paracrine potency, and thus therapeutic efficacy, of transplanted MSCs, including determining the appropriate source and preconditioning strategy for MSCs and the route and timing of their administration.
Collapse
|
16
|
Hou C, Peng D, Gao L, Tian D, Dai J, Luo Z, Liu E, Chen H, Zou L, Fu Z. Human umbilical cord-derived mesenchymal stem cells protect from hyperoxic lung injury by ameliorating aberrant elastin remodeling in the lung of O 2-exposed newborn rat. Biochem Biophys Res Commun 2017; 495:1972-1979. [PMID: 29242152 DOI: 10.1016/j.bbrc.2017.12.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 12/10/2017] [Indexed: 01/08/2023]
Abstract
The incidence and mortality rates of bronchopulmonary dysplasia (BPD) remain very high. Therefore, novel therapies are imminently needed to improve the outcome of this disease. Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) show promising therapeutic effects on oxygen-induced model of BPD. In our experiment, UC-MSCs were intratracheally delivered into the newborn rats exposed to hyperoxia, a well-established BPD model. This study demonstrated that UC-MSCs reduce elastin expression stimulated by 90% O2 in human lung fibroblasts-a (HLF-a), and inhibit HLF-a transdifferentiation into myofibroblasts. In addition, the therapeutic effects of UC-MSCs in neonatal rats with BPD, UC-MSCs could inhibit lung elastase activity and reduce aberrant elastin expression and deposition in the lung of BPD rats. Overall, this study suggested that UC-MSCs could ameliorate aberrant elastin expression in the lung of hyperoxia-induced BPD model which may be associated with suppressing increased TGFβ1 activation.
Collapse
Affiliation(s)
- Chen Hou
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Chongqing Key Laboratory of Pediatrics, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, China
| | - Danyi Peng
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Key Laboratory of Pediatrics, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, China
| | - Li Gao
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Otorhinolaryngology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, China
| | - Daiyin Tian
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jihong Dai
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhengxiu Luo
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Enmei Liu
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hong Chen
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Chongqing Key Laboratory of Pediatrics, China; Department of Pediatrics, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Lin Zou
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, China.
| | - Zhou Fu
- Pediatrics Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China; Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, China.
| |
Collapse
|
17
|
Shima Y, Kumasaka S, Nishimaki S. Urinary β2-microglobulin and bronchopulmonary dysplasia: Trends in preterm infants. Pediatr Int 2017; 59:1169-1173. [PMID: 28833913 DOI: 10.1111/ped.13407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/06/2017] [Accepted: 08/14/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND The developmental process of bronchopulmonary dysplasia (BPD) is not identical between very preterm infants born small for gestational age (SGA) and those born appropriate for gestational age (AGA). In this study, we compared the pattern of the inflammatory response in infants of each group, by measuring urinary β2-microglobulin (Uβ2M) as an alternative, concise, and less-invasive biomarker. METHODS Uβ2M and clinical details were examined at birth and at 4 weeks of age in 146 very preterm infants. RESULTS Of the 57 infants diagnosed with BPD, 18 were SGA, and 39 were AGA. Uβ2M at birth was significantly lower in SGA BPD infants than in AGA BPD infants, but it increased with time. The prevalence of chorioamnionitis (CAM) was significantly lower in SGA BPD infants than in AGA BPD infants, while that of pregnancy-induced hypertension was the opposite. CONCLUSIONS Exposure to prenatal factors other than CAM may sensitize fetal lungs to become vulnerable to postnatal inflammation in very preterm SGA infants with BPD.
Collapse
Affiliation(s)
- Yoshio Shima
- Department of Neonatal Medicine, Nippon Medical School, Musashikosugi Hospital, Kanagawa, Japan.,Department of Pediatrics, Japanese Red Cross Maternity Hospital, Tokyo, Japan
| | - Sakae Kumasaka
- Department of Pediatrics, Japanese Red Cross Maternity Hospital, Tokyo, Japan
| | | |
Collapse
|
18
|
Schanton M, Maymó JL, Pérez-Pérez A, Sánchez-Margalet V, Varone CL. Involvement of leptin in the molecular physiology of the placenta. Reproduction 2017; 155:R1-R12. [PMID: 29018059 DOI: 10.1530/rep-17-0512] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 12/26/2022]
Abstract
Leptin is a homeostatic regulator in the placenta where it promotes proliferation, protein synthesis and the expression of tolerogenic maternal response molecules such as HLA-G. Leptin also exerts an anti-apoptotic action in placenta controlling the expression of p53 master cell cycle regulator under different stress conditions. On the other hand, leptin is an integrative target of different placental stimuli. The expression of leptin in placenta is regulated by hCG, insulin, steroids, hypoxia and many other growth hormones, suggesting that it might have an important endocrine function in the trophoblastic cells. The leptin expression is induced involving the cAMP/PKA or cAMP/Epac pathways which have profound actions upon human trophoblast function. The activation of PI3K and MAPK pathways also participates in the leptin expression. Estrogens play a central role during pregnancy, particularly 17β-estradiol upregulates the leptin expression in placental cells through genomic and non-genomic actions. The leptin promoter analysis reveals specific elements that are active in placental cells. The transcription factors CREB, AP1, Sp1, NFκB and the coactivator CBP are involved in the placental leptin expression. Moreover, placental leptin promoter is a target of epigenetic marks such as DNA methylation and histone acetylation that regulates not only the leptin expression in placenta during pregnancy but also determines the predisposition of acquiring adult metabolism diseases. Taken together, all these results allow a better understanding of leptin function and regulatory mechanisms of leptin expression in human placental trophoblasts, and support the importance of leptin during pregnancy and in programming adult health.
Collapse
Affiliation(s)
- Malena Schanton
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Julieta L Maymó
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Antonio Pérez-Pérez
- Departamento de Bioquímica Médica y Biología MolecularHospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Víctor Sánchez-Margalet
- Departamento de Bioquímica Médica y Biología MolecularHospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Cecilia L Varone
- Departamento de Química BiológicaUniversidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina .,Universidad de Buenos AiresCONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
19
|
Menden HL, Xia S, Mabry SM, Navarro A, Nyp MF, Sampath V. Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 Regulates LPS-Induced Inflammation and Alveolar Remodeling in the Developing Lung. Am J Respir Cell Mol Biol 2017; 55:767-778. [PMID: 27438994 DOI: 10.1165/rcmb.2016-0006oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In premature infants, sepsis is associated with alveolar simplification manifesting as bronchopulmonary dysplasia. The redox-dependent mechanisms underlying sepsis-induced inflammation and alveolar remodeling in the immature lung remain unclear. We developed a neonatal mouse model of sepsis-induced lung injury to investigate whether nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) regulates Toll-like receptor (TLR)-mediated inflammation and alveolar remodeling. Six-day-old NOX2+/+ and NOX2-/- mice were injected with intraperitoneal LPS to induce sepsis. Lung inflammation and canonical TLR signaling were assessed 24 hours after LPS. Alveolar development was examined in 15-day-old mice after LPS on Day 6. The in vivo efficacy of a NOX2 inhibitor (NOX2-I) on NOX2 complex assembly and sepsis-induced lung inflammation were examined. Lung cytokine expression and neutrophil influx induced with sepsis in NOX2+/+ mice was decreased by >50% in NOX2-/- mice. LPS-induced TLR4 signaling evident by inhibitor of NF-κB kinase-β and mitogen-activated protein kinase phosphorylation, and nuclear factor-κB/AP-1 translocation were attenuated in NOX2-/- mice. LPS increased matrix metalloproteinase 9 while decreasing elastin and keratinocyte growth factor levels in NOX2+/+ mice. An LPS-induced increase in matrix metalloproteinase 9 and decrease in fibroblast growth factor 7 and elastin were not evident in NOX2-/- mice. An LPS-induced reduction in radial alveolar counts and increased mean linear intercepts were attenuated in NOX2-/- mice. LPS-induced NOX2 assembly evident by p67phox/gp91phox coimmunoprecipitation was disrupted with NOX2-I. NOX2-I also mitigated LPS-induced cytokine expression, TLR pathway signaling, and alveolar simplification. In a mouse model of neonatal sepsis, NOX2 regulates proinflammatory TLR signaling and alveolar remodeling induced by a single dose of LPS. Our results provide mechanistic insight into the regulation of sepsis-induced alveolar remodeling in the developing lung.
Collapse
Affiliation(s)
- Heather L Menden
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Sheng Xia
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Angels Navarro
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Michael F Nyp
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Venkatesh Sampath
- Department of Pediatrics, Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri
| |
Collapse
|
20
|
Zhang L, Zhao S, Yuan L, Wu H, Jiang H, Luo G. Hyperoxia-mediated LC3B activation contributes to the impaired transdifferentiation of type II alveolar epithelial cells (AECIIs) to type I cells (AECIs). Clin Exp Pharmacol Physiol 2017; 43:834-43. [PMID: 27187184 DOI: 10.1111/1440-1681.12592] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/05/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022]
Abstract
Life-saving mechanical ventilation can also cause lung injury through the overproduction of reactive oxygen species (ROS), leading to bronchopulmonary dysplasia (BPD)-like symptoms in preterm infants. It is reported that the autophagic protein microtubule-associated protein-1 light chain (LC)-3B can confer protection against hyperoxia-induced DNA damage in lung alveolar epithelium. However, its role in the transdifferentiation of type II alveolar epithelial cells (AECIIs) to type I cells (AECIs) is unclear and requires further investigation. In this study, newborn Sprague-Dawley rats were exposed to 90% oxygen for up to 14 days to mimic BPD in human infants, with neonatal pups exposed to room air (21% oxygen) as controls. Primary rat AECIIs were cultured under hyperoxic conditions for up to 24 hours to further investigate the underlying mechanisms. This study found that hyperoxia promoted a significant and time-dependent increase of AECII marker surfactant protein (SP)-C in the lung. The increase of AECI marker T1α was repressed by hyperoxia during lung development. These results indicated an impaired AECII transdifferentiation. Pulmonary ROS concentration and expression of autophagic protein LC-3B were increased gradually in response to hyperoxia exposure. Furthermore, AECIIs produced more ROS when cultured under hyperoxic conditions in vitro. Both the LC3B expression and the conversion from LC3BI to LC3BII were enhanced in hyperoxic AECs. Interestingly, inhibition of LC3B either by ROS inhibitor N-acetyl-l-cysteine (NAC) or adenovirus-mediated LC3B shRNA could partly restore AECII transdifferentiation under hyperoxia condition. In summary, the current study reveals a novel role of activated LC3B induced by hyperoxia in AECII transdifferentiation.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Zhao
- Department of Paediatrics, Shenyang Fourth People's Hospital, Shenyang, China
| | - Lijie Yuan
- Department of Biochemistry and Molecular Biology, Harbin Medical University (Daqing Campus), Daqing, China
| | - Hongmin Wu
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hong Jiang
- Department of Paediatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Gang Luo
- Department of Paediatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Mills DR, Mao Q, Chu S, Falcon Girard K, Kraus M, Padbury JF, De Paepe ME. Effects of human umbilical cord blood mononuclear cells on respiratory system mechanics in a murine model of neonatal lung injury. Exp Lung Res 2017; 43:66-81. [PMID: 28353351 DOI: 10.1080/01902148.2017.1300713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mononuclear cells (MNCs) have well-documented beneficial effects in a wide range of adult pulmonary diseases. The effects of human umbilical cord blood-derived MNCs on neonatal lung injury, highly relevant for potential autologous application in preterm newborns at risk for bronchopulmonary dysplasia (BPD), remain incompletely established. The aim of this study was to determine the long-term morphologic and functional effects of systemically delivered MNCs in a murine model of neonatal lung injury. MATERIALS AND METHODS MNCs from cryopreserved cord blood (1 × 106 cells per pup) were given intravenously to newborn mice exposed to 90% O2 from birth; controls received cord blood total nucleated cells (TNCs) or granular cells, or equal volume vehicle buffer (sham controls). In order to avoid immune rejection, we used SCID mice as recipients. Lung mechanics (flexiVent™), engraftment, growth, and alveolarization were evaluated eight weeks postinfusion. RESULTS Systemic MNC administration to hyperoxia-exposed newborn mice resulted in significant attenuation of methacholine-induced airway hyperreactivity, leading to reduction of central airway resistance to normoxic levels. These bronchial effects were associated with mild improvement of alveolarization, lung compliance, and elastance. TNCs had no effects on alveolar remodeling and were associated with worsened methacholine-induced bronchial hyperreactivity. Granular cell administration resulted in a marked morphologic and functional emphysematous phenotype, associated with high mortality. Pulmonary donor cell engraftment was sporadic in all groups. CONCLUSIONS These results suggest that cord blood MNCs may have a cell type-specific role in therapy of pulmonary conditions characterized by increased airway resistance, such as BPD and asthma. Future studies need to determine the active MNC subtype(s), their mechanisms of action, and optimal purification methods to minimize granular cell contamination.
Collapse
Affiliation(s)
- David R Mills
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA
| | - Quanfu Mao
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | - Sharon Chu
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | | | - Morey Kraus
- c ViaCord LLC, a Perkin Elmer Company , Cambridge , Massachusetts , USA
| | - James F Padbury
- d Department of Pediatrics , Women and Infants Hospital , Providence , Rhode Island , USA.,e Department of Pediatrics , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | - Monique E De Paepe
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| |
Collapse
|
22
|
Abstract
Despite recent advances in neonatal medicine, neonatal disorders, such as bronchopulmonary dysplasia and intraventricular hemorrhage in preterm neonates and hypoxic ischemic encephalopathy in term neonates, remain major causes of mortality and morbidities. Promising preclinical research results suggest that stem cell therapies represent the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies. This review focuses primarily on the potential role of stem cell therapy in the above mentioned neonatal disorders, highlighting the results of human clinical trials and the challenges that remain to be addressed for their safe and successful translation into clinical care of newborn infants.
Collapse
Affiliation(s)
- Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sein Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Choi YE, Cho HJ, Song ES, Jeong IS, Yoon N, Choi YY, Ma JS, Cho YK. Clinical Utility of Echocardiography for the Diagnosis and Prognosis in Children with Bronchopulmonary Dsyplasia. J Cardiovasc Ultrasound 2016; 24:278-284. [PMID: 28090255 PMCID: PMC5234346 DOI: 10.4250/jcu.2016.24.4.278] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 09/06/2016] [Accepted: 11/30/2016] [Indexed: 11/29/2022] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) may result in chronic pulmonary artery hypertension and right ventricular (RV) dysfunction. Various echocardiographic assessments of RV dysfunction have been used to determine whether echocardiographic measurements of premature infants with BPD could provide sensitive measures of RV function that correlates with BPD severity. Methods Twenty-eight control subjects without BPD (non BPD group), 28 patients with mild BPD, 11 patients with moderate BPD, and six patients with severe BPD underwent echocardiograms with standard measurement such as ejection fraction by M-mode, tricuspid regurgitation pressure gradient, myocardial performance index (MPI) derived from pulse Doppler, and tissue Doppler imaging (TDI) measurements. BPD severity was classified by the NICHD/NHLBI/ORD workshop rating scale. Twenty-eight control subjects without BPD (non BPD group), 28 patients with mild BPD, 11 patients with moderate BPD, and six patients with severe BPD underwent echocardiograms with standard measurement such as ejection fraction by M-mode, tricuspid regurgitation pressure gradient, myocardial performance index (MPI) derived from pulse Doppler, and TDI measurements. BPD severity was classified by the NICHD/NHLBI/ORD workshop rating scale. Results None of the standard echocardiographic findings was significantly different between the control group and BPD groups. However, mean septal TDI-MPI of the severe BPD group (0.68 ± 0.06) was significantly (p < 0.01) higher than that of the non-BPD (0.58 ± 0.10) or the mild BPD group (0.59 ± 0.12). In addition, mean RV TDI-MPI of the severe BPD group (0.71 ± 0.13) was significantly (p < 0.05) higher than that of the non-BPD group (0.56 ± 0.08) or the mild BPD group (0.60 ± 0.125). Linear regression showed a good correlation between the severity of BPD and RV TDI-MPI (p = 0.01, R = 0.30) or septal TDI-MPI (p = 0.04, R = 0.24). Conclusion Echocardiographic evaluation of RV function based on an assessment of RV TDI-MPI can provide RV dysfunction parameter in premature infants with BPD.
Collapse
Affiliation(s)
- Young Earl Choi
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Hwa Jin Cho
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Eun Song Song
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - In Seok Jeong
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Namsik Yoon
- The Heart Center of Chonnam National University Hospital, Chonnam National University Medical School and The Research Institute of Medical Sciences of Chonnam National University, Gwangju, Korea
| | - Young Youn Choi
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Jae Sook Ma
- Department of Pediatrics, KS Hospital, Gwangju, Korea
| | - Young Kuk Cho
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
24
|
Xu YP. Bronchopulmonary Dysplasia in Preterm Infants Born at Less Than 32 Weeks Gestation. Glob Pediatr Health 2016; 3:2333794X16668773. [PMID: 27689102 PMCID: PMC5028015 DOI: 10.1177/2333794x16668773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/11/2016] [Indexed: 01/24/2023] Open
Abstract
Objectives: Bronchopulmonary dysplasia (BPD) is a chronic pulmonary disorder affecting preterm infants. We studied the factors and echocardiographic evidence of early pulmonary hypertension (PH) associated with moderate or severe BPD. Methods: We retrospectively reviewed preterm infants who were born at <32 weeks gestation and admitted to the neonatal intensive care unit at the Children’s Hospital of Zhejiang University School of Medicine between July 2013 and July 2015. Results: Forty-two preterm infants were enrolled in the study. All the patients received oxygen treatment for a mean of 62.5 ± 28.0 days. The grades of BPD were classified as follows: severe, 35.7%; moderate, 40.5%; and mild, 23.8%. The time of ventilator and oxygen supplementation was longer in infants who developed PH. Severe BPD was related to PH at 28 days. Conclusions: These findings support the notion that early pulmonary vascular disease and long-term infection in preterm infants contributes to increased susceptibility for severe BPD.
Collapse
Affiliation(s)
- Yan-Ping Xu
- The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
Benjamin JT, van der Meer R, Im AM, Plosa EJ, Zaynagetdinov R, Burman A, Havrilla ME, Gleaves LA, Polosukhin VV, Deutsch GH, Yanagisawa H, Davidson JM, Prince LS, Young LR, Blackwell TS. Epithelial-Derived Inflammation Disrupts Elastin Assembly and Alters Saccular Stage Lung Development. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1786-1800. [PMID: 27181406 DOI: 10.1016/j.ajpath.2016.02.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 12/22/2022]
Abstract
The highly orchestrated interactions between the epithelium and mesenchyme required for normal lung development can be disrupted by perinatal inflammation in preterm infants, although the mechanisms are incompletely understood. We used transgenic (inhibitory κB kinase β transactivated) mice that conditionally express an activator of the NF-κB pathway in airway epithelium to investigate the impact of epithelial-derived inflammation during lung development. Epithelial NF-κB activation selectively impaired saccular stage lung development, with a phenotype comprising rapidly progressive distal airspace dilation, impaired gas exchange, and perinatal lethality. Epithelial-derived inflammation resulted in disrupted elastic fiber organization and down-regulation of elastin assembly components, including fibulins 4 and 5, lysyl oxidase like-1, and fibrillin-1. Fibulin-5 expression by saccular stage lung fibroblasts was consistently inhibited by treatment with bronchoalveolar lavage fluid from inhibitory κB kinase β transactivated mice, Escherichia coli lipopolysaccharide, or tracheal aspirates from preterm infants exposed to chorioamnionitis. Expression of a dominant NF-κB inhibitor in fibroblasts restored fibulin-5 expression after lipopolysaccharide treatment, whereas reconstitution of fibulin-5 rescued extracellular elastin assembly by saccular stage lung fibroblasts. Elastin organization was disrupted in saccular stage lungs of preterm infants exposed to systemic inflammation. Our study reveals a critical window for elastin assembly during the saccular stage that is disrupted by inflammatory signaling and could be amenable to interventions that restore elastic fiber assembly in the developing lung.
Collapse
Affiliation(s)
- John T Benjamin
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Riet van der Meer
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Amanda M Im
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Erin J Plosa
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rinat Zaynagetdinov
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ankita Burman
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Madeline E Havrilla
- Department of Pediatrics, Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Linda A Gleaves
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vasiliy V Polosukhin
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital, Seattle, Washington
| | - Hiromi Yanagisawa
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey M Davidson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lawrence S Prince
- Department of Pediatrics, Division of Neonatology, University of California-San Diego, San Diego, California
| | - Lisa R Young
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pediatrics, Division of Pulmonary Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy S Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
26
|
Programmierung durch intrauterine Mangelversorgung. Monatsschr Kinderheilkd 2015. [DOI: 10.1007/s00112-015-3420-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
27
|
Sung DK, Chang YS, Ahn SY, Sung SI, Yoo HS, Choi SJ, Kim SY, Park WS. Optimal Route for Human Umbilical Cord Blood-Derived Mesenchymal Stem Cell Transplantation to Protect Against Neonatal Hyperoxic Lung Injury: Gene Expression Profiles and Histopathology. PLoS One 2015; 10:e0135574. [PMID: 26305093 PMCID: PMC4549285 DOI: 10.1371/journal.pone.0135574] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/23/2015] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to determine the optimal route of mesenchymal stem cell (MSC) transplantation. To this end, gene expression profiling was performed to compare the effects of intratracheal (IT) versus intravenous (IV) MSC administration. Furthermore, the therapeutic efficacy of each route to protect against neonatal hyperoxic lung injury was also determined. Newborn Sprague-Dawley rats were exposed to hyperoxia (90% oxygen) from birth for 14 days. Human umbilical cord blood-derived MSCs labeling with PKH26 were transplanted through either the IT (5×105) or IV (2×106) route at postnatal day (P) 5. At P14, lungs were harvested for histological, biochemical and microarray analyses. Hyperoxic conditions induced an increase in the mean linear intercept and mean alveolar volume (MAV), indicative of impaired alveolarization. The number of ED-1 positive cells was significantly decreased by both IT and IV transplantations. However, IT administration of MSCs resulted in a greater decrease in MAV and ED-1 positive cells compared to IV administration. Moreover, the number of TUNEL-positive cells was significantly decreased in the IT group, but not in the IV group. Although the IT group received only one fourth of the number of MSCs that the IV group did, a significantly higher number of donor cell-derived red PKH 26 positivity were recovered in the IT group. Hyperoxic conditions induced the up regulation of genes associated with the inflammatory response, such as macrophage inflammatory protein-1 α, tumor necrosis factor-α and inter leukin-6; genes associated with cell death, such as p53 and caspases; and genes associated with fibrosis, such as connective tissue growth factor. In contrast, hyperoxic conditions induced the dwon-regulation of vascular endothelial growth factor and hepatocyte growth factor. These hyperoxia-induced changes in gene expression were decreased in the IT group, but not in the IV group. Thus, local IT MSC transplantation was more effective than systemic IV MSC administration in protecting against neonatal hyperoxic lung injury.
Collapse
Affiliation(s)
- Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Soo Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Soo Yoon Kim
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
28
|
Kovachy VN, Adams JN, Tamaresis JS, Feldman HM. Reading abilities in school-aged preterm children: a review and meta-analysis. Dev Med Child Neurol 2015; 57:410-9. [PMID: 25516105 PMCID: PMC4397135 DOI: 10.1111/dmcn.12652] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2014] [Indexed: 12/19/2022]
Abstract
AIM Children born preterm (at ≤32wks) are at risk of developing deficits in reading ability. This meta-analysis aims to determine whether or not school-aged preterm children perform worse than those born at term in single-word reading (decoding) and reading comprehension. METHOD Electronic databases were searched for studies published between 2000 and 2013, which assessed decoding or reading comprehension performance in English-speaking preterm and term-born children aged between 6 years and 13 years, and born after 1990. Standardized mean differences in decoding and reading comprehension scores were calculated. RESULTS Nine studies were suitable for analysis of decoding, and five for analysis of reading comprehension. Random-effects meta-analyses showed that children born preterm had significantly lower scores (reported as Cohen's d values [d] with 95% confidence intervals [CIs]) than those born at term for decoding (d=-0.42, 95% CI -0.57 to -0.27, p<0.001) and reading comprehension (d=-0.57, 95% CI -0.68 to -0.46, p<0.001). Meta-regressions showed that lower gestational age was associated with larger differences in decoding (Q[1]=5.92, p=0.02) and reading comprehension (Q[1]=4.69, p=0.03) between preterm and term groups. Differences between groups increased with age for reading comprehension (Q[1]=5.10, p=0.02) and, although not significant, there was also a trend for increased group differences for decoding (Q[1]=3.44, p=0.06). INTERPRETATION Preterm children perform worse than peers born at term on decoding and reading comprehension. These findings suggest that preterm children should receive more ongoing monitoring for reading difficulties throughout their education.
Collapse
Affiliation(s)
- Vanessa N Kovachy
- Department of Pediatrics, Stanford School of Medicine, Palo Alto, CA, USA
| | | | | | | |
Collapse
|
29
|
Ahn SY, Chang YS, Sung DK, Yoo HS, Sung SI, Choi SJ, Park WS. Cell type-dependent variation in paracrine potency determines therapeutic efficacy against neonatal hyperoxic lung injury. Cytotherapy 2015; 17:1025-35. [PMID: 25863963 DOI: 10.1016/j.jcyt.2015.03.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS The aim of this study was to determine the optimal cell type for transplantation to protect against neonatal hyperoxic lung injury. To this end, the in vitro and in vivo therapeutic efficacies and paracrine potencies of human umbilical cord blood-derived mesenchymal stromal cells (HUMs), human adipose tissue-derived mesenchymal stromal cells (HAMs) and human umbilical cord blood mononuclear cells (HMNs) were compared. METHODS Hyperoxic injury was induced in vitro in A549 cells by challenge with H2O2. Alternatively, hyperoxic injury was induced in newborn Sprague-Dawley rats in vivo by exposure to hyperoxia (90% oxygen) for 14 days. HUMs, HAMs or HMNs (5 × 10(5) cells) were given intratracheally at postnatal day 5. RESULTS Hyperoxia-induced increases in in vitro cell death and in vivo impaired alveolarization were significantly attenuated in both the HUM and HAM groups but not in the HMN group. Hyperoxia impaired angiogenesis, increased the cell death and pulmonary macrophages and elevated inflammatory cytokine levels. These effects were significantly decreased in the HUM group but not in the HAM or HMN groups. The levels of human vascular endothelial growth factor and hepatocyte growth factor produced by donor cells were highest in HUM group, followed by HAM group and then HMN group. CONCLUSIONS HUMs exhibited the best therapeutic efficacy and paracrine potency than HAMs or HMNs in protecting against neonatal hyperoxic lung injury. These cell type-dependent variations in therapeutic efficacy might be associated or mediated with the paracrine potency of the transplanted donor cells.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Soo Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
30
|
Borg AJ, Yong HEJ, Lappas M, Degrelle SA, Keogh RJ, Da Silva-Costa F, Fournier T, Abumaree M, Keelan JA, Kalionis B, Murthi P. Decreased STAT3 in human idiopathic fetal growth restriction contributes to trophoblast dysfunction. Reproduction 2015; 149:523-32. [PMID: 25713425 DOI: 10.1530/rep-14-0622] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Abnormal trophoblast function is associated with fetal growth restriction (FGR). The JAK-STAT pathway is one of the principal signalling mechanisms by which cytokines and growth factors modulate cell proliferation, differentiation, cell migration and apoptosis. The expression of placental JAK-STAT genes in human idiopathic FGR is unknown. In this study, we propose the hypothesis that JAK-STAT pathway genes are differentially expressed in idiopathic FGR-affected pregnancies and contribute to abnormal feto-placental growth by modulating the expression of the amino acid transporter SNAT2, differentiation marker CGB/human chorionic gonadotrophin beta-subunit (β-hCG) and apoptosis markers caspases 3 and 8, and TP53. Expression profiling of FGR-affected placentae revealed that mRNA levels of STAT3, STAT2 and STAT5B decreased by 69, 52 and 50%, respectively, compared with gestational-age-matched controls. Further validation by real-time PCR and immunoblotting confirmed significantly lower STAT3 mRNA and STAT3 protein (total and phosphorylated) levels in FGR placentae. STAT3 protein was localised to the syncytiotrophoblast (ST) in both FGR and control placentae. ST differentiation was modelled by in vitro differentiation of primary villous trophoblast cells from first-trimester and term placentae, and by treating choriocarcinoma-derived BeWo cells with forskolin in cell culture. Differentiation in these models was associated with increased STAT3 mRNA and protein levels. In BeWo cells treated with siRNA targeting STAT3, the mRNA and protein levels of CGB/β-hCG, caspases 3 and 8, and TP53 were significantly increased, while that of SNAT2 was significantly decreased compared with the negative control siRNA. In conclusion, we report that decreased STAT3 expression in placentae may contribute to abnormal trophoblast function in idiopathic FGR-affected pregnancies.
Collapse
Affiliation(s)
- A J Borg
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - H E J Yong
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - M Lappas
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - S A Degrelle
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesP
| | - R J Keogh
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - F Da Silva-Costa
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - T Fournier
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - M Abumaree
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - J A Keelan
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - B Kalionis
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| | - P Murthi
- Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia Department of Perinatal MedicinePregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria, AustraliaDepartment of Obstetrics and GynaecologyUniversity of Melbourne, Melbourne, Victoria, AustraliaDepartment of Obstetrics and GynaecologyMercy Hospital for Women, Heidelberg, Victoria, AustraliaINSERM-U767Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, FranceUniversite Paris DescartesParis F-75006, FrancePremUp FoundationParis F-75006, FranceCollege of Science and Health ProfessionsKing Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi ArabiaSchool of Women's and Infants' HealthKing Edward Memorial Hospital, University of Western Australia, Subiaco, Western Australia, Australia
| |
Collapse
|
31
|
Liu L, Mao Q, Chu S, Mounayar M, Abdi R, Fodor W, Padbury JF, De Paepe ME. Intranasal versus intraperitoneal delivery of human umbilical cord tissue-derived cultured mesenchymal stromal cells in a murine model of neonatal lung injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3344-58. [PMID: 25455688 DOI: 10.1016/j.ajpath.2014.08.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/18/2014] [Accepted: 08/05/2014] [Indexed: 12/19/2022]
Abstract
Clinical trials investigating mesenchymal stromal cell (MSC) therapy for bronchopulmonary dysplasia have been initiated; however, the optimal delivery route and functional effects of MSC therapy in newborns remain incompletely established. We studied the morphologic and functional effects of intranasal versus i.p. MSC administration in a rodent model of neonatal lung injury. Cultured human cord tissue MSCs (0.1, 0.5, or 1 × 10(6) cell per pup) were given intranasally or i.p. to newborn severe combined immunodeficiency-beige mice exposed to 90% O2 from birth; sham controls received an equal volume of phosphate-buffered saline. Lung mechanics, engraftment, lung growth, and alveolarization were evaluated 8 weeks after transplantation. High-dose i.p. MSC administration to newborn mice exposed to 90% O2 resulted in the restoration of normal lung compliance, elastance, and pressure-volume loops (tissue recoil). Histologically, high-dose i.p. MSC administration was associated with alveolar septal widening, suggestive of interstitial matrix modification. Intranasal MSC or lower-dose i.p. administration had no significant effects on lung function or alveolar remodeling. Pulmonary engraftment was rare in all the groups. These findings suggest that high-dose systemic administration of human cultured MSCs can restore normal compliance in neonatally injured lungs, possibly by paracrine modulation of the interstitial matrix. Intranasal delivery had no obvious pulmonary effects.
Collapse
Affiliation(s)
- Liansheng Liu
- Department of Pathology, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Quanfu Mao
- Department of Pathology, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, Rhode Island; Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharon Chu
- Department of Pathology, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, Rhode Island; Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Marwan Mounayar
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Reza Abdi
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - James F Padbury
- Department of Pediatrics, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, Rhode Island; Department of Pediatrics, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Monique E De Paepe
- Department of Pathology, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, Rhode Island; Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
32
|
Mirza H, Ziegler J, Ford S, Padbury J, Tucker R, Laptook A. Pulmonary hypertension in preterm infants: prevalence and association with bronchopulmonary dysplasia. J Pediatr 2014; 165:909-14.e1. [PMID: 25189821 DOI: 10.1016/j.jpeds.2014.07.040] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 07/03/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To determine whether early pulmonary hypertension (PH) at 10-14 days of life in preterm infants is associated with bronchopulmonary dysplasia (BPD) at 36 weeks' postmenstrual age (PMA). STUDY DESIGN This was a prospective observational cohort study of infants <28 weeks' gestation. Exclusion criteria were any major anomaly, genetic syndrome, or death before the initial echocardiogram. Echocardiograms were performed between 10 and 14 days of life and at 36 weeks' PMA to assess PH. BPD and its severity were determined at 36 weeks PMA by the National Institutes of Health workshop definition. RESULTS From March 2011 to April 2013, of 146 consecutively admitted infants <28 weeks, 120 were enrolled. One infant was excluded, 17 did not consent, and 8 died before undergoing a study echocardiogram. At 10-14 days of life, 10 infants had early PH (8%). Male sex (56% vs 40%), gestational age (26(+2) ± 1(+2) vs 25(+6) ± 1(+4) weeks), birth weight (837 ± 205 g vs 763 ± 182 g), and small for gestational age (14% vs 20%) were not significantly different among infants with no PH and early PH, respectively. Infants with early PH required >0.3 fraction of inspired oxygen by day 10 of life (70% vs 27%, P < .01). Moderate/severe BPD or death was greater among infants with early PH (90%) compared with no PH (47%, relative risk 1.9, 95% CI 1.43-2.53). CONCLUSION In this prospective, single-center cohort, early PH was associated with moderate/severe BPD or death at 36 weeks' PMA.
Collapse
Affiliation(s)
- Hussnain Mirza
- Department of Pediatrics, Women & Infants Hospital/The Alpert Medical School of Brown University, Providence, RI
| | - James Ziegler
- Division of Pediatric Cardiology, Hasbro Children's Hospital/The Alpert Medical School of Brown University, Providence, RI
| | - Sara Ford
- Division of Pediatric Cardiology, Hasbro Children's Hospital/The Alpert Medical School of Brown University, Providence, RI
| | - James Padbury
- Department of Pediatrics, Women & Infants Hospital/The Alpert Medical School of Brown University, Providence, RI
| | - Richard Tucker
- Department of Pediatrics, Women & Infants Hospital/The Alpert Medical School of Brown University, Providence, RI
| | - Abbot Laptook
- Department of Pediatrics, Women & Infants Hospital/The Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
33
|
Chang YS, Ahn SY, Jeon HB, Sung DK, Kim ES, Sung SI, Yoo HS, Choi SJ, Oh WI, Park WS. Critical role of vascular endothelial growth factor secreted by mesenchymal stem cells in hyperoxic lung injury. Am J Respir Cell Mol Biol 2014; 51:391-9. [PMID: 24669883 DOI: 10.1165/rcmb.2013-0385oc] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Intratracheal transplantation of human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) protects against neonatal hyperoxic lung injury by a paracrine rather than a regenerative mechanism. However, the role of paracrine factors produced by the MSCs, such as vascular endothelial growth factor (VEGF), has not been delineated. This study examined whether VEGF secreted by MSCs plays a pivotal role in protecting against neonatal hyperoxic lung injury. VEGF was knocked down in human UCB-derived MSCs by transfection with small interfering RNA specific for human VEGF. The in vitro effects of MSCs with or without VEGF knockdown or neutralizing antibody were evaluated in a rat lung epithelial (L2) cell line challenged with H2O2. To confirm these results in vivo, newborn Sprague-Dawley rats were exposed to hyperoxia (90% O2) for 14 days. MSCs (1 × 10(5) cells) with or without VEGF knockdown were administered intratracheally at postnatal Day 5. Lungs were serially harvested for biochemical and histologic analyses. VEGF knockdown and antibody abolished the in vitro benefits of MSCs on H2O2-induced cell death and the up-regulation of inflammatory cytokines in L2 cells. VEGF knockdown also abolished the in vivo protective effects of MSCs in hyperoxic lung injury, such as the attenuation of impaired alveolarization and angiogenesis, reduction in the number of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive and ED-1-positive cells, and down-regulation of proinflammatory cytokine levels. Our data indicate that VEGF secreted by transplanted MSCs is one of the critical paracrine factors that play seminal roles in attenuating hyperoxic lung injuries in neonatal rats.
Collapse
Affiliation(s)
- Yun Sil Chang
- 1 Department of Pediatrics, Samsung Medical Center, and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yoder BA, Coalson JJ. Animal models of bronchopulmonary dysplasia. The preterm baboon models. Am J Physiol Lung Cell Mol Physiol 2014; 307:L970-7. [PMID: 25281639 DOI: 10.1152/ajplung.00171.2014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Much of the progress in improved neonatal care, particularly management of underdeveloped preterm lungs, has been aided by investigations of multiple animal models, including the neonatal baboon (Papio species). In this article we highlight how the preterm baboon model at both 140 and 125 days gestation (term equivalent 185 days) has advanced our understanding and management of the immature human infant with neonatal lung disease. Not only is the 125-day baboon model extremely relevant to the condition of bronchopulmonary dysplasia but there are also critical neurodevelopmental and other end-organ pathological features associated with this model not fully discussed in this limited forum. We also describe efforts to incorporate perinatal infection into these preterm models, both fetal and neonatal, and particularly associated with Ureaplasma/Mycoplasma organisms. Efforts to rekindle the preterm primate model for future evaluations of therapies such as stem cell replacement, early lung recruitment interventions coupled with noninvasive surfactant and high-frequency nasal ventilation, and surfactant therapy coupled with antioxidant or anti-inflammatory medications, to name a few, should be undertaken.
Collapse
Affiliation(s)
- Bradley A Yoder
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Jacqueline J Coalson
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
35
|
Mesenchymal stem cells for bronchopulmonary dysplasia: phase 1 dose-escalation clinical trial. J Pediatr 2014; 164:966-972.e6. [PMID: 24508444 DOI: 10.1016/j.jpeds.2013.12.011] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/13/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To assess the safety and feasibility of allogeneic human umbilical cord blood (hUCB)-derived mesenchymal stem cell (MSC) transplantation in preterm infants. STUDY DESIGN In a phase I dose-escalation trial, we assessed the safety and feasibility of a single, intratracheal transplantation of hUCB-derived MSCs in preterm infants at high risk for bronchopulmonary dysplasia (BPD). The first 3 patients were given a low dose (1 × 10(7) cells/kg) of cells, and the next 6 patients were given a high dose (2 × 10(7) cells/kg). We compared their adverse outcomes, including BPD severity, with those of historical case-matched comparison group. RESULTS Intratracheal MSC transplantation was performed in 9 preterm infants, with a mean gestational age of 25.3 ± 0.9 weeks and a mean birth weight of 793 ± 127 g, at a mean of 10.4 ± 2.6 days after birth. The treatments were well tolerated, without serious adverse effects or dose-limiting toxicity attributable to the transplantation. Levels of interleukin-6, interleukin-8, matrix metalloproteinase-9, tumor necrosis factor α, and transforming growth factor β1 in tracheal aspirates at day 7 were significantly reduced compared with those at baseline or at day 3 posttransplantation. BPD severity was lower in the transplant recipients, and rates of other adverse outcomes did not differ between the comparison group and transplant recipients. CONCLUSION Intratracheal transplantation of allogeneic hUCB-derived MSCs in preterm infants is safe and feasible, and warrants a larger and controlled phase II study.
Collapse
|
36
|
HE HUA, CHEN FEI, NI WENSI, LI JIANHUI, ZHANG YONGJUN. Theophylline improves lipopolysaccharide-induced alveolarization arrest through inflammatory regulation. Mol Med Rep 2014; 10:269-75. [DOI: 10.3892/mmr.2014.2188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 03/11/2014] [Indexed: 11/06/2022] Open
|
37
|
Lacaze-Masmonteil T. That chorioamnionitis is a risk factor for bronchopulmonary dysplasia--the case against. Paediatr Respir Rev 2014; 15:53-5. [PMID: 24120077 DOI: 10.1016/j.prrv.2013.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most frequent long term sequelae in infants born at less than 29 weeks of gestational age (GA) and histological chorioamnionitis (CA) is the most frequent condition associated with very preterm birth. Numerous studies have explored the association between BPD and CA with conflicting results. This inconsistency may be attributable to differences in populations, definitions, methods, and whether potential confounding factors such as GA, antenatal steroids, and post natal events were considered. A recent systematic review and meta-analysis shows some evidence of an association between BPD and CA; however, results adjusting for important confounders show more conservative measures of association. In addition, there was evidence of publication bias: when controlling for publication bias the results were more conservative and adjusted results were no longer significant. Recent large cohort studies not included in the systematic review do not support the belief that CA is associated with an increased risk of BPD. Despite a large body of evidence, CA cannot be definitively considered a risk factor for BPD.
Collapse
Affiliation(s)
- Thierry Lacaze-Masmonteil
- Department of Pediatrics and Research Institute, Children's Hospital of Eastern Ontario, University of Ottawa, Canada.
| |
Collapse
|
38
|
Thomas W, Speer CP. Chorioamnionitis is essential in the evolution of bronchopulmonary dysplasia--the case in favour. Paediatr Respir Rev 2014; 15:49-52. [PMID: 24128984 DOI: 10.1016/j.prrv.2013.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a major sequel of extremely premature birth. Multiple ante- and postnatal factors act in concert to injure the immature lung in the pathogenesis of the disease. Among them, chorioamnionitis--according to current evidence--plays a pivotal role. Pulmonary inflammatory processes seen in animal models of chorioamnionitis resemble those seen in premature infants who developed BPD. Chorioamnionitis can doubtlessly induce extremely preterm birth, thus contributing to a gestation-dependent risk of BPD. A gestation-independent association of chorioamnionitis with an increased risk of developing BPD has been demonstrated by a recent systematic review of clinical observational studies. Antenatal inflammation with signs of a systemic fetal response reduces the response to exogenous surfactant in infants with respiratory distress syndrome, leading to a longer need for mechanical ventilation. Moreover, chorioamnionitis increases the risk of early onset sepsis. Both mechanical ventilation and sepsis are, however, major postnatal risk factors for BPD.
Collapse
Affiliation(s)
- Wolfgang Thomas
- Mutterhaus der Borromaeerinnen, Department of Pediatrics, Feldstr. 6, 54290 Trier, Germany.
| | - Christian P Speer
- University of Würzburg, University Children's Hospital, Josef-Schneider-Str. 2, 97080 Würzburg, Germany.
| |
Collapse
|
39
|
Sezer RG, Aydemir G, Bozaykut A, Hira S, Tanju IA, Ozcan O. The relationship between the first episode of wheezing and matrix metalloproteinases-9 and MMP-2 and tissue inhibitors of MMP-1 levels in preterm infants. Ann Thorac Med 2013; 8:209-13. [PMID: 24250734 PMCID: PMC3821280 DOI: 10.4103/1817-1737.118506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 07/04/2013] [Indexed: 11/04/2022] Open
Abstract
AIMS Matrix metalloproteinases (MMP) have been associated with neonatal lung morbidity and MMP dysregulation contributes to the pathology of chronic and acute lung disorders. Most of the previous studies were performed in the 1(st) weeks of life of the preterm newborns. There are no data on the serum levels of MMP-2, MMP-9 or tissue inhibitors of matrix metalloproteinases (TIMP-1) from preterm infants recovering from lung morbidities. We aimed to compare MMP-2, MMP-9 and TIMP-1 levels in preterm and term infants hospitalized with their first episode of wheezing. METHODS We prospectively evaluated 18 preterm infants with a history of chronic lung disease, respiratory distress syndrome or oxygen therapy and 14 age- and sex-matched term infants who were admitted for a first episode of wheezing. We quantified total serum concentrations of MMP-2, MMP-9 and TIMP-1 to assess whether these serum markers levels were associated with the first episode of wheezing in infants with a history of oxygen therapy during the neonatal period. RESULTS Upon hospitalization, MMP-2 and TIMP-1 levels were higher in preterm infants than in term infants. In contrast, there was no significant relationship between MMP-9 levels or the MMP-9/TIMP-1 ratio between preterm and term infants. The area under the receiver operating characteristic curve for MMP-2 was 0.70 (95% confidence interval [CI] 0.51-0.89). The area under the curve for TIMP-1 was 0.78 (95% CI 0.61-0.94). MMP-9, MMP-2 and TIMP-1 levels did not correlate with gestational age, gender or severity of wheezing. CONCLUSION The negative proportion of MMP-9 to TIMP-1 that we detected in term infants was not present in preterm infants. The balance of MMP-9 to TIMP-1 may have been disrupted by lung damage in the premature infants. Overproduction of MMP-2 and TIMP-1 in the serum may be associated with the pathogenesis of wheezing in preterm infants.
Collapse
Affiliation(s)
- Rabia Gonul Sezer
- Department of Pediatrics, Zeynep Kamil Maternity and Children's Disease Training and Research Hospital, Uskudar 34668, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
40
|
Madurga A, Mizíková I, Ruiz-Camp J, Morty RE. Recent advances in late lung development and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2013; 305:L893-905. [PMID: 24213917 DOI: 10.1152/ajplung.00267.2013] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In contrast to early lung development, a process exemplified by the branching of the developing airways, the later development of the immature lung remains very poorly understood. A key event in late lung development is secondary septation, in which secondary septa arise from primary septa, creating a greater number of alveoli of a smaller size, which dramatically expands the surface area over which gas exchange can take place. Secondary septation, together with architectural changes to the vascular structure of the lung that minimize the distance between the inspired air and the blood, are the objectives of late lung development. The process of late lung development is disturbed in bronchopulmonary dysplasia (BPD), a disease of prematurely born infants in which the structural development of the alveoli is blunted as a consequence of inflammation, volutrauma, and oxygen toxicity. This review aims to highlight notable recent developments in our understanding of late lung development and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Alicia Madurga
- Dept. of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, D-61231 Bad Nauheim, Germany.
| | | | | | | |
Collapse
|
41
|
Harijith A, Pendyala S, Reddy NM, Bai T, Usatyuk PV, Berdyshev E, Gorshkova I, Huang LS, Mohan V, Garzon S, Kanteti P, Reddy SP, Raj JU, Natarajan V. Sphingosine kinase 1 deficiency confers protection against hyperoxia-induced bronchopulmonary dysplasia in a murine model: role of S1P signaling and Nox proteins. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1169-1182. [PMID: 23933064 DOI: 10.1016/j.ajpath.2013.06.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/05/2013] [Accepted: 06/24/2013] [Indexed: 12/23/2022]
Abstract
Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| | - Srikanth Pendyala
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Narsa M Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Tao Bai
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Peter V Usatyuk
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Evgeny Berdyshev
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Irina Gorshkova
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Long Shuang Huang
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Vijay Mohan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Steve Garzon
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois
| | - Prasad Kanteti
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Sekhar P Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - J Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
42
|
Choi CW, Kim BI, Mason SN, Potts-Kant EN, Brahmajothi MV, Auten RL. Intra-amniotic LPS amplifies hyperoxia-induced airway hyperreactivity in neonatal rats. Pediatr Res 2013; 74:11-8. [PMID: 23563192 PMCID: PMC3707085 DOI: 10.1038/pr.2013.58] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 01/17/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND We previously showed that intra-amniotic lipopolysaccharide (LPS) amplifies alveolar hypoplasia induced by postnatal hyperoxia. We determined whether the priming effect of intra-amniotic LPS amplifies hyperoxia-induced airway hyperreactivity (AHR). METHODS LPS or normal saline was injected into the amniotic cavities of pregnant rats at the 20th day of gestation. After birth, rat pups were exposed to 60% O₂ or air for 14 d. On postnatal day 14, rat pups underwent forced oscillometry, which included a challenge with nebulized methacholine, and the lungs were harvested for morphological studies. RESULTS Hyperoxia significantly increased airway reactivity and decreased compliance. Intra-amniotic LPS further increased hyperoxia-induced AHR but did not further impair respiratory system compliance. Hyperoxia-induced changes in lung parenchymal and small airway morphology were not further altered by intra-amniotic LPS. However, combined exposure to intra-amniotic LPS and hyperoxia increased the proportion of degranulating mast cells in the hilar airways. CONCLUSION Intra-amniotic LPS amplified postnatal hyperoxia-induced AHR. This was associated with increased airway mast cell degranulation, which has previously been linked with hyperoxia-induced AHR. There were no morphologic changes of parenchyma or airways that would account for the LPS augmentation of hyperoxia-induced AHR.
Collapse
Affiliation(s)
- Chang Won Choi
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Beyong Il Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea,Department of Pediatrics and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Stanley N. Mason
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Erin N. Potts-Kant
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | | | - Richard L. Auten
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
43
|
Iliodromiti Z, Zygouris D, Sifakis S, Pappa KI, Tsikouras P, Salakos N, Daniilidis A, Siristatidis C, Vrachnis N. Acute lung injury in preterm fetuses and neonates: mechanisms and molecular pathways. J Matern Fetal Neonatal Med 2013; 26:1696-704. [PMID: 23611524 DOI: 10.3109/14767058.2013.798284] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acute lung injury (ALI) results in high morbidity and mortality among preterm neonates and efforts have therefore been devoted to both antenatal and postnatal prevention of the disease. ALI is the result of an inflammatory response which is triggered by a variety of different mechanisms. It mostly affects the fetal lung and, in particular, causes damage to the integrity of the lung's alveolar-capillary unit while weakening its cellular linings. Chemotactic activity and inflammatory products, such as proinflammatory cytokines TNF-α, IL-1, IL-6, IL-11, VEGF,TGF-α and TGF-β, provoke serious damage to the capillary endothelium and the alveolar epithelium, resulting in hyaline membrane formation and leakage of protein-rich edema fluid into the alveoli. Chorioamnionitis plays a major part in triggering fetal lung inflammation, while mechanical ventilation, the application of which is frequently necessary in preterm neonates, also causes ALI by inducing proinflammatory cytokines. Many different ventilation-strategies have been developed in order to reduce potential lung injury. Furthermore, tissue injury may occur as a result of injurious oxygen by-products (Reactive Oxygen Species, ROS), secondary to hyperoxia. Knowledge of the inflammatory pathways that connect intra-amniotic inflammation and ALI can lead to the formulation of novel interventional procedures. Future research should concentrate on the pathophysiology of ALI in preterm neonates and οn possible pharmaceutical interventions targeting prevention and/or resolution of ALI.
Collapse
Affiliation(s)
- Zoe Iliodromiti
- 2nd Department of Obstetrics and Gynecology, University of Athens Medical School, Aretaieio Hospital , Athens , Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ahn SY, Chang YS, Kim SY, Sung DK, Kim ES, Rime SY, Yu WJ, Choi SJ, Oh WI, Park WS. Long-term (postnatal day 70) outcome and safety of intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells in neonatal hyperoxic lung injury. Yonsei Med J 2013; 54:416-24. [PMID: 23364976 PMCID: PMC3575965 DOI: 10.3349/ymj.2013.54.2.416] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This study was performed to evaluate the long-term effects and safety of intratracheal (IT) transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) in neonatal hyperoxic lung injury at postnatal day (P)70 in a rat model. MATERIALS AND METHODS Newborn Sprague Dawley rat pups were subjected to 14 days of hyperoxia (90% oxygen) within 10 hours after birth and allowed to recover at room air until sacrificed at P70. In the transplantation groups, hUCB-MSCs (5×10⁵) were administered intratracheally at P5. At P70, various organs including the heart, lung, liver, and spleen were histologically examined, and the harvested lungs were assessed for morphometric analyses of alveolarization. ED-1, von Willebrand factor, and human-specific nuclear mitotic apparatus protein (NuMA) staining in the lungs and the hematologic profile of blood were evaluated. RESULTS Impaired alveolar and vascular growth, which evidenced by an increased mean linear intercept and decreased amount of von Willebrand factor, respectively, and the hyperoxia-induced inflammatory responses, as evidenced by inflammatory foci and ED-1 positive alveolar macrophages, were attenuated in the P70 rat lungs by IT transplantation of hUCB-MSCs. Although rare, donor cells with human specific NuMA staining were persistently present in the P70 rat lungs. There were no gross or microscopic abnormal findings in the heart, liver, or spleen, related to the MSCs transplantation. CONCLUSION The protective and beneficial effects of IT transplantation of hUCB-MSCs in neonatal hyperoxic lung injuries were sustained for a prolonged recovery period without any long-term adverse effects up to P70.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Yoon Kim
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Kyung Sung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Sun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yub Rime
- Korea Institute of Toxicology, Daejeon, Korea
| | | | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Won Il Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
45
|
Westover AJ, Moss TJM. Effects of intrauterine infection or inflammation on fetal lung development. Clin Exp Pharmacol Physiol 2013; 39:824-30. [PMID: 22816773 DOI: 10.1111/j.1440-1681.2012.05742.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Intrauterine infection or inflammation is common in cases of preterm birth. Preterm infants are at risk of acute respiratory distress as a result of lung immaturity; evidence of exposure to infection and/or inflammation before birth is associated with a reduced risk of neonatal respiratory distress syndrome (RDS). Experimentally induced intrauterine inflammation or infection in sheep causes a precocious increase in pulmonary surfactant in the preterm lungs that improves preterm lung function, consistent with the reduced risk of RDS in human infants exposed to infection and/or inflammation before birth. 2. The effects of intrauterine inflammation on fetal lung development appear to result from direct action of proinflammatory stimuli within the lungs rather than by systemic signals, such as the classical glucocorticoid-mediated lung maturation pathway. However, paracrine and/or autocrine production and/or metabolism of glucocorticoids in fetal lung tissue may occur as a result of inflammation-induced changes in the expression of 11β-hydroxysteroid dehydrogenase (types 1 and 2). 3. Likely candidates that mediate inflammation-induced surfactant production by the preterm lung include prostaglandin E₂ and/or other arachidonic acid metabolites. Intrauterine inflammation induces the expression of enzymes responsible for prostaglandin production in fetal lung tissue. Inhibition of prostaglandin production prevents, at least in part, the effects of inflammation on fetal lungs. 4. Our experiments are identifying mechanisms of surfactant production by the preterm lungs that may be exploited as novel therapies for preventing respiratory distress in preterm infants. Elucidation of the effects of inflammation on the fetal lungs and other organs will allow more refined approaches to the care of preterm infants exposed to inflammation in utero.
Collapse
Affiliation(s)
- Alana J Westover
- The Ritchie Centre, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | | |
Collapse
|
46
|
Chorioamnionitis and lung injury in preterm newborns. Crit Care Res Pract 2013; 2013:890987. [PMID: 23365731 PMCID: PMC3556412 DOI: 10.1155/2013/890987] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 12/13/2012] [Indexed: 11/18/2022] Open
Abstract
There is a strong evidence that histologic chorioamnionitis is associated with a reduction of incidence and severity of respiratory distress syndrome (RDS). Short-term maturational effects on the lungs of extremely premature infants seem to be, however, accompanied by a greater susceptibility of the lung, eventually contributing to an increased risk of bronchopulmonary dysplasia (BPD). Genetic susceptibility to BPD is an evolving area of research and several studies have directly related the risk of BPD to genomic variants. There is a substantial heterogeneity across the studies in the magnitude of the association between chorioamnionitis and BPD, and whether or not the association is statistically significant. Considerable variation is largely dependent on differences of inclusion and exclusion criteria, as well as on clinical and histopathological definitions. The presence of significant publication bias may exaggerate the magnitude of the association. Controlling for publication bias may conduct to adjusted results that are no longer significant. Recent studies generally seem to confirm the effect of chorioamnionitis on RDS incidence, while no effect on BPD is seen. Recent data suggest susceptibility for subsequent asthma to be increased on long-term followup. Additional research on this field is needed.
Collapse
|
47
|
Choi CW, Kim BI. Epidemiology, Clinical Characteristics, and Pathophysiology of Bronchopulmonary Dysplasia. NEONATAL MEDICINE 2013. [DOI: 10.5385/nm.2013.20.3.283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Chang Won Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Beyong Il Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Hartman WR, Smelter DF, Sathish V, Karass M, Kim S, Aravamudan B, Thompson MA, Amrani Y, Pandya HC, Martin RJ, Prakash YS, Pabelick CM. Oxygen dose responsiveness of human fetal airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2012; 303:L711-9. [PMID: 22923637 DOI: 10.1152/ajplung.00037.2012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Maintenance of blood oxygen saturation dictates supplemental oxygen administration to premature infants, but hyperoxia predisposes survivors to respiratory diseases such as asthma. Although much research has focused on oxygen effects on alveoli in the setting of bronchopulmonary dysplasia, the mechanisms by which oxygen affects airway structure or function relevant to asthma are still under investigation. We used isolated human fetal airway smooth muscle (fASM) cells from 18-20 postconceptual age lungs (canalicular stage) to examine oxygen effects on intracellular Ca(2+) ([Ca(2+)](i)) and cellular proliferation. fASM cells expressed substantial smooth muscle actin and myosin and several Ca(2+) regulatory proteins but not fibroblast or epithelial markers, profiles qualitatively comparable to adult human ASM. Fluorescence Ca(2+) imaging showed robust [Ca(2+)](i) responses to 1 μM acetylcholine (ACh) and 10 μM histamine (albeit smaller and slower than adult ASM), partly sensitive to zero extracellular Ca(2+). Compared with adult, fASM showed greater baseline proliferation. Based on this validation, we assessed fASM responses to 10% hypoxia through 90% hyperoxia and found enhanced proliferation at <60% oxygen but increased apoptosis at >60%, effects accompanied by appropriate changes in proliferative vs. apoptotic markers and enhanced mitochondrial fission at >60% oxygen. [Ca(2+)](i) responses to ACh were enhanced for <60% but blunted at >60% oxygen. These results suggest that hyperoxia has dose-dependent effects on structure and function of developing ASM, which could have consequences for airway diseases of childhood. Thus detrimental effects on ASM should be an additional consideration in assessing risks of supplemental oxygen in prematurity.
Collapse
|
49
|
Ahmed MN, Zhang Y, Codipilly C, Zaghloul N, Patel D, Wolin M, Miller EJ. Extracellular superoxide dismutase overexpression can reverse the course of hypoxia-induced pulmonary hypertension. Mol Med 2012; 18:38-46. [PMID: 22045221 DOI: 10.2119/molmed.2011.00339] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 10/27/2011] [Indexed: 01/07/2023] Open
Abstract
Hypoxia leads to free radical production, which has a pivotal role in the pathophysiology of pulmonary hypertension (PH). We hypothesized that treatment with extracellular superoxide dismutase (EC-SOD) could ameliorate the development of PH induced by hypoxia. In vitro studies using pulmonary microvascular endothelial cells showed that cells transfected with EC-SOD had significantly less accumulation of xanthine oxidase and reactive oxygen species than nontransfected cells after hypoxia exposure for 24 h. To study the prophylactic role of EC-SOD, adult male wild-type (WT) and transgenic (TG) mice, with lung-specific overexpression of human EC-SOD (hEC-SOD), were exposed to fraction of inspired oxygen (FiO(2)) 10% for 10 d. After exposure, right ventricular systolic pressure (RVSP), right ventricular mass (RV/S + LV), pulmonary vascular wall thickness (PVWT) and pulmonary artery contraction/relaxation were assessed. TG mice were protected against PH compared with WT mice with significantly lower RVSP (23.9 ± 1.24 versus 47.2 ± 3.4), RV/S + LV (0.287 ± 0.015 versus 0.335 ± 0.022) and vascular remodeling, indicated by PVWT (14.324 ± 1.107 versus 18.885 ± 1.529). Functional studies using pulmonary arteries isolated from mice indicated that EC-SOD prevents hypoxia-mediated attenuation of nitric oxide-induced relaxation. Therapeutic potential was assessed by exposing WT mice to FiO(2) 10% for 10 d. Half of the group was transfected with plasmid containing cDNA encoding human EC-SOD. The remaining animals were transfected with empty vector. Both groups were exposed to FiO(2) 10% for a further 10 d. Transfected mice had significantly reduced RVSP (18.97 ± 1.12 versus 41.3 ± 1.5), RV/S + LV (0.293 ± 0.012 versus 0.372 ± 0.014) and PVWT (12.51 ± 0.72 versus 18.98 ± 1.24). On the basis of these findings, we concluded that overexpression of EC-SOD prevents the development of PH and ameliorates established PH.
Collapse
Affiliation(s)
- Mohamed N Ahmed
- Cohen Children's Medical Center, North Shore-Long Island Jewish Health System, New Hyde Park, New York, United States of America.
| | | | | | | | | | | | | |
Collapse
|
50
|
Jiang JS, Lang YD, Chou HC, Shih CM, Wu MY, Chen CM, Wang LF. Activation of the renin-angiotensin system in hyperoxia-induced lung fibrosis in neonatal rats. Neonatology 2012; 101:47-54. [PMID: 21791939 DOI: 10.1159/000329451] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 05/16/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND Oxygen toxicity plays an important role in lung injury and may lead to bronchopulmonary dysplasia. We previously demonstrated that hyperoxia activated the renin-angiotensin system (RAS) in cultured human fetal lung fibroblasts. OBJECTIVE To examine whether the upregulation of RAS components is associated with hyperoxia-induced lung fibrosis in neonatal Sprague-Dawley rats. METHODS Experimental rat pups were exposed to 1 week of >95% O(2) and a further 2 weeks of 60% O(2). Control pups were exposed to room air over the same periods. Lung tissues were taken for biochemical and histochemical assays on postnatal days 7 and 21. RESULTS Hyperoxia significantly increased total collagen content and the expression of type I collagen and alpha smooth muscle actin when compared to control rats. RAS components including angiotensinogen, angiotensin-converting enzyme, angiotensin II, and angiotensin II type 1 receptor were significantly upregulated by hyperoxia. The results also demonstrated that only the extracellular signal-regulated kinase (ERK) signaling pathway was activated by hyperoxia exposure. p38 mitogen-activated protein kinase and c-Jun N-terminal kinase were not activated. CONCLUSIONS Local RAS activation is involved in the pathogenesis of hyperoxia-induced lung fibrosis in neonatal rats. ERK phosphorylation might mediate angiotensin II type 1 receptor activation.
Collapse
Affiliation(s)
- Jiunn-Song Jiang
- Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|