1
|
Liang Y, Ruan W, Jiang Y, Smalling R, Yuan X, Eltzschig HK. Interplay of hypoxia-inducible factors and oxygen therapy in cardiovascular medicine. Nat Rev Cardiol 2023; 20:723-737. [PMID: 37308571 PMCID: PMC11014460 DOI: 10.1038/s41569-023-00886-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 06/14/2023]
Abstract
Mammals have evolved to adapt to differences in oxygen availability. Although systemic oxygen homeostasis relies on respiratory and circulatory responses, cellular adaptation to hypoxia involves the transcription factor hypoxia-inducible factor (HIF). Given that many cardiovascular diseases involve some degree of systemic or local tissue hypoxia, oxygen therapy has been used liberally over many decades for the treatment of cardiovascular disorders. However, preclinical research has revealed the detrimental effects of excessive use of oxygen therapy, including the generation of toxic oxygen radicals or attenuation of endogenous protection by HIFs. In addition, investigators in clinical trials conducted in the past decade have questioned the excessive use of oxygen therapy and have identified specific cardiovascular diseases in which a more conservative approach to oxygen therapy could be beneficial compared with a more liberal approach. In this Review, we provide numerous perspectives on systemic and molecular oxygen homeostasis and the pathophysiological consequences of excessive oxygen use. In addition, we provide an overview of findings from clinical studies on oxygen therapy for myocardial ischaemia, cardiac arrest, heart failure and cardiac surgery. These clinical studies have prompted a shift from liberal oxygen supplementation to a more conservative and vigilant approach to oxygen therapy. Furthermore, we discuss the alternative therapeutic strategies that target oxygen-sensing pathways, including various preconditioning approaches and pharmacological HIF activators, that can be used regardless of the level of oxygen therapy that a patient is already receiving.
Collapse
Affiliation(s)
- Yafen Liang
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Wei Ruan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yandong Jiang
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard Smalling
- Department of Cardiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaoyi Yuan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Holger K Eltzschig
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- Outcomes Research Consortium, Cleveland, OH, USA
| |
Collapse
|
2
|
Rao S, Liu M, Iosef C, Knutsen C, Alvira CM. Endothelial-specific loss of IKKβ disrupts pulmonary endothelial angiogenesis and impairs postnatal lung growth. Am J Physiol Lung Cell Mol Physiol 2023; 325:L299-L313. [PMID: 37310763 PMCID: PMC10625829 DOI: 10.1152/ajplung.00034.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023] Open
Abstract
Pulmonary angiogenesis drives alveolarization, but the transcriptional regulators directing pulmonary angiogenesis remain poorly defined. Global, pharmacological inhibition of nuclear factor-kappa B (NF-κB) impairs pulmonary angiogenesis and alveolarization. However, establishing a definitive role for NF-κB in pulmonary vascular development has been hindered by embryonic lethality induced by constitutive deletion of NF-κB family members. We created a mouse model allowing inducible deletion of the NF-κB activator, IKKβ, in endothelial cells (ECs) and assessed the effect on lung structure, endothelial angiogenic function, and the lung transcriptome. Embryonic deletion of IKKβ permitted lung vascular development but resulted in a disorganized vascular plexus, while postnatal deletion significantly decreased radial alveolar counts, vascular density, and proliferation of both endothelial and nonendothelial lung cells. Loss of IKKβ impaired survival, proliferation, migration, and angiogenesis in primary lung ECs in vitro, in association with decreased expression of VEGFR2 and activation of downstream effectors. Loss of endothelial IKKβ in vivo induced broad changes in the lung transcriptome with downregulation of genes related to mitotic cell cycle, extracellular matrix (ECM)-receptor interaction, and vascular development, and the upregulation of genes related to inflammation. Computational deconvolution suggested that loss of endothelial IKKβ decreased general capillary, aerocyte capillary, and alveolar type I cell abundance. Taken together, these data definitively establish an essential role for endogenous endothelial IKKβ signaling during alveolarization. A deeper understanding of the mechanisms directing this developmental, physiological activation of IKKβ in the lung vasculature may provide novel targets for the development of strategies to enhance beneficial proangiogenic signaling in lung development and disease.NEW & NOTEWORTHY This study highlights the cell-specific complexity of nuclear factor kappa B signaling in the developing lung by demonstrating that inducible loss of IKKβ in endothelial cells impairs alveolarization, disrupts EC angiogenic function, and broadly represses genes important for vascular development.
Collapse
Affiliation(s)
- Shailaja Rao
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Min Liu
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristiana Iosef
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Carsten Knutsen
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| |
Collapse
|
3
|
Tessema B, Sack U, Serebrovska Z, König B, Egorov E. Effects of Hyperoxia on Aging Biomarkers: A Systematic Review. FRONTIERS IN AGING 2022; 2:783144. [PMID: 35822043 PMCID: PMC9261365 DOI: 10.3389/fragi.2021.783144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022]
Abstract
The effects of short-term hyperoxia on age-related diseases and aging biomarkers have been reported in animal and human experiments using different protocols; however, the findings of the studies remain conflicting. In this systematic review, we summarized the existing reports in the effects of short-term hyperoxia on age-related diseases, hypoxia-inducible factor 1α (HIF-1α), and other oxygen-sensitive transcription factors relevant to aging, telomere length, cellular senescence, and its side effects. This review was done as described in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. A systematic search was done in PubMed, Google Scholar, and Cochrane Library and from the references of selected articles to identify relevant studies until May 2021. Of the total 1,699 identified studies, 17 were included in this review. Most of the studies have shown significant effects of short-term hyperoxia on age-related diseases and aging biomarkers. The findings of the studies suggest the potential benefits of short-term hyperoxia in several clinical applications such as for patients undergoing stressful operations, restoration of cognitive function, and the treatment of severe traumatic brain injury. Short-term hyperoxia has significant effects in upregulation or downregulation of transcription factors relevant to aging such as HIF-1α, nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB), and nuclear factor (erythroid-derived 2)-like 2 (NRF2) among others. Short-term hyperoxia also has significant effects to increase antioxidant enzymes, and increase telomere length and clearance of senescent cells. Some of the studies have also reported adverse consequences including mitochondrial DNA damage and nuclear cataract formation depending on the dose and duration of oxygen exposure. In conclusion, short-term hyperoxia could be a feasible treatment option to treat age-related disease and to slow aging because of its ability to increase antioxidant enzymes, significantly increase telomere length and clearance of senescent cells, and improve cognitive function, among others. The reported side effects of hyperoxia vary depending on the dose and duration of exposure. Therefore, it seems that additional studies for better understanding the beneficial effects of short-term hyperoxia and for minimizing side effects are necessary for optimal clinical application.
Collapse
Affiliation(s)
- Belay Tessema
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Department of Medical Microbiology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Ulrich Sack
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Zoya Serebrovska
- Department of Hypoxic States Investigation, Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Brigitte König
- Institute of Medical Microbiology and Epidemiology of Infectious Diseases, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Egor Egorov
- Ipam Institute for Preventive and Anti-Aging Medicine, Berlin, Germany
| |
Collapse
|
4
|
Huang X, Han D, Wei Y, Lin B, Zeng D, Zhang Y, Wei B, Huang Z, Chen X, Yang C. Decreased plasma levels of PDGF-BB, VEGF-A, and HIF-2α in preterm infants after ibuprofen treatment. Front Pediatr 2022; 10:919879. [PMID: 35958170 PMCID: PMC9361044 DOI: 10.3389/fped.2022.919879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Ibuprofen is one of the most common non-steroidal anti-inflammatory drugs used to close patent ductus arteriosus (PDA) in preterm infants. PDA is associated with bronchopulmonary dysplasia (BPD), while PDA closure by ibuprofen did not reduce the incidence of BPD or death. Previous studies have indicated an anti-angiogenesis effect of ibuprofen. This study investigated the change of angiogenic factors after ibuprofen treatment in preterm infants. METHODS Preterm infants with hemodynamically significant PDA (hsPDA) were included. After confirmed hsPDA by color doppler ultrasonography within 1 week after birth, infants received oral ibuprofen for three continuous days. Paired plasma before and after the ibuprofen treatment was collected and measured by ELISA to determine the concentrations of platelet-derived growth factor-BB (PDGF-BB) and vascular endothelial growth factor A (VEGF-A), and hypoxia-inducible factor-2α (HIF-2α). RESULTS 17 paired plasma from infants with hsPDA were collected. The concentration of PDGF-BB and VEGF-A significantly decreased after ibuprofen treatment (1,908 vs. 442 pg/mL for PDGF-BB, 379 vs. 174 pg/mL for VEGF-A). HIF-2α level showed a tendency to decrease after ibuprofen treatment, although the reduction was not statistically significant (p = 0.077). CONCLUSION This study demonstrated decreased vascular growth factors after ibuprofen exposure in hsPDA infants.
Collapse
Affiliation(s)
- Xuemei Huang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China.,Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, China
| | - Dongshan Han
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yanfei Wei
- Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, China
| | - Bingchun Lin
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Dingyuan Zeng
- Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, China.,Guangxi Health Commission Key Laboratory of Birth Cohort Study in Pregnant Women of Advanced Age, Liuzhou, China
| | - Yu Zhang
- Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, China.,Guangxi Health Commission Key Laboratory of Birth Cohort Study in Pregnant Women of Advanced Age, Liuzhou, China
| | - Ba Wei
- Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Liuzhou, China
| | - Zhifeng Huang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Xueyu Chen
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Chuanzhong Yang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| |
Collapse
|
5
|
Sudhadevi T, Jafri A, Ha AW, Basa P, Thomas JM, Fu P, Wary K, Mehta D, Natarajan V, Harijith A. Hyperoxia-induced S1P 1 signaling reduced angiogenesis by suppression of TIE-2 leading to experimental bronchopulmonary dysplasia. Cell Biochem Biophys 2021; 79:561-573. [PMID: 34176100 PMCID: PMC8551021 DOI: 10.1007/s12013-021-01014-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 01/16/2023]
Abstract
INTRODUCTION We have earlier shown that hyperoxia (HO)-induced sphingosine kinase 1 (SPHK1)/sphingosine-1-phosphate (S1P) signaling contribute to bronchopulmonary dysplasia (BPD). S1P acts through G protein-coupled receptors, S1P1 through S1P5. Further, we noted that heterozygous deletion of S1pr1 ameliorated the HO-induced BPD in the murine model. The mechanism by which S1P1 signaling contributes to HO-induced BPD was explored. METHODS S1pr1+/+ and S1pr1+/- mice pups were exposed to either room air (RA) or HO (75% oxygen) for 7 days from PN 1-7. Lung injury and alveolar simplification was evaluated. Lung protein expression was determined by Western blotting and immunohistochemistry (IHC). In vitro experiments were performed using human lung microvascular endothelial cells (HLMVECs) with S1P1 inhibitor, NIBR0213 to interrogate the S1P1 signaling pathway. RESULTS HO increased the expression of S1pr1 gene as well as S1P1 protein in both neonatal lungs and HLMVECs. The S1pr1+/- neonatal mice showed significant protection against HO-induced BPD which was accompanied by reduced inflammation markers in the bronchoalveolar lavage fluid. HO-induced reduction in ANG-1, TIE-2, and VEGF was rescued in S1pr1+/- mouse, accompanied by an improvement in the number of arterioles in the lung. HLMVECs exposed to HO increased the expression of KLF-2 accompanied by reduced expression of TIE-2, which was reversed with S1P1 inhibition. CONCLUSION HO induces S1P1 followed by reduced expression of angiogenic factors. Reduction of S1P1 signaling restores ANG-1/ TIE-2 signaling leading to improved angiogenesis and alveolarization thus protecting against HO-induced neonatal lung injury.
Collapse
Affiliation(s)
- Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alison W Ha
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Jaya M Thomas
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Panfeng Fu
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Kishore Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
6
|
Li Y, Luo NC, Zhang X, Hara T, Inadomi C, Li TS. Prolonged oxygen exposure causes the mobilization and functional damage of stem or progenitor cells and exacerbates cardiac ischemia or reperfusion injury in healthy mice. J Cell Physiol 2021; 236:6657-6665. [PMID: 33554327 DOI: 10.1002/jcp.30317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/31/2020] [Accepted: 01/27/2021] [Indexed: 11/09/2022]
Abstract
Oxygen is often administered to patients and occasionally to healthy individuals as well; however, the cellular toxicity of oxygen, especially following prolonged exposure, is widely known. To evaluate the potential effect of oxygen exposure on circulating stem/progenitor cells and cardiac ischemia/reperfusion (I/R) injury, we exposed healthy adult mice to 100% oxygen for 20 or 60 min. We then examined the c-kit-positive stem/progenitor cells and colony-forming cells and measured the cytokine/chemokine levels in peripheral blood. We also induced cardiac I/R injury in mice at 3 h after 60 min of oxygen exposure and examined the recruitment of inflammatory cells and the fibrotic area in the heart. The proportion of c-kit-positive stem/progenitor cells significantly increased in peripheral blood at 3 and 24 h after oxygen exposure for either 20 or 60 min (p < .01 vs. control). However, the abundance of colony-forming cells in peripheral blood conversely decreased at 3 and 24 h after oxygen exposure for only 60 min (p < .05 vs. control). Oxygen exposure for either 20 or 60 min resulted in significantly decreased plasma vascular endothelial growth factor levels at 3 h, whereas oxygen exposure for only 60 min reduced plasma insulin-like growth factor 1 levels at 24 h (p < .05 vs. control). Protein array indicated the increase in the levels of some cytokines/chemokines, such as CXCL6 (GCP-2) at 24 h after 60 min of oxygen exposure. Moreover, oxygen exposure for 60 min enhanced the recruitment of Ly6g- and CD11c-positive inflammatory cells at 3 days (p < .05 vs. control) and increased the fibrotic area at 14 days in the heart after I/R injury (p < .05 vs. control). Prolonged oxygen exposure induced the mobilization and functional impairment of stem/progenitor cells and likely enhanced inflammatory responses to exacerbate cardiac I/R injury in healthy mice.
Collapse
Affiliation(s)
- Yu Li
- School of Medicine, Nanchang University, Nanchang, Jiangxi, China
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Na-Chuan Luo
- School of Medicine, Nanchang University, Nanchang, Jiangxi, China
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Xu Zhang
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tetsuya Hara
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chiaki Inadomi
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
7
|
Wang Y, Jiang L. Role of vitamin D-vitamin D receptor signaling on hyperoxia-induced bronchopulmonary dysplasia in neonatal rats. Pediatr Pulmonol 2021; 56:2335-2344. [PMID: 33878208 DOI: 10.1002/ppul.25418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/25/2021] [Accepted: 04/03/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Vitamin D exerts therapeutic effects on bronchopulmonary dysplasia (BPD), but its underlying mechanisms remain unclear. The present study was designed to investigate the effects of vitamin D on hyperoxia-induced BPD and elucidate the underlying mechanisms. METHODS Neonatal rats were exposed to either room air (control) or 75% O2 (hyperoxia) and intraperitoneally injected with vitamin D3. After 14 days, a histopathological examination was performed in the lungs of rats. Serum 25-hydroxyvitamin D (25OHD) was measured by liquid chromatography-tandom mass spectrometry (LC-MS)/MS. Interleukin 1 beta (IL-1β) and interferon gamma (IFN-γ) were measured by specific enzyme-linked immunosorbent assays. The messenger RNA and protein levels of vitamin D receptor (VDR), vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), and hypoxia-inducible factor 1α (HIF-1α) were determined by real-time quantitative reverse transcription polymerase chain reaction and immunoblot analysis, respectively. RESULTS Treatment with vitamin D3 increased serum 25OHD and upregulated VDR in lung tissues with or without hyperoxia. In addition, treatment with vitamin D3 attenuated alveolar simplification, increased VEGF and VEGFR2, and protected alveolar simplification induced by hyperoxia. Furthermore, treatment with vitamin D3 resulted in a decrease of IL-1β and IFN-γ and an increase of HIF-1α in lung tissues under hyperoxia conditions. CONCLUSION Vitamin D exerts protective effects on hyperoxia-induced BPD in neonatal rats by regulating vitamin D-VDR signaling pathways.
Collapse
Affiliation(s)
- Yuchun Wang
- Department of Pediatrics, Hebei Medical University, Shijiazhuang, Hebei, China.,Department of Neonatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lian Jiang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Systemic Factors Associated with a Thinner Choroid in Preterm Infants. OPHTHALMOLOGY SCIENCE 2021; 1:100032. [PMID: 36249299 PMCID: PMC9559969 DOI: 10.1016/j.xops.2021.100032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/30/2022]
Abstract
Purpose To identify systemic health factors associated with a thinner choroid, which has been hypothesized as a cause of poor visual outcomes in low–birth weight infants. Design The prospective, observational Study of Eye Imaging in Preterm Infants (BabySTEPS) enrolled infants recommended for retinopathy of prematurity screening based on the American Association of Pediatrics guidelines. Participants Infants who underwent imaging with investigational handheld OCT at 36 ± 1 weeks’ postmenstrual age (PMA) as part of BabySTEPS. Methods Average choroidal thickness was measured across the central subfoveal 1 mm. We concurrently collected maternal and infant clinical health data. Univariate and multivariate linear regression analyses were performed to evaluate factors associated with choroidal thickness. The left and right eyes showed similar thicknesses, so their average was used for analysis. Main Outcomes Measures Association between infant health factors and subfoveal choroidal thickness. Results Subfoveal choroidal thickness was measurable in 82 of 85 infants and 94% of eyes. Mean choroidal thickness was 231 ± 78 μm. In the univariate analysis, a thinner choroid was associated with decreased growth velocity (P < 0.001), lower birth weight (P < 0.001), smaller head circumference (P < 0.001), younger gestational age (P = 0.01), the presence of patent ductus arteriosus (P = 0.05), sepsis or necrotizing enterocolitis (P = 0.03), bronchopulmonary dysplasia (P = 0.03), pulmonary interstitial emphysema (P = 0.002), more days on oxygen support (P < 0.001), and being on oxygen support at 36 weeks (P < 0.001) and at the time of imaging (P < 0.001). In the multivariate analysis, growth velocity (P = 0.002) and oxygen support at the time of OCT imaging (P = 0.004) remained associated with a thinner choroid. Conclusions A thinner choroid is associated independently with growth velocity and receiving oxygen support at 36 ± 1 weeks PMA. This suggests that choroidal development in preterm infants may be related to growth rate in the first weeks of life and the prolonged use of supplemental oxygen. Longitudinal studies are needed to assess differences in choroidal thickness before 36 weeks PMA and to assess their impact on visual outcomes.
Collapse
|
9
|
Roberts K, Stepanovich G, Bhatt-Mehta V, Donn SM. New Pharmacologic Approaches to Bronchopulmonary Dysplasia. J Exp Pharmacol 2021; 13:377-396. [PMID: 33790663 PMCID: PMC8006962 DOI: 10.2147/jep.s262350] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary Dysplasia is the most common long-term respiratory morbidity of preterm infants, with the risk of development proportional to the degree of prematurity. While its pathophysiologic and histologic features have changed over time as neonatal demographics and respiratory therapies have evolved, it is now thought to be characterized by impaired distal lung growth and abnormal pulmonary microvascular development. Though the exact sequence of events leading to the development of BPD has not been fully elucidated and likely varies among patients, it is thought to result from inflammatory and mechanical/oxidative injury from chronic ventilatory support in fragile, premature lungs susceptible to injury from surfactant deficiency, structural abnormalities, inadequate antioxidant defenses, and a chest wall that is more compliant than the lung. In addition, non-pulmonary issues may adversely affect lung development, including systemic infections and insufficient nutrition. Once BPD has developed, its management focuses on providing adequate gas exchange while promoting optimal lung growth. Pharmacologic strategies to ameliorate or prevent BPD continue to be investigated. A variety of agents, to be reviewed henceforth, have been developed or re-purposed to target different points in the pathways that lead to BPD, including anti-inflammatories, diuretics, steroids, pulmonary vasodilators, antioxidants, and a number of molecules involved in the cell signaling cascade thought to be involved in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Katelyn Roberts
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gretchen Stepanovich
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Varsha Bhatt-Mehta
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- College of Pharmacy, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Steven M Donn
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Amarelle L, Quintela L, Hurtado J, Malacrida L. Hyperoxia and Lungs: What We Have Learned From Animal Models. Front Med (Lausanne) 2021; 8:606678. [PMID: 33768102 PMCID: PMC7985075 DOI: 10.3389/fmed.2021.606678] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Although oxygen (O2) is essential for aerobic life, it can also be an important source of cellular damage. Supra-physiological levels of O2 determine toxicity due to exacerbated reactive oxygen species (ROS) production, impairing the homeostatic balance of several cellular processes. Furthermore, injured cells activate inflammation cascades, amplifying the tissue damage. The lung is the first (but not the only) organ affected by this condition. Critically ill patients are often exposed to several insults, such as mechanical ventilation, infections, hypo-perfusion, systemic inflammation, and drug toxicity. In this scenario, it is not easy to dissect the effect of oxygen toxicity. Translational investigations with animal models are essential to explore injuring stimuli in controlled experimental conditions, and are milestones in understanding pathological mechanisms and developing therapeutic strategies. Animal models can resemble what happens in critical care or anesthesia patients under mechanical ventilation and hyperoxia, but are also critical to explore the effect of O2 on lung development and the role of hyperoxic damage on bronchopulmonary dysplasia. Here, we set out to review the hyperoxia effects on lung pathology, contributing to the field by describing and analyzing animal experimentation's main aspects and its implications on human lung diseases.
Collapse
Affiliation(s)
- Luciano Amarelle
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Lucía Quintela
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Javier Hurtado
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Leonel Malacrida
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay.,Advanced Bioimaging Unit, Institut Pasteur Montevideo and Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
11
|
Mandell EW, Ryan S, Seedorf GJ, Gonzalez T, Smith BJ, Fleet JC, Abman SH. Maternal Vitamin D Deficiency Causes Sustained Impairment of Lung Structure and Function and Increases Susceptibility to Hyperoxia-induced Lung Injury in Infant Rats. Am J Respir Cell Mol Biol 2020; 63:79-91. [PMID: 32135073 DOI: 10.1165/rcmb.2019-0295oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vitamin D deficiency (VDD) during pregnancy is associated with increased respiratory morbidities and risk for chronic lung disease after preterm birth. However, the direct effects of maternal VDD on perinatal lung structure and function and whether maternal VDD increases the susceptibility of lung injury due to hyperoxia are uncertain. In the present study, we sought to determine whether maternal VDD is sufficient to impair lung structure and function and whether VDD increases the impact of hyperoxia on the developing rat lung. Four-week-old rats were fed VDD chow and housed in a room shielded from ultraviolet A/B light to achieve 25-hydroxyvitamin D concentrations <10 ng/ml at mating and throughout lactation. Lung structure was assessed at 2 weeks for radial alveolar count, mean linear intercept, pulmonary vessel density, and lung function (lung compliance and resistance). The effects of hyperoxia for 2 weeks after birth were assessed after exposure to fraction of inspired oxygen of 0.95. At 2 weeks, VDD offspring had decreased alveolar and vascular growth and abnormal airway reactivity and lung function. Impaired lung structure and function in VDD offspring were similar to those observed in control rats exposed to postnatal hyperoxia alone. Maternal VDD causes sustained abnormalities of distal lung growth, increases in airway hyperreactivity, and abnormal lung mechanics during infancy. These changes in VDD pups were as severe as those measured after exposure to postnatal hyperoxia alone. We speculate that antenatal disruption of vitamin D signaling increases the risk for late-childhood respiratory disease.
Collapse
Affiliation(s)
| | - Sharon Ryan
- Pediatric Heart Lung Center.,Section of Neonatology, and
| | - Gregory J Seedorf
- Pediatric Heart Lung Center.,Section of Pulmonary Medicine, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Tania Gonzalez
- Pediatric Heart Lung Center.,Section of Neonatology, and
| | - Bradford J Smith
- Department of Bioengineering, College of Engineering and Applied Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
| | - James C Fleet
- Department of Foods and Nutrition, and.,Interdepartmental Nutrition Program, Purdue University, West Lafayette, Indiana
| | - Steven H Abman
- Pediatric Heart Lung Center.,Section of Pulmonary Medicine, Department of Pediatrics, Children's Hospital Colorado and University of Colorado Anschutz Medical Center, Aurora, Colorado
| |
Collapse
|
12
|
Paturi B, Ryan RM, Nielsen L, Wang H, Kumar VHS. Effects of antioxidant MnTBAP on angiogenesis in newborn mice with hyperoxic lung injury. J Neonatal Perinatal Med 2020; 14:53-60. [PMID: 32804105 DOI: 10.3233/npm-200483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Oxygen toxicity mediated by reactive oxygen species (ROS) plays an essential role in the development of bronchopulmonary dysplasia in premature infants. By reducing oxidative stress, antioxidants protect the immature lung. We studied the effects of MnTBAP, a catalytic antioxidant on angiogenesis and alveolar growth following neonatal hyperoxia. METHODS Newborn mouse litters randomized to room air (RA) or >95% O2 for 72 hours from day 4 (D4) to D7 to receive either MnTBAP (10 mg/kg/d) or saline intraperitoneally (every 24 h for three doses). Lungs harvested for angiogenic gene expression, protein expression, and histopathology post-hyperoxia exposure. Radial alveolar count (RAC), mean linear intercept (MLI) and vessel density assessed by histopathology. RESULTS Angiogenic gene expression was significantly lower in the hyperoxia group compared to the RA group. The protein expression for VEGF and its receptor, VEGFR1, was significantly lower following treatment with MnTBAP compared to hyperoxia alone. Expression of VEGFR2, Angiopoietin-1 and TIE2, were substantially higher in the RA groups compared to hyperoxia groups with or without MnTBAP. Hyperoxia groups demonstrated alveolar simplification. MnTBAP reduced vessel density and failed to improve alveolar growth following hyperoxia. CONCLUSIONS MnTBAP, a catalytic antioxidant, does not offer protection from hyperoxia-induced alveolar impairment. The lack of angiogenic upregulation by MnTBAP may contribute to alveolar simplification in newborn mice.
Collapse
Affiliation(s)
- B Paturi
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - R M Ryan
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - L Nielsen
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - H Wang
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| | - V H S Kumar
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
13
|
Salaets T, Aertgeerts M, Gie A, Vignero J, de Winter D, Regin Y, Jimenez J, Vande Velde G, Allegaert K, Deprest J, Toelen J. Preterm birth impairs postnatal lung development in the neonatal rabbit model. Respir Res 2020; 21:59. [PMID: 32085773 PMCID: PMC7035772 DOI: 10.1186/s12931-020-1321-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
Background Bronchopulmonary dysplasia continues to cause important respiratory morbidity throughout life, and new therapies are needed. The common denominator of all BPD cases is preterm birth, however most preclinical research in this area focusses on the effect of hyperoxia or mechanical ventilation. In this study we investigated if and how prematurity affects lung structure and function in neonatal rabbits. Methods Pups were delivered on either day 28 or day 31. For each gestational age a group of pups was harvested immediately after birth for lung morphometry and surfactant protein B and C quantification. All other pups were hand raised and harvested on day 4 for the term pups and day 7 for the preterm pups (same corrected age) for lung morphometry, lung function testing and qPCR. A subset of pups underwent microCT and dark field imaging on day 0, 2 and 4 for terms and on day 0, 3, 5 and 7 for preterms. Results Preterm pups assessed at birth depicted a more rudimentary lung structure (larger alveoli and thicker septations) and a lower expression of surfactant proteins in comparison to term pups. MicroCT and dark field imaging revealed delayed lung aeration in preterm pups, in comparison to term pups. Preterm birth led to smaller pups, with smaller lungs with a lower alveolar surface area on day 7/day 4. Furthermore, preterm birth affected lung function with increased tissue damping, tissue elastance and resistance and decreased dynamic compliance. Expression of vascular endothelial growth factor (VEGFA) was significantly decreased in preterm pups, however in the absence of structural vascular differences. Conclusions Preterm birth affects lung structure and function at birth, but also has persistent effects on the developing lung. This supports the use of a preterm animal model, such as the preterm rabbit, for preclinical research on BPD. Future research that focuses on the identification of pathways that are involved in in-utero lung development and disrupted by pre-term birth, could lead to novel therapeutic strategies for BPD.
Collapse
Affiliation(s)
- Thomas Salaets
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Margo Aertgeerts
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - André Gie
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Janne Vignero
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Derek de Winter
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Yannick Regin
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Julio Jimenez
- Facultad de Medicina, Universidad del Desarollo, Clínica Alemana, Santiago de Chile, Chile
| | | | - Karel Allegaert
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.,Department of Clinical Pharmacy, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jan Deprest
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.,Institute for Women's Health, University College London Hospital, London, UK
| | - Jaan Toelen
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
14
|
Guerra K, Bryan C, Dapaah-Siakwan F, Sammour I, Drummond S, Zambrano R, Chen P, Huang J, Sharma M, Shrager S, Benny M, Wu S, Young KC. Intra-tracheal administration of a naked plasmid expressing stromal derived factor-1 improves lung structure in rodents with experimental bronchopulmonary dysplasia. Respir Res 2019; 20:255. [PMID: 31718614 PMCID: PMC6852969 DOI: 10.1186/s12931-019-1224-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is characterized by alveolar simplification and disordered angiogenesis. Stromal derived factor-1 (SDF-1) is a chemokine which modulates cell migration, proliferation, and angiogenesis. Here we tested the hypothesis that intra-tracheal (IT) administration of a naked plasmid DNA expressing SDF-1 would attenuate neonatal hyperoxia-induced lung injury in an experimental model of BPD, by promoting angiogenesis. Design/methods Newborn Sprague-Dawley rat pups (n = 18–20/group) exposed to room air (RA) or hyperoxia (85% O2) from postnatal day (P) 1 to 14 were randomly assigned to receive IT a naked plasmid expressing SDF-1, JVS-100 (Juventas Therapeutics, Cleveland, Ohio) or placebo (PL) on P3. Lung alveolarization, angiogenesis, inflammation, vascular remodeling and pulmonary hypertension (PH) were assessed on P14. PH was determined by measuring right ventricular systolic pressure (RVSP) and the weight ratio of the right to left ventricle + septum (RV/LV + S). Capillary tube formation in SDF-1 treated hyperoxia-exposed human pulmonary microvascular endothelial cells (HPMEC) was determined by matrigel assay. Data is expressed as mean ± SD and analyzed by two-way ANOVA. Results Exposure of neonatal pups to 14 days of hyperoxia decreased lung SDF-1 gene expression. Moreover, whilst hyperoxia exposure inhibited capillary tube formation in HPMEC, SDF-1 treatment increased tube length and branching in HPMEC. PL-treated hyperoxia-exposed pups had decreased alveolarization and lung vascular density. This was accompanied by an increase in RVSP, RV/LV + S, pulmonary vascular remodeling and inflammation. In contrast, IT JVS-100 improved lung structure, reduced inflammation, PH and vascular remodeling. Conclusions Intratracheal administration of a naked plasmid expressing SDF-1 improves alveolar and vascular structure in an experimental model of BPD. These findings suggest that therapies which modulate lung SDF-1 expression may have beneficial effects in preterm infants with BPD.
Collapse
Affiliation(s)
- Kasonya Guerra
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Carleene Bryan
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Frederick Dapaah-Siakwan
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Ibrahim Sammour
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Shelly Drummond
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Sebastian Shrager
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA. .,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA. .,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue RM-344, Miami, FL, 33136, USA.
| |
Collapse
|
15
|
Ofman G, Tipple TE. Antioxidants & bronchopulmonary dysplasia: Beating the system or beating a dead horse? Free Radic Biol Med 2019; 142:138-145. [PMID: 30769161 DOI: 10.1016/j.freeradbiomed.2019.01.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/13/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Preterm birth is a primary cause of worldwide childhood mortality. Bronchopulmonary dysplasia, characterized by impaired alveolar and lung vascular development, affects 25-50% of extremely low birth weight (BW; <1 kg) infants. Abnormalities in lung function persist into childhood in affected infants and are second only to asthma in terms of childhood respiratory disease healthcare costs. While advances in the medical care of preterm infants have reduced mortality, the incidence of BPD has not decreased in the past 10 years. Reactive oxygen intermediates play a key role in the development of lung disease but, despite promising preclinical therapies, antioxidants have failed to translate into meaningful clinical interventions to decrease the incidence of lung disease in premature infants. In this review we will summarize the state of the art research developments in regards to antioxidants and premature lung disease and discuss the limitations of antioxidant therapies in order to more fully comprehend the reasons why therapeutic antioxidant administration failed to prevent BPD. Finally we will review promising therapeutic strategies and targets.
Collapse
Affiliation(s)
- Gaston Ofman
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Trent E Tipple
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
16
|
Dapaah-Siakwan F, Zambrano R, Luo S, Duncan MR, Kerr N, Donda K, Vaccari JPDR, Keane RW, Dietrich WD, Benny M, Young K, Wu S. Caspase-1 Inhibition Attenuates Hyperoxia-induced Lung and Brain Injury in Neonatal Mice. Am J Respir Cell Mol Biol 2019; 61:341-354. [DOI: 10.1165/rcmb.2018-0192oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Fredrick Dapaah-Siakwan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Ronald Zambrano
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shihua Luo
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Matthew R. Duncan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Nadine Kerr
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - Keyur Donda
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Juan Pablo de Rivero Vaccari
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Robert W. Keane
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - W. Dalton Dietrich
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Merline Benny
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Karen Young
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shu Wu
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| |
Collapse
|
17
|
Dumpa V, Nielsen L, Wang H, Kumar VHS. Caffeine is associated with improved alveolarization and angiogenesis in male mice following hyperoxia induced lung injury. BMC Pulm Med 2019; 19:138. [PMID: 31362742 PMCID: PMC6668145 DOI: 10.1186/s12890-019-0903-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background Caffeine therapy for apnea of prematurity reduces the incidence of bronchopulmonary dysplasia (BPD) in premature neonates. Several mechanisms, including improvement in pulmonary mechanics underly beneficial effects of caffeine in BPD. As vascular development promotes alveologenesis, we hypothesized that caffeine might enhance angiogenesis in the lung, promoting lung growth, thereby attenuating BPD. Methods C57Bl/6 mice litters were randomized within 12 h of birth to room air (RA) or 95%O2 to receive caffeine (20 mg/kg/day) or placebo for 4 days and recovered in RA for 12wks. The lung mRNA and protein expression for hypoxia-inducible factors (HIF) and angiogenic genes performed on day 5. Lung morphometry and vascular remodeling assessed on inflation fixed lungs at 12wks. Results Caffeine and hyperoxia in itself upregulate HIF-2α and vascular endothelial growth factor gene expression. Protein expression of HIF-2α and VEGFR1 were higher in hyperoxia/caffeine and angiopoietin-1 lower in hyperoxia. An increase in radial alveolar count, secondary septal count, and septal length with a decrease in mean linear intercept indicate an amelioration of hyperoxic lung injury by caffeine. An increase in vessel surface area and a significant reduction in smooth muscle thickness of the pulmonary arterioles may suggest a beneficial effect of caffeine on vascular remodeling in hyperoxia, especially in male mice. Conclusions Postnatal caffeine by modulating angiogenic gene expression early in lung development may restore the pulmonary microvasculature and alveolarization in adult lung.
Collapse
Affiliation(s)
| | - Lori Nielsen
- Division of Neonatology, Department of Pediatrics, John R Oishei Children's Hospital, University at Buffalo, 1001 5th Floor Main Street, Buffalo, NY, 14203, USA
| | - Huamei Wang
- Division of Neonatology, Department of Pediatrics, John R Oishei Children's Hospital, University at Buffalo, 1001 5th Floor Main Street, Buffalo, NY, 14203, USA
| | - Vasantha H S Kumar
- Division of Neonatology, Department of Pediatrics, John R Oishei Children's Hospital, University at Buffalo, 1001 5th Floor Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
18
|
Wang J, Zhang A, Li Y, Xu J, Huang F, Zhao M, Wu B, He S. Effect of intermittent hypoxia or hyperoxia on lung development in preterm rat neonates during constant oxygen therapy. J Cell Biochem 2019; 120:17545-17554. [PMID: 31245867 DOI: 10.1002/jcb.29019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
Impaired lung development is a major negative factor in the survival of preterm neonates. The present study was aimed to investigate the impact of constant oxygen, intermittent hyperoxia, and hypoxia on the lung development in preterm rat neonates. Neonatal rats were exposed to 40% O2 with or without brief hyperoxia episodes (95% O2 ) or brief hypoxia episodes (10% O2 ) from day 0 to day 14, or to room air. The body weight, radical alveolar count (RAC), and total antioxidant capacity (TAOC) were significantly lower whereas the lung coefficient and malondialdehyde (MDA) were significantly higher in the hyperoxia and hypoxia groups than the air control and constant oxygen group at day 7, day 14, and day 21 after birth. The lung function indexes were reduced by intermittent hyperoxia and hypoxia. In contrast, the constant oxygen therapy increased the lung function. HIF-1α and VEGF expression were significantly increased by hypoxia and decreased by hyperoxia. The constant oxygen therapy only decreased the HIF-1α expression at day 14 and 21. In summary, the constant oxygen treatment promoted lung function without affecting the antioxidative capacity in preterm rat neonates. While intermittent hyperoxia and hypoxia inhibited lung development, decreased antioxidative capacity, and dysregulated HIF-1α/VEGF signaling in preterm rat neonates.
Collapse
Affiliation(s)
- Juanmei Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neonatology, Guangdong General Hospital, Guangzhou, Guangdong, China.,Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Aimin Zhang
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Yun Li
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Jun Xu
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Furong Huang
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Menghua Zhao
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Bufei Wu
- Department of Pediatrics, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Shaoru He
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neonatology, Guangdong General Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
20
|
Lecarpentier Y, Gourrier E, Gobert V, Vallée A. Bronchopulmonary Dysplasia: Crosstalk Between PPARγ, WNT/β-Catenin and TGF-β Pathways; The Potential Therapeutic Role of PPARγ Agonists. Front Pediatr 2019; 7:176. [PMID: 31131268 PMCID: PMC6509750 DOI: 10.3389/fped.2019.00176] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/16/2019] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a serious pulmonary disease which occurs in preterm infants. Mortality remains high due to a lack of effective treatment, despite significant progress in neonatal resuscitation. In BPD, a persistently high level of canonical WNT/β-catenin pathway activity at the canalicular stage disturbs the pulmonary maturation at the saccular and alveolar stages. The excessive thickness of the alveolar wall impairs the normal diffusion of oxygen and carbon dioxide, leading to hypoxia. Transforming growth factor (TGF-β) up-regulates canonical WNT signaling and inhibits the peroxysome proliferator activated receptor gamma (PPARγ). This profile is observed in BPD, especially in animal models. Following a premature birth, hypoxia activates the canonical WNT/TGF-β axis at the expense of PPARγ. This gives rise to the differentiation of fibroblasts into myofibroblasts, which can lead to pulmonary fibrosis that impairs the respiratory function after birth, during childhood and even adulthood. Potential therapeutic treatment could target the inhibition of the canonical WNT/TGF-β pathway and the stimulation of PPARγ activity, in particular by the administration of nebulized PPARγ agonists.
Collapse
Affiliation(s)
- Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Elizabeth Gourrier
- Service de néonatologie, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Vincent Gobert
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| | - Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hôtel-Dieu Hospital, AP-HP Paris, Paris-Descartes University, Paris, France
| |
Collapse
|
21
|
Fu X, Zhang F. Role of the HIF-1 signaling pathway in chronic obstructive pulmonary disease. Exp Ther Med 2018; 16:4553-4561. [PMID: 30542404 PMCID: PMC6257248 DOI: 10.3892/etm.2018.6785] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/25/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most common cause of chronic morbidity and mortality. However, the molecular mechanisms underlying COPD remain largely unknown. The purpose of the present study was to investigate the expression patterns of hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and VEGF receptor 2 (R2) in regard to the HIF-1 signaling pathway in COPD. The expressions of HIF-1α, VEGF and VEGFR2 were examined and quantified in the human lung tissues of 102 subjects with a defined smoking status, with or without COPD. The expressions of HIF-1α, VEGF and VEGFR2 were observed to be increased in the lung tissues collected from smoking COPD subjects when compared with those tissues from smoking subjects without COPD and non-smoking subjects without COPD. The expression of HIF-1α was shown to be positively associated with the expression of VEGF and VEGFR2. In addition, increased expression of HIF-1α, VEGF and VEGFR2 reflected the disease severity of COPD. The key findings obtained from the present study indicated that high expression of HIF-1α, VEGF and VEGFR2 may be associated with decreased lung function and reduced quality of life, contributing to disease progression in COPD.
Collapse
Affiliation(s)
- Xiang Fu
- Department of Respiratory Medicine, The No. 5 Hospital of Xiamen, Xiamen, Fujian 361100, P.R. China
| | - Fengling Zhang
- Department of Respiratory Medicine, The No. 5 Hospital of Xiamen, Xiamen, Fujian 361100, P.R. China
| |
Collapse
|
22
|
Iosef C, Liu M, Ying L, Rao SP, Concepcion KR, Chan WK, Oman A, Alvira CM. Distinct roles for IκB kinases alpha and beta in regulating pulmonary endothelial angiogenic function during late lung development. J Cell Mol Med 2018; 22:4410-4422. [PMID: 29993183 PMCID: PMC6111877 DOI: 10.1111/jcmm.13741] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 05/13/2018] [Indexed: 01/01/2023] Open
Abstract
Pulmonary angiogenesis is essential for alveolarization, the final stage of lung development that markedly increases gas exchange surface area. We recently demonstrated that activation of the nuclear factor kappa-B (NFκB) pathway promotes pulmonary angiogenesis during alveolarization. However, the mechanisms activating NFκB in the pulmonary endothelium, and its downstream targets are not known. In this study, we sought to delineate the specific roles for the NFκB activating kinases, IKKα and IKKβ, in promoting developmental pulmonary angiogenesis. Microarray analysis of primary pulmonary endothelial cells (PECs) after silencing IKKα or IKKβ demonstrated that the 2 kinases regulate unique panels of genes, with few shared targets. Although silencing IKKα induced mild impairments in angiogenic function, silencing IKKβ induced more severe angiogenic defects and decreased vascular cell adhesion molecule expression, an IKKβ regulated target essential for both PEC adhesion and migration. Taken together, these data show that IKKα and IKKβ regulate unique genes in PEC, resulting in differential effects on angiogenesis upon inhibition, and identify IKKβ as the predominant regulator of pulmonary angiogenesis during alveolarization. These data suggest that therapeutic strategies to specifically enhance IKKβ activity in the pulmonary endothelium may hold promise to enhance lung growth in diseases marked by altered alveolarization.
Collapse
Affiliation(s)
- Cristiana Iosef
- Department of Pharmacology, Faculty of Medicine, University of Nevada Reno, Reno, NV, USA.,Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Min Liu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lihua Ying
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shailaja P Rao
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Katherine R Concepcion
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Westin K Chan
- Department of Pharmacology, Faculty of Medicine, University of Nevada Reno, Reno, NV, USA
| | - Andrew Oman
- Department of Pharmacology, Faculty of Medicine, University of Nevada Reno, Reno, NV, USA
| | - Cristina M Alvira
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
23
|
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
|
24
|
Zhong Y, Catheline D, Houeijeh A, Sharma D, Du L, Besengez C, Deruelle P, Legrand P, Storme L. Maternal omega-3 PUFA supplementation prevents hyperoxia-induced pulmonary hypertension in the offspring. Am J Physiol Lung Cell Mol Physiol 2018; 315:L116-L132. [PMID: 29597832 DOI: 10.1152/ajplung.00527.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary hypertension (PH) and right ventricular hypertrophy (RVH) affect 16-25% of premature infants with bronchopulmonary dysplasia (BPD), contributing significantly to perinatal morbidity and mortality. Omega-3 polyunsaturated fatty acids (PUFA ω-3) can improve vascular remodeling, angiogenesis, and inflammation under pathophysiological conditions. However, the effects of PUFA ω-3 supplementation in BPD-associated PH are unknown. The present study aimed to evaluate the effects of PUFA ω-3 on pulmonary vascular remodeling, angiogenesis, and inflammatory response in a hyperoxia-induced rat model of PH. From embryonic day 15, pregnant Sprague-Dawley rats were supplemented daily with PUFA ω-3, PUFA ω-6, or normal saline (0.2 ml/day). After birth, pups were pooled, assigned as 12 per litter, randomly assigned to either air or continuous oxygen exposure (fraction of inspired oxygen = 85%) for 20 days, and then euthanized for pulmonary hemodynamic and morphometric analysis. We found that PUFA ω-3 supplementation improved survival, decreased right ventricular systolic pressure and RVH caused by hyperoxia, and significantly improved alveolarization, vascular remodeling, and vascular density. PUFA ω-3 supplementation produced a higher level of total ω-3 in lung tissue and breast milk and was found to reverse the reduced levels of VEGFA, VEGF receptor 2, angiopoietin-1 (ANGPT1), endothelial TEK tyrosine kinase, endothelial nitric oxide synthase, and nitric oxide concentrations in lung tissue and the increased ANGPT2 levels in hyperoxia-exposed rats. The beneficial effects of PUFA ω-3 in improving lung injuries were also associated with an inhibition of leukocyte infiltration and reduced expression of the proinflammatory cytokines IL-1β, IL-6, and TNF-α. These data indicate that maternal PUFA ω-3 supplementation strategies could effectively protect against infant PH induced by hyperoxia.
Collapse
Affiliation(s)
- Ying Zhong
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Daniel Catheline
- Laboratoire de Biochimie et Nutrition Humaine, Institut National de la Recherche Agronomique USC 2012, Institut Supérieur des Sciences Agronomiques, Agroalimentaires, Horticoles et du Paysage, Rennes , France
| | - Ali Houeijeh
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Dyuti Sharma
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Pediatric Surgery, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Lizhong Du
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Capucine Besengez
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Philippe Deruelle
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Obstetrics and Gynecology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Philippe Legrand
- Laboratoire de Biochimie et Nutrition Humaine, Institut National de la Recherche Agronomique USC 2012, Institut Supérieur des Sciences Agronomiques, Agroalimentaires, Horticoles et du Paysage, Rennes , France
| | - Laurent Storme
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| |
Collapse
|
25
|
Luan Y, Zhang L, Chao S, Liu X, Li K, Wang Y, Zhang Z. Mesenchymal stem cells in combination with erythropoietin repair hyperoxia-induced alveoli dysplasia injury in neonatal mice via inhibition of TGF-β1 signaling. Oncotarget 2018; 7:47082-47094. [PMID: 27191651 PMCID: PMC5216925 DOI: 10.18632/oncotarget.9314] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study is to investigate the protection effects of bone marrow mesenchymal stem cells (MSCs) in combination with EPO against hyperoxia-induced bronchopulmonary dysplasia (BPD) injury in neonatal mice. BPD model was prepared by continuous high oxygen exposure, 1×106 bone marrow MSCs and 5000U/kg recombinant human erythropoietin (EPO) were injected respectively. Results showed that administration of MSCs, EPO especially MSCs+EPO significant attenuated hyperoxia-induced lung damage with a decrease of fibrosis, radical alveolar counts and inhibition of the occurrence of epithelial-mesenchymal transition (EMT). Furthermore, MSCs+EPO co-treatment more significantly suppressed the levels of transforming growth factor-β1(TGF-β1) than MSCs or EPO alone. Collectively, these results suggested that MSCs, EPO in particular MSCs+EPO co-treatment could promote lung repair in hyperoxia-induced alveoli dysplasia injury via inhibition of TGF-β1 signaling pathway to further suppress EMT process and may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Luan Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Sun Chao
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, China
| | - Kaili Li
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Yibiao Wang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Zhaohua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
26
|
Alvira CM, Morty RE. Can We Understand the Pathobiology of Bronchopulmonary Dysplasia? J Pediatr 2017; 190:27-37. [PMID: 29144252 PMCID: PMC5726414 DOI: 10.1016/j.jpeds.2017.08.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/28/2017] [Accepted: 08/16/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Cristina M. Alvira
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California 94305
| | - Rory E. Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center campus of the German Center for Lung Research, Giessen, Germany,Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
27
|
Coarfa C, Zhang Y, Maity S, Perera DN, Jiang W, Wang L, Couroucli X, Moorthy B, Lingappan K. Sexual dimorphism of the pulmonary transcriptome in neonatal hyperoxic lung injury: identification of angiogenesis as a key pathway. Am J Physiol Lung Cell Mol Physiol 2017; 313:L991-L1005. [PMID: 28818871 DOI: 10.1152/ajplung.00230.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by impaired alveolar secondary septation and vascular growth. Exposure to high concentrations of oxygen (hyperoxia) contributes to the development of BPD. The male sex is considered an independent risk factor for the development of BPD. The reasons underlying sexually dimorphic outcomes in premature neonates are not known. We hypothesized that sex-specific modulation of biological processes in the lung under hyperoxic conditions contributes to sex-based differences. Neonatal male and female mice (C57BL/6) were exposed to hyperoxia [95% [Formula: see text], postnatal day (PND) 1-5: saccular stage of lung development] and euthanized on PND 7 or 21. Pulmonary gene expression was studied using RNA-Seq on the Illumina HiSeq 2500 platform. Analysis of the pulmonary transcriptome revealed differential sex-specific modulation of crucial pathways such as angiogenesis, response to hypoxia, inflammatory response, and p53 pathway. Candidate genes from these pathways were validated at the mRNA level by qPCR. Analysis also revealed sex-specific differences in the modulation of crucial transcription factors. Focusing on the differential modulation of the angiogenesis pathway, we also showed sex-specific differential activation of Hif-1α-regulated genes using ChIP-qPCR and differences in expression of crucial genes (Vegf, VegfR2, and Phd2) modulating angiogenesis. We demonstrate the translational relevance of our findings by showing that our murine sex-specific differences in gene expression correlate with those from a preexisting human BPD data set. In conclusion, we provide novel molecular insights into differential sex-specific modulation of the pulmonary transcriptome in neonatal hyperoxic lung injury and highlight angiogenesis as one of the crucial differentially modulated pathways.
Collapse
Affiliation(s)
- Cristian Coarfa
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas; and.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Yuhao Zhang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Suman Maity
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas; and
| | - Dimuthu N Perera
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas; and
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Lihua Wang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Xanthi Couroucli
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Bhagavatula Moorthy
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas;
| |
Collapse
|
28
|
Zhang Y, Jiang W, Wang L, Lingappan K. Sex-specific differences in the modulation of Growth Differentiation Factor 15 (GDF15) by hyperoxia in vivo and in vitro: Role of Hif-1α. Toxicol Appl Pharmacol 2017; 332:8-14. [PMID: 28734801 DOI: 10.1016/j.taap.2017.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/01/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022]
Abstract
Male premature neonates are more susceptible than females to the development of bronchopulmonary dysplasia (BPD). The reasons underlying sexually dimorphic outcomes in premature neonates are not known. GDF15 (Growth and differentiation factor 15) is a secreted cytokine and plays a role in cell proliferation, apoptosis, and angiogenesis. In this study, we sought to elucidate the sex-specific expression of Gdf15 in the lung in vivo in neonatal hyperoxic lung injury and its regulation by Hif-1α, and to delineate the differences in GDF15 expression in male and female human umbilical venous endothelial cells in an in vitro model of oxygen toxicity. Following hyperoxia exposure (95% FiO2, PND (postnatal day 1-5: saccular stage of lung development), neonatal male mice (C57BL/6) show increased GDF15 and decreased HIF-1α expression compared to female mice. For the in vitro experiments, male and female HUVECs were exposed to room air condition (21% O2, 5% CO2) or in hyperoxia condition (95% O2, 5% CO2) for up to 72h. Male HUVECs had greater expression of GDF15 mRNA and protein. To study the inter-relationship between GDF15 and HIF-1α, we measured the expression of GDF15 in H441 cells after HIF-1α knockdown using promoter dual luciferase reporter assay, which showed that HIF-1α and GDF15 expression are inversely related under normoxia and hyperoxia. The results indicate that sex differences exist in the expression and modulation of GDF15 by HIF-1α in neonatal hyperoxic injury both in vivo and in vitro. These differences could explain in part the mechanisms behind sex-specific differences in BPD.
Collapse
Affiliation(s)
- Yuhao Zhang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Lihua Wang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
29
|
Schmiedl A, Roolfs T, Tutdibi E, Gortner L, Monz D. Influence of prenatal hypoxia and postnatal hyperoxia on morphologic lung maturation in mice. PLoS One 2017; 12:e0175804. [PMID: 28426693 PMCID: PMC5398543 DOI: 10.1371/journal.pone.0175804] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 03/31/2017] [Indexed: 01/15/2023] Open
Abstract
Background Oxygen supply as a lifesaving intervention is frequently used to treat preterm infants suffering additionally from possible prenatal or perinatal pathogen features. The impact of oxygen and/or physical lung injury may influence the morphological lung development, leading to a chronic postnatal lung disease called bronchopulmonary dysplasia (BPD). At present different experimental BPD models are used. However, there are no systematic comparative studies regarding different influences of oxygen on morphological lung maturation. Objective We investigated the influence of prenatal hypoxia and/or postnatal hyperoxia on morphological lung maturation based on stereological parameters, to find out which model best reflects morphological changes in lung development comparable with alterations found in BPD. Methods Pregnant mice were exposed to normoxia, the offspring to normoxia (No/No) or to hyperoxia (No/Hyper). Furthermore, pregnant mice were exposed to hypoxia and the offspring to normoxia (Hypo/No) or to hyperoxia (Hypo/Hyper). Stereological investigations were performed on all pups at 14 days after birth. Results Compared to controls (No/No) 1) the lung volume was significantly reduced in the No/Hyper and Hypo/Hyper groups, 2) the volume weighted mean volume of the parenchymal airspaces was significantly higher in the Hypo/Hyper group, 3) the total air space volume was significantly lower in the No/Hyper and Hypo/Hyper groups, 4) the total septal surface showed significantly lower values in the No/Hyper and Hypo/Hyper groups, 5) the wall thickness of septa showed the highest values in the Hypo/Hyper group without reaching significance, 6) the volume density and the volume weighted mean volume of lamellar bodies in alveolar epithelial cells type II (AEII) were significantly lower in the Hypo/Hyper group. Conclusion Prenatal hypoxia and postnatal hyperoxia differentially influence the maturation of lung parenchyma. In 14 day old mice a significant retardation of morphological lung development leading to BPD-like alterations indicated by different parameters was only seen after hypoxia and hyperoxia.
Collapse
Affiliation(s)
- Andreas Schmiedl
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage und Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Torge Roolfs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Erol Tutdibi
- Department of Pediatrics and Neonatology, Saarland University, Homburg/Saar, Germany
| | - Ludwig Gortner
- Department of Pediatrics and Neonatology, Saarland University, Homburg/Saar, Germany
| | - Dominik Monz
- Department of Pediatrics and Neonatology, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
30
|
Role of Cytochrome P450 (CYP)1A in Hyperoxic Lung Injury: Analysis of the Transcriptome and Proteome. Sci Rep 2017; 7:642. [PMID: 28377578 PMCID: PMC5428698 DOI: 10.1038/s41598-017-00516-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/28/2017] [Indexed: 01/22/2023] Open
Abstract
Hyperoxia contributes to lung injury in experimental animals and diseases such as acute respiratory distress syndrome in humans. Cytochrome P450 (CYP)1A enzymes are protective against hyperoxic lung injury (HLI). The molecular pathways and differences in gene expression that modulate these protective effects remain largely unknown. Our objective was to characterize genotype specific differences in the transcriptome and proteome of acute hyperoxic lung injury using the omics platforms: microarray and Reverse Phase Proteomic Array. Wild type (WT), Cyp1a1−/− and Cyp1a2−/− (8–10 wk, C57BL/6J background) mice were exposed to hyperoxia (FiO2 > 0.95) for 48 hours. Comparison of transcriptome changes in hyperoxia-exposed animals (WT versus knock-out) identified 171 genes unique to Cyp1a1−/− and 119 unique to Cyp1a2−/− mice. Gene Set Enrichment Analysis revealed pathways including apoptosis, DNA repair and early estrogen response that were differentially regulated between WT, Cyp1a1−/− and Cyp1a2−/− mice. Candidate genes from these pathways were validated at the mRNA and protein level. Quantification of oxidative DNA adducts with 32P-postlabeling also revealed genotype specific differences. These findings provide novel insights into mechanisms behind the differences in susceptibility of Cyp1a1−/− and Cyp1a2−/− mice to HLI and suggest novel pathways that need to be investigated as possible therapeutic targets for acute lung injury.
Collapse
|
31
|
Differential sex-specific effects of oxygen toxicity in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2017; 486:431-437. [PMID: 28315681 DOI: 10.1016/j.bbrc.2017.03.058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/14/2017] [Indexed: 11/21/2022]
Abstract
Despite the well-established sex-specific differences in the incidence of bronchopulmonary dysplasia (BPD), the molecular mechanism(s) behind these are not completely understood. Pulmonary angiogenesis is critical for alveolarization and arrest in vascular development adversely affects lung development. Human neonatal umbilical vein endothelial cells (HUVECs) provide a robust in vitro model for the study of endothelial cell physiology and function. Male and Female HUVECs were exposed to room air (21% O2, 5% CO2) or hyperoxia (95% O2, 5% CO2) for up to 72 h. Cell viability, proliferation, H2O2 production and angiogenesis were analyzed. Sex-specific differences in the expression of VEGFR2 and modulation of NF-kappa B pathway were measured. Male HUVECs have decreased survival, greater oxidative stress and impairment in angiogenesis compared to similarly exposed female cells. There is differential expression of VEGFR2 between male and female HUVECs and greater activation of the NF-kappa B pathway in female HUVECs under hyperoxic conditions. The results indicate that sex differences exist between male and female HUVECs in vitro after hyperoxia exposure. Since endothelial dysfunction has a major role in the pathogenesis of BPD, these differences could explain in part the mechanisms behind sex-specific differences in the incidence of this disease.
Collapse
|
32
|
Wedgwood S, Warford C, Agvateesiri SC, Thai P, Berkelhamer SK, Perez M, Underwood MA, Steinhorn RH. Postnatal growth restriction augments oxygen-induced pulmonary hypertension in a neonatal rat model of bronchopulmonary dysplasia. Pediatr Res 2016; 80:894-902. [PMID: 27509009 DOI: 10.1038/pr.2016.164] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/16/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Prematurity and fetal growth restriction are risk factors for pulmonary hypertension (PH) in infants with bronchopulmonary dysplasia (BPD). Neonatal rats develop PH and vascular remodeling when exposed to hyperoxia. We hypothesize that postnatal growth restriction (PNGR) due to under-nutrition increases the severity of PH induced by hyperoxia in neonatal rats. METHODS Pups were randomized at birth to litters maintained in room air or 75% oxygen (hyperoxia), together with litters of normal milk intake (10 pups) or PNGR (17 pups). After 14 d, right ventricular hypertrophy (RVH) was assessed by Fulton's index (right ventricular weight/left ventricular plus septal weight) and PH by echocardiography. Lungs were analyzed by immunohistochemistry, morphometrics, western blotting, and metabolomics. RESULTS Hyperoxia and PNGR each significantly increased pulmonary arterial pressure, RVH and pulmonary arterial medial wall thickness, and significantly decreased pulmonary vessel number. These changes were significantly augmented in pups exposed to both insults. Hyperoxia and PNGR both significantly decreased expression of proteins involved in lung development and vasodilation. CONCLUSION PNGR induces right ventricular and pulmonary vascular remodeling and augments the effects of oxygen in neonatal rats. This may be a powerful tool to investigate the mechanisms that induce PH in low-birth-weight preterm infants with BPD.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, UC Davis Medical Center, Sacramento, California
| | - Cris Warford
- Department of Pediatrics, UC Davis Medical Center, Sacramento, California
| | | | - Phung Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, UC Davis Health System, Sacramento, California
| | | | - Marta Perez
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Mark A Underwood
- Department of Pediatrics, UC Davis Medical Center, Sacramento, California
| | - Robin H Steinhorn
- Department of Hospitalist Medicine, Children's National Health System, Washington, DC
| |
Collapse
|
33
|
Elberson VD, Nielsen LC, Wang H, Kumar HSV. Effects of intermittent hypoxia and hyperoxia on angiogenesis and lung development in newborn mice. J Neonatal Perinatal Med 2016; 8:313-22. [PMID: 26836820 DOI: 10.3233/npm-15814134] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Premature birth disrupts hypoxia driven microvascular development that directs alveolar and lung growth. Changes in oxygen exposure after birth can perturb the regulation of angiogenesis leading to bronchopulmonary dysplasia (BPD). We studied the effects of intermittent hypoxia or hyperoxia on HIF and angiogenic gene expression and lung development in newborn mice. METHODS Newborn litters were randomized within 12 h of birth to 12% O2 (4 h), 50% O2 (4 h) or 12% O2 (2 h)/50% O2 (2 h) followed by room air (RA) recovery for 20 h. Mice in RA were the control group. The mice were exposed to 6 such cycles (D1-D6) and sacrifice on D7. Whole lung mRNA was isolated and gene expression performed by qRT-PCR (HIF1α/2α/1β; PHD2, Ang1, Tie2, Vegf, VegfR1 & VegfR2) and analyzed by PCR array data analysis web portal. HIF-1α, prolyl hydroxylase-2 and VEGF protein were analyzed in whole lung by ELISA. Lung morphology was assessed by H&E sections and radial alveolar counts; cell proliferation by Ki67 immunostaining. RESULTS HIF-1α mRNA and VEGF protein were significantly downregulated in the 50% O2 group; VEGF mRNA and protein were significantly downregulated in the 12% O2-50% O2 group; Ang-1 and its receptor mRNA expression were downregulated in 12% O2 and 12% O2-50% O2 groups. 50% O2 (hyperoxia) and 12% O2-50% O2 (hypoxia-hyperoxia) groups demonstrated alveolar simplification by RAC and the same groups had decreased cell proliferation by Ki67 staining compared to RA and hypoxia (12% O2) groups. CONCLUSIONS Downregulation of HIF and angiogenic gene expression with associated changes in lung histology following intermittent hypoxia-hyperoxia is likely an important contributing factor in the development of BPD.
Collapse
|
34
|
Merchant SJ, Crocker IP, Baker PN, Tansinda D, Davidge ST, Guilbert LJ. Matrix Metalloproteinase Release From Placental Explants of Pregnancies Complicated by Intrauterine Growth Restriction. ACTA ACUST UNITED AC 2016; 11:97-103. [PMID: 14980311 DOI: 10.1016/j.jsgi.2003.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE There is evidence of impaired placental development in intrauterine growth restriction (IUGR). Matrix metalloproteinases (MMPs) are extracellular matrix-degrading enzymes that are released by placental cells during tissue remodeling processes. We hypothesized 1) that release of MMP-2 and -9 is decreased and/or release of tissue inhibitors of metalloproteinases (TIMPs) is increased from placental explants in pregnancies complicated by IUGR and 2) that oxygen levels affect such release. METHODS Placental villous explants from normal (n = 7) and IUGR (n = 7) pregnancies were cultured at high (20%) and low (3%) oxygen levels for 24 hours. Supernatants were analyzed for MMP-2 and MMP-9 by zymography and for TIMP-1 and -2 by western blot analysis. RESULTS : At 20% oxygen there was significantly reduced MMP-2 (P < .05) and TIMP-1 (P < .01) release and a trend for decreased MMP-9 release (P = .07) in explants from IUGR pregnancies compared with normal pregnancies; however, there were no differences at 3% oxygen. TIMP-2 was below detectable levels in all samples. Although MMP-2 and TIMP-1 release was significantly reduced at 3% compared with 20% oxygen in explants from both normal (P < .001; P < .05) and IUGR (P < .05) pregnancies, MMP-2 release changed less in IUGR compared with normal explant cultures. There were no significant effects of oxygen on MMP-9 release. CONCLUSION Placental explants from IUGR pregnancies demonstrated reduced MMP-2, MMP-9, and TIMP-1 release compared with explants from normal pregnancies at high (20%) but not low (3%) oxygen.
Collapse
Affiliation(s)
- S J Merchant
- Perinatal Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
35
|
Lingappan K, Jiang W, Wang L, Moorthy B. Sex-specific differences in neonatal hyperoxic lung injury. Am J Physiol Lung Cell Mol Physiol 2016; 311:L481-93. [PMID: 27343189 DOI: 10.1152/ajplung.00047.2016] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/24/2016] [Indexed: 11/22/2022] Open
Abstract
Male sex is considered an independent predictor for the development of bronchopulmonary dysplasia (BPD) after adjusting for other confounders. BPD is characterized by an arrest in lung development with marked impairment of alveolar septation and vascular development. The reasons underlying sexually dimorphic outcomes in premature neonates are not known. In this investigation, we tested the hypothesis that male neonatal mice will be more susceptible to hyperoxic lung injury and will display larger arrest in lung alveolarization. Neonatal male and female mice (C57BL/6) were exposed to hyperoxia [95% FiO2, postnatal day (PND) 1-5] and euthanized on PND 7 and 21. Extent of alveolarization, pulmonary vascularization, inflammation, and modulation of the NF-κB pathway were determined and compared with room air controls. Macrophage and neutrophil infiltration was significantly increased in hyperoxia-exposed animals but was increased to a larger extent in males compared with females. Lung morphometry showed a higher mean linear intercept (MLI) and a lower radial alveolar count (RAC) and therefore greater arrest in lung development in male mice. This was accompanied by a significant decrease in the expression of markers of angiogenesis (PECAM1 and VEGFR2) in males after hyperoxia exposure compared with females. Interestingly, female mice showed increased activation of the NF-κB pathway in the lungs compared with males. These results support the hypothesis that sex plays a crucial role in hyperoxia-mediated lung injury in this model. Elucidation of the sex-specific molecular mechanisms may aid in the development of novel individualized therapies to prevent/treat BPD.
Collapse
Affiliation(s)
- Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Lihua Wang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Bhagavatula Moorthy
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
36
|
Artese L, Perrotti V, Di Giulio C, Bianchi G, Piccirilli M, Piattelli A. Vascular Endothelial Growth Factor Expression (VEGF) in Salivary Glands of Young and Old Hyperoxic Rats. EUR J INFLAMM 2016. [DOI: 10.1177/1721727x0600400203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The aim of this study is to evaluate whether hyperoxia and age can influence the expression of vascular endothelial growth factor (VEGF) in the salivary glands of rats. Our study was carried out on four groups of male Wistar rats (total 24 rats). One group was composed of 6 young rats (3 months old); another group by 6 old rats (24 months old). The rats belonging to the first two groups were exposed to hyperoxia (98–100% O2) for a period of 60–65 hours in a large Plexiglas chamber; the other two groups were control groups. The rats were then anaesthetized with Nembutal (30 mg/kg) intraperitoneally and then killed. The submandibular glands were removed and processed for immunohistochemical analysis of VEGF. The exposure to hyperoxia decreased salivary gland VEGF expression in rats. The tissues analyzed (nervous tissue, vascular endothelium, myoepithelial cells, ductal endothelium, mucinous glands) always expressed VEGF, thus demonstrating that not only vascular endothelial cells, but also the other elements evaluated, have a role in the neoangiogenesis. Only in the serous glands, in both normoxic and hyperoxic young and old rats, was the VEGF expression constantly negative and it did not influence the neoangiogenesis. The vascular growth is a fundamental part of normal salivary gland development, so we speculated that strategies aimed at preservation or promotion of salivary gland VEGF expression may mitigate or attenuate hyperoxia-induced gland microvascular injury. Further studies specifically aimed at investigating these prospects are warranted.
Collapse
Affiliation(s)
| | | | - C. Di Giulio
- Department of Biomedical Sciences, University of Chieti-Pescara, Italy
| | - G. Bianchi
- Department of Biomedical Sciences, University of Chieti-Pescara, Italy
| | | | | |
Collapse
|
37
|
Boucherat O, Morissette MC, Provencher S, Bonnet S, Maltais F. Bridging Lung Development with Chronic Obstructive Pulmonary Disease. Relevance of Developmental Pathways in Chronic Obstructive Pulmonary Disease Pathogenesis. Am J Respir Crit Care Med 2016; 193:362-75. [PMID: 26681127 DOI: 10.1164/rccm.201508-1518pp] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic airflow limitation. This generic term encompasses emphysema and chronic bronchitis, two common conditions, each having distinct but also overlapping features. Recent epidemiological and experimental studies have challenged the traditional view that COPD is exclusively an adult disease occurring after years of inhalational insults to the lungs, pinpointing abnormalities or disruption of the pathways that control lung development as an important susceptibility factor for adult COPD. In addition, there is growing evidence that emphysema is not solely a destructive process because it is also characterized by a failure in cell and molecular maintenance programs necessary for proper lung development. This leads to the concept that tissue regeneration required stimulation of signaling pathways that normally operate during development. We undertook a review of the literature to outline the contribution of developmental insults and genes in the occurrence and pathogenesis of COPD, respectively.
Collapse
Affiliation(s)
- Olivier Boucherat
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Mathieu C Morissette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Steeve Provencher
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - Sébastien Bonnet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - François Maltais
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| |
Collapse
|
38
|
Zhang ZH, Pan YY, Jing RS, Luan Y, Zhang L, Sun C, Kong F, Li KL, Wang YB. Protective effects of BMSCs in combination with erythropoietin in bronchopulmonary dysplasia-induced lung injury. Mol Med Rep 2016; 14:1302-8. [PMID: 27279073 DOI: 10.3892/mmr.2016.5378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 05/12/2016] [Indexed: 11/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common type of chronic lung disease in infancy, for which no effective therapy is currently available. The aim of the present study was to investigate the effect of treatment with bone marrow mesenchymal stem cells (BMSCs) in combination with recombinant human erythropoietin (rHuEPO) on BPD‑induced mouse lung injury, and discuss the underlying mechanism. The BPD model was established by the exposure of neonatal mice to continuous high oxygen exposure for 14 days, following which 1x106 BMSCs and 5,000 U/kg rHuEPO were injected into the mice 1 h prior to and 7 days following exposure to hyperoxia. The animals received four treatments in total (n=10 in each group). After 14 days, the body weights, airway structure, and levels of matrix metalloproteinase‑9 (MMP‑9) and vascular endothelial growth factor (VEGF) were detected using histological and immunohistochemical analyses. The effect on cell differentiation was observed by examining the presence of platelet endothelial cell adhesion molecule (PECAM) and VEGF using immunofluorescence. Compared with the administration of BMSCs alone, the body weight, airway structure, and the levels of MMP‑9 and VEGF were significantly improved in the BMSCs/rHuEPO group. The results of the present study demonstrated that the intravenous injection of BMSCs significantly improved lung damage in the hyperoxia‑exposed neonatal mouse model. Furthermore, the injection of BMSCs in combination with intraperitoneal injection of rHuEPO had a more marked effect, compared with BMSCs alone, and the mechanism may be mediated by the promoting effects of BMSCs and EPO. The results of the present study provided information, which may assist in future clinical trials.
Collapse
Affiliation(s)
- Zhao-Hua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan-Yan Pan
- Department of Pediatrics, Qilu Children's Hospital of Shandong University, Jinan, Shandong 066600, P.R. China
| | - Rui-Sheng Jing
- Department of Internal Medicine, Xinji Central Hospital, Changli, Hebei 250000, P.R. China
| | - Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Luan Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Chao Sun
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Kai-Lin Li
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yi-Biao Wang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
39
|
Alvira CM. Aberrant Pulmonary Vascular Growth and Remodeling in Bronchopulmonary Dysplasia. Front Med (Lausanne) 2016; 3:21. [PMID: 27243014 PMCID: PMC4873491 DOI: 10.3389/fmed.2016.00021] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/28/2016] [Indexed: 01/12/2023] Open
Abstract
In contrast to many other organs, a significant portion of lung development occurs after birth during alveolarization, thus rendering the lung highly susceptible to injuries that may disrupt this developmental process. Premature birth heightens this susceptibility, with many premature infants developing the chronic lung disease, bronchopulmonary dysplasia (BPD), a disease characterized by arrested alveolarization. Over the past decade, tremendous progress has been made in the elucidation of mechanisms that promote postnatal lung development, including extensive data suggesting that impaired pulmonary angiogenesis contributes to the pathogenesis of BPD. Moreover, in addition to impaired vascular growth, patients with BPD also frequently demonstrate alterations in pulmonary vascular remodeling and tone, increasing the risk for persistent hypoxemia and the development of pulmonary hypertension. In this review, an overview of normal lung development will be presented, and the pathologic features of arrested development observed in BPD will be described, with a specific emphasis on the pulmonary vascular abnormalities. Key pathways that promote normal pulmonary vascular development will be reviewed, and the experimental and clinical evidence demonstrating alterations of these essential pathways in BPD summarized.
Collapse
Affiliation(s)
- Cristina M Alvira
- Department of Pediatrics, Division of Critical Care Medicine, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
40
|
Snoek KG, Reiss IKM, Tibboel J, van Rosmalen J, Capolupo I, van Heijst A, Schaible T, Post M, Tibboel D. Sphingolipids in Congenital Diaphragmatic Hernia; Results from an International Multicenter Study. PLoS One 2016; 11:e0155136. [PMID: 27159222 PMCID: PMC4861280 DOI: 10.1371/journal.pone.0155136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/25/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Congenital diaphragmatic hernia is a severe congenital anomaly with significant mortality and morbidity, for instance chronic lung disease. Sphingolipids have shown to be involved in lung injury, but their role in the pathophysiology of chronic lung disease has not been explored. We hypothesized that sphingolipid profiles in tracheal aspirates could play a role in predicting the mortality/ development of chronic lung disease in congenital diaphragmatic hernia patients. Furthermore, we hypothesized that sphingolipid profiles differ between ventilation modes; conventional mechanical ventilation versus high-frequency oscillation. METHODS Sphingolipid levels in tracheal aspirates were determined at days 1, 3, 7 and 14 in 72 neonates with congenital diaphragmatic hernia, born after > 34 weeks gestation at four high-volume congenital diaphragmatic hernia centers. Data were collected within a multicenter trial of initial ventilation strategy (NTR 1310). RESULTS 36 patients (50.0%) died or developed chronic lung disease, 34 patients (47.2%) by stratification were initially ventilated by conventional mechanical ventilation and 38 patients (52.8%) by high-frequency oscillation. Multivariable logistic regression analysis with correction for side of the defect, liver position and observed-to-expected lung-to-head ratio, showed that none of the changes in sphingolipid levels were significantly associated with mortality /development of chronic lung disease. At day 14, long-chain ceramides 18:1 and 24:0 were significantly elevated in patients initially ventilated by conventional mechanical ventilation compared to high-frequency oscillation. CONCLUSIONS We could not detect significant differences in temporal sphingolipid levels in congenital diaphragmatic hernia infants with mortality/development of chronic lung disease versus survivors without development of CLD. Elevated levels of ceramides 18:1 and 24:0 in the conventional mechanical ventilation group when compared to high-frequency oscillation could probably be explained by high peak inspiratory pressures and remodeling of the alveolar membrane.
Collapse
Affiliation(s)
- Kitty G. Snoek
- Intensive Care and Department of Paediatric Surgery, Erasmus Medical Center- Sophia Children’s Hospital, Rotterdam, The Netherlands
- * E-mail:
| | - Irwin K. M. Reiss
- Department of Neonatology, Erasmus Medical Center- Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Jeroen Tibboel
- Intensive Care and Department of Paediatric Surgery, Erasmus Medical Center- Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Joost van Rosmalen
- Department of Biostatistics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Irma Capolupo
- Department of Medical and Surgical Neonatology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Arno van Heijst
- Department of Neonatology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Thomas Schaible
- Department of Neonatology, Universitätsklinikum Mannheim, Mannheim, Germany
| | - Martin Post
- Program of Physiology and Experimental Medicine, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Dick Tibboel
- Intensive Care and Department of Paediatric Surgery, Erasmus Medical Center- Sophia Children’s Hospital, Rotterdam, The Netherlands
| |
Collapse
|
41
|
Shivanna B, Zhang S, Patel A, Jiang W, Wang L, Welty SE, Moorthy B. Omeprazole Attenuates Pulmonary Aryl Hydrocarbon Receptor Activation and Potentiates Hyperoxia-Induced Developmental Lung Injury in Newborn Mice. Toxicol Sci 2015; 148:276-87. [PMID: 26272953 DOI: 10.1093/toxsci/kfv183] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hyperoxia contributes to the development of bronchopulmonary dysplasia (BPD) in human preterm infants and a similar lung phenotype characterized by alveolar simplification in newborn mice. Omeprazole (OM) is a proton pump inhibitor that is used to treat humans with gastric acid related disorders. OM-mediated aryl hydrocarbon receptor (AhR) activation attenuates acute hyperoxic lung injury (HLI) in adult mice. Whether OM activates pulmonary AhR and protects C57BL/6J newborn mice against hyperoxia-induced developmental lung (alveolar and pulmonary vascular simplification, inflammation, and oxidative stress) injury (HDLI) is unknown. Therefore, we tested the hypothesis that OM will activate pulmonary AhR and mitigate HDLI in newborn mice. Newborn mice were treated daily with i.p. injections of OM at doses of 10 (OM10) or 25 (OM25) mg/kg while being exposed to air or hyperoxia (FiO2 of 85%) for 14 days, following which their lungs were harvested to determine alveolarization, pulmonary vascularization, inflammation, oxidative stress, vascular injury, and AhR activation. To our surprise, hyperoxia-induced alveolar and pulmonary vascular simplification, inflammation, oxidative stress, and vascular injury were augmented in OM25-treated animals. These findings were associated with attenuated pulmonary vascular endothelial growth factor receptor 2 expression and decreased pulmonary AhR activation in the OM25 group. We conclude that contrary to our hypothesis, OM decreases functional activation of pulmonary AhR and potentiates HDLI in newborn mice. These observations are consistent with our previous findings, which suggest that AhR activation plays a protective role in HDLI in newborn mice.
Collapse
Affiliation(s)
- Binoy Shivanna
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Shaojie Zhang
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Ananddeep Patel
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Weiwu Jiang
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Lihua Wang
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Stephen E Welty
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| | - Bhagavatula Moorthy
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
42
|
Hou Y, Liu M, Husted C, Chen C, Thiagarajan K, Johns JL, Rao SP, Alvira CM. Activation of the nuclear factor-κB pathway during postnatal lung inflammation preserves alveolarization by suppressing macrophage inflammatory protein-2. Am J Physiol Lung Cell Mol Physiol 2015; 309:L593-604. [PMID: 26163511 DOI: 10.1152/ajplung.00029.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/06/2015] [Indexed: 11/22/2022] Open
Abstract
A significant portion of lung development is completed postnatally during alveolarization, rendering the immature lung vulnerable to inflammatory stimuli that can disrupt lung structure and function. Although the NF-κB pathway has well-recognized pro-inflammatory functions, novel anti-inflammatory and developmental roles for NF-κB have recently been described. Thus, to determine how NF-κB modulates alveolarization during inflammation, we exposed postnatal day 6 mice to vehicle (PBS), systemic lipopolysaccharide (LPS), or the combination of LPS and the global NF-κB pathway inhibitor BAY 11-7082 (LPS + BAY). LPS impaired alveolarization, decreased lung cell proliferation, and reduced epithelial growth factor expression. BAY exaggerated these detrimental effects of LPS, further suppressing proliferation and disrupting pulmonary angiogenesis, an essential component of alveolarization. The more severe pathology induced by LPS + BAY was associated with marked increases in lung and plasma levels of macrophage inflammatory protein-2 (MIP-2). Experiments using primary neonatal pulmonary endothelial cells (PEC) demonstrated that MIP-2 directly impaired neonatal PEC migration in vitro; and neutralization of MIP-2 in vivo preserved lung cell proliferation and pulmonary angiogenesis and prevented the more severe alveolar disruption induced by the combined treatment of LPS + BAY. Taken together, these studies demonstrate a key anti-inflammatory function of the NF-κB pathway in the early alveolar lung that functions to mitigate the detrimental effects of inflammation on pulmonary angiogenesis and alveolarization. Furthermore, these data suggest that neutralization of MIP-2 may represent a novel therapeutic target that could be beneficial in preserving lung growth in premature infants exposed to inflammatory stress.
Collapse
Affiliation(s)
- Yanli Hou
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Cristiana Husted
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California; Department of Biochemistry, Faculty of Medicine, University of Nevada/Reno, Reno, Nevada; and
| | - Chihhsin Chen
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Kavitha Thiagarajan
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Jennifer L Johns
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California
| | - Shailaja P Rao
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California; Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California;
| |
Collapse
|
43
|
Abstract
Sphingolipids are a diverse class of signaling molecules implicated in many important aspects of cellular biology, including growth, differentiation, apoptosis, and autophagy. Autophagy and apoptosis are fundamental physiological processes essential for the maintenance of cellular and tissue homeostasis. There is great interest into the investigation of sphingolipids and their roles in regulating these key physiological processes as well as the manifestation of several disease states. With what is known to date, the entire scope of sphingolipid signaling is too broad, and a single review would hardly scratch the surface. Therefore, this review attempts to highlight the significance of sphingolipids in determining cell fate (e.g. apoptosis, autophagy, cell survival) in the context of the healthy lung, as well as various respiratory diseases including acute lung injury, acute respiratory distress syndrome, bronchopulmonary dysplasia, asthma, chronic obstructive pulmonary disease, emphysema, and cystic fibrosis. We present an overview of the latest findings related to sphingolipids and their metabolites, provide a short introduction to autophagy and apoptosis, and then briefly highlight the regulatory roles of sphingolipid metabolites in switching between cell survival and cell death. Finally, we describe functions of sphingolipids in autophagy and apoptosis in lung homeostasis, especially in the context of the aforementioned diseases.
Collapse
Affiliation(s)
- Joyce Lee
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada
- Institute of Medical Science, University of Toronto, Toronto, ON Canada
| | - Behzad Yeganeh
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada
| | - Leonardo Ermini
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada
| | - Martin Post
- Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada
- Institute of Medical Science, University of Toronto, Toronto, ON Canada
| |
Collapse
|
44
|
Lee DD, Schwarz MA. Adapted approach to profile genes while reconciling Vegf-a mRNA expression in the developing and injured lung. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1202-11. [PMID: 25868151 DOI: 10.1152/ajplung.00053.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/09/2015] [Indexed: 12/12/2022] Open
Abstract
During lung development and injury, messenger RNA (mRNA) transcript levels of genes fluctuate over both space and time. Quantitative PCR (qPCR) is a highly sensitive, widely used technique to measure the mRNA levels. The sensitivity of this technique can be disadvantageous and errors amplified when each qPCR assay is not validated. In contrast to other organs, lungs have high RNase activity, resulting in less than optimal RNA integrity. We implemented a strategy to address these limitations in developing and injured lungs. Parameters were established and a filter designed that optimized amplicon length and included or excluded samples based on RNA integrity. This approach was illustrated and validated by measuring mRNA levels including Vegf-a in newborn mouse lungs that were injured by 85% oxygen (hyperoxia) for 12 days and compared with control (normoxia). We demonstrate that, in contrast to contradictory Vegf-a expression when normalized to the least suitable housekeeping genes, application of this filter and normalization to most suitable three housekeeping genes, Hprt, Eef2, and Rpl13a, gave reproducible Vegf-a expression, thus corroborating the sample filter. Accordingly, both short amplicon length and proper normalization to ranked, evaluated genes minimized erroneous fluctuation and qPCR amplification issues associated with nonideal RNA integrity in injured and developing lungs. Furthermore, our work uncovers how RNA integrity, purity, amplicon length, and discovery of stable candidate reference genes enhance precision of qPCR results and utilizes the advantages of qPCR in developmental studies.
Collapse
Affiliation(s)
- Daniel D Lee
- Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| |
Collapse
|
45
|
Tibboel J, Groenman FA, Selvaratnam J, Wang J, Tseu I, Huang Z, Caniggia I, Luo D, van Tuyl M, Ackerley C, de Jongste JC, Tibboel D, Post M. Hypoxia-Inducible Factor-1 Stimulates Postnatal Lung Development but Does Not Prevent O2-Induced Alveolar Injury. Am J Respir Cell Mol Biol 2015; 52:448-58. [DOI: 10.1165/rcmb.2014-0037oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
46
|
Porzionato A, Sfriso MM, Mazzatenta A, Macchi V, De Caro R, Di Giulio C. Effects of hyperoxic exposure on signal transduction pathways in the lung. Respir Physiol Neurobiol 2015; 209:106-14. [DOI: 10.1016/j.resp.2014.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 12/18/2022]
|
47
|
Sabnis A, Carrasco R, Liu SXL, Yan X, Managlia E, Chou PM, Tan XD, De Plaen IG. Intestinal vascular endothelial growth factor is decreased in necrotizing enterocolitis. Neonatology 2015; 107:191-8. [PMID: 25659996 PMCID: PMC4354688 DOI: 10.1159/000368879] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/02/2014] [Indexed: 01/06/2023]
Abstract
BACKGROUND Decreased intestinal perfusion may contribute to the development of necrotizing enterocolitis (NEC). Vascular endothelial growth factor (VEGF) is an angiogenic protein necessary for the development and maintenance of capillary networks. Whether VEGF is dysregulated in NEC remains unknown. OBJECTIVES The objective of this study was to determine whether intestinal VEGF expression is altered in a neonatal mouse model of NEC and in human NEC patients. METHODS We first assessed changes of intestinal VEGF mRNA and protein in a neonatal mouse NEC model before significant injury occurs. We then examined whether exposure to formula feeding, bacterial inoculation, cold stress and/or intermittent hypoxia affected intestinal VEGF expression. Last, we visualized VEGF protein in intestinal tissues of murine and human NEC and control cases by immunohistochemistry. RESULTS Intestinal VEGF protein and mRNA were significantly decreased in pups exposed to the NEC protocol compared to controls. Hypoxia, cold stress and commensal bacteria, when administered together, significantly downregulated intestinal VEGF expression, while they had no significant effect when given alone. VEGF was localized to a few single intestinal epithelial cells and some cells of the lamina propria and myenteric plexus. VEGF staining was decreased in murine and human NEC intestines when compared to control tissues. CONCLUSION Intestinal VEGF protein is reduced in human and experimental NEC. Decreased VEGF production might contribute to NEC pathogenesis.
Collapse
Affiliation(s)
- Animesh Sabnis
- Division of Neonatology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinberg School of Medicine, Chicago, Ill., USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Luan Y, Ding W, Ju ZY, Zhang ZH, Zhang X, Kong F. Bone marrow-derived mesenchymal stem cells protect against lung injury in a mouse model of bronchopulmonary dysplasia. Mol Med Rep 2014; 11:1945-50. [PMID: 25406024 DOI: 10.3892/mmr.2014.2959] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 11/04/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect of bone marrow‑derived mesenchymal stem cells (BMSCs) in the treatment of lung injury in a mouse model of bronchopulmonary dysplasia (BPD) and examine the underlying mechanisms. A mouse model of BPD was created using continuous exposure to high oxygen levels for 14 days. BMSCs were isolated, cultured and then labeled with green fluorescent protein. Cells (1x106) were subsequently injected intravenously 1 h prior to high oxygen treatment. Animals were randomly divided into three groups (n=5 in each): Control group, BPD model group and BMSC injection group. At two weeks post‑treatment, the expression of transforming growth factor‑β1 (TGF‑β1), vascular endothelial growth factor (VEGF) and von Willebrand factor (vWF) was detected using immunohistochemical staining and immunofluorescence. Compared with the BPD model group, the body weight, airway structure and levels of TGF‑β1 and VEGF were significantly improved in the BMSC‑treated group. Immunofluorescence observations indicated that BMSCs were able to differentiate into cells expressing vWF and VEGF, which are markers of vascular tissues. The present study demonstrated that intravenous injection of BMSCs significantly improved lung damage in a neonatal mouse model of BPD at 14 days following hyperoxia‑induced injury. This provides novel information which may be used to guide further investigation into the use of stem cells in BPD.
Collapse
Affiliation(s)
- Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Wei Ding
- Department of Radiology, Rizhao City People's Hospital, Rizhao, Shandong 276807, P.R. China
| | - Zhi-Ye Ju
- Department of Radiology, Rizhao City People's Hospital, Rizhao, Shandong 276807, P.R. China
| | - Zhao-Hua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Xue Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
49
|
Wedgwood S, Steinhorn RH. Role of reactive oxygen species in neonatal pulmonary vascular disease. Antioxid Redox Signal 2014; 21:1926-42. [PMID: 24350610 PMCID: PMC4202910 DOI: 10.1089/ars.2013.5785] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Abnormal lung development in the perinatal period can result in severe neonatal complications, including persistent pulmonary hypertension (PH) of the newborn and bronchopulmonary dysplasia. Reactive oxygen species (ROS) play a substantive role in the development of PH associated with these diseases. ROS impair the normal pulmonary artery (PA) relaxation in response to vasodilators, and ROS are also implicated in pulmonary arterial remodeling, both of which can increase the severity of PH. RECENT ADVANCES PA ROS levels are elevated when endogenous ROS-generating enzymes are activated and/or when endogenous ROS scavengers are inactivated. Animal models have provided valuable insights into ROS generators and scavengers that are dysregulated in different forms of neonatal PH, thus identifying potential therapeutic targets. CRITICAL ISSUES General antioxidant therapy has proved ineffective in reversing PH, suggesting that it is necessary to target specific signaling pathways for successful therapy. FUTURE DIRECTIONS Development of novel selective pharmacologic inhibitors along with nonantioxidant therapies may improve the treatment outcomes of patients with PH, while further investigation of the underlying mechanisms may enable earlier detection of the disease.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, University of California Davis Medical Center , Sacramento, California
| | | |
Collapse
|
50
|
D'Angio CT, Ryan RM. Animal models of bronchopulmonary dysplasia. The preterm and term rabbit models. Am J Physiol Lung Cell Mol Physiol 2014; 307:L959-69. [PMID: 25326582 DOI: 10.1152/ajplung.00228.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is an important lung developmental pathophysiology that affects many premature infants each year. Newborn animal models employing both premature and term animals have been used over the years to study various components of BPD. This review describes some of the neonatal rabbit studies that have contributed to the understanding of BPD, including those using term newborn hyperoxia exposure models, premature hyperoxia models, and a term newborn hyperoxia model with recovery in moderate hyperoxia, all designed to emulate aspects of BPD in human infants. Some investigators perturbed these models to include exposure to neonatal infection/inflammation or postnatal malnutrition. The similarities to lung injury in human premature infants include an acute inflammatory response with the production of cytokines, chemokines, and growth factors that have been implicated in human disease, abnormal pulmonary function, disordered lung architecture, and alveolar simplification, development of fibrosis, and abnormal vascular growth factor expression. Neonatal rabbit models have the drawback of limited access to reagents as well as the lack of readily available transgenic models but, unlike smaller rodent models, are able to be manipulated easily and are significantly less expensive than larger animal models.
Collapse
Affiliation(s)
- Carl T D'Angio
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York and
| | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|