1
|
Bdaiwi AS, Svoboda AM, Murdock KE, Hendricks A, Hossain MM, Kramer EL, Brewington JJ, Willmering MM, Woods JC, Walkup LL, Cleveland ZI. Quantifying abnormal alveolar microstructure in cystic fibrosis lung disease via hyperpolarized 129Xe diffusion MRI. J Cyst Fibros 2024; 23:926-935. [PMID: 38997823 PMCID: PMC11410525 DOI: 10.1016/j.jcf.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
RATIONALE Cystic Fibrosis (CF) progresses through recurrent infection and inflammation, causing permanent lung function loss and airway remodeling. CT scans reveal abnormally low-density lung parenchyma in CF, but its microstructural nature remains insufficiently explored due to clinical CT limitations. To this end, diffusion-weighted 129Xe MRI is a non-invasive and validated measure of lung microstructure. In this work, we investigate microstructural changes in people with CF (pwCF) relative to age-matched, healthy subjects using comprehensive imaging and analysis involving pulmonary-function tests (PFTs), and 129Xe MRI. METHODS 38 healthy subjects (age 6-40; 17.2 ± 9.5 years) and 39 pwCF (age 6-40; 15.6 ± 8.0 years) underwent 129Xe-diffusion MRI and PFTs. The distribution of diffusion measurements (i.e., apparent diffusion coefficients (ADC) and morphometric parameters) was assessed via linear binning (LB). The resulting volume percentages of bins were compared between controls and pwCF. Mean ADC and morphometric parameters were also correlated with PFTs. RESULTS Mean whole-lung ADC correlated significantly with age (P < 0.001) for both controls and CF, and with PFTs (P < 0.05) specifically for pwCF. Although there was no significant difference in mean ADC between controls and pwCF (P = 0.334), age-adjusted LB indicated significant voxel-level diffusion (i.e., ADC and morphometric parameters) differences in pwCF compared to controls (P < 0.05). CONCLUSIONS 129Xe diffusion MRI revealed microstructural abnormalities in CF lung disease. Smaller microstructural size may reflect compression from overall higher lung density due to interstitial inflammation, fibrosis, or other pathological changes. While elevated microstructural size may indicate emphysema-like remodeling due to chronic inflammation and infection.
Collapse
Affiliation(s)
- Abdullah S Bdaiwi
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States
| | - Alexandra M Svoboda
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; College of Medicine, University of Cincinnati, Cincinnati, OH 45221, United States
| | - Kyle E Murdock
- Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Alexandra Hendricks
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States
| | - Md M Hossain
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Elizabeth L Kramer
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - John J Brewington
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew M Willmering
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Jason C Woods
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States; Department of Physics, University of Cincinnati, Cincinnati, United States
| | - Laura L Walkup
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States; Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Zackary I Cleveland
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States; Imaging Research Center, Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States.
| |
Collapse
|
2
|
Wang Y, Yi Y. CISD2 downregulation participates in the ferroptosis process of human ovarian SKOV-3 cells through modulating the wild type p53-mediated GLS2/SAT1/SLC7A11 and Gpx4/TRF signaling pathway. Tissue Cell 2024; 89:102458. [PMID: 38991271 DOI: 10.1016/j.tice.2024.102458] [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: 05/07/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
CISD2 and ferroptosis participate in cancer development, but are rarely reported in ovarian cancer. This study aimed to clarify interaction between CISD2 and ferroptosis and evaluate related mechanisms. si-CISD2, wt-p53 and mut-p53 lentiviruses were transfected into SKOV-3 cells. CISD2 and p53 (wild/mutant p53) gene transcriptions were evaluated by RT-PCR. Cell viability, invasion ability, and migration capacity were determined. Expressions of ferroptosis-associated CISD2, p53, elastin, β-catenin and levels of Gpx4 and TRF were examined. CISD2 downregulation (si-CISD2) has a significant inhibitory effect on cell activity and exerts a synergistic effect with p53. si-CISD2 and Wt-p53 markedly inhibited SKOV-3 invasion and migration capacity, compared to the downregulation control (si-NC) and overexpression control (ov-NC) group (p < 0.001). p53 expression was increased significantly in si-CISD2 treated SKOV-3, compared to si-NC treated cells (p < 0.05). si-CISD2 markedly decreased elastin and β-catenin expression compared to the si-NC and ov-NC group (p < 0.001). si-CISD2 modulated ferroptosis-associated molecules (CDKN1A, GLS2, SAT1, SLC7A11), decreased Gpx4 and increased TRF levels in SKOV-3. si-CISD2 and Wt-p53 played an obvious synergistic role in regulating ferroptosis-associated molecules and Gpx4/TRF pathway molecules. In conclusion, CISD2 downregulation was involved in ferroptosis process of SKOV-3 cells. This effect of CISD2 was mediated by wild-type p53-associated GLS2/SAT1/SLC7A11 and Gpx4/TRF pathway.
Collapse
Affiliation(s)
- Yaqin Wang
- School of Basic Medicine, Chongqing Medical University, Chongqing, China; School of Basic Medicine, North Sichuan Medical College, Nanchong, China
| | - Yongfen Yi
- School of Basic Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Li J, Cao J, Yan C, Gong X. TGF-α/EGFR signaling promotes lipopolysaccharide-induced abnormal elastin deposition and alveolar simplification. Exp Cell Res 2024; 437:113997. [PMID: 38508328 DOI: 10.1016/j.yexcr.2024.113997] [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: 11/29/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by shortened secondary septa and fewer, larger alveoli. Elastin deposition to the distal tips of the secondary septa is critical for elongation of the secondary septa. Alveolar myofibroblasts, which are thought to migrate to the septal tips during alveolarization, are mainly responsible for elastin production and deposition. Antenatal exposure to inflammation induces abnormal elastin deposition, thereby increasing the risk of developing BPD. Here, we found that lipopolysaccharide (LPS) significantly increased the expression of transforming growth factor-α (TGF-α) in an LPS-induced rat model of BPD and in LPS-treated human pulmonary epithelial cells (BEAS-2B). In addition, in vitro experiments suggested that LPS upregulated TGF-α expression via toll-like receptor 4 (TLR4)/tumor necrosis factor α-converting enzyme (TACE) signaling. Increased TGF-α levels via its receptor epidermal growth factor receptor (EGFR)-induced lysyl oxidase (LOX) overactivation and cell division cycle 42 (Cdc42) activity inhibition of myofibroblasts. Similarly, in vivo LOX overactivation and inhibition of Cdc42 activity were observed in the lungs of LPS-exposed pups. LOX overactivation led to abnormal elastin deposition, and inhibition of Cdc42 activity disturbed the directional migration of myofibroblasts and disrupted elastin localization. Most importantly, the EGFR inhibitor erlotinib partially rescued LOX overactivation and Cdc42 activity inhibition, and improved elastin deposition and alveolar development in antenatal LPS-treated rats. Taken together, our data suggest that TGF-α/EGFR signaling is critically involved in the regulation of elastin deposition and represents a novel therapeutic target.
Collapse
Affiliation(s)
- Jianhui Li
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, 355 Lu Ding Road, 200062, Shanghai, China.
| | - Jian Cao
- Department of Respiratory Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, 355 Lu Ding Road, 200062, Shanghai, China
| | - Chongbing Yan
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, 355 Lu Ding Road, 200062, Shanghai, China
| | - Xiaohui Gong
- Department of Neonatology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, 355 Lu Ding Road, 200062, Shanghai, China.
| |
Collapse
|
4
|
Kikuchi K, Otsuka S, Takada S, Nakanishi K, Setoyama K, Sakakima H, Tanaka E, Maruyama I. 1,5-anhydro-D-fructose induces anti-aging effects on aging-associated brain diseases by increasing 5'-adenosine monophosphate-activated protein kinase activity via the peroxisome proliferator-activated receptor-γ co-activator-1α/brain-derived neurotrophic factor pathway. Aging (Albany NY) 2023; 15:11740-11763. [PMID: 37950725 PMCID: PMC10683599 DOI: 10.18632/aging.205228] [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: 12/05/2022] [Accepted: 10/11/2023] [Indexed: 11/13/2023]
Abstract
5'-Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor that serves as a cellular housekeeper; it also controls energy homeostasis and stress resistance. Thus, correct regulation of this factor can enhance health and survival. AMPK signaling may have a critical role in aging-associated brain diseases. Some in vitro studies have shown that 1,5-anhydro-D-fructose (1,5-AF) induces AMPK activation. In the present study, we experimentally evaluated the effects of 1,5-AF on aging-associated brain diseases in vivo using an animal model of acute ischemic stroke (AIS), stroke-prone spontaneously hypertensive rats (SHRSPs), and the spontaneous senescence-accelerated mouse-prone 8 (SAMP8) model. In the AIS model, intraperitoneal injection of 1,5-AF reduced cerebral infarct volume, neurological deficits, and mortality. In SHRSPs, oral administration of 1,5-AF reduced blood pressure and prolonged survival. In the SAMP8 model, oral administration of 1,5-AF alleviated aging-related decline in motor cognitive function. Although aging reduced the expression levels of peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) and brain-derived neurotrophic factor (BDNF), we found that 1,5-AF activated AMPK, which led to upregulation of the PGC-1α/BDNF pathway. Our results suggest that 1,5-AF can induce endogenous neurovascular protection, potentially preventing aging-associated brain diseases. Clinical studies are needed to determine whether 1,5-AF can prevent aging-associated brain diseases.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Fukuoka 830-0011, Japan
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka 830-0011, Japan
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan
| | - Shotaro Otsuka
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan
| | - Seiya Takada
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan
| | - Kazuki Nakanishi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima 890-8544, Japan
| | - Kentaro Setoyama
- Division of Laboratory Animal Resources and Research, Center for Advanced Science Research and Promotion, Kagoshima University, Kagoshima 890-8520, Japan
| | - Harutoshi Sakakima
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima 890-8544, Japan
| | - Eiichiro Tanaka
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Fukuoka 830-0011, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan
| |
Collapse
|
5
|
Mižíková I, Thébaud B. Perinatal origins of bronchopulmonary dysplasia-deciphering normal and impaired lung development cell by cell. Mol Cell Pediatr 2023; 10:4. [PMID: 37072570 PMCID: PMC10113423 DOI: 10.1186/s40348-023-00158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a multifactorial disease occurring as a consequence of premature birth, as well as antenatal and postnatal injury to the developing lung. BPD morbidity and severity depend on a complex interplay between prenatal and postnatal inflammation, mechanical ventilation, and oxygen therapy as well as associated prematurity-related complications. These initial hits result in ill-explored aberrant immune and reparative response, activation of pro-fibrotic and anti-angiogenic factors, which further perpetuate the injury. Histologically, the disease presents primarily by impaired lung development and an arrest in lung microvascular maturation. Consequently, BPD leads to respiratory complications beyond the neonatal period and may result in premature aging of the lung. While the numerous prenatal and postnatal stimuli contributing to BPD pathogenesis are relatively well known, the specific cell populations driving the injury, as well as underlying mechanisms are still not well understood. Recently, an effort to gain a more detailed insight into the cellular composition of the developing lung and its progenitor populations has unfold. Here, we provide an overview of the current knowledge regarding perinatal origin of BPD and discuss underlying mechanisms, as well as novel approaches to study the perturbed lung development.
Collapse
Affiliation(s)
- I Mižíková
- Experimental Pulmonology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - B Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO), CHEO Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
6
|
Koc C, Cakir A, Salman B, Ocalan B, Alkan T, Kafa IM, Cetinkaya M, Cansev M. Preventive effects of antenatal CDP-choline in a rat model of neonatal hyperoxia-induced lung injury. Can J Physiol Pharmacol 2023; 101:65-73. [PMID: 36524681 DOI: 10.1139/cjpp-2022-0321] [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: 12/23/2022]
Abstract
Antenatal steroid administration to pregnant women at risk of prematurity provides pulmonary maturation in infants, while it has limited effects on incidence of bronchopulmonary dysplasia (BPD), the clinical expression of hyperoxia-induced lung injury (HILI). Cytidine-5'-diphosphate choline (CDP-choline) was shown to alleviate HILI when administered to newborn rats. Therefore, we investigated effects of maternal administration of CDP-choline, alone or in combination with betamethasone, on lung maturation in neonatal rats subjected to HILI immediately after birth. Pregnant rats were randomly assigned to one of the four treatments: saline (1 mL/kg), CDP-choline (300 mg/kg), betamethasone (0.4 mg/kg), or CDP-choline plus betamethasone (combination therapy). From postnatal day 1 to 11, pups born to mothers in the same treatment group were pooled and randomly assigned to either normoxia or hyperoxia group. Biochemical an d histopathological effects of CDP-choline on neonatal lung tissue were evaluated. Antenatal CDP-choline treatment increased levels of phosphatidylcholine and total lung phospholipids, decreased apoptosis, and improved alveolarization. The outcomes were further improved with combination therapy compared to the administration of CDP-choline or betamethasone alone. These results demonstrate that antenatal CDP-choline treatment provides benefit in experimental HILI either alone or more intensively when administered along with a steroid, suggesting a possible utility for CDP-choline against BPD.
Collapse
Affiliation(s)
- Cansu Koc
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Aysen Cakir
- Department of Physiology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Berna Salman
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Busra Ocalan
- Department of Physiology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Tulin Alkan
- Department of Physiology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ilker Mustafa Kafa
- Department of Anatomy, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Merih Cetinkaya
- Department of Neonatology, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkey
| | - Mehmet Cansev
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
7
|
Validation of disease-specific biomarkers for the early detection of bronchopulmonary dysplasia. Pediatr Res 2023; 93:625-632. [PMID: 35595912 PMCID: PMC9988689 DOI: 10.1038/s41390-022-02093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/23/2022] [Accepted: 04/25/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To demonstrate and validate the improvement of current risk stratification for bronchopulmonary dysplasia (BPD) early after birth by plasma protein markers (sialic acid-binding Ig-like lectin 14 (SIGLEC-14), basal cell adhesion molecule (BCAM), angiopoietin-like 3 protein (ANGPTL-3)) in extremely premature infants. METHODS AND RESULTS Proteome screening in first-week-of-life plasma samples of n = 52 preterm infants <32 weeks gestational age (GA) on two proteomic platforms (SomaLogic®, Olink-Proteomics®) confirmed three biomarkers with significant predictive power: BCAM, SIGLEC-14, and ANGPTL-3. We demonstrate high sensitivity (0.92) and specificity (0.86) under consideration of GA, show the proteins' critical contribution to the predictive power of known clinical risk factors, e.g., birth weight and GA, and predicted the duration of mechanical ventilation, oxygen supplementation, as well as neonatal intensive care stay. We confirmed significant predictive power for BPD cases when switching to a clinically applicable method (enzyme-linked immunosorbent assay) in an independent sample set (n = 25, p < 0.001) and demonstrated disease specificity in different cohorts of neonatal and adult lung disease. CONCLUSION While successfully addressing typical challenges of clinical biomarker studies, we demonstrated the potential of BCAM, SIGLEC-14, and ANGPTL-3 to inform future clinical decision making in the preterm infant at risk for BPD. TRIAL REGISTRATION Deutsches Register Klinische Studien (DRKS) No. 00004600; https://www.drks.de . IMPACT The urgent need for biomarkers that enable early decision making and personalized monitoring strategies in preterm infants with BPD is challenged by targeted marker analyses, cohort size, and disease heterogeneity. We demonstrate the potential of the plasma proteins BCAM, SIGLEC-14, and ANGPTL-3 to identify infants with BPD early after birth while improving the predictive power of clinical variables, confirming the robustness toward proteome assays and proving disease specificity. Our comprehensive analysis enables a phase-III clinical trial that allows full implementation of the biomarkers into clinical routine to enable early risk stratification in preterms with BPD.
Collapse
|
8
|
Zhong Y, Mahoney RC, Khatun Z, Chen HH, Nguyen CT, Caravan P, Roberts JD. Lysyl oxidase regulation and protein aldehydes in the injured newborn lung. Am J Physiol Lung Cell Mol Physiol 2022; 322:L204-L223. [PMID: 34878944 PMCID: PMC8794022 DOI: 10.1152/ajplung.00158.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
During newborn lung injury, excessive activity of lysyl oxidases (LOXs) disrupts extracellular matrix (ECM) formation. Previous studies indicate that TGFβ activation in the O2-injured mouse pup lung increases lysyl oxidase (LOX) expression. But how TGFβ regulates this, and whether the LOXs generate excess pulmonary aldehydes are unknown. First, we determined that O2-mediated lung injury increases LOX protein expression in TGFβ-stimulated pup lung interstitial fibroblasts. This regulation appeared to be direct; this is because TGFβ treatment also increased LOX protein expression in isolated pup lung fibroblasts. Then using a fibroblast cell line, we determined that TGFβ stimulates LOX expression at a transcriptional level via Smad2/3-dependent signaling. LOX is translated as a pro-protein that requires secretion and extracellular cleavage before assuming amine oxidase activity and, in some cells, reuptake with nuclear localization. We found that pro-LOX is processed in the newborn mouse pup lung. Also, O2-mediated injury was determined to increase pro-LOX secretion and nuclear LOX immunoreactivity particularly in areas populated with interstitial fibroblasts and exhibiting malformed ECM. Then, using molecular probes, we detected increased aldehyde levels in vivo in O2-injured pup lungs, which mapped to areas of increased pro-LOX secretion in lung sections. Increased activity of LOXs plays a critical role in the aldehyde generation; an inhibitor of LOXs prevented the elevation of aldehydes in the O2-injured pup lung. These results reveal new mechanisms of TGFβ and LOX in newborn lung disease and suggest that aldehyde-reactive probes might have utility in sensing the activation of LOXs in vivo during lung injury.
Collapse
Affiliation(s)
- Ying Zhong
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Rose C. Mahoney
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts
| | - Zehedina Khatun
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Howard H. Chen
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christopher T. Nguyen
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter Caravan
- 4Harvard Medical School, Harvard University, Cambridge, Massachusetts,5Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts,6Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts,7The Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Jesse D. Roberts
- 1Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, Massachusetts,2Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts,3Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts,4Harvard Medical School, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
9
|
Ha AW, Bai T, Ebenezer DL, Sethi T, Sudhadevi T, Mangio LA, Garzon S, Pryhuber GS, Natarajan V, Harijith A. Sphingosine kinase 1 regulates lysyl oxidase through STAT3 in hyperoxia-mediated neonatal lung injury. Thorax 2022; 77:47-57. [PMID: 33883249 PMCID: PMC9115769 DOI: 10.1136/thoraxjnl-2020-216469] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Neonatal lung injury as a consequence of hyperoxia (HO) therapy and ventilator care contribute to the development of bronchopulmonary dysplasia (BPD). Increased expression and activity of lysyl oxidase (LOX), a key enzyme that cross-links collagen, was associated with increased sphingosine kinase 1 (SPHK1) in human BPD. We, therefore, examined closely the link between LOX and SPHK1 in BPD. METHOD The enzyme expression of SPHK1 and LOX were assessed in lung tissues of human BPD using immunohistochemistry and quantified (Halo). In vivo studies were based on Sphk1-/- and matched wild type (WT) neonatal mice exposed to HO while treated with PF543, an inhibitor of SPHK1. In vitro mechanistic studies used human lung microvascular endothelial cells (HLMVECs). RESULTS Both SPHK1 and LOX expressions were increased in lungs of patients with BPD. Tracheal aspirates from patients with BPD had increased LOX, correlating with sphingosine-1-phosphate (S1P) levels. HO-induced increase of LOX in lungs were attenuated in both Sphk1-/- and PF543-treated WT mice, accompanied by reduced collagen staining (sirius red). PF543 reduced LOX activity in both bronchoalveolar lavage fluid and supernatant of HLMVECs following HO. In silico analysis revealed STAT3 as a potential transcriptional regulator of LOX. In HLMVECs, following HO, ChIP assay confirmed increased STAT3 binding to LOX promoter. SPHK1 inhibition reduced phosphorylation of STAT3. Antibody to S1P and siRNA against SPNS2, S1P receptor 1 (S1P1) and STAT3 reduced LOX expression. CONCLUSION HO-induced SPHK1/S1P signalling axis plays a critical role in transcriptional regulation of LOX expression via SPNS2, S1P1 and STAT3 in lung endothelium.
Collapse
Affiliation(s)
- Alison W Ha
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tao Bai
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - David L Ebenezer
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tanvi Sethi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Lizar Ace Mangio
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Steven Garzon
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gloria S Pryhuber
- Department of Pediatrics, University of Rochester, Rochester, New York, USA
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Panahabadi S, Heindel K, Mueller A, Holdenrieder S, Kipfmueller F. Increased circulating cytokeratin 19 fragment levels in preterm neonates receiving mechanical ventilation are associated with poor outcome. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1036-L1043. [PMID: 34585605 DOI: 10.1152/ajplung.00176.2021] [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: 11/22/2022] Open
Abstract
Invasive mechanical ventilation and oxygen toxicity are postnatal contributors to chronic lung disease of prematurity, also known as bronchopulmonary dysplasia (BPD). Cyfra 21-1 is a soluble fragment of cytokeratin 19, which belongs to the cytoskeleton stabilizing epithelial intermediate filaments. As a biomarker of structural integrity, Cyfra 21-1 might be associated with airway injury and lung hypoplasia in neonates. Serum Cyfra 21-1 concentrations for 80 preterm and 80 healthy term newborns were measured within 48 h after birth. Preterm infants with the combined endpoint BPD/mortality had significantly higher Cyfra 21-1 levels compared with those without fulfilling BPD/mortality criteria (P = 0.01). Also, severe RDS (>grade III) was associated with higher Cyfra levels (P = 0.01). Total duration of oxygen therapy was more than five times longer in neonates with high Cyfra 21-1 levels (P = 0.01). Infants with higher Cyfra 21-1 values were more likely to receive mechanical ventilation (50% vs. 17.5%). However, the duration of mechanical ventilation was similar between groups. The median Cyfra value was 1.93 ng/mL (IQR: 1.68-2.53 ng/mL) in healthy term neonates and 8.5 ng/mL (IQR: 3.6-16.0 ng/mL) in preterm infants. Using ROC analysis, we calculated a Cyfra cutoff > 8.5 ng/mL to predict BPD/death with an AUC of 0.795 (P = 0.004), a sensitivity of 88.9%, and a specificity of 55%. Mortality was predicted with a cutoff > 17.4 ng/mL (AUC: 0.94; P = 0.001), a sensitivity of 100%, and a specificity of 84%. These findings suggest that Cyfra 21-1 concentration might be useful to predict poor outcome in premature infants.
Collapse
Affiliation(s)
- Sarah Panahabadi
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany.,Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Katrin Heindel
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Andreas Mueller
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | - Stefan Holdenrieder
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.,Institute for Laboratory Medicine, German Heart Center of the State of Bavaria and the Technical University Munich, Munich, Germany
| | - Florian Kipfmueller
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
Mereness JA, Mariani TJ. The critical role of collagen VI in lung development and chronic lung disease. Matrix Biol Plus 2021; 10:100058. [PMID: 34195595 PMCID: PMC8233475 DOI: 10.1016/j.mbplus.2021.100058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 01/20/2023] Open
Abstract
Type VI collagen (collagen VI) is an obligate extracellular matrix component found mainly in the basement membrane region of many mammalian tissues and organs, including skeletal muscle and throughout the respiratory system. Collagen VI is probably most recognized in medicine as the genetic cause of a spectrum of muscular dystrophies, including Ullrich Congenital Myopathy and Bethlem Myopathy. Collagen VI is thought to contribute to myopathy, at least in part, by mediating muscle fiber integrity by anchoring myoblasts to the muscle basement membrane. Interestingly, collagen VI myopathies present with restrictive respiratory insufficiency, thought to be due primarily to thoracic muscular weakening. Although it was recently recognized as one of the (if not the) most abundant collagens in the mammalian lung, there is a substantive knowledge gap concerning its role in respiratory system development and function. A few studies have suggested that collagen VI insufficiency is associated with airway epithelial cell survival and altered lung function. Our recent work suggested collagen VI may be a genomic risk factor for chronic lung disease in premature infants. Using this as motivation, we thoroughly assessed the role of collagen VI in lung development and in lung epithelial cell biology. Here, we describe the state-of-the-art for collagen VI cell and developmental biology within the respiratory system, and reveal its essential roles in normal developmental processes and airway epithelial cell phenotype and intracellular signaling.
Collapse
Affiliation(s)
- Jared A. Mereness
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Thomas J. Mariani
- Corresponding author. Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, 601 Elmwood Ave, Box 850, Rochester, NY 14642, USA.
| |
Collapse
|
12
|
Signaling Pathways Involved in the Development of Bronchopulmonary Dysplasia and Pulmonary Hypertension. CHILDREN-BASEL 2020; 7:children7080100. [PMID: 32824651 PMCID: PMC7465273 DOI: 10.3390/children7080100] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/31/2022]
Abstract
The alveolar and vascular developmental arrest in the premature infants poses a major problem in the management of these infants. Although, with the current management, the survival rate has improved in these infants, but bronchopulmonary dysplasia (BPD) is a serious complication associated with a high mortality rate. During the neonatal developmental period, these infants are vulnerable to stress. Hypoxia, hyperoxia, and ventilation injury lead to oxidative and inflammatory stress, which induce further damage in the lung alveoli and vasculature. Development of pulmonary hypertension (PH) in infants with BPD worsens the prognosis. Despite considerable progress in the management of premature infants, therapy to prevent BPD is not yet available. Animal experiments have shown deregulation of multiple signaling factors such as transforming growth factorβ (TGFβ), connective tissue growth factor (CTGF), fibroblast growth factor 10 (FGF10), vascular endothelial growth factor (VEGF), caveolin-1, wingless & Int-1 (WNT)/β-catenin, and elastin in the pathogenesis of BPD. This article reviews the signaling pathways entailed in the pathogenesis of BPD associated with PH and the possible management.
Collapse
|
13
|
Yue H, Ji X, Ku T, Li G, Sang N. Sex difference in bronchopulmonary dysplasia of offspring in response to maternal PM 2.5 exposure. JOURNAL OF HAZARDOUS MATERIALS 2020; 389:122033. [PMID: 32004849 DOI: 10.1016/j.jhazmat.2020.122033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/05/2020] [Indexed: 06/10/2023]
Abstract
The adverse effects of fine particulate matters (PM2.5) on respiratory diseases start in utero. In order to investigate whether maternal PM2.5 exposure could lead to bronchopulmonary dysplasia (BPD) in offspring, PM2.5 was collected in Taiyuan, Shanxi, China during the annual heating period. Mice were mated and gestation day 0 (GD0) was considered the day on which a vaginal plug was observed. The plug-positive mice received 3 mg/kg b.w. PM2.5 by oropharyngeal aspiration every other day starting on GD0 and throughout the gestation period. Offspring were sacrificed at postnatal days (PNDs) 1, 7, 14 and 21. We assessed some typical BPD-like symptoms in offspring. The results showed that maternal PM2.5 exposure caused low birth weight, hypoalveolarization, decreased angiogenesis, suppressed production of secretory and surfactant proteins, and increased inflammation in the lungs of male offspring. However, maternal PM2.5 exposure induced only hypoalveolarization and inflammation in the lungs of female offspring. Furthermore, these alterations were reversed during postnatal development. Our results demonstrated that maternal exposure to PM2.5 caused reversible BPD-related consequences in offspring, and male offspring were more sensitive than females. However, these alterations were reversed during postnatal development.
Collapse
Affiliation(s)
- Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
14
|
Allison BJ, Youn H, Malhotra A, McDonald CA, Castillo-Melendez M, Pham Y, Sutherland AE, Jenkin G, Polglase GR, Miller SL. Is Umbilical Cord Blood Therapy an Effective Treatment for Early Lung Injury in Growth Restriction? Front Endocrinol (Lausanne) 2020; 11:86. [PMID: 32194502 PMCID: PMC7063054 DOI: 10.3389/fendo.2020.00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/11/2020] [Indexed: 11/22/2022] Open
Abstract
Fetal growth restriction (FGR) and prematurity are often co-morbidities, and both are risk factors for lung disease. Despite advances in early delivery combined with supportive ventilation, rates of ventilation-induced lung injury (VILI) remain high. There are currently no protective treatments or interventions available that target lung morbidities associated with FGR preterm infants. Stem cell therapy, such as umbilical cord blood (UCB) cell administration, demonstrates an ability to attenuate inflammation and injury associated with VILI in preterm appropriately grown animals. However, no studies have looked at the effects of stem cell therapy in growth restricted newborns. We aimed to determine if UCB treatment could attenuate acute inflammation in the first 24 h of ventilation, comparing effects in lambs born preterm following FGR with those born preterm but appropriately grown (AG). Placental insufficiency (FGR) was induced by single umbilical artery ligation in twin-bearing ewes at 88 days gestation, with twins used as control (appropriately grown, AG). Lambs were delivered preterm at ~126 days gestation (term is 150 days) and randomized to either immediate euthanasia (unventilated controls, AGUVC and FGRUVC) or commenced on 24 h of gentle supportive ventilation (AGV and FGRV) with additional cohorts receiving UCB treatment at 1 h (AGCELLS, FGRCELLS). Lungs were collected at post-mortem for histological and biochemical examination. Ventilation caused lung injury in AG lambs, as indicated by decreased septal crests and elastin density, as well as increased inflammation. Lung injury in AG lambs was attenuated with UCB therapy. Ventilated FGR lambs also sustained lung injury, albeit with different indices compared to AG lambs; in FGR, ventilation reduced septal crest density, reduced alpha smooth muscle actin density and reduced cell proliferation. UCB treatment in ventilated FGR lambs further decreased septal crest density and increased collagen deposition, however, it increased angiogenesis as evidenced by increased vascular endothelial growth factor (VEGF) expression and vessel density. This is the first time that a cell therapy has been investigated in the lungs of growth restricted animals. We show that the uterine environment can alter the response to both secondary stress (ventilation) and therapy (UCB). This study highlights the need for further research on the potential impact of novel therapies on a growth restricted offspring.
Collapse
Affiliation(s)
- Beth J. Allison
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
- *Correspondence: Beth J. Allison
| | - Hannah Youn
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Atul Malhotra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Monash Newborn, Monash Medical Centre, Clayton, VIC, Australia
| | - Courtney A. McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Amy E. Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Graeme R. Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology and Paediatrics, Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Li C, Lee MK, Gao F, Webster S, Di H, Duan J, Yang CY, Bhopal N, Peinado N, Pryhuber G, Smith SM, Borok Z, Bellusci S, Minoo P. Secondary crest myofibroblast PDGFRα controls the elastogenesis pathway via a secondary tier of signaling networks during alveologenesis. Development 2019; 146:dev.176354. [PMID: 31331942 PMCID: PMC6703710 DOI: 10.1242/dev.176354] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/11/2019] [Indexed: 12/16/2022]
Abstract
Postnatal alveolar formation is the most important and the least understood phase of lung development. Alveolar pathologies are prominent in neonatal and adult lung diseases. The mechanisms of alveologenesis remain largely unknown. We inactivated Pdgfra postnatally in secondary crest myofibroblasts (SCMF), a subpopulation of lung mesenchymal cells. Lack of Pdgfra arrested alveologenesis akin to bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. The transcriptome of mutant SCMF revealed 1808 altered genes encoding transcription factors, signaling and extracellular matrix molecules. Elastin mRNA was reduced, and its distribution was abnormal. Absence of Pdgfra disrupted expression of elastogenic genes, including members of the Lox, Fbn and Fbln families. Expression of EGF family members increased when Tgfb1 was repressed in mouse. Similar, but not identical, results were found in human BPD lung samples. In vitro, blocking PDGF signaling decreased elastogenic gene expression associated with increased Egf and decreased Tgfb family mRNAs. The effect was reversible by inhibiting EGF or activating TGFβ signaling. These observations demonstrate the previously unappreciated postnatal role of PDGFA/PDGFRα in controlling elastogenic gene expression via a secondary tier of signaling networks composed of EGF and TGFβ.
Collapse
Affiliation(s)
- Changgong Li
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Matt K Lee
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Feng Gao
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Sha Webster
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Helen Di
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Jiang Duan
- Department of Pediatrics, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Chang-Yo Yang
- Department of Pediatrics, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan
| | - Navin Bhopal
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Neil Peinado
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Susan M Smith
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Zea Borok
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Saverio Bellusci
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA.,Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), 35392, Giessen, Germany
| | - Parviz Minoo
- Department of Pediatrics, Division of Newborn Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| |
Collapse
|
16
|
Deng S, Zhang H, Han W, Guo C, Deng C. Transforming Growth Factor-β-Neutralizing Antibodies Improve Alveolarization in the Oxygen-Exposed Newborn Mouse Lung. J Interferon Cytokine Res 2019; 39:106-116. [PMID: 30657417 DOI: 10.1089/jir.2018.0080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Abnormal alveolar formation and excessive disordered elastin accumulation are key pathological features in bronchopulmonary dysplasia. Transforming growth factor (TGF)-β is an important regulator of the extracellular matrix in the developing lung. To determine if increased TGF-β would injure alveolar development by activating TGF-β signaling and by influencing the expression of elastogenesis-related protein, we performed intraperitoneal injection of newborn mice with the TGF-β-neutralizing antibody 1D11 and observed whether 1D11 had a protective role in the oxygen (O2)-exposed newborn mouse lung. The newborn mice were exposed to 85% O2 for 14 and 21 days. 1D11 was administered by intraperitoneal injection every day from postnatal days 3 to 20. Alveolar morphology was assessed by hematoxylin and eosin staining. The expression and distribution of elastin were evaluated by immunohistochemistry. The level of TGF-β signaling-related proteins were measured by immunohistochemistry, enzyme-linked immunosorbent assay, and Western blot. The expression levels of elastogenesis-related proteins, including tropoelastin, fibulin-5, and neutrophil elastase (NE), which participate in the synthesis, assembly, and degradation of elastin, were detected by real-time PCR and Western blot. In this research, impaired alveolar development and elastin deposition as well as the excessive activation of TGF-β signaling were observed in the newborn mouse lung exposed to hyperoxia. 1D11 improved alveolarization as well as the distribution of elastin in the newborn lung with hyperoxia exposure. The expression levels of tropoelastin, fibulin-5, and NE, which are important components of elastogenesis, were decreased by treatment with 1D11 in the injured newborn lung. These data demonstrate that 1D11 improved alveolarization by blocking the TGF-β signaling pathway and by reducing the abnormal expression of elastogenesis-related proteins in the O2-exposed newborn mouse lung. 1D11 may become a new therapeutic method to prevent the development of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Sijun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Han Zhang
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wenli Han
- 2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.,4 Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China.,5 Department of Hepatology and Liver Transplantation Center, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,2 China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,3 Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Alejandre Alcazar MA, Kaschwich M, Ertsey R, Preuss S, Milla C, Mujahid S, Masumi J, Khan S, Mokres LM, Tian L, Mohr J, Hirani DV, Rabinovitch M, Bland RD. Elafin Treatment Rescues EGFR-Klf4 Signaling and Lung Cell Survival in Ventilated Newborn Mice. Am J Respir Cell Mol Biol 2018; 59:623-634. [PMID: 29894205 PMCID: PMC6236693 DOI: 10.1165/rcmb.2017-0332oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
Mechanical ventilation with O2-rich gas (MV-O2) inhibits alveologenesis and lung growth. We previously showed that MV-O2 increased elastase activity and apoptosis in lungs of newborn mice, whereas elastase inhibition by elafin suppressed apoptosis and enabled lung growth. Pilot studies suggested that MV-O2 reduces lung expression of prosurvival factors phosphorylated epidermal growth factor receptor (pEGFR) and Krüppel-like factor 4 (Klf4). Here, we sought to determine whether apoptosis and lung growth arrest evoked by MV-O2 reflect disrupted pEGFR-Klf4 signaling, which elafin treatment preserves, and to assess potential biomarkers of bronchopulmonary dysplasia (BPD). Five-day-old mice underwent MV with air or 40% O2 for 8-24 hours with or without elafin treatment. Unventilated pups served as controls. Immunoblots were used to assess lung pEGFR and Klf4 proteins. Cultured MLE-12 cells were exposed to AG1478 (EGFR inhibitor), Klf4 siRNA, or vehicle to assess effects on proliferation, apoptosis, and EGFR regulation of Klf4. Plasma elastase and elafin levels were measured in extremely premature infants. In newborn mice, MV with air or 40% O2 inhibited EGFR phosphorylation and suppressed Klf4 protein content in lungs (vs. unventilated controls), yielding increased apoptosis. Elafin treatment inhibited elastase, preserved lung pEGFR and Klf4, and attenuated the apoptosis observed in lungs of vehicle-treated mice. In MLE-12 studies, pharmacological inhibition of EGFR and siRNA suppression of Klf4 increased apoptosis and reduced proliferation, and EGFR inhibition decreased Klf4. Plasma elastase levels were more than twofold higher, without a compensating increase of plasma elafin, in infants with BPD, compared to infants without BPD. These findings indicate that pEGFR-Klf4 is a novel prosurvival signaling pathway in lung epithelium that MV disrupts. Elafin preserves pEGFR-Klf4 signaling and inhibits apoptosis, thereby enabling lung growth during MV. Together, our animal and human data raise the question: would elastase inhibition prevent BPD in high-risk infants exposed to MV-O2?
Collapse
Affiliation(s)
- Miguel A. Alejandre Alcazar
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
- Department of Pediatric and Adolescent Medicine, Center of Molecular Medicine Cologne, University Hospital of Cologne, Cologne, Germany
| | - Mark Kaschwich
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Robert Ertsey
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Stefanie Preuss
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Carlos Milla
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Sana Mujahid
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Juliet Masumi
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Suleman Khan
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Lucia M. Mokres
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Lu Tian
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Jasmine Mohr
- Department of Pediatric and Adolescent Medicine, Center of Molecular Medicine Cologne, University Hospital of Cologne, Cologne, Germany
| | - Dharmesh V. Hirani
- Department of Pediatric and Adolescent Medicine, Center of Molecular Medicine Cologne, University Hospital of Cologne, Cologne, Germany
| | - Marlene Rabinovitch
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| | - Richard D. Bland
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and
| |
Collapse
|
18
|
Tang X. Interleukin-33 (IL-33) Increases Hyperoxia-Induced Bronchopulmonary Dysplasia in Newborn Mice by Regulation of Inflammatory Mediators. Med Sci Monit 2018; 24:6717-6728. [PMID: 30244258 PMCID: PMC6266634 DOI: 10.12659/msm.910851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Interleukin-33 (IL-33) has been reported to affect chronic inflammation of the lungs, but its impact on hyperoxia-injured lungs in newborns remains obscure. This study aimed to investigate the role of IL-33 in the lungs of neonatal mice with hyperoxia-induced bronchopulmonary dysplasia (BPD). Material/Methods Twenty-four C57BL/6 baby mice were randomly separated into three groups: the on-air group (N=16); the O2 group (N=8); and the O2 + anti-IL-33 group (N=8). Forced mechanical ventilation with oxygen-rich air (MV-O2) was used in 16 mouse pups. The mouse pups were incubated in containers with either air or 85% O2 for 1, 3, 7, 14, 21, and 28 days after birth. At the end of the treatment period, the mouse lungs were studied by histology, Western blot, and quantitative real-time polymerase chain reaction (qRT-PCR) to examine the expression of the pro-inflammatory mediators, including interleukin (IL)-1β, chemokine (CC motif) ligand 1 (CXCL-1), and monocyte chemoattractant protein-1 (MCP-1). Results Following forced MV-O2, increased levels of IL-33 in whole mouse lungs were associated with impaired alveolar growth and with changes consistent with BPD, including reduced numbers of enlarged alveoli, increased apoptosis, and increased expression of IL-1β, CXCL-1, and MCP-1. IL-33 inhibition improved alveolar development in hyperoxia-impaired lungs and suppressed IL-1β and MCP-1 expression and was associated with increased transforming growth factor-β (TGF-β) signaling, reduced pulmonary NF-κB activity and decreased expression of the TGF-β inhibitor SMAD-7 in forced MV-O2 exposed mouse pups. Conclusions IL-33 increased hyperoxia-induced BPD in newborn mice by regulation of the expression of inflammatory mediators.
Collapse
Affiliation(s)
- Xiqin Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
19
|
Mižíková I, Pfeffer T, Nardiello C, Surate Solaligue DE, Steenbock H, Tatsukawa H, Silva DM, Vadász I, Herold S, Pease RJ, Iismaa SE, Hitomi K, Seeger W, Brinckmann J, Morty RE. Targeting transglutaminase 2 partially restores extracellular matrix structure but not alveolar architecture in experimental bronchopulmonary dysplasia. FEBS J 2018; 285:3056-3076. [PMID: 29935061 DOI: 10.1111/febs.14596] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/12/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022]
Abstract
The generation, maturation and remodelling of the extracellular matrix (ECM) are essential for the formation of alveoli during lung development. Alveoli formation is disturbed in preterm infants that develop bronchopulmonary dysplasia (BPD), where collagen fibres are malformed, and perturbations to lung ECM structures may underlie BPD pathogenesis. Malformed ECM structures might result from abnormal protein cross-linking, in part attributable to the increased expression and activity of transglutaminase 2 (TGM2) that have been noted in affected patient lungs, as well as in hyperoxia-based BPD animal models. The objective of the present study was to assess whether TGM2 plays a causal role in normal and aberrant lung alveolarization. Targeted deletion of Tgm2 in C57BL/6J mice increased septal thickness and reduced gas-exchange surface area in otherwise normally developing lungs. During aberrant lung alveolarization that occurred under hyperoxic conditions, collagen structures in Tgm2-/- mice were partially protected from the impact of hyperoxia, where normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance was restored; however, the lung alveolar architecture remained abnormal. Inhibition of transglutaminases (including TGM2) with cysteamine appreciably reduced transglutaminase activity in vivo, as assessed by Nε -(γ-l-glutamyl)-l-lysine abundance and TGM catalytic activity, and restored normal dihydroxylysinonorleucine and hydroxylysylpiridinoline collagen cross-link abundance under pathological conditions. Furthermore, a moderate improvement in alveoli size and gas-exchange surface density was noted in cysteamine-treated mouse lungs in which BPD was modelled. These data indicate that TGM2 plays a role in normal lung alveolarization, and contributes to the formation of aberrant ECM structures during disordered lung alveolarization.
Collapse
Affiliation(s)
- Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Tilman Pfeffer
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Heiko Steenbock
- Institute of Virology and Cell Biology, University of Lübeck, Germany
| | - Hideki Tatsukawa
- Graduate School of Pharmaceutical Sciences, Nagoya University, Japan
| | - Diogo M Silva
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Richard J Pease
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Siiri E Iismaa
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Kiyotaka Hitomi
- Graduate School of Pharmaceutical Sciences, Nagoya University, Japan
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| | - Jürgen Brinckmann
- Institute of Virology and Cell Biology, University of Lübeck, Germany.,Department of Dermatology, University of Lübeck, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Giessen, Germany, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Germany
| |
Collapse
|
20
|
Oak P, Pritzke T, Thiel I, Koschlig M, Mous DS, Windhorst A, Jain N, Eickelberg O, Foerster K, Schulze A, Goepel W, Reicherzer T, Ehrhardt H, Rottier RJ, Ahnert P, Gortner L, Desai TJ, Hilgendorff A. Attenuated PDGF signaling drives alveolar and microvascular defects in neonatal chronic lung disease. EMBO Mol Med 2018; 9:1504-1520. [PMID: 28923828 PMCID: PMC5666314 DOI: 10.15252/emmm.201607308] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Neonatal chronic lung disease (nCLD) affects a significant number of neonates receiving mechanical ventilation with oxygen-rich gas (MV-O2). Regardless, the primary molecular driver of the disease remains elusive. We discover significant enrichment for SNPs in the PDGF-Rα gene in preterms with nCLD and directly test the effect of PDGF-Rα haploinsufficiency on the development of nCLD using a preclinical mouse model of MV-O2 In the context of MV-O2, attenuated PDGF signaling independently contributes to defective septation and endothelial cell apoptosis stemming from a PDGF-Rα-dependent reduction in lung VEGF-A. TGF-β contributes to the PDGF-Rα-dependent decrease in myofibroblast function. Remarkably, endotracheal treatment with exogenous PDGF-A rescues both the lung defects in haploinsufficient mice undergoing MV-O2 Overall, our results establish attenuated PDGF signaling as an important driver of nCLD pathology with provision of PDGF-A as a protective strategy for newborns undergoing MV-O2.
Collapse
Affiliation(s)
- Prajakta Oak
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Tina Pritzke
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Isabella Thiel
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Markus Koschlig
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Daphne S Mous
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Anita Windhorst
- Institute for Medical Informatics, Justus-Liebig-University, Giessen, Germany
| | - Noopur Jain
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Kai Foerster
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Andreas Schulze
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Wolfgang Goepel
- Department of General Pediatrics, University Clinic of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Tobias Reicherzer
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Ludwig Gortner
- Department of Pediatrics and Neonatology, Medical University Vienna, Vienna, Austria
| | - Tushar J Desai
- Department of Internal Medicine, Pulmonary and Critical Care, Stanford University School of Medicine, Stanford, CA, USA
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany .,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany.,Center for Comprehensive Developmental Care, Dr. von Haunersches Children's Hospital University Hospital Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
21
|
Yin J, Wang X, Zhang L, Wang X, Liu H, Hu Y, Yan X, Tang Y, Wang J, Li Z, Yu Z, Cao Y, Han S. Peptidome analysis of lung tissues from a hyperoxia‐induced bronchopulmonary dysplasia mouse model: Insights into the pathophysiological process of bronchopulmonary dysplasia. J Cell Physiol 2018; 233:7101-7112. [PMID: 29741761 DOI: 10.1002/jcp.26633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/30/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Jing Yin
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Xingyun Wang
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Le Zhang
- Department of Neonatology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Heng Liu
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Yin Hu
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Xiangyun Yan
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Yongfeng Tang
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Juan Wang
- Department of Pediatrics, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Zhengyin Li
- Department of Neonatology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhangbin Yu
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| | - Shuping Han
- Department of Pediatrics, The Affiliated Obsterics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
23
|
Oak P, Hilgendorff A. The BPD trio? Interaction of dysregulated PDGF, VEGF, and TGF signaling in neonatal chronic lung disease. Mol Cell Pediatr 2017; 4:11. [PMID: 29116547 PMCID: PMC5676585 DOI: 10.1186/s40348-017-0076-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022] Open
Abstract
The development of neonatal chronic lung disease (nCLD), i.e., bronchopulmonary dysplasia (BPD) in preterm infants, significantly determines long-term outcome in this patient population. Risk factors include mechanical ventilation and oxygen toxicity impacting on the immature lung resulting in impaired alveolarization and vascularization. Disease development is characterized by inflammation, extracellular matrix remodeling, and apoptosis, closely intertwined with the dysregulation of growth factor signaling. This review focuses on the causes and consequences of altered signaling in central pathways like transforming growth factor (TGF), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF) driving these above indicated processes, i.e., inflammation, matrix remodeling, and vascular development. We emphasize the shared and distinct role of these pathways as well as their interconnection in disease initiation and progression, generating important knowledge for the development of future treatment strategies.
Collapse
Affiliation(s)
- Prajakta Oak
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany.
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany.
- Center for Comprehensive Developmental Care, Dr. von Haunersches Children's Hospital University, Hospital Ludwig-Maximilians University, Munich, Germany.
| |
Collapse
|
24
|
Recombinant human elafin promotes alveologenesis in newborn mice exposed to chronic hyperoxia. Int J Biochem Cell Biol 2017; 92:173-182. [DOI: 10.1016/j.biocel.2017.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 01/20/2023]
|
25
|
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: 4.9] [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
|
26
|
Durrani-Kolarik S, Pool CA, Gray A, Heyob KM, Cismowski MJ, Pryhuber G, Lee LJ, Yang Z, Tipple TE, Rogers LK. miR-29b supplementation decreases expression of matrix proteins and improves alveolarization in mice exposed to maternal inflammation and neonatal hyperoxia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L339-L349. [PMID: 28473324 PMCID: PMC5582933 DOI: 10.1152/ajplung.00273.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 04/28/2017] [Accepted: 04/30/2017] [Indexed: 02/06/2023] Open
Abstract
Even with advances in the care of preterm infants, chronic lung disease or bronchopulmonary dysplasia (BPD) continues to be a significant pulmonary complication. Among those diagnosed with BPD, a subset of infants develop severe BPD with disproportionate pulmonary morbidities. In addition to decreased alveolarization, these infants develop obstructive and/or restrictive lung function due to increases in or dysregulation of extracellular matrix proteins. Analyses of plasma obtained from preterm infants during the first week of life indicate that circulating miR-29b is suppressed in infants that subsequently develop BPD and that decreased circulating miR-29b is inversely correlated with BPD severity. Our mouse model mimics the pathophysiology observed in infants with severe BPD, and we have previously reported decreased pulmonary miR-29b expression in this model. The current studies tested the hypothesis that adeno-associated 9 (AAV9)-mediated restoration of miR-29b in the developing lung will improve lung alveolarization and minimize the deleterious changes in matrix deposition. Pregnant C3H/HeN mice received an intraperitoneal LPS injection on embryonic day 16 and newborn pups were exposed to 85% oxygen from birth to 14 days of life. On postnatal day 3, AAV9-miR-29b or AAV9-control was administered intranasally. Mouse lung tissues were then analyzed for changes in miR-29 expression, alveolarization, and matrix protein levels and localization. Although only modest improvements in alveolarization were detected in the AAV9-miR29b-treated mice at postnatal day 28, treatment completely attenuated defects in matrix protein expression and localization. Our data suggest that miR-29b restoration may be one component of a novel therapeutic strategy to treat or prevent severe BPD in prematurely born infants.
Collapse
Affiliation(s)
- Shaheen Durrani-Kolarik
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Caylie A Pool
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ashley Gray
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Kathryn M Heyob
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Mary J Cismowski
- Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - L James Lee
- The Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio
| | - Zhaogang Yang
- The Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio
| | - Trent E Tipple
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Lynette K Rogers
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio;
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
27
|
Mižíková I, Palumbo F, Tábi T, Herold S, Vadász I, Mayer K, Seeger W, Morty RE. Perturbations to lysyl oxidase expression broadly influence the transcriptome of lung fibroblasts. Physiol Genomics 2017; 49:416-429. [DOI: 10.1152/physiolgenomics.00026.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 01/05/2023] Open
Abstract
Lysyl oxidases are credited with pathogenic roles in lung diseases, including cancer, fibrosis, pulmonary hypertension, congenital diaphragmatic hernia, and bronchopulmonary dysplasia (BPD). Lysyl oxidases facilitate the covalent intra- and intermolecular cross-linking of collagen and elastin fibers, thereby imparting tensile strength to the extracellular matrix (ECM). Alternative ECM-independent roles have recently been proposed for lysyl oxidases, including regulation of growth factor signaling, chromatin remodeling, and transcriptional regulation, all of which impact cell phenotype. We demonstrate here that three of the five lysyl oxidase family members, Lox, Loxl1, and Loxl2, are highly expressed in primary mouse lung fibroblasts compared with other constituent cell types of the lung. Microarray analyses revealed that small interfering RNA knockdown of Lox, Loxl1, and Loxl2 was associated with apparent changes in the expression of 134, 3,761, and 3,554 genes, respectively, in primary mouse lung fibroblasts. The impact of lysyl oxidase expression on steady-state Mmp3, Mmp9, Eln, Rarres1, Gdf10, Ifnb1, Csf2, and Cxcl9 mRNA levels was validated, which is interesting, since the corresponding gene products are relevant to lung development and BPD, where lysyl oxidases play a functional role. In vivo, the expression of these genes broadly correlated with Lox, Loxl1, and Loxl2 expression in a mouse model of BPD. Furthermore, β-aminopropionitrile (BAPN), a selective lysyl oxidase inhibitor, did not affect the steady-state mRNA levels of lysyl oxidase target genes, in vitro in lung fibroblasts or in vivo in BAPN-treated mice. This study is the first to report that lysyl oxidases broadly influence the cell transcriptome.
Collapse
Affiliation(s)
- Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Francesco Palumbo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Tamás Tábi
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Konstantin Mayer
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| | - Rory E. Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany; and
| |
Collapse
|
28
|
Nardiello C, Mižíková I, Morty RE. Looking ahead: where to next for animal models of bronchopulmonary dysplasia? Cell Tissue Res 2016; 367:457-468. [PMID: 27917436 PMCID: PMC5320021 DOI: 10.1007/s00441-016-2534-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth, with appreciable morbidity and mortality in a neonatal intensive care setting. Much interest has been shown in the identification of pathogenic pathways that are amenable to pharmacological manipulation (1) to facilitate the development of novel therapeutic and medical management strategies and (2) to identify the basic mechanisms of late lung development, which remains poorly understood. A number of animal models have therefore been developed and continue to be refined with the aim of recapitulating pathological pulmonary hallmarks noted in lungs from neonates with BPD. These animal models rely on several injurious stimuli, such as mechanical ventilation or oxygen toxicity and infection and sterile inflammation, as applied in mice, rats, rabbits, pigs, lambs and nonhuman primates. This review addresses recent developments in modeling BPD in experimental animals and highlights important neglected areas that demand attention. Additionally, recent progress in the quantitative microscopic analysis of pathology tissue is described, together with new in vitro approaches of value for the study of normal and aberrant alveolarization. The need to examine long-term sequelae of damage to the developing neonatal lung is also considered, as is the need to move beyond the study of the lungs alone in experimental animal models of BPD.
Collapse
Affiliation(s)
- Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany. .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
| |
Collapse
|
29
|
Zhang J, Liu Z, Zhang T, Lin Z, Li Z, Zhang A, Sun X, Gao J. Loss of Lysyl Oxidase-like 3 Attenuates Embryonic Lung Development in Mice. Sci Rep 2016; 6:33856. [PMID: 27645581 PMCID: PMC5029289 DOI: 10.1038/srep33856] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 09/05/2016] [Indexed: 12/31/2022] Open
Abstract
Lysyl oxidase-like 3 (LOXL3), a human disease gene candidate, is a member of the lysyl oxidase (LOX) family and is indispensable for mouse palatogenesis and vertebral column development. Our previous study showed that the loss of LOXL3 resulted in a severe cleft palate and spinal deformity. In this study, we investigated a possible role for LOXL3 in mouse embryonic lung development. LOXL3-deficient mice displayed reduced lung volumes and weights, diminished saccular spaces, and deformed and smaller thoracic cavities. Excess elastic fibres were detected in LOXL3-deficient lungs, which might be related to the increased LOXL4 expression. Increased transforming growth factor β1 (TGFβ1) expression might be involved in the up-regulation of LOXL4 in LOXL3-deficient lungs. We concluded that the loss of LOXL3 attenuates mouse embryonic lung development.
Collapse
Affiliation(s)
- Jian Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Ziyi Liu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Tingting Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Zhuchun Lin
- Jinan First People's Hospital, Jinan 250011, China
| | - Zhenzu Li
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Aizhen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xiaoyang Sun
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| |
Collapse
|
30
|
Meiners S, Hilgendorff A. Early injury of the neonatal lung contributes to premature lung aging: a hypothesis. Mol Cell Pediatr 2016; 3:24. [PMID: 27406259 PMCID: PMC4942446 DOI: 10.1186/s40348-016-0052-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
Chronic lung disease of the newborn, also known as bronchopulmonary dysplasia (BPD), is the most common chronic lung disease in early infancy and results in an increased risk for long-lasting pulmonary impairment in the adult. BPD develops upon injury of the immature lung by oxygen toxicity, mechanical ventilation, and infections which trigger sustained inflammatory immune responses and extensive remodeling of the extracellular matrix together with dysregulated growth factor signaling. Histopathologically, BPD is characterized by impaired alveolarization, disrupted vascular development, and saccular wall fibrosis. Here, we explore the hypothesis that development of BPD involves disturbance of conserved pathways of molecular aging that may contribute to premature aging of the lung and an increased susceptibility to chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), Ludwig-Maximilians University, Helmholtz Zentrum München, German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377, München, Germany.
| | - Anne Hilgendorff
- Comprehensive Pneumology Center (CPC), Ludwig-Maximilians University, Helmholtz Zentrum München, German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377, München, Germany.,Perinatal Center Grosshadern, Dr. von Haunersches Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
31
|
Shahzad T, Radajewski S, Chao CM, Bellusci S, Ehrhardt H. Pathogenesis of bronchopulmonary dysplasia: when inflammation meets organ development. Mol Cell Pediatr 2016; 3:23. [PMID: 27357257 PMCID: PMC4927524 DOI: 10.1186/s40348-016-0051-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/20/2016] [Indexed: 01/12/2023] Open
Abstract
Bronchopulmonary dysplasia is a chronic lung disease of preterm infants. It is caused by the disturbance of physiologic lung development mainly in the saccular stage with lifelong restrictions of pulmonary function and an increased risk of abnormal somatic and psychomotor development. The contributors to this disease’s entity are multifactorial with pre- and postnatal origin. Central to the pathogenesis of bronchopulmonary is the induction of a massive pulmonary inflammatory response due to mechanical ventilation and oxygen toxicity. The extent of the pro-inflammatory reaction and the disturbance of further alveolar growth and vasculogenesis vary largely and can be modified by prenatal infections, antenatal steroids, and surfactant application. This minireview summarizes the important recent research findings on the pulmonary inflammatory reaction obtained in patient cohorts and in experimental models. Unfortunately, recent changes in clinical practice based on these findings had only limited impact on the incidence of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Tayyab Shahzad
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Sarah Radajewski
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Cho-Ming Chao
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany.,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Saverio Bellusci
- University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany
| | - Harald Ehrhardt
- 1Department of General Pediatrics and Neonatology, Center for Pediatrics and Youth Medicine, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Giessen, Germany. .,University of Giessen Lung Center, Excellence Cluster Cardio-Pulmonary Systems, Member of the German Lung Center, Department of Internal Medicine II, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|
32
|
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: 5.2] [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
|
33
|
Yaroshenko A, Pritzke T, Koschlig M, Kamgari N, Willer K, Gromann L, Auweter S, Hellbach K, Reiser M, Eickelberg O, Pfeiffer F, Hilgendorff A. Visualization of neonatal lung injury associated with mechanical ventilation using x-ray dark-field radiography. Sci Rep 2016; 6:24269. [PMID: 27072871 PMCID: PMC4829826 DOI: 10.1038/srep24269] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/23/2016] [Indexed: 02/01/2023] Open
Abstract
Mechanical ventilation (MV) and supplementation of oxygen-enriched gas, often needed in postnatal resuscitation procedures, are known to be main risk factors for impaired pulmonary development in the preterm and term neonates. Unfortunately, current imaging modalities lack in sensitivity for the detection of early stage lung injury. The present study reports a new imaging approach for diagnosis and staging of early lung injury induced by MV and hyperoxia in neonatal mice. The imaging method is based on the Talbot-Lau x-ray grating interferometry that makes it possible to quantify the x-ray small-angle scattering on the air-tissue interfaces. This so-called dark-field signal revealed increasing loss of x-ray small-angle scattering when comparing images of neonatal mice undergoing hyperoxia and MV-O2 with animals kept at room air. The changes in the dark field correlated well with histologic findings and provided superior differentiation than conventional x-ray imaging and lung function testing. The results suggest that x-ray dark-field radiography is a sensitive tool for assessing structural changes in the developing lung. In the future, with further technical developments x-ray dark-field imaging could be an important tool for earlier diagnosis and sensitive monitoring of lung injury in neonates requiring postnatal oxygen or ventilator therapy.
Collapse
Affiliation(s)
- Andre Yaroshenko
- Lehrstuhl für Biomedizinische Physik, Physik-Department &Institut für Medizintechnik, Technische Universität München, Garching, Germany
| | - Tina Pritzke
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Markus Koschlig
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Nona Kamgari
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Konstantin Willer
- Lehrstuhl für Biomedizinische Physik, Physik-Department &Institut für Medizintechnik, Technische Universität München, Garching, Germany
| | - Lukas Gromann
- Lehrstuhl für Biomedizinische Physik, Physik-Department &Institut für Medizintechnik, Technische Universität München, Garching, Germany
| | - Sigrid Auweter
- Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital Munich, Munich
| | - Katharina Hellbach
- Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital Munich, Munich
| | - Maximilian Reiser
- Institute for Clinical Radiology, Ludwig-Maximilians-University Hospital Munich, Munich
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Franz Pfeiffer
- Lehrstuhl für Biomedizinische Physik, Physik-Department &Institut für Medizintechnik, Technische Universität München, Garching, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany, Member of the German Center for Lung Research (DZL).,Department of Neonatology, Perinatal Center, Dr. von Haunersches Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
34
|
Ehrhardt H, Pritzke T, Oak P, Kossert M, Biebach L, Förster K, Koschlig M, Alvira CM, Hilgendorff A. Absence of TNF-α enhances inflammatory response in the newborn lung undergoing mechanical ventilation. Am J Physiol Lung Cell Mol Physiol 2016; 310:L909-18. [PMID: 27016588 DOI: 10.1152/ajplung.00367.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/18/2016] [Indexed: 12/25/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), characterized by impaired alveolarization and vascularization in association with lung inflammation and apoptosis, often occurs after mechanical ventilation with oxygen-rich gas (MV-O2). As heightened expression of the proinflammatory cytokine TNF-α has been described in infants with BPD, we hypothesized that absence of TNF-α would reduce pulmonary inflammation, and attenuate structural changes in newborn mice undergoing MV-O2 Neonatal TNF-α null (TNF-α(-/-)) and wild type (TNF-α(+/+)) mice received MV-O2 for 8 h; controls spontaneously breathed 40% O2 Histologic, mRNA, and protein analysis in vivo were complemented by in vitro studies subjecting primary pulmonary myofibroblasts to mechanical stretch. Finally, TNF-α level in tracheal aspirates from preterm infants were determined by ELISA. Although MV-O2 induced larger and fewer alveoli in both, TNF-α(-/-) and TNF-α(+/+) mice, it caused enhanced lung apoptosis (TUNEL, caspase-3/-6/-8), infiltration of macrophages and neutrophils, and proinflammatory mediator expression (IL-1β, CXCL-1, MCP-1) in TNF-α(-/-) mice. These differences were associated with increased pulmonary transforming growth factor-β (TGF-β) signaling, decreased TGF-β inhibitor SMAD-7 expression, and reduced pulmonary NF-κB activity in ventilated TNF-α(-/-) mice. Preterm infants who went on to develop BPD showed significantly lower TNF-α levels at birth. Our results suggest a critical balance between TNF-α and TGF-β signaling in the developing lung, and underscore the critical importance of these key pathways in the pathogenesis of BPD. Future treatment strategies need to weigh the potential benefits of inhibiting pathologic cytokine expression against the potential of altering key developmental pathways.
Collapse
Affiliation(s)
- Harald Ehrhardt
- Department of General Pediatrics and Neonatology, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Tina Pritzke
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany
| | - Prajakta Oak
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany
| | - Melina Kossert
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany
| | - Luisa Biebach
- Department of Neonatology, Dr. von Haunersches Children's Hospital, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Kai Förster
- Department of Neonatology, Dr. von Haunersches Children's Hospital, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Markus Koschlig
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany
| | | | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Zentrum Muenchen, Munich, Germany; Department of Neonatology, Dr. von Haunersches Children's Hospital, Ludwig-Maximilians University of Munich, Munich, Germany;
| |
Collapse
|
35
|
Ahlfeld SK, Wang J, Gao Y, Snider P, Conway SJ. Initial Suppression of Transforming Growth Factor-β Signaling and Loss of TGFBI Causes Early Alveolar Structural Defects Resulting in Bronchopulmonary Dysplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:777-93. [PMID: 26878215 DOI: 10.1016/j.ajpath.2015.11.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 11/02/2015] [Accepted: 11/18/2015] [Indexed: 12/22/2022]
Abstract
Septation of the gas-exchange saccules of the morphologically immature mouse lung requires regulated timing, spatial direction, and dosage of transforming growth factor (TGF)-β signaling. We found that neonatal hyperoxia acutely initially diminished saccular TGF-β signaling coincident with alveolar simplification. However, sustained hyperoxia resulted in a biphasic response and subsequent up-regulation of TGF-β signaling, ultimately resulting in bronchopulmonary dysplasia. Significantly, we found that the TGF-β-induced matricellular protein (TGFBI) was similarly biphasically altered in response to hyperoxia. Moreover, genetic ablation revealed that TGFBI was required for normal alveolar structure and function. Although the phenotype was not neonatal lethal, Tgfbi-deficient lungs were morphologically abnormal. Mutant septal tips were stunted, lacked elastin-positive tips, exhibited reduced proliferation, and contained abnormally persistent alveolar α-smooth muscle actin myofibroblasts. In addition, Tgfbi-deficient lungs misexpressed TGF-β-responsive follistatin and serpine 1, and transiently suppressed myofibroblast platelet-derived growth factor α differentiation marker. Finally, despite normal lung volume, Tgfbi-null lungs displayed diminished elastic recoil and gas exchange efficiency. Combined, these data demonstrate that initial suppression of the TGF-β signaling apparatus, as well as loss of key TGF-β effectors (like TGFBI), underlies early alveolar structural defects, as well as long-lasting functional deficits routinely observed in chronic lung disease of infancy patients. These studies underline the complex (and often contradictory) role of TGF-β and indicate a need to design studies to associate alterations with initial appearance of phenotypical changes suggestive of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Shawn K Ahlfeld
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jian Wang
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yong Gao
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Paige Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
36
|
Yu B, Li X, Wan Q, Han W, Deng C, Guo C. High-Mobility Group Box-1 Protein Disrupts Alveolar Elastogenesis of Hyperoxia-Injured Newborn Lungs. J Interferon Cytokine Res 2016; 36:159-68. [PMID: 26982166 DOI: 10.1089/jir.2015.0080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although high-mobility group box-1 (HMGB1) levels in tracheal aspirates are associated with the pathological features of bronchopulmonary dysplasia (BPD), the role of HMGB1 in the terminal stage of abnormal alveologenesis has not yet been understood. In this study, we addressed the role of HMGB1 in the elastogenesis disruption in the lungs of newborn mice with BPD. We found that elevations of whole lung HMGB1 level were associated with impaired alveolar development and aberrant elastin production in 85% O2-exposed lungs. HMGB1 neutralizing antibody attenuated the structural disintegration developed in hyperoxia-damaged lungs. Furthermore, HMGB1 inhibition rescued the neutrophil influx in hyperoxia-injured lung and partially abolished the mRNA level of the proinflammatory mediators, interleukin (IL)-1β and transforming growth factor (TGF)-β1. These data suggested that pulmonary HMGB1 plays an important role in the disruption of elastogenesis in the terminal stage of lung development through reduced pulmonary inflammatory response.
Collapse
Affiliation(s)
- Benli Yu
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Xiaoyu Li
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Qiufeng Wan
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Wenli Han
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,3 Department of Pharmacology, Chongqing Medical University , Chongqing, P.R. China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,4 Department of Hepatology and Liver Transplantation Center, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| |
Collapse
|
37
|
Mižíková I, Morty RE. The Extracellular Matrix in Bronchopulmonary Dysplasia: Target and Source. Front Med (Lausanne) 2015; 2:91. [PMID: 26779482 PMCID: PMC4688343 DOI: 10.3389/fmed.2015.00091] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/08/2015] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication of preterm birth that contributes significantly to morbidity and mortality in neonatal intensive care units. BPD results from life-saving interventions, such as mechanical ventilation and oxygen supplementation used to manage preterm infants with acute respiratory failure, which may be complicated by pulmonary infection. The pathogenic pathways driving BPD are not well-delineated but include disturbances to the coordinated action of gene expression, cell-cell communication, physical forces, and cell interactions with the extracellular matrix (ECM), which together guide normal lung development. Efforts to further delineate these pathways have been assisted by the use of animal models of BPD, which rely on infection, injurious mechanical ventilation, or oxygen supplementation, where histopathological features of BPD can be mimicked. Notable among these are perturbations to ECM structures, namely, the organization of the elastin and collagen networks in the developing lung. Dysregulated collagen deposition and disturbed elastin fiber organization are pathological hallmarks of clinical and experimental BPD. Strides have been made in understanding the disturbances to ECM production in the developing lung, but much still remains to be discovered about how ECM maturation and turnover are dysregulated in aberrantly developing lungs. This review aims to inform the reader about the state-of-the-art concerning the ECM in BPD, to highlight the gaps in our knowledge and current controversies, and to suggest directions for future work in this exciting and complex area of lung development (patho)biology.
Collapse
Affiliation(s)
- Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Pulmonology, Department of Internal Medicine, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Pulmonology, Department of Internal Medicine, University of Giessen and Marburg Lung Center, Giessen, Germany
| |
Collapse
|
38
|
Balany J, Bhandari V. Understanding the Impact of Infection, Inflammation, and Their Persistence in the Pathogenesis of Bronchopulmonary Dysplasia. Front Med (Lausanne) 2015; 2:90. [PMID: 26734611 PMCID: PMC4685088 DOI: 10.3389/fmed.2015.00090] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
The concerted interaction of genetic and environmental factors acts on the preterm human immature lung with inflammation being the common denominator leading to the multifactorial origin of the most common chronic lung disease in infants – bronchopulmonary dysplasia (BPD). Adverse perinatal exposure to infection/inflammation with added insults like invasive mecha nical ventilation, exposure to hyperoxia, and sepsis causes persistent immune dysregulation. In this review article, we have attempted to analyze and consolidate current knowledge about the role played by persistent prenatal and postnatal inflammation in the pathogenesis of BPD. While some parameters of the early inflammatory response (neutrophils, cytokines, etc.) may not be detectable after days to weeks of exposure to noxious stimuli, they have already initiated the signaling pathways of the inflammatory process/immune cascade and have affected permanent defects structurally and functionally in the BPD lungs. Hence, translational research aimed at prevention/amelioration of BPD needs to focus on dampening the inflammatory response at an early stage to prevent the cascade of events leading to lung injury with impaired healing resulting in the pathologic pulmonary phenotype of alveolar simplification and dysregulated vascularization characteristic of BPD.
Collapse
Affiliation(s)
- Jherna Balany
- Section of Neonatology, Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine , Philadelphia, PA , USA
| | - Vineet Bhandari
- Section of Neonatology, Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine , Philadelphia, PA , USA
| |
Collapse
|
39
|
Silva DMG, Nardiello C, Pozarska A, Morty RE. Recent advances in the mechanisms of lung alveolarization and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1239-72. [PMID: 26361876 DOI: 10.1152/ajplung.00268.2015] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/09/2015] [Indexed: 02/08/2023] Open
Abstract
Alveolarization is the process by which the alveoli, the principal gas exchange units of the lung, are formed. Along with the maturation of the pulmonary vasculature, alveolarization is the objective of late lung development. The terminal airspaces that were formed during early lung development are divided by the process of secondary septation, progressively generating an increasing number of alveoli that are of smaller size, which substantially increases the surface area over which gas exchange can take place. Disturbances to alveolarization occur in bronchopulmonary dysplasia (BPD), which can be complicated by perturbations to the pulmonary vasculature that are associated with the development of pulmonary hypertension. Disturbances to lung development may also occur in persistent pulmonary hypertension of the newborn in term newborn infants, as well as in patients with congenital diaphragmatic hernia. These disturbances can lead to the formation of lungs with fewer and larger alveoli and a dysmorphic pulmonary vasculature. Consequently, affected lungs exhibit a reduced capacity for gas exchange, with important implications for morbidity and mortality in the immediate postnatal period and respiratory health consequences that may persist into adulthood. It is the objective of this Perspectives article to update the reader about recent developments in our understanding of the molecular mechanisms of alveolarization and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Diogo M G Silva
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Claudio Nardiello
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Agnieszka Pozarska
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rory E Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
40
|
Jobe AH. Animal Models, Learning Lessons to Prevent and Treat Neonatal Chronic Lung Disease. Front Med (Lausanne) 2015; 2:49. [PMID: 26301222 PMCID: PMC4528292 DOI: 10.3389/fmed.2015.00049] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/14/2015] [Indexed: 11/23/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a unique injury syndrome caused by prolonged injury and repair imposed on an immature and developing lung. The decreased septation and decreased microvascular development phenotype of BPD can be reproduced in newborn rodents with increased chronic oxygen exposure and in premature primates and sheep with oxygen and/or mechanical ventilation. The inflammation caused by oxidants, inflammatory agonists, and/or stretch injury from mechanical ventilation seems to promote the anatomic abnormalities. Multiple interventions targeted to specific inflammatory cells or pathways or targeted to decreasing ventilation-mediated injury can substantially prevent the anatomic changes associated with BPD in term rodents and in preterm sheep or primate models. Most of the anti-inflammatory therapies with benefit in animal models have not been tested clinically. None of the interventions that have been tested clinically are as effective as anticipated from the animal models. These inconsistencies in responses likely are explained by the antenatal differences in lung exposures of the developing animals relative to very preterm humans. The animals generally have normal lungs while the lungs of preterm infants are exposed variably to intrauterine inflammation, growth abnormalities, antenatal corticosteroids, and poorly understood effects from the causes of preterm delivery. The animal models have been essential for the definition of the mediators that can cause a BPD phenotype. These models will be necessary to develop and test future-targeted interventions to prevent and treat BPD.
Collapse
Affiliation(s)
- Alan H Jobe
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, OH , USA
| |
Collapse
|
41
|
Mižíková I, Ruiz-Camp J, Steenbock H, Madurga A, Vadász I, Herold S, Mayer K, Seeger W, Brinckmann J, Morty RE. Collagen and elastin cross-linking is altered during aberrant late lung development associated with hyperoxia. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1145-58. [DOI: 10.1152/ajplung.00039.2015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/03/2015] [Indexed: 12/31/2022] Open
Abstract
Maturation of the lung extracellular matrix (ECM) plays an important role in the formation of alveolar gas exchange units. A key step in ECM maturation is cross-linking of collagen and elastin, which imparts stability and functionality to the ECM. During aberrant late lung development in bronchopulmonary dysplasia (BPD) patients and animal models of BPD, alveolarization is blocked, and the function of ECM cross-linking enzymes is deregulated, suggesting that perturbed ECM cross-linking may impact alveolarization. In a hyperoxia (85% O2)-based mouse model of BPD, blunted alveolarization was accompanied by alterations to lung collagen and elastin levels and cross-linking. Total collagen levels were increased (by 63%). The abundance of dihydroxylysinonorleucine collagen cross-links and the dihydroxylysinonorleucine-to-hydroxylysinonorleucine ratio were increased by 11 and 18%, respectively, suggestive of a profibrotic state. In contrast, insoluble elastin levels and the abundance of the elastin cross-links desmosine and isodesmosine in insoluble elastin were decreased by 35, 30, and 21%, respectively. The lung collagen-to-elastin ratio was threefold increased. Treatment of hyperoxia-exposed newborn mice with the lysyl oxidase inhibitor β-aminopropionitrile partially restored normal collagen levels, normalized the dihydroxylysinonorleucine-to-hydroxylysinonorleucine ratio, partially normalized desmosine and isodesmosine cross-links in insoluble elastin, and partially restored elastin foci structure in the developing septa. However, β-aminopropionitrile administration concomitant with hyperoxia exposure did not improve alveolarization, evident from unchanged alveolar surface area and alveoli number, and worsened septal thickening (increased by 12%). These data demonstrate that collagen and elastin cross-linking are perturbed during the arrested alveolarization of developing mouse lungs exposed to hyperoxia.
Collapse
Affiliation(s)
- Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jordi Ruiz-Camp
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Heiko Steenbock
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany; and
| | - Alicia Madurga
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Konstantin Mayer
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Jürgen Brinckmann
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany; and
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Rory E. Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
42
|
Niedermaier S, Hilgendorff A. Bronchopulmonary dysplasia - an overview about pathophysiologic concepts. Mol Cell Pediatr 2015; 2:2. [PMID: 26542292 PMCID: PMC4530566 DOI: 10.1186/s40348-015-0013-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/25/2015] [Indexed: 12/27/2022] Open
Abstract
Neonatal chronic lung disease in the preterm infant, i.e. bronchopulmonary dysplasia (BPD) is characterized by impaired pulmonary development with its effects persisting into adulthood. Triggered in the immature lung by infectious complications, oxygen toxicity and the impact of mechanical ventilation, a sustained inflammatory response, extensive remodeling of the extracellular matrix, increased apoptosis as well as altered growth factor signaling characterize the disease. The current review focuses on selected pathophysiologic processes and their interplay in disease development. Furthermore, the potential of both, acute and long-term changes to the pulmonary scaffold and the cellular interface in concert with dysregulated growth factor signaling to affect aging and repair processes in the adult lung is discussed.
Collapse
Affiliation(s)
- Sophie Niedermaier
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich Max-Lebsche-Platz 31, 81377, Munich, Germany. .,Dr. von Hauner Children's Hospital, Ludwig-Maximilians University Munich, Munich, Germany.
| | - Anne Hilgendorff
- Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich Max-Lebsche-Platz 31, 81377, Munich, Germany. .,Dr. von Hauner Children's Hospital, Ludwig-Maximilians University Munich, Munich, Germany.
| |
Collapse
|
43
|
Hilgendorff A, O'Reilly MA. Bronchopulmonary dysplasia early changes leading to long-term consequences. Front Med (Lausanne) 2015; 2:2. [PMID: 25729750 PMCID: PMC4325927 DOI: 10.3389/fmed.2015.00002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 01/05/2015] [Indexed: 12/05/2022] Open
Abstract
Neonatal chronic lung disease, i.e., bronchopulmonary dysplasia, is characterized by impaired pulmonary development resulting from the impact of different risk factors including infections, hyperoxia, and mechanical ventilation on the immature lung. Remodeling of the extracellular matrix, apoptosis as well as altered growth factor signaling characterize the disease. The immediate consequences of these early insults have been studied in different animal models supported by results from in vitro approaches leading to the successful application of some findings to the clinical setting in the past. Nonetheless, existing information about long-term consequences of the identified early and most likely sustained changes to the developing lung is limited. Interesting results point towards a tremendous impact of these early injuries on the pulmonary repair capacity as well as aging related processes in the adult lung.
Collapse
Affiliation(s)
- Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL) , Munich , Germany ; Neonatology, Perinatal Center Grosshadern, Dr. von Hauner Children's Hospital, Ludwig-Maximilians University , Munich , Germany
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester , Rochester, NY , USA
| |
Collapse
|
44
|
Kroon AA, Wang J, Post M. Alterations in expression of elastogenic and angiogenic genes by different conditions of mechanical ventilation in newborn rat lung. Am J Physiol Lung Cell Mol Physiol 2015; 308:L639-49. [PMID: 25617376 DOI: 10.1152/ajplung.00293.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/22/2015] [Indexed: 11/22/2022] Open
Abstract
Mechanical ventilation is an important risk factor for development of bronchopulmonary dysplasia. Here we investigated the effects of different tidal volumes (VT) and duration of ventilation on expression of genes involved in alveolarization [tropoelastin (Eln), lysyloxidase-like 1 (Loxl1), fibulin5 (Fbln5), and tenascin-C (Tnc)] and angiogenesis [platelet derived growth factors (Pdgf) and vascular endothelial growth factors (Vegf) and their receptors] in 8-day-old rats. First, pups were ventilated for 8 h with low (LVT: 3.5 ml/kg), moderate (MVT: 8.5 ml/kg), or high (HVT: 25 ml/kg) tidal volumes. LVT and MVT decreased Tnc expression, whereas HVT increased expression of all three elastogenic genes and Tnc. PDGF α-receptor mRNA was increased in all ventilation groups, while Pdgfb expression was decreased after MVT and HVT ventilation. Only HVT ventilation upregulated Vegf expression. Independent of VT, ventilation upregulated Vegfr1 expression, while MVT and HVT downregulated Vegfr2 expression. Next, we evaluated duration (0-24 h) of MVT ventilation on gene expression. Although expression of all elastogenic genes peaked at 12 h of ventilation, only Fbln5 was negatively affected at 24 h. Tnc expression decreased with duration of ventilation. Changes in expression of Pdgfr and Vegfr were maximal at 8 h of ventilation. Disturbed elastin fiber deposition and decrease in small vessel density was only observed after 24 h. Thus, an imbalance between Fbln5 and Eln expression may trigger dysregulated elastin fiber deposition during the first 24 h of mechanical ventilation. Furthermore, ventilation-induced alterations in Pdgf and Vegf receptor expression are tidal volume dependent and may affect pulmonary vessel formation.
Collapse
Affiliation(s)
- Andreas A Kroon
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Canada; Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | - Jinxia Wang
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Martin Post
- Physiology and Experimental Medicine Program, Hospital for Sick Children Research Institute, Toronto, Canada; Department of Pediatrics University of Toronto, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada; and
| |
Collapse
|
45
|
Hilgendorff A, Parai K, Ertsey R, Navarro E, Jain N, Carandang F, Peterson J, Mokres L, Milla C, Preuss S, Alcazar MA, Khan S, Masumi J, Ferreira-Tojais N, Mujahid S, Starcher B, Rabinovitch M, Bland R. Lung matrix and vascular remodeling in mechanically ventilated elastin haploinsufficient newborn mice. Am J Physiol Lung Cell Mol Physiol 2014; 308:L464-78. [PMID: 25539853 DOI: 10.1152/ajplung.00278.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Elastin plays a pivotal role in lung development. We therefore queried if elastin haploinsufficient newborn mice (Eln(+/-)) would exhibit abnormal lung structure and function related to modified extracellular matrix (ECM) composition. Because mechanical ventilation (MV) has been linked to dysregulated elastic fiber formation in the newborn lung, we also asked if elastin haploinsufficiency would accentuate lung growth arrest seen after prolonged MV of neonatal mice. We studied 5-day-old wild-type (Eln(+/+)) and Eln(+/-) littermates at baseline and after MV with air for 8-24 h. Lungs of unventilated Eln(+/-) mice contained ∼50% less elastin and ∼100% more collagen-1 and lysyl oxidase compared with Eln(+/+) pups. Eln(+/-) lungs contained fewer capillaries than Eln(+/+) lungs, without discernible differences in alveolar structure. In response to MV, lung tropoelastin and elastase activity increased in Eln(+/+) neonates, whereas tropoelastin decreased and elastase activity was unchanged in Eln(+/-) mice. Fibrillin-1 protein increased in lungs of both groups during MV, more in Eln(+/-) than in Eln(+/+) pups. In both groups, MV caused capillary loss, with larger and fewer alveoli compared with unventilated controls. Respiratory system elastance, which was less in unventilated Eln(+/-) compared with Eln(+/+) mice, was similar in both groups after MV. These results suggest that elastin haploinsufficiency adversely impacts pulmonary angiogenesis and that MV dysregulates elastic fiber integrity, with further loss of lung capillaries, lung growth arrest, and impaired respiratory function in both Eln(+/+) and Eln(+/-) mice. Paucity of lung capillaries in Eln(+/-) newborns might help explain subsequent development of pulmonary hypertension previously reported in adult Eln(+/-) mice.
Collapse
Affiliation(s)
- Anne Hilgendorff
- Department of Pediatrics, Stanford University, Stanford, California; Comprehensive Pneumology Center, Ludwig-Maximilian University, Munich, Germany; and
| | - Kakoli Parai
- Department of Pediatrics, Stanford University, Stanford, California
| | - Robert Ertsey
- Department of Pediatrics, Stanford University, Stanford, California
| | - Edwin Navarro
- Department of Pediatrics, Stanford University, Stanford, California
| | - Noopur Jain
- Department of Pediatrics, Stanford University, Stanford, California
| | | | - Joanna Peterson
- Department of Pediatrics, Stanford University, Stanford, California
| | - Lucia Mokres
- Department of Pediatrics, Stanford University, Stanford, California
| | - Carlos Milla
- Department of Pediatrics, Stanford University, Stanford, California
| | - Stefanie Preuss
- Department of Pediatrics, Stanford University, Stanford, California
| | | | - Suleman Khan
- Department of Pediatrics, Stanford University, Stanford, California
| | - Juliet Masumi
- Department of Pediatrics, Stanford University, Stanford, California
| | | | - Sana Mujahid
- Department of Pediatrics, Stanford University, Stanford, California
| | - Barry Starcher
- Department of Biochemistry, University of Texas, Tyler, Texas
| | | | - Richard Bland
- Department of Pediatrics, Stanford University, Stanford, California;
| |
Collapse
|
46
|
Aberrant elastin remodeling in the lungs of O2-exposed newborn mice; primarily results from perturbed interaction between integrins and elastin. Cell Tissue Res 2014; 359:589-603. [DOI: 10.1007/s00441-014-2035-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/13/2014] [Indexed: 01/06/2023]
|
47
|
Collins JJP, Thébaud B. Lung mesenchymal stromal cells in development and disease: to serve and protect? Antioxid Redox Signal 2014; 21:1849-62. [PMID: 24350665 DOI: 10.1089/ars.2013.5781] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Bronchopulmonary dysplasia (BPD) is a disease of the developing lung that afflicts extreme preterm infants in the neonatal intensive care unit. Follow-up studies into adulthood show that BPD is not merely a problem of the neonatal period, as it also may predispose to early-onset emphysema and poor lung function in later life. RECENT ADVANCES The increasing promise of bone marrow- or umbilical cord-derived mesenchymal stromal cells (MSCs) to repair neonatal and adult lung diseases may for the first time offer the chance to make substantial strides in improving the outcome of extreme premature infants at risk of developing BPD. As more knowledge has been obtained on MSCs over the past decades, it has become clear that each organ has its own reservoir of endogenous MSCs, including the lung. CRITICAL ISSUES We have only barely scratched the surface on what resident lung MSCs exactly are and what their role and function in lung development may be. Moreover, what happens to these putative repair cells in BPD when alveolar development goes awry and why do their counterparts from the bone marrow and umbilical cord succeed in restoring normal alveolar development when they themselves do not? FUTURE DIRECTIONS Much work remains to be carried out to validate lung MSCs, but with the high potential of MSC-based treatment for BPD and other lung diseases, a thorough understanding of the endogenous lung MSC will be pivotal to get to the bottom of these diseases.
Collapse
Affiliation(s)
- Jennifer J P Collins
- 1 Regenerative Medicine Program, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, University of Ottawa , Ottawa, Canada
| | | |
Collapse
|
48
|
Berger J, Bhandari V. Animal models of bronchopulmonary dysplasia. The term mouse models. Am J Physiol Lung Cell Mol Physiol 2014; 307:L936-47. [PMID: 25305249 DOI: 10.1152/ajplung.00159.2014] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The etiology of bronchopulmonary dysplasia (BPD) is multifactorial, with genetics, ante- and postnatal sepsis, invasive mechanical ventilation, and exposure to hyperoxia being well described as contributing factors. Much of what is known about the pathogenesis of BPD is derived from animal models being exposed to the environmental factors noted above. This review will briefly cover the various mouse models of BPD, focusing mainly on the hyperoxia-induced lung injury models. We will also include hypoxia, hypoxia/hyperoxia, inflammation-induced, and transgenic models in room air. Attention to the stage of lung development at the timing of the initiation of the environmental insult and the duration of lung injury is critical to attempt to mimic the human disease pulmonary phenotype, both in the short term and in outcomes extending into childhood, adolescence, and adulthood. The various indexes of alveolar and vascular development as well as pulmonary function including pulmonary hypertension will be highlighted. The advantages (and limitations) of using such approaches will be discussed in the context of understanding the pathogenesis of and targeting therapeutic interventions to ameliorate human BPD.
Collapse
Affiliation(s)
- Jessica Berger
- Division of Perinatal Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Vineet Bhandari
- Division of Perinatal Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
49
|
Kneyber MCJ, Zhang H, Slutsky AS. Ventilator-induced lung injury. Similarity and differences between children and adults. Am J Respir Crit Care Med 2014; 190:258-65. [PMID: 25003705 DOI: 10.1164/rccm.201401-0168cp] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
It is well established that mechanical ventilation can injure the lung, producing an entity known as ventilator-induced lung injury (VILI). There are various forms of VILI, including volutrauma (i.e., injury caused by overdistending the lung), atelectrauma (injury due to repeated opening/closing of lung units), and biotrauma (release of mediators that can induce lung injury or aggravate pre-existing injury, potentially leading to multiple organ failure). Experimental data in the pediatric context are in accord with the importance of VILI, and appear to show age-related susceptibility to VILI, although a conclusive link between use of large Vts and mortality has not been demonstrated in this population. The relevance of VILI in the pediatric intensive care unit population is thus unclear. Given the physiological and biological differences in the respiratory systems of infants, children, and adults, it is difficult to directly extrapolate clinical practice from adults to children. This Critical Care Perspective analyzes the relevance of VILI to the pediatric population, and addresses why pediatric patients might be less susceptible than adults to VILI.
Collapse
Affiliation(s)
- Martin C J Kneyber
- 1 Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | |
Collapse
|
50
|
Hilgendorff A, Reiss I, Ehrhardt H, Eickelberg O, Alvira CM. Chronic lung disease in the preterm infant. Lessons learned from animal models. Am J Respir Cell Mol Biol 2014; 50:233-45. [PMID: 24024524 DOI: 10.1165/rcmb.2013-0014tr] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neonatal chronic lung disease, also known as bronchopulmonary dysplasia (BPD), is the most common complication of premature birth, affecting up to 30% of very low birth weight infants. Improved medical care has allowed for the survival of the most premature infants and has significantly changed the pathology of BPD from a disease marked by severe lung injury to the "new" form characterized by alveolar hypoplasia and impaired vascular development. However, increased patient survival has led to a paucity of pathologic specimens available from infants with BPD. This, combined with the lack of a system to model alveolarization in vitro, has resulted in a great need for animal models that mimic key features of the disease. To this end, a number of animal models have been created by exposing the immature lung to injuries induced by hyperoxia, mechanical stretch, and inflammation and most recently by the genetic modification of mice. These animal studies have 1) allowed insight into the mechanisms that determine alveolar growth, 2) delineated factors central to the pathogenesis of neonatal chronic lung disease, and 3) informed the development of new therapies. In this review, we summarize the key findings and limitations of the most common animal models of BPD and discuss how knowledge obtained from these studies has informed clinical care. Future studies should aim to provide a more complete understanding of the pathways that preserve and repair alveolar growth during injury, which might be translated into novel strategies to treat lung diseases in infants and adults.
Collapse
Affiliation(s)
- Anne Hilgendorff
- 1 Department of Perinatology Grosshadern, Ludwig-Maximilian-University, Munich, Germany
| | | | | | | | | |
Collapse
|