1
|
Khan IS, Molina C, Ren X, Auyeung VC, Cohen M, Tsukui T, Atakilit A, Sheppard D. Impaired myofibroblast proliferation is a central feature of pathologic post-natal alveolar simplification. eLife 2024; 13:RP94425. [PMID: 39660606 PMCID: PMC11634066 DOI: 10.7554/elife.94425] [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] [Indexed: 12/12/2024] Open
Abstract
Premature infants with bronchopulmonary dysplasia (BPD) have impaired alveolar gas exchange due to alveolar simplification and dysmorphic pulmonary vasculature. Advances in clinical care have improved survival for infants with BPD, but the overall incidence of BPD remains unchanged because we lack specific therapies to prevent this disease. Recent work has suggested a role for increased transforming growth factor-beta (TGFβ) signaling and myofibroblast populations in BPD pathogenesis, but the functional significance of each remains unclear. Here, we utilize multiple murine models of alveolar simplification and comparative single-cell RNA sequencing to identify shared mechanisms that could contribute to BPD pathogenesis. Single-cell RNA sequencing reveals a profound loss of myofibroblasts in two models of BPD and identifies gene expression signatures of increased TGFβ signaling, cell cycle arrest, and impaired proliferation in myofibroblasts. Using pharmacologic and genetic approaches, we find no evidence that increased TGFβ signaling in the lung mesenchyme contributes to alveolar simplification. In contrast, this is likely a failed compensatory response, since none of our approaches to inhibit TGFβ signaling protect mice from alveolar simplification due to hyperoxia while several make simplification worse. In contrast, we find that impaired myofibroblast proliferation is a central feature in several murine models of BPD, and we show that inhibiting myofibroblast proliferation is sufficient to cause pathologic alveolar simplification. Our results underscore the importance of impaired myofibroblast proliferation as a central feature of alveolar simplification and suggest that efforts to reverse this process could have therapeutic value in BPD.
Collapse
Affiliation(s)
- Imran S Khan
- Division of Neonatology, Department of Pediatrics, UCSFSan FranciscoUnited States
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
| | - Christopher Molina
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Xin Ren
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Vincent C Auyeung
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
| | - Max Cohen
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Tatsuya Tsukui
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Amha Atakilit
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Dean Sheppard
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| |
Collapse
|
2
|
Zhang J, Du W, Zhang Z, Li T, Li X, Xi S. Research progress of microvascular development in bronchopulmonary dysplasia. Pediatr Investig 2024; 8:299-312. [PMID: 39720284 PMCID: PMC11664543 DOI: 10.1002/ped4.12441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 06/06/2024] [Indexed: 12/26/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that arises during the neonatal period, and its underlying mechanisms are still not fully understood. The disorder of microvascular development plays a significant role in the development of BPD. This article presents a comprehensive review of the advancements made in understanding the mechanisms and treatment approaches related to microvascular development in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of PediatricsTaihe HospitalHubei University of MedicineShiyanChina
| | - Weiwei Du
- Department of PediatricsThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxiChina
| | - Zongli Zhang
- Department of PediatricsTaihe HospitalHubei University of MedicineShiyanChina
- Institute of Pediatric DiseaseTaihe HospitalHubei University of MedicineShiyanChina
| | - Tao Li
- Department of PediatricsTaihe HospitalHubei University of MedicineShiyanChina
| | - Xingchao Li
- Department of PediatricsTaihe HospitalHubei University of MedicineShiyanChina
- Institute of Pediatric DiseaseTaihe HospitalHubei University of MedicineShiyanChina
| | - Shibing Xi
- Department of PediatricsTaihe HospitalHubei University of MedicineShiyanChina
- Institute of Pediatric DiseaseTaihe HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
3
|
Behnke J, Goetz MJ, Holzfurtner L, Korte P, Weiss A, Shahzad T, Wilhelm J, Schermuly RT, Rivetti S, Bellusci S, Ehrhardt H. Senescence of lung mesenchymal stem cells of preterm infants by cyclic stretch and hyperoxia via p21. Am J Physiol Lung Cell Mol Physiol 2024; 327:L694-L711. [PMID: 39316679 PMCID: PMC11563592 DOI: 10.1152/ajplung.00355.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/19/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
Phenotype distortion of lung resident mesenchymal stem cells (MSC) in preterm infants is a hallmark event in the pathogenesis of bronchopulmonary dysplasia (BPD). Here, we evaluated the impact of cyclic mechanical stretch (CMS) and hyperoxia (HOX). The negative action of HOX on proliferation and cell death was more pronounced at 80% than at 40%. Although the impact of CMS alone was modest, CMS plus HOX displayed the strongest effect sizes. Exposure to CMS and/or HOX induced the downregulation of PDGFRα, and cellular senescence preceded by p21 accumulation. p21 interference interfered with cellular senescence and resulted in aggravated cell death, arguing for a prosurvival mechanism. HOX 40% and limited exposure to HOX 80% prevailed in a reversible phenotype with reuptake of proliferation, while prolonged exposure to HOX 80% resulted in definite MSC growth arrest. Our mechanistic data explain how HOX and CMS induce the effects on MSC phenotype disruption. The results are congruent with the clinical observation that preterm infants requiring supplemental oxygen plus mechanical ventilation are at particular risk for BPD. Although inhibiting p21 is not a feasible approach, limiting the duration and magnitude of the exposures is promising.NEW & NOTEWORTHY Rarefication of lung mesenchymal stem cells (MSC) due to exposure to cyclic mechanical stretch (CMS) during mechanical ventilation with oxygen-rich gas is a hallmark of bronchopulmonary dysplasia in preterm infants, but the pathomechanistic understanding is incomplete. Our studies identify a common signaling mechanism mediated by p21 accumulation, leading to cellular senescence and cell death, most pronounced during the combined exposure with in principle reversible phenotype change depending on strength and duration of exposures.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Maurizio J Goetz
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Lena Holzfurtner
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Pauline Korte
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Astrid Weiss
- Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jochen Wilhelm
- Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Ralph T Schermuly
- Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Stefano Rivetti
- Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Saverio Bellusci
- Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University Giessen and Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Division of Neonatology and Pediatric Intensive Care Medicine, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
4
|
Khan IS, Molina C, Ren X, Auyeung VC, Cohen M, Tsukui T, Atakilit A, Sheppard D. Impaired Myofibroblast Proliferation is a Central Feature of Pathologic Post-Natal Alveolar Simplification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572766. [PMID: 38187712 PMCID: PMC10769348 DOI: 10.1101/2023.12.21.572766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Premature infants with bronchopulmonary dysplasia (BPD) have impaired alveolar gas exchange due to alveolar simplification and dysmorphic pulmonary vasculature. Advances in clinical care have improved survival for infants with BPD, but the overall incidence of BPD remains unchanged because we lack specific therapies to prevent this disease. Recent work has suggested a role for increased transforming growth factor-beta (TGFβ) signaling and myofibroblast populations in BPD pathogenesis, but the functional significance of each remains unclear. Here, we utilize multiple murine models of alveolar simplification and comparative single-cell RNA sequencing to identify shared mechanisms that could contribute to BPD pathogenesis. Single-cell RNA sequencing reveals a profound loss of myofibroblasts in two models of BPD and identifies gene expression signatures of increased TGFβ signaling, cell cycle arrest, and impaired proliferation in myofibroblasts. Using pharmacologic and genetic approaches, we find no evidence that increased TGFβ signaling in the lung mesenchyme contributes to alveolar simplification. In contrast, this is likely a failed compensatory response, since none of our approaches to inhibit TGFb signaling protect mice from alveolar simplification due to hyperoxia while several make simplification worse. In contrast, we find that impaired myofibroblast proliferation is a central feature in several murine models of BPD, and we show that inhibiting myofibroblast proliferation is sufficient to cause pathologic alveolar simplification. Our results underscore the importance of impaired myofibroblast proliferation as a central feature of alveolar simplification and suggest that efforts to reverse this process could have therapeutic value in BPD.
Collapse
Affiliation(s)
- Imran S. Khan
- Division of Neonatology, Department of Pediatrics, UCSF
- Cardiovascular Research Institute, UCSF
| | - Christopher Molina
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Xin Ren
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Vincent C. Auyeung
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Max Cohen
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Tatsuya Tsukui
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Amha Atakilit
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Dean Sheppard
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| |
Collapse
|
5
|
Kumar N, Bidkhori HR, Yawno T, Lim R, Inocencio IM. Therapeutic potential of extracellular vesicles derived from human amniotic epithelial cells for perinatal cerebral and pulmonary injury. Stem Cells Transl Med 2024; 13:711-723. [PMID: 38895873 PMCID: PMC11328935 DOI: 10.1093/stcltm/szae037] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.
Collapse
Affiliation(s)
- Naveen Kumar
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Hamid Reza Bidkhori
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton 3168, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Ishmael Miguel Inocencio
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| |
Collapse
|
6
|
Shahzad T, Dong Y, Behnke NK, Brandner J, Hilgendorff A, Chao CM, Behnke J, Bellusci S, Ehrhardt H. Anti-CCL2 therapy reduces oxygen toxicity to the immature lung. Cell Death Discov 2024; 10:311. [PMID: 38961074 PMCID: PMC11222519 DOI: 10.1038/s41420-024-02073-5] [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: 05/11/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
Oxygen toxicity constitutes a key contributor to bronchopulmonary dysplasia (BPD). Critical step in the pathogenesis of BPD is the inflammatory response in the immature lung with the release of pro-inflammatory cytokines and the influx of innate immune cells. Identification of efficient therapies to alleviate the inflammatory response remains an unmet research priority. First, we studied macrophage and neutrophil profiles in tracheal aspirates of n = 103 preterm infants <29 weeks´ gestation requiring mechanical ventilation. While no differences were present at birth, a higher fraction of macrophages, the predominance of the CD14+CD16+ subtype on day 5 of life was associated with moderate/severe BPD. Newborn CCL-2-/- mice insufficient in pulmonary macrophage recruitment had a reduced influx of neutrophils, lower apoptosis induction in the pulmonary tissue and better-preserved lung morphometry with higher counts of type II cells, mesenchymal stem cells and vascular endothelial cells when exposed to hyperoxia for 7 days. To study the benefit of a targeted approach to prevent the pulmonary influx of macrophages, wildtype mice were repeatedly treated with CCL-2 blocking antibodies while exposed to hyperoxia for 7 days. Congruent with the results in CCL-2-/- animals, the therapeutic intervention reduced the pulmonary inflammatory response, attenuated cell death in the lung tissue and better-preserved lung morphometry. Overall, our preclinical and clinical datasets document the predominant role of macrophage recruitment to the pathogenesis of BPD and establish the abrogation of CCL-2 function as novel approach to protect the immature lung from hyperoxic injury.
Collapse
Affiliation(s)
- Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), German Lung Research Center (DZL), Feulgenstrasse 12, Giessen, Germany
| | - Ying Dong
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), German Lung Research Center (DZL), Feulgenstrasse 12, Giessen, Germany
| | - Nina K Behnke
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Großhadern, Marchioninistrasse 15, Munich, Germany
| | - Julia Brandner
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Großhadern, Marchioninistrasse 15, Munich, Germany
| | - Anne Hilgendorff
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Großhadern, Marchioninistrasse 15, Munich, Germany
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Cho-Ming Chao
- Department of Pediatrics, Helios University Medical Center, Witten/Herdecke University, Heusnerstrasse 40, 42283, Wuppertal, Germany
| | - Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), German Lung Research Center (DZL), Feulgenstrasse 12, Giessen, Germany
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Germany German Lung Research Center (DZL), Aulweg 130, Giessen, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center (UGMLC), German Lung Research Center (DZL), Feulgenstrasse 12, Giessen, Germany.
- Division of Neonatology and Pediatric Intensive Care Medicine, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.
| |
Collapse
|
7
|
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common complication of premature birth, imposing a significant and potentially life-long burden on patients and their families. Despite advances in our understanding of the mechanisms that contribute to patterns of lung injury and dysfunctional repair, current therapeutic strategies remain non-specific with limited success. Contemporary definitions of BPD continue to rely on clinician prescribed respiratory support requirements at specific time points. While these criteria may be helpful in broadly identifying infants at higher risk of adverse outcomes, they do not offer any precise information regarding the degree to which each compartment of the lung is affected. In this review we will outline the different pulmonary phenotypes of BPD and discuss important features in the pathogenesis, clinical presentation, and management of these frequently overlapping scenarios.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Division of Neonatology, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| | - Vineet Bhandari
- Division of Neonatology, The Children's Regional Hospital at Cooper/Cooper Medical School of Rowan University, Camden, NJ 08103, USA.
| |
Collapse
|
8
|
Yie TA, Loomis CA, Nowatzky J, Khodadadi-Jamayran A, Lin Z, Cammer M, Barnett C, Mezzano V, Alu M, Novick JA, Munger JS, Kugler MC. Hedgehog and Platelet-derived Growth Factor Signaling Intersect during Postnatal Lung Development. Am J Respir Cell Mol Biol 2023; 68:523-536. [PMID: 36693140 PMCID: PMC10174164 DOI: 10.1165/rcmb.2022-0269oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 01/24/2023] [Indexed: 01/26/2023] Open
Abstract
Normal lung development critically depends on HH (Hedgehog) and PDGF (platelet-derived growth factor) signaling, which coordinate mesenchymal differentiation and proliferation. PDGF signaling is required for postnatal alveolar septum formation by myofibroblasts. Recently, we demonstrated a requirement for HH in postnatal lung development involving alveolar myofibroblast differentiation. Given shared features of HH signaling and PDGF signaling and their impact on this key cell type, we sought to clarify their relationship during murine postnatal lung development. Timed experiments revealed that HH inhibition phenocopies the key lung myofibroblast phenotypes of Pdgfa (platelet-derived growth factor subunit A) and Pdgfra (platelet-derived growth factor receptor alpha) knockouts during secondary alveolar septation. Using a dual signaling reporter, Gli1lZ;PdgfraEGFP, we show that HH and PDGF pathway intermediates are concurrently expressed during alveolar septal myofibroblast accumulation, suggesting pathway convergence in the generation of lung myofibroblasts. Consistent with this hypothesis, HH inhibition reduces Pdgfra expression and diminishes the number of Pdgfra-positive and Pdgfra-lineage cells in postnatal lungs. Bulk RNA sequencing data of Pdgfra-expressing cells from Postnatal Day 8 (P8) lungs show that HH inhibition alters the expression not only of well-established HH targets but also of several putative PDGF target genes. This, together with the presence of Gli-binding sites in PDGF target genes, suggests HH input into PDGF signaling. We identified these HH/PDGF targets in several postnatal lung mesenchymal cell populations, including myofibroblasts, using single-cell transcriptomic analysis. Collectively, our data indicate that HH signaling and PDGF signaling intersect to support myofibroblast/fibroblast function during secondary alveolar septum formation. Moreover, they provide a molecular foundation relevant to perinatal lung diseases associated with impaired alveolarization.
Collapse
Affiliation(s)
- Ting-An Yie
- Division of Pulmonary, Critical Care and Sleep Medicine and
| | | | - Johannes Nowatzky
- Division of Rheumatology, Department of Medicine
- Department of Pathology
| | | | | | | | - Clea Barnett
- Division of Pulmonary, Critical Care and Sleep Medicine and
| | | | | | | | - John S. Munger
- Division of Pulmonary, Critical Care and Sleep Medicine and
- Department of Cell Biology, School of Medicine and Langone Medical Center, New York University, New York, New York
| | | |
Collapse
|
9
|
Damianos A, Kalinichenko VV. Hedgehog and Platelet-derived Growth Factor Collaborate to Guide Fibroblasts during Alveolarization. Am J Respir Cell Mol Biol 2023; 68:472-474. [PMID: 36796088 PMCID: PMC10174160 DOI: 10.1165/rcmb.2023-0031ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Affiliation(s)
- Andreas Damianos
- Division of Neonatology and Pulmonary Biology Perinatal Institute, Department of Pediatrics Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology Perinatal Institute, Department of Pediatrics and Center of Lung Regenerative Medicine Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, Ohio
| |
Collapse
|
10
|
Postnatal serum IGF-1 levels associate with brain volumes at term in extremely preterm infants. Pediatr Res 2023; 93:666-674. [PMID: 35681088 PMCID: PMC9988684 DOI: 10.1038/s41390-022-02134-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Growth factors important for normal brain development are low in preterm infants. This study investigated the link between growth factors and preterm brain volumes at term. MATERIAL/METHODS Infants born <28 weeks gestational age (GA) were included. Endogenous levels of insulin-like growth factor (IGF)-1, brain-derived growth factor, vascular endothelial growth factor, and platelet-derived growth factor (expressed as area under the curve [AUC] for serum samples from postnatal days 1, 7, 14, and 28) were utilized in a multivariable linear regression model. Brain volumes were determined by magnetic resonance imaging (MRI) at term equivalent age. RESULTS In total, 49 infants (median [range] GA 25.4 [22.9-27.9] weeks) were included following MRI segmentation quality assessment and AUC calculation. IGF-1 levels were independently positively associated with the total brain (p < 0.001, β = 0.90), white matter (p = 0.007, β = 0.33), cortical gray matter (p = 0.002, β = 0.43), deep gray matter (p = 0.008, β = 0.05), and cerebellar (p = 0.006, β = 0.08) volume adjusted for GA at birth and postmenstrual age at MRI. No associations were seen for other growth factors. CONCLUSIONS Endogenous exposure to IGF-1 during the first 4 weeks of life was associated with total and regional brain volumes at term. Optimizing levels of IGF-1 might improve brain growth in extremely preterm infants. IMPACT High serum levels of insulin-like growth factor (IGF)-1 during the first month of life were independently associated with increased total brain volume, white matter, gray matter, and cerebellar volume at term equivalent age in extremely preterm infants. IGF-1 is a critical regulator of neurodevelopment and postnatal levels are low in preterm infants. The effects of IGF-1 levels on brain development in extremely preterm infants are not fully understood. Optimizing levels of IGF-1 may benefit early brain growth in extremely preterm infants. The effects of systemically administered IGF-1/IGFBP3 in extremely preterm infants are now being investigated in a randomized controlled trial (Clinicaltrials.gov: NCT03253263).
Collapse
|
11
|
Heydarian M, Oak P, Zhang X, Kamgari N, Kindt A, Koschlig M, Pritzke T, Gonzalez-Rodriguez E, Förster K, Morty RE, Häfner F, Hübener C, Flemmer AW, Yildirim AO, Sudheendra D, Tian X, Petrera A, Kirsten H, Ahnert P, Morrell N, Desai TJ, Sucre J, Spiekerkoetter E, Hilgendorff A. Relationship between impaired BMP signalling and clinical risk factors at early-stage vascular injury in the preterm infant. Thorax 2022; 77:1176-1186. [PMID: 35580897 PMCID: PMC9685723 DOI: 10.1136/thoraxjnl-2021-218083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/11/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Prajakta Oak
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Zhang
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nona Kamgari
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alida Kindt
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Markus Koschlig
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Tina Pritzke
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Erika Gonzalez-Rodriguez
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kai Förster
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, University Hospital Heidelberg, Translational Lung Research Center campus of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Friederike Häfner
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Andreas W Flemmer
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Ali Oender Yildirim
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Deepti Sudheendra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Xuefei Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Nick Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tushar J Desai
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Jennifer Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Anne Hilgendorff
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU), Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| |
Collapse
|
12
|
Heydarian M, Schulz C, Stoeger T, Hilgendorff A. Association of immune cell recruitment and BPD development. Mol Cell Pediatr 2022; 9:16. [PMID: 35917002 PMCID: PMC9346035 DOI: 10.1186/s40348-022-00148-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
In the neonatal lung, exposure to both prenatal and early postnatal risk factors converge into the development of injury and ultimately chronic disease, also known as bronchopulmonary dysplasia (BPD). The focus of many studies has been the characteristic inflammatory responses provoked by these exposures. Here, we review the relationship between immaturity and prenatal conditions, as well as postnatal exposure to mechanical ventilation and oxygen toxicity, with the imbalance of pro- and anti-inflammatory regulatory networks. In these conditions, cytokine release, protease activity, and sustained presence of innate immune cells in the lung result in pathologic processes contributing to lung injury. We highlight the recruitment and function of myeloid innate immune cells, in particular, neutrophils and monocyte/macrophages in the BPD lung in human patients and animal models. We also discuss dissimilarities between the infant and adult immune system as a basis for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christian Schulz
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Tobias Stoeger
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany. .,Center for Comprehensive Developmental Care (CDeCLMU) at the interdisciplinary Social Pediatric Center, (iSPZ), University Hospital Ludwig-Maximilian University, Munich, Germany.
| |
Collapse
|
13
|
TRAIL protects the immature lung from hyperoxic injury. Cell Death Dis 2022; 13:614. [PMID: 35840556 PMCID: PMC9287454 DOI: 10.1038/s41419-022-05072-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
The hyperoxia-induced pro-inflammatory response and tissue damage constitute pivotal steps leading to bronchopulmonary dysplasia (BPD) in the immature lung. The pro-inflammatory cytokines are considered attractive candidates for a directed intervention but the complex interplay between inflammatory and developmental signaling pathways requires a comprehensive evaluation before introduction into clinical trials as studied here for the death inducing ligand TRAIL. At birth and during prolonged exposure to oxygen and mechanical ventilation, levels of TRAIL were lower in tracheal aspirates of preterm infants <29 weeks of gestation which developed moderate/severe BPD. These findings were reproduced in the newborn mouse model of hyperoxic injury. The loss of TRAIL was associated with increased inflammation, apoptosis induction and more pronounced lung structural simplification after hyperoxia exposure for 7 days while activation of NFκB signaling during exposure to hyperoxia was abrogated. Pretreatment with recombinant TRAIL rescued the developmental distortions in precision cut lung slices of both wildtype and TRAIL-/- mice exposed to hyperoxia. Of importance, TRAIL preserved alveolar type II cells, mesenchymal progenitor cells and vascular endothelial cells. In the situation of TRAIL depletion, our data ascribe oxygen toxicity a more injurious impact on structural lung development. These data are not surprising taking into account the diverse functions of TRAIL and its stimulatory effects on NFκB signaling as central driver of survival and development. TRAIL exerts a protective role in the immature lung as observed for the death inducing ligand TNF-α before.
Collapse
|
14
|
Holzfurtner L, Shahzad T, Dong Y, Rekers L, Selting A, Staude B, Lauer T, Schmidt A, Rivetti S, Zimmer KP, Behnke J, Bellusci S, Ehrhardt H. When inflammation meets lung development-an update on the pathogenesis of bronchopulmonary dysplasia. Mol Cell Pediatr 2022; 9:7. [PMID: 35445327 PMCID: PMC9021337 DOI: 10.1186/s40348-022-00137-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/14/2022] [Indexed: 12/28/2022] Open
Abstract
Even more than 50 years after its initial description, bronchopulmonary dysplasia (BPD) remains one of the most important and lifelong sequelae following premature birth. Tremendous efforts have been undertaken since then to reduce this ever-increasing disease burden but a therapeutic breakthrough preventing BPD is still not in sight. The inflammatory response provoked in the immature lung is a key driver of distorted lung development and impacts the formation of alveolar, mesenchymal, and vascular structures during a particularly vulnerable time-period. During the last 5 years, new scientific insights have led to an improved pathomechanistic understanding of BPD origins and disease drivers. Within the framework of current scientific progress, concepts involving disruption of the balance of key inflammatory and lung growth promoting pathways by various stimuli, take center stage. Still today, the number of efficient therapeutics available to prevent BPD is limited to a few, well-established pharmacological interventions including postnatal corticosteroids, early caffeine administration, and vitamin A. Recent advances in the clinical care of infants in the neonatal intensive care unit (NICU) have led to improvements in survival without a consistent reduction in the incidence of BPD. Our update provides latest insights from both preclinical models and clinical cohort studies and describes novel approaches to prevent BPD.
Collapse
Affiliation(s)
- Lena Holzfurtner
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Ying Dong
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Lisa Rekers
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Ariane Selting
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Birte Staude
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Tina Lauer
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Annesuse Schmidt
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Stefano Rivetti
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392, Giessen, Germany
| | - Klaus-Peter Zimmer
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Judith Behnke
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392, Giessen, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Justus-Liebig-University, Feulgenstrasse 12, 35392, Giessen, Germany.
| |
Collapse
|
15
|
Yallapragada SG, Savani RC, Goss KN. Cardiovascular impact and sequelae of bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:3453-3463. [PMID: 33756045 DOI: 10.1002/ppul.25370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
The development, growth, and function of the cardiac, pulmonary, and vascular systems are closely intertwined during both fetal and postnatal life. In utero, placental, environmental, and genetic insults may contribute to abnormal pulmonary alveolarization and vascularization that increase susceptibility to the development of bronchopulmonary dysplasia (BPD) in preterm infants. However, the shared milieu of stressors may also contribute to abnormal cardiac or vascular development in the fetus and neonate, leading to the potential for cardiovascular dysfunction. Further, cardiac or pulmonary maladaptation can potentiate dysfunction in the other organ, amplify the risk for BPD in the neonate, and increase the trajectory for overall neonatal morbidity. Beyond infancy, there is an increased risk for systemic and pulmonary vascular disease including hypertension, as well as potential cardiac dysfunction, particularly within the right ventricle. This review will focus on the cardiovascular antecedents of BPD in the fetus, cardiovascular consequences of preterm birth in the neonate including associations with BPD, and cardiovascular impact of prematurity and BPD throughout the lifespan.
Collapse
Affiliation(s)
- Sushmita G Yallapragada
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kara N Goss
- Division of Pulmonary and Critical Care, Departments of Medicine and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
Oxygen Toxicity to the Immature Lung-Part I: Pathomechanistic Understanding and Preclinical Perspectives. Int J Mol Sci 2021; 22:ijms222011006. [PMID: 34681665 PMCID: PMC8540649 DOI: 10.3390/ijms222011006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/13/2023] Open
Abstract
In utero, the fetus and its lungs develop in a hypoxic environment, where HIF-1α and VEGFA signaling constitute major determinants of further development. Disruption of this homeostasis after preterm delivery and extrauterine exposure to high fractions of oxygen are among the key events leading to bronchopulmonary dysplasia (BPD). Reactive oxygen species (ROS) production constitutes the initial driver of pulmonary inflammation and cell death, altered gene expression, and vasoconstriction, leading to the distortion of further lung development. From preclinical studies mainly performed on rodents over the past two decades, the deleterious effects of oxygen toxicity and the injurious insults and downstream cascades arising from ROS production are well recognized. This article provides a concise overview of disease drivers and different therapeutic approaches that have been successfully tested within experimental models. Despite current studies, clinical researchers are still faced with an unmet clinical need, and many of these strategies have not proven to be equally effective in clinical trials. In light of this challenge, adapting experimental models to the complexity of the clinical situation and pursuing new directions constitute appropriate actions to overcome this dilemma. Our review intends to stimulate research activities towards the understanding of an important issue of immature lung injury.
Collapse
|
17
|
Behnke J, Dippel CM, Choi Y, Rekers L, Schmidt A, Lauer T, Dong Y, Behnke J, Zimmer KP, Bellusci S, Ehrhardt H. Oxygen Toxicity to the Immature Lung-Part II: The Unmet Clinical Need for Causal Therapy. Int J Mol Sci 2021; 22:10694. [PMID: 34639034 PMCID: PMC8508961 DOI: 10.3390/ijms221910694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen toxicity continues to be one of the inevitable injuries to the immature lung. Reactive oxygen species (ROS) production is the initial step leading to lung injury and, subsequently, the development of bronchopulmonary dysplasia (BPD). Today, BPD remains the most important disease burden following preterm delivery and results in life-long restrictions in lung function and further important health sequelae. Despite the tremendous progress in the pathomechanistic understanding derived from preclinical models, the clinical needs for preventive or curative therapies remain unmet. This review summarizes the clinical progress on guiding oxygen delivery to the preterm infant and elaborates future directions of research that need to take into account both hyperoxia and hypoxia as ROS sources and BPD drivers. Many strategies have been tested within clinical trials based on the mechanistic understanding of ROS actions, but most have failed to prove efficacy. The majority of these studies were tested in an era before the latest modes of non-invasive respiratory support and surfactant application were introduced or were not appropriately powered. A comprehensive re-evaluation of enzymatic, antioxidant, and anti-inflammatory therapies to prevent ROS injury is therefore indispensable. Strategies will only succeed if they are applied in a timely and vigorous manner and with the appropriate outcome measures.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Constanze M. Dippel
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Yesi Choi
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Lisa Rekers
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Annesuse Schmidt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Tina Lauer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Ying Dong
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Jonas Behnke
- Department of Internal Medicine V, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Klinikstrasse 33, 35392 Giessen, Germany;
| | - Klaus-Peter Zimmer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany;
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Research Center (DZL), Feulgenstrasse 12, 35392 Giessen, Germany; (J.B.); (C.M.D.); (Y.C.); (L.R.); (A.S.); (T.L.); (Y.D.); (K.-P.Z.)
| |
Collapse
|
18
|
Gokey JJ, Snowball J, Green J, Waltamath M, Spinney JJ, Black KE, Hariri LP, Xu Y, Perl AK. Pretreatment of aged mice with retinoic acid supports alveolar regeneration via upregulation of reciprocal PDGFA signalling. Thorax 2021; 76:456-467. [PMID: 33479039 PMCID: PMC8070612 DOI: 10.1136/thoraxjnl-2020-214986] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Idiopathic pulmonary fibrosis (IPF) primarily affects the aged population and is characterised by failure of alveolar regeneration, leading to loss of alveolar type 1 (AT1) cells. Aged mouse models of lung repair have demonstrated that regeneration fails with increased age. Mouse and rat lung repair models have shown retinoic acid (RA) treatment can restore alveolar regeneration. Herein, we seek to determine the signalling mechanisms that become activated on RA treatment prior to injury, which support alveolar differentiation. DESIGN Partial pneumonectomy lung injury model and next-generation sequencing of sorted cell populations were used to uncover molecular targets regulating alveolar repair. In vitro organoids generated from epithelial cells of mouse or patient with IPF co-cultured with young, aged or RA-pretreated murine fibroblasts were used to test potential targets. MAIN OUTCOME MEASUREMENTS Known alveolar epithelial cell differentiation markers, including HOPX and AGER for AT1 cells, were used to assess outcome of treatments. RESULTS Gene expression analysis of sorted fibroblasts and epithelial cells isolated from lungs of young, aged and RA-pretreated aged mice predicted increased platelet-derived growth factor subunit A (PDGFA) signalling that coincided with regeneration and alveolar epithelial differentiation. Addition of PDGFA induced AT1 and AT2 differentiation in both mouse and human IPF lung organoids generated with aged fibroblasts, and PDGFA monoclonal antibody blocked AT1 cell differentiation in organoids generated with young murine fibroblasts. CONCLUSIONS Our data support the concept that RA indirectly induces reciprocal PDGFA signalling, which activates regenerative fibroblasts that support alveolar epithelial cell differentiation and repair, providing a potential therapeutic strategy to influence the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jason J Gokey
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - John Snowball
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jenna Green
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Marion Waltamath
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jillian J Spinney
- Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Katharine E Black
- Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Lida P Hariri
- Division of Pulmonary and Critical Care Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yan Xu
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anne Karina Perl
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
19
|
Behnke J, Windhorst A, Oehmke F, Berthold LD, Zimmer K, Waitz M, Ehrhardt H. Preeclampsia was a risk factor for pulmonary interstitial emphysema in preterm infants born ≤32 weeks of gestational age. Acta Paediatr 2021; 110:134-140. [PMID: 32369221 DOI: 10.1111/apa.15338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
Abstract
AIM This study determined the prenatal and postnatal risk factors for pulmonary interstitial emphysema (PIE) in preterm infants born at up to 32 weeks of gestational age (GA) and their contribution to severe complications. METHODS We studied 179 preterm infants, who had undergone chest X-rays during the first five days of life at Justus Liebig University Giessen, Germany, between 2016 and 2017. Of these, 33 were retrospectively classified as PIE and 146 as non-PIE. The PIE cases were also matched with 33 non-PIE cases by GA and gender. Risk factors were identified by univariate analyses and multivariable logistic regression. RESULTS Previously known risk factors for pulmonary interstitial emphysema were confirmed, including GA and birthweight and the associations with adverse outcomes like intraventricular haemorrhage and mortality. We identified preeclampsia and haemolysis, elevated liver enzymes and low platelet count (HELLP) syndrome as additional risk factors for PIE (P = .027), and lung impairment was associated with respiratory distress syndrome (P = .001), higher maximum inspired oxygen (P = .014) and needing surfactant (P = .006). CONCLUSION Preeclampsia and HELLP syndrome were identified as possible additional risk factors for PIE in preterm infants. These conditions should be included in future studies, to identify preterm infants at risk of PIE straight after birth.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics & Neonatology Justus Liebig University and Universities of Giessen and Marburg Lung Center Giessen Germany
- German Center for Lung Research Giessen Germany
| | - Anita Windhorst
- Department of Medical Statistics Justus Liebig University of Giessen Giessen Germany
| | - Frank Oehmke
- Department of Gynecology and Obstetrics Justus Liebig University of Giessen Germany
| | - Lars D. Berthold
- Department of Pediatric Radiology Institute for Diagnostic and Interventional Radiology Justus Liebig University of Giessen Giessen Germany
| | - Klaus‐Peter Zimmer
- Department of General Pediatrics & Neonatology Justus Liebig University and Universities of Giessen and Marburg Lung Center Giessen Germany
| | - Markus Waitz
- Department of General Pediatrics & Neonatology Justus Liebig University and Universities of Giessen and Marburg Lung Center Giessen Germany
| | - Harald Ehrhardt
- Department of General Pediatrics & Neonatology Justus Liebig University and Universities of Giessen and Marburg Lung Center Giessen Germany
- German Center for Lung Research Giessen Germany
| |
Collapse
|
20
|
Chao CM, Chong L, Chu X, Shrestha A, Behnke J, Ehrhardt H, Zhang J, Chen C, Bellusci S. Targeting Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension (BPD-PH): Potential Role of the FGF Signaling Pathway in the Development of the Pulmonary Vascular System. Cells 2020; 9:cells9081875. [PMID: 32796770 PMCID: PMC7464452 DOI: 10.3390/cells9081875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
More than 50 years after the first description of Bronchopulmonary dysplasia (BPD) by Northway, this chronic lung disease affecting many preterm infants is still poorly understood. Additonally, approximately 40% of preterm infants suffering from severe BPD also suffer from Bronchopulmonary dysplasia-associated pulmonary hypertension (BPD-PH), leading to a significant increase in total morbidity and mortality. Until today, there is no curative therapy for both BPD and BPD-PH available. It has become increasingly evident that growth factors are playing a central role in normal and pathologic development of the pulmonary vasculature. Thus, this review aims to summarize the recent evidence in our understanding of BPD-PH from a basic scientific point of view, focusing on the potential role of Fibroblast Growth Factor (FGF)/FGF10 signaling pathway contributing to disease development, progression and resolution.
Collapse
Affiliation(s)
- Cho-Ming Chao
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
- Correspondence: (C.-M.C.); (S.B.)
| | - Lei Chong
- Institute of Pediatrics, National Key Clinical Specialty of Pediatric Respiratory Medicine, Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China;
| | - Xuran Chu
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Amit Shrestha
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
| | - Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
| | - Jinsan Zhang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- International Collaborative Center on Growth Factor Research, Life Science Institute, Wenzhou University, Wenzhou 325035, China
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
| | - Saverio Bellusci
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- Correspondence: (C.-M.C.); (S.B.)
| |
Collapse
|
21
|
Ruiz-Camp J, Quantius J, Lignelli E, Arndt PF, Palumbo F, Nardiello C, Surate Solaligue DE, Sakkas E, Mižíková I, Rodríguez-Castillo JA, Vadász I, Richardson WD, Ahlbrecht K, Herold S, Seeger W, Morty RE. Targeting miR-34a/ Pdgfra interactions partially corrects alveologenesis in experimental bronchopulmonary dysplasia. EMBO Mol Med 2020; 11:emmm.201809448. [PMID: 30770339 PMCID: PMC6404112 DOI: 10.15252/emmm.201809448] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication of preterm birth characterized by arrested lung alveolarization, which generates lungs that are incompetent for effective gas exchange. We report here deregulated expression of miR‐34a in a hyperoxia‐based mouse model of BPD, where miR‐34a expression was markedly increased in platelet‐derived growth factor receptor (PDGFR)α‐expressing myofibroblasts, a cell type critical for proper lung alveolarization. Global deletion of miR‐34a; and inducible, conditional deletion of miR‐34a in PDGFRα+ cells afforded partial protection to the developing lung against hyperoxia‐induced perturbations to lung architecture. Pdgfra mRNA was identified as the relevant miR‐34a target, and using a target site blocker in vivo, the miR‐34a/Pdgfra interaction was validated as a causal actor in arrested lung development. An antimiR directed against miR‐34a partially restored PDGFRα+ myofibroblast abundance and improved lung alveolarization in newborn mice in an experimental BPD model. We present here the first identification of a pathology‐relevant microRNA/mRNA target interaction in aberrant lung alveolarization and highlight the translational potential of targeting the miR‐34a/Pdgfra interaction to manage arrested lung development associated with preterm birth.
Collapse
Affiliation(s)
- Jordi Ruiz-Camp
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - Jennifer Quantius
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ettore Lignelli
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - Philipp F Arndt
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - 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), 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
| | - 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), 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
| | - Elpidoforos Sakkas
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - 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), 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
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - 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
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), 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
| | - 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
| | - 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), 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
| | - 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), 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
| |
Collapse
|
22
|
Gouveia L, Kraut S, Hadzic S, Vazquéz-Liébanas E, Kojonazarov B, Wu CY, Veith C, He L, Mermelekas G, Schermuly RT, Weissmann N, Betsholtz C, Andrae J. Lung developmental arrest caused by PDGF-A deletion: consequences for the adult mouse lung. Am J Physiol Lung Cell Mol Physiol 2020; 318:L831-L843. [PMID: 32186397 DOI: 10.1152/ajplung.00295.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PDGF-A is a key contributor to lung development in mice. Its expression is needed for secondary septation of the alveoli and deletion of the gene leads to abnormally enlarged alveolar air spaces in mice. In humans, the same phenotype is the hallmark of bronchopulmonary dysplasia (BPD), a disease that affects premature babies and may have long lasting consequences in adulthood. So far, the knowledge regarding adult effects of developmental arrest in the lung is limited. This is attributable to few follow-up studies of BPD survivors and lack of good experimental models that could help predict the outcomes of this early age disease for the adult individual. In this study, we used the constitutive lung-specific Pdgfa deletion mouse model to analyze the consequences of developmental lung defects in adult mice. We assessed lung morphology, physiology, cellular content, ECM composition and proteomics data in mature mice, that perinatally exhibited lungs with a BPD-like morphology. Histological and physiological analyses both revealed that enlarged alveolar air spaces remained until adulthood, resulting in higher lung compliance and higher respiratory volume in knockout mice. Still, no or only small differences were seen in cellular, ECM and protein content when comparing knockout and control mice. Taken together, our results indicate that Pdgfa deletion-induced lung developmental arrest has consequences for the adult lung at the morphological and functional level. In addition, these mice can reach adulthood with a BPD-like phenotype, which makes them a robust model to further investigate the pathophysiological progression of the disease and test putative regenerative therapies.
Collapse
Affiliation(s)
- Leonor Gouveia
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Simone Kraut
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Elisa Vazquéz-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Baktybek Kojonazarov
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Cheng-Yu Wu
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christine Veith
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Georgios Mermelekas
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ralph Theo Schermuly
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Norbert Weissmann
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Ubags ND, Baker J, Boots A, Costa R, El-Merhie N, Fabre A, Faiz A, Heijink IH, Hiemstra PS, Lehmann M, Meiners S, Rolandsson Enes S, Bartel S. ERS International Congress, Madrid, 2019: highlights from the Basic and Translational Science Assembly. ERJ Open Res 2020; 6:00350-2019. [PMID: 32154289 PMCID: PMC7049707 DOI: 10.1183/23120541.00350-2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/26/2020] [Indexed: 11/15/2022] Open
Abstract
In this review, the Basic and Translational Sciences Assembly of the European Respiratory Society (ERS) provides an overview of the 2019 ERS International Congress highlights. In particular, we discuss how the novel and very promising technology of single cell sequencing has led to the development of a comprehensive map of the human lung, the lung cell atlas, including the discovery of novel cell types and new insights into cellular trajectories in lung health and disease. Further, we summarise recent insights in the field of respiratory infections, which can aid in a better understanding of the molecular mechanisms underlying these infections in order to develop novel vaccines and improved treatment options. Novel concepts delineating the early origins of lung disease are focused on the effects of pre- and post-natal exposures on neonatal lung development and long-term lung health. Moreover, we discuss how these early life exposures can affect the lung microbiome and respiratory infections. In addition, the importance of metabolomics and mitochondrial function analysis to subphenotype chronic lung disease patients according to their metabolic program is described. Finally, basic and translational respiratory science is rapidly moving forward and this will be beneficial for an advanced molecular understanding of the mechanisms underlying a variety of lung diseases. In the long-term this will aid in the development of novel therapeutic targeting strategies in the field of respiratory medicine. Highlights of basic and translational science presented at #ERSCongress 2019 summarising latest research on the lung cell atlas, lung infections, early origins of lung disease and the importance of metabolic alterations in the lunghttp://bit.ly/2UbdBs4
Collapse
Affiliation(s)
- Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland
| | - Jonathan Baker
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Agnes Boots
- Dept of Pharmacology and Toxicology, Maastricht University, Maastricht, the Netherlands
| | - Rita Costa
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Natalia El-Merhie
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Member of the DZL and the Airway Research Center North (ARCN), Borstel, Germany
| | - Aurélie Fabre
- St Vincent's University Hospital, Dublin, Ireland.,University College Dublin School of Medicine, Dublin, Ireland
| | - Alen Faiz
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, Sydney, Australia
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Depts of Pathology & Medical Biology and Pulmonology, Groningen, The Netherlands
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the DZL, Munich, Germany
| | - Sara Rolandsson Enes
- University of Vermont, Dept of Medicine, Larner College of Medicine, Burlington, VT, USA.,Lund University, Dept of Experimental Medical Science, Lund, Sweden
| | - Sabine Bartel
- University of Groningen, University Medical Center Groningen, Depts of Pathology & Medical Biology and Pulmonology, Groningen, The Netherlands
| |
Collapse
|
24
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
25
|
Nikolić MZ, Sun D, Rawlins EL. Human lung development: recent progress and new challenges. Development 2018; 145:145/16/dev163485. [PMID: 30111617 PMCID: PMC6124546 DOI: 10.1242/dev.163485] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent studies have revealed biologically significant differences between human and mouse lung development, and have reported new in vitro systems that allow experimental manipulation of human lung models. At the same time, emerging clinical data suggest that the origins of some adult lung diseases are found in embryonic development and childhood. The convergence of these research themes has fuelled a resurgence of interest in human lung developmental biology. In this Review, we discuss our current understanding of human lung development, which has been profoundly influenced by studies in mice and, more recently, by experiments using in vitro human lung developmental models and RNA sequencing of human foetal lung tissue. Together, these approaches are helping to shed light on the mechanisms underlying human lung development and disease, and may help pave the way for new therapies. Summary: This Review describes how recent technological advances have shed light on the mechanisms underlying human lung development and disease, and outlines the future challenges in this field.
Collapse
Affiliation(s)
- Marko Z Nikolić
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK.,University of Cambridge School of Clinical Medicine, Department of Medicine, Cambridge CB2 0QQ, UK
| | - Dawei Sun
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK
| | - Emma L Rawlins
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK
| |
Collapse
|
26
|
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
|
27
|
Reicherzer T, Häffner S, Shahzad T, Gronbach J, Mysliwietz J, Hübener C, Hasbargen U, Gertheiss J, Schulze A, Bellusci S, Morty RE, Hilgendorff A, Ehrhardt H. Activation of the NF-κB pathway alters the phenotype of MSCs in the tracheal aspirates of preterm infants with severe BPD. Am J Physiol Lung Cell Mol Physiol 2018; 315:L87-L101. [PMID: 29644893 DOI: 10.1152/ajplung.00505.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are released into the airways of preterm infants following lung injury. These cells display a proinflammatory phenotype and are associated with development of severe bronchopulmonary dysplasia (BPD). We aimed to characterize the functional properties of MSCs obtained from tracheal aspirates of 50 preterm infants who required invasive ventilation. Samples were separated by disease severity. The increased proliferative capacity of MSCs was associated with longer duration of mechanical ventilation and higher severity of BPD. Augmented growth depended on nuclear accumulation of NFκBp65 and was accompanied by reduced expression of cytosolic α-smooth muscle actin (α-SMA). The central role of NF-κB signaling was confirmed by inhibition of IκBα phosphorylation. The combined score of proliferative capacity, accumulation of NFκBp65, and expression of α-SMA was used to predict the development of severe BPD with an area under the curve (AUC) of 0.847. We mimicked the clinical situation in vitro, and stimulated MSCs with IL-1β and TNF-α. Both cytokines induced similar and persistent changes as was observed in MSCs obtained from preterm infants with severe BPD. RNA interference was employed to investigate the mechanistic link between NFκBp65 accumulation and alterations in phenotype. Our data indicate that determining the phenotype of resident pulmonary MSCs represents a promising biomarker-based approach. The persistent alterations in phenotype, observed in MSCs from preterm infants with severe BPD, were induced by the pulmonary inflammatory response. NFκBp65 accumulation was identified as a central regulatory mechanism. Future preclinical and clinical studies, aimed to prevent BPD, should focus on phenotype changes in pulmonary MSCs.
Collapse
Affiliation(s)
- Tobias Reicherzer
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Grosshadern, Munich , Germany.,Comprehensive Pneumology Center, Ludwig-Maximilians-University, Asklepios Hospital, and Helmholtz Center Munich , Munich , Germany
| | - Susanne Häffner
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Grosshadern, Munich , Germany.,Comprehensive Pneumology Center, Ludwig-Maximilians-University, Asklepios Hospital, and Helmholtz Center Munich , Munich , Germany
| | - Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center, Member of the German Lung Research Center (DZL) , Giessen , Germany
| | - Judith Gronbach
- Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center, Member of the German Lung Research Center (DZL) , Giessen , Germany
| | - Josef Mysliwietz
- Institute of Molecular Immunology, Helmholtz Center Munich , Munich , Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, Perinatal Center, University Hospital, Ludwig-Maximilians-University, Munich , Germany
| | - Uwe Hasbargen
- Department of Obstetrics and Gynecology, Perinatal Center, University Hospital, Ludwig-Maximilians-University, Munich , Germany
| | - Jan Gertheiss
- Institute of Applied Stochastics and Operations Research, Research Group Applied Statistics, Clausthal University of Technology , Clausthal-Zellerfeld , Germany
| | - Andreas Schulze
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Grosshadern, Munich , Germany
| | - Saverio Bellusci
- Universities of Giessen and Marburg Lung Center, Excellence Cluster Cardio-Pulmonary System, Member of the German Center for Lung Research (DZL), Department of Internal Medicine II , Giessen , Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Lung Center (DZL) , Bad Nauheim , Germany
| | - Anne Hilgendorff
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Grosshadern, Munich , Germany.,Comprehensive Pneumology Center, Ludwig-Maximilians-University, Asklepios Hospital, and Helmholtz Center Munich , Munich , Germany
| | - Harald Ehrhardt
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilians-University, Campus Grosshadern, Munich , Germany.,Department of General Pediatrics and Neonatology, Justus-Liebig-University and Universities of Giessen and Marburg Lung Center, Member of the German Lung Research Center (DZL) , Giessen , Germany
| |
Collapse
|
28
|
The Potentials and Caveats of Mesenchymal Stromal Cell-Based Therapies in the Preterm Infant. Stem Cells Int 2018; 2018:9652897. [PMID: 29765429 PMCID: PMC5911321 DOI: 10.1155/2018/9652897] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/04/2018] [Indexed: 02/06/2023] Open
Abstract
Preponderance of proinflammatory signals is a characteristic feature of all acute and resulting long-term morbidities of the preterm infant. The proinflammatory actions are best characterized for bronchopulmonary dysplasia (BPD) which is the chronic lung disease of the preterm infant with lifelong restrictions of pulmonary function and severe consequences for psychomotor development and quality of life. Besides BPD, the immature brain, eye, and gut are also exposed to inflammatory injuries provoked by infection, mechanical ventilation, and oxygen toxicity. Despite the tremendous progress in the understanding of disease pathologies, therapeutic interventions with proven efficiency remain restricted to a few drug therapies with restricted therapeutic benefit, partially considerable side effects, and missing option of applicability to the inflamed brain. The therapeutic potential of mesenchymal stromal cells (MSCs)—also known as mesenchymal stem cells—has attracted much attention during the recent years due to their anti-inflammatory activities and their secretion of growth and development-promoting factors. Based on a molecular understanding, this review summarizes the positive actions of exogenous umbilical cord-derived MSCs on the immature lung and brain and the therapeutic potential of reprogramming resident MSCs. The pathomechanistic understanding of MSC actions from the animal model is complemented by the promising results from the first phase I clinical trials testing allogenic MSC transplantation from umbilical cord blood. Despite all the enthusiasm towards this new therapeutic option, the caveats and outstanding issues have to be critically evaluated before a broad introduction of MSC-based therapies.
Collapse
|
29
|
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
|