1
|
Predella C, Lapsley L, Ni K, Murray JW, Liu HY, Motelow JE, Snoeck HW, Glasser SW, Saqi A, Dorrello NV. Engraftment of wild-type alveolar type II epithelial cells in surfactant protein C deficient mice. RESEARCH SQUARE 2024:rs.3.rs-4673915. [PMID: 39315275 PMCID: PMC11419168 DOI: 10.21203/rs.3.rs-4673915/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Childhood interstitial lung disease (chILD) secondary to pulmonary surfactant deficiency is a devastating chronic lung disease in children. Clinical presentation includes mild to severe respiratory failure and fibrosis. There is no specific treatment, except lung transplantation, which is hampered by a severe shortage of donor organs, especially for young patients. Repair of lungs with chILD represents a longstanding therapeutic challenge but cell therapy is a promising strategy. As surfactant is produced by alveolar type II epithelial (ATII) cells, engraftment with normal or gene-corrected ATII cells might provide an avenue to cure. Here we used a chILD disease-like model, Sftpc -/- mice, to provide proof-of-principle for this approach. Sftpc -/- mice developed chronic interstitial lung disease with age and were hypersensitive to bleomycin. We could engraft wild-type ATII cells after low dose bleomycin conditioning. Transplanted ATII cells produced mature SPC and attenuated bleomycin-induced lung injury up to two months post-transplant. This study demonstrates that partial replacement of mutant ATII cells can promote lung repair in a mouse model of chILD-like disease.
Collapse
Affiliation(s)
- Camilla Predella
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico of Milan, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico of Milan, Milan, Italy
| | - Lauren Lapsley
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Keyue Ni
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - John W. Murray
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Hsiao-Yun Liu
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua E. Motelow
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephan W. Glasser
- Medical Sciences Program, Department of Medical Education, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian, New York, NY, USA
| | - N. Valerio Dorrello
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| |
Collapse
|
2
|
Lucas JH, Wang Q, Meehan-Atrash J, Pang C, Rahman I. Developmental PFOS exposure alters lung inflammation and barrier integrity in juvenile mice. Toxicol Sci 2024; 201:48-60. [PMID: 38830033 PMCID: PMC11347778 DOI: 10.1093/toxsci/kfae073] [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: 06/05/2024] Open
Abstract
Emerging epidemiological evidence indicates perfluorooctane sulfonic acid (PFOS) is increasingly associated with asthma and respiratory viral infections. Animal studies suggest PFOS disrupts lung development and immuno-inflammatory responses, but little is known about the potential consequences on respiratory health and disease risk. Importantly, PFOS exposure during the critical stages of lung development may increase disease risk later in life. Thus, we hypothesized that developmental PFOS exposure will affect lung inflammation and alveolar/airway development in a sex-dependent manner. To address this knowledge gap, timed pregnant Balb/cJ dams were orally dosed with a PFOS (1.0 or 2.0 mg/kg/d) injected mealworm or a vehicle control daily from gestational day (GD) 0.5 to postnatal day (PND) 21, and offspring were sacrificed at PND 22-23. PFOS-exposed male offspring displayed increased alveolar septa thickness. Occludin was also downregulated in the lungs after PFOS exposure in mice, indicative of barrier dysfunction. BALF macrophages were significantly elevated at 2.0 mg/kg/d PFOS in both sexes compared with vehicles, whereas BALF cytokines (TNF-α, IL-6, KC, MIP-1α, MIP-1β, and MCP-1) were suppressed in PFOS-exposed male offspring compared with vehicle controls. Multiplex nucleic acid hybridization assay showed male-specific downregulation of cytokine gene expression in PFOS-exposed mice compared with vehicle mice. Overall, these results demonstrate PFOS exposure exhibits male-specific adverse effects on lung development and inflammation in juvenile offspring, possibly predisposing them to later-in-life respiratory disease. Further research is required to elucidate the mechanisms underlying the sex-differentiated pulmonary toxicity of PFOS.
Collapse
Affiliation(s)
- Joseph H Lucas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Jiries Meehan-Atrash
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Cortney Pang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States
| |
Collapse
|
3
|
Eddens T, Parks OB, Zhang Y, Manni ML, Casanova JL, Ogishi M, Williams JV. PD-1 signaling in neonates restrains CD8 + T cell function and protects against respiratory viral immunopathology. Mucosal Immunol 2024; 17:476-490. [PMID: 38176655 PMCID: PMC11180597 DOI: 10.1016/j.mucimm.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Respiratory viral infections, including human metapneumovirus (HMPV), remain a leading cause of morbidity and mortality in neonates and infants. However, the mechanisms behind the increased sensitivity to those respiratory viral infections in neonates are poorly understood. Neonates, unlike adults, have several anti-inflammatory mechanisms in the lung, including elevated baseline expression of programmed death ligand 1 (PD-L1), a ligand for the inhibitory receptor programmed cell death protein 1 (PD-1). We thus hypothesized that neonates would rely on PD-1:PD-L1 signaling to restrain antiviral CD8 responses. To test this, we developed a neonatal primary HMPV infection model using wild-type C57BL/6 (B6) and Pdcd1-/- (lacking PD-1) mice. HMPV-infected neonatal mice had increased PD-L1/PD-L2 co-expression on innate immune cells but a similar number of antigen-specific CD8+ T cells and upregulation of PD-1 to that of adult B6 mice. Neonatal CD8+ T cells had reduced interferon-gamma (IFN-γ), granzyme B, and interleukin-2 production compared with B6 adults. Pdcd1-/- neonatal CD8+ T cells had markedly increased production of IFN-γ and granzyme B compared with B6 neonates. Pdcd1-/- neonates had increased acute pathology with HMPV or influenza. Pdcd1-/- neonates infected with HMPV had long-term changes in pulmonary physiology with evidence of immunopathology and a persistent CD8+ T-cell response with increased granzyme B production. Using single-cell ribonucleic acid sequencing from a child lacking PD-1 signaling, a similar activated CD8+ T-cell signature with increased granzyme B expression was observed. These data indicate that PD-1 signaling critically limits CD8+ T-cell effector functions and prevents immunopathology in response to neonatal respiratory viral infections.
Collapse
Affiliation(s)
- Taylor Eddens
- Division of Allergy and Immunology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA
| | - Olivia B Parks
- University of Pittsburgh Medical Scientist Training Program, Pittsburgh, Pennsylvania, USA
| | - Yu Zhang
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA
| | - Michelle L Manni
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France; University Paris Cité, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA; Howard Hughes Medical Institute, New York, New York, USA; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - John V Williams
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA; Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
4
|
Slama N, Abdellatif A, Bahria K, Gasmi S, Khames M, Hadji A, Birkmayer G, Oumouna M, Amrani Y, Benachour K. NADH Intraperitoneal Injection Prevents Lung Inflammation in a BALB/C Mice Model of Cigarette Smoke-Induced Chronic Obstructive Pulmonary Disease. Cells 2024; 13:881. [PMID: 38786103 PMCID: PMC11120028 DOI: 10.3390/cells13100881] [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: 03/28/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Cigarette smoke is one of the main factors in Chronic Obstructive Pulmonary Disease (COPD), a respiratory syndrome marked by persistent respiratory symptoms and increasing airway obstruction. Perturbed NAD+/NADH levels may play a role in various diseases, including lung disorders like COPD. In our study, we investigated the preventive effect of NADH supplementation in an experimental model of COPD induced by cigarette smoke extract (CSE). N = 64 mice randomly distributed in eight groups were injected with NADH (two doses of 100 mg/kg or 200 mg/kg) or dexamethasone (2 mg/kg) before being exposed to CSE for up to 9 weeks. Additionally, NADH supplementation preserved lung antioxidant defenses by preventing the functional loss of key enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase, and the expression levels of glutathione (GSH) (n = 4, p < 0.001). It also reduced oxidative damage markers, such as malondialdehyde (MDA) and nitrites (n = 4, p < 0.001). A marked increase in tissue myeloperoxidase activity was assessed (MPO), confirming neutrophils implication in the inflammatory process. The latter was significantly ameliorated in the NADH-treated groups (p < 0.001). Finally, NADH prevented the CSE-induced secretion of cytokines such as Tumor Necrosis Factor alpha (TNF-α), IL-17, and IFN-y (n = 4, p < 0.001). Our study shows, for the first time, the clinical potential of NADH supplementation in preventing key features of COPD via its unique anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Nada Slama
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Amina Abdellatif
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Karima Bahria
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Sara Gasmi
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Maamar Khames
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Abderrahmene Hadji
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - George Birkmayer
- Department of Medical Chemistry, University of Graz, 8020 Graz, Austria
- Birkmayer Laboratories, 1090 Vienna, Austria
| | - Mustapha Oumouna
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| | - Yassine Amrani
- Department of Respiratory Sciences, Institute of Lung Health and NIHR Leicester BRC-Respiratory, Glenfield Hospital, University of Leicester, Leicester LE1 7RH, UK;
| | - Karine Benachour
- Laboratory of Experimental Biology and Pharmacology, Faculty of Sciences, Dr. Yahia Fares University, Medea 26000, Algeria; (N.S.); (A.A.); (K.B.); (S.G.); (M.K.); (A.H.); (M.O.)
| |
Collapse
|
5
|
De Bie FR, Regin Y, Dubois A, Scuglia M, Arai T, Muylle E, Basurto D, Regin M, Croubels S, Cherlet M, Partridge EA, Allegaert K, Russo FM, Deprest JA. Prenatal treprostinil improves pulmonary arteriolar hypermuscularization in the rabbit model of congenital diaphragmatic hernia. Biomed Pharmacother 2024; 170:115996. [PMID: 38086148 DOI: 10.1016/j.biopha.2023.115996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a congenital malformation characterized by pulmonary hypoplasia, pulmonary hypertension, and cardiac dysfunction. Pulmonary hypertension represents the major cause of neonatal mortality and morbidity. Prenatal diagnosis allows assessment of severity and selection of foetal surgery candidates. We have shown that treprostinil, a prostacyclin analogue with an anti-remodelling effect, attenuates the relative hypermuscularization of the pulmonary vasculature in rats with nitrofen-induced CDH. Here we confirm these observations in a large animal model of surgically-created CDH. In the rabbit model, subcutaneous maternal administration of treprostinil at 150 ng/kg/min consistently reached target foetal concentrations without demonstrable detrimental foetal or maternal adverse effects. In pups with CDH, prenatal treprostinil reduced pulmonary arteriolar proportional medial wall thickness and downregulated inflammation and myogenesis pathways. No effect on alveolar morphometry or lung mechanics was observed. These findings provide further support towards clinical translation of prenatal treprostinil for CDH.
Collapse
Affiliation(s)
- Felix R De Bie
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium
| | - Yannick Regin
- Unit of Woman and Child, Department of Development and Regeneration, KU Leuven, Belgium
| | - Antoine Dubois
- Unit of Abdominal Transplantation, Department of Microbiology, Immunology and Transplantation, KU Leuven, Belgium
| | - Marianna Scuglia
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium
| | - Tomohiro Arai
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium
| | - Ewout Muylle
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium
| | - David Basurto
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium
| | - Marius Regin
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel, Belgium
| | - Siska Croubels
- Department of Pathobiology, Pharmacology and Zoological Medicine, Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Marc Cherlet
- Department of Pathobiology, Pharmacology and Zoological Medicine, Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Emily A Partridge
- Center for Fetal Research, The Children's Hospital of Philadelphia, United States
| | - Karel Allegaert
- Unit of Woman and Child, Department of Development and Regeneration, KU Leuven, Belgium; Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Hospital Pharmacy, Erasmus MC, Rotterdam, the Netherlands
| | - Francesca M Russo
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium; Division of Obstetrics and Gynecology, University Hospitals Leuven, Belgium
| | - Jan A Deprest
- Unit of Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Belgium; Division of Obstetrics and Gynecology, University Hospitals Leuven, Belgium; Institute for Women's Health, University College London, United Kingdom.
| |
Collapse
|
6
|
Riccetti MR, Green J, Taylor TJ, Perl AKT. Prenatal FGFR2 Signaling via PI3K/AKT Specifies the PDGFRA + Myofibroblast. Am J Respir Cell Mol Biol 2024; 70:63-77. [PMID: 37734036 PMCID: PMC10768833 DOI: 10.1165/rcmb.2023-0245oc] [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: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
It is well known that FGFR2 (fibroblast growth factor receptor 2) signaling is critical for proper lung development. Recent studies demonstrate that epithelial FGFR2 signaling during the saccular phase of lung development (sacculation) regulates alveolar type 1 (AT1) and AT2 cell differentiation. During sacculation, PDGFRA (platelet-derived growth factor receptor-α)-positive lung fibroblasts exist as three functional subtypes: contractile myofibroblasts, extracellular matrix-producing matrix fibroblasts, and lipofibroblasts. All three subtypes are required during alveolarization to establish a niche that supports AT2 epithelial cell self-renewal and AT1 epithelial cell differentiation. FGFR2 signaling directs myofibroblast differentiation in PDGFRA+ fibroblasts during alveolar reseptation after pneumonectomy. However, it remains unknown if FGFR2 signaling regulates PDGFRA+ myo-, matrix, or lipofibroblast differentiation during sacculation. In this study, FGFR2 signaling was inhibited by temporal expression of a secreted dominant-negative FGFR2b (dnFGFR2) by AT2 cells from embryonic day (E) 16.5 to E18.5. Fibroblast and epithelial differentiation were analyzed at E18.5 and postnatal days 7 and 21. At all time points, the number of myofibroblasts was reduced and the number of lipo-/matrix fibroblasts was increased. AT2 cells are increased and AT1 cells are reduced postnatally, but not at E18.5. Similarly, in organoids made with PDGFRA+ fibroblasts from dnFGFR2 lungs, increased AT2 cells and reduced AT1 cells were observed. In vitro treatment of primary wild-type E16.5 adherent saccular lung fibroblasts with recombinant dnFGFR2b/c resulted in reduced myofibroblast contraction. Treatment with the PI3K/AKT activator 740 Y-P rescued the lack of myofibroblast differentiation caused by dnFGFR2b/2c. Moreover, treatment with the PI3K/AKT activator 740 Y-P rescued myofibroblast differentiation in E18.5 fibroblasts isolated from dnFGFR2 lungs.
Collapse
Affiliation(s)
- Matthew R. Riccetti
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Jenna Green
- Division of Neonatology and Pulmonary Biology and
| | - Thomas J. Taylor
- Division of Neonatology and Pulmonary Biology and
- College of Arts and Sciences, University of Cincinnati, Cincinnati, Ohio; and
| | - Anne-Karina T. Perl
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
7
|
Valenzuela I, Regin Y, Gie A, Basurto D, Emam D, Scuglia M, Zapletalova K, Greyling M, Deprest J, van der Merwe J. Long-term pulmonary and neurodevelopmental impairment in a fetal growth restriction rabbit model. Sci Rep 2023; 13:20966. [PMID: 38017239 PMCID: PMC10684490 DOI: 10.1038/s41598-023-48174-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
Fetal growth restriction (FGR) remains one of the main obstetrical problems worldwide, with consequences beyond perinatal life. Animal models with developmental and structural similarities to the human are essential to understand FGR long-term consequences and design novel therapeutic strategies aimed at preventing or ameliorating them. Herein, we described the long-term consequences of FGR in pulmonary function, structure, and gene expression, and characterized neurodevelopmental sequelae up to preadolescence in a rabbit model. FGR was induced at gestational day 25 by surgically reducing placental blood supply in one uterine horn, leaving the contralateral horn as internal control. Neonatal rabbits born near term were assigned to foster care in mixed groups until postnatal day (PND) 21. At that time, one group underwent pulmonary biomechanical testing followed by lung morphometry and gene expression analysis. A second group underwent longitudinal neurobehavioral assessment until PND 60 followed by brain harvesting for multiregional oligodendrocyte and microglia quantification. FGR was associated with impaired pulmonary function and lung development at PND 21. FGR rabbits had higher respiratory resistance and altered parenchymal biomechanical properties in the lungs. FGR lungs presented thicker alveolar septal walls and reduced alveolar space. Furthermore, the airway smooth muscle content was increased, and the tunica media of the intra-acinar pulmonary arteries was thicker. In addition, FGR was associated with anxiety-like behavior, impaired memory and attention, and lower oligodendrocyte proportion in the frontal cortex and white matter. In conclusion, we documented and characterized the detrimental pulmonary function and structural changes after FGR, independent of prematurity, and beyond the neonatal period for the first time in the rabbit model, and describe the oligodendrocyte alteration in pre-adolescent rabbit brains. This characterization will allow researchers to develop and test therapies to treat FGR and prevent its sequelae.
Collapse
Affiliation(s)
- Ignacio Valenzuela
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium.
| | - Yannick Regin
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
| | - Andre Gie
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - David Basurto
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
| | - Doaa Emam
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Department of Obstetrics and Gynaecology, University Hospitals Tanta, Tanta, Egypt
| | - Marianna Scuglia
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Katerina Zapletalova
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Third Faculty of Medicine, Institute for the Care of Mother and Child, Charles University, Prague, Czech Republic
| | - Marnel Greyling
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Johannes van der Merwe
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Herestraat 49, Box 805, B-3000, Leuven, Belgium
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
8
|
Zini T, Miselli F, Berardi A. Noninvasive Monitoring Strategies for Bronchopulmonary Dysplasia or Post-Prematurity Respiratory Disease: Current Challenges and Future Prospects. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1753. [PMID: 38002844 PMCID: PMC10670116 DOI: 10.3390/children10111753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023]
Abstract
Definitions of bronchopulmonary dysplasia (BPD) or post-prematurity respiratory disease (PPRD) aim to stratify the risk of mortality and morbidity, with an emphasis on long-term respiratory outcomes. There is no univocal classification of BPD due to its complex multifactorial nature and the substantial heterogeneity of clinical presentation. Currently, there is no definitive treatment available for extremely premature very-low-birth-weight infants with BPD, and challenges in finding targeted preventive therapies persist. However, innovative stem cell-based postnatal therapies targeting BPD-free survival are emerging, which are likely to be offered in the first few days of life to high-risk premature infants. Hence, we need easy-to-use noninvasive tools for a standardized, precise, and reliable BPD assessment at a very early stage, to support clinical decision-making and to predict the response to treatment. In this non-systematic review, we present an overview of strategies for monitoring preterm infants with early and evolving BPD-PPRD, and we make some remarks on future prospects, with a focus on near-infrared spectroscopy (NIRS).
Collapse
Affiliation(s)
- Tommaso Zini
- Department of Medical and Surgical Sciences of Mothers, Children and Adults, Post-Graduate School of Pediatrics, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Francesca Miselli
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences of Mothers, Children and Adults, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| | - Alberto Berardi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Sciences of Mothers, Children and Adults, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| |
Collapse
|
9
|
Zapletalova K, Valenzuela I, Greyling M, Regin Y, Frigolett C, Krofta L, Deprest J, van der Merwe J. The Effects of Prenatal Pravastatin Treatment in the Rabbit Fetal Growth Restriction Model. Biomedicines 2023; 11:2685. [PMID: 37893059 PMCID: PMC10604497 DOI: 10.3390/biomedicines11102685] [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: 08/22/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Fetal growth restriction (FGR) remains without an effective prenatal treatment. Evidence from murine FGR models suggests a beneficial effect of prenatal pravastatin. Since the rabbit hemodichorial placenta more closely resembles the human condition, we investigated the effects of prenatal maternal pravastatin administration in the rabbit FGR model. At a gestational age of 25 days (term 31d), pregnant dams underwent partial uteroplacental vessel ligation (UPVL) in one uterine horn to induce FGR, leaving the other horn as a control. Dams were randomized to either receive 5 mg/kg/d pravastatin dissolved in their drinking water or normal drinking water until delivery. At GA 30d, the rabbits were delivered and were divided into four groups: control without pravastatin (C/NoPrav), FGR without pravastatin (FGR/NoPrav), FGR with pravastatin (FGR/Prav), and controls with pravastatin (C/Prav). The newborn rabbits underwent pulmonary functional assessment and neurobehavioral assessment, and they were harvested for alveolar morphometry or neuropathology. The placentas underwent histology examination and RNA expression. Birth weight was lower in the FGR groups (FGR/Prav, FGR/NoPrav), but there was no difference between FGR/Prav and C/NoPrav. No differences were noted in placental zone proportions, but eNOS in FGR/Prav placentas and VEGFR-2 in FGR/Prav and C/Prav were upregulated. There were no differences in pulmonary function assessment and alveolar morphometry. FGR/Prav kittens had increased neurosensory scores, but there were no differences in neuromotor tests, neuron density, apoptosis, and astrogliosis. In conclusion, in the rabbit FGR model, pravastatin upregulated the expression of VEGFR-2 and eNOS in FGR placentas and was associated with higher neurosensory scores, without measurable effects on birthweight, pulmonary function and morphology, and neuron density.
Collapse
Affiliation(s)
- Katerina Zapletalova
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
- Institute for the Care of Mother and Child, Third Faculty of Medicine, Charles University, 147 10 Prague, Czech Republic
| | - Ignacio Valenzuela
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
| | - Marnel Greyling
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
| | - Yannick Regin
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
| | - Cristian Frigolett
- Department of Public Health and Primary Care, Leuven Statistics Research Centre, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Ladislav Krofta
- Institute for the Care of Mother and Child, Third Faculty of Medicine, Charles University, 147 10 Prague, Czech Republic
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Johannes van der Merwe
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium; (K.Z.); (I.V.)
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
10
|
Pu J, Leme AS, de Lima e Silva C, Beeche C, Nyunoya T, Königshoff M, Chandra D. Deep-Masker: A Deep Learning-based Tool to Assess Chord Length from Murine Lung Images. Am J Respir Cell Mol Biol 2023; 69:126-134. [PMID: 37236629 PMCID: PMC10399147 DOI: 10.1165/rcmb.2023-0051ma] [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: 02/10/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Chord length is an indirect measure of alveolar size and a critical endpoint in animal models of chronic obstructive pulmonary disease (COPD). In assessing chord length, the lumens of nonalveolar structures are eliminated from measurement by various methods, including manual masking. However, manual masking is resource intensive and can introduce variability and bias. We created a fully automated deep learning-based tool to mask murine lung images and assess chord length to facilitate mechanistic and therapeutic discovery in COPD called Deep-Masker (available at http://47.93.0.75:8110/login). We trained the deep learning algorithm for Deep-Masker using 1,217 images from 137 mice from 12 strains exposed to room air or cigarette smoke for 6 months. We validated this algorithm against manual masking. Deep-Masker demonstrated high accuracy with an average difference in chord length compared with manual masking of -0.3 ± 1.4% (rs = 0.99) for room-air-exposed mice and 0.7 ± 1.9% (rs = 0.99) for cigarette-smoke-exposed mice. The difference between Deep-Masker and manually masked images for change in chord length because of cigarette smoke exposure was 6.0 ± 9.2% (rs = 0.95). These values exceed published estimates for interobserver variability for manual masking (rs = 0.65) and the accuracy of published algorithms by a significant margin. We validated the performance of Deep-Masker using an independent set of images. Deep-Masker can be an accurate, precise, fully automated method to standardize chord length measurement in murine models of lung disease.
Collapse
Affiliation(s)
- Jiantao Pu
- Department of Radiology
- Department of Bioengineering, and
| | - Adriana S. Leme
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Camilla de Lima e Silva
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Toru Nyunoya
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Divay Chandra
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
11
|
Ng B, Xie C, Su L, Kuthubudeen FF, Kwek XY, Yeong D, Pua CJ, Cook SA, Lim WW. IL11 (Interleukin-11) Causes Emphysematous Lung Disease in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:739-754. [PMID: 36924234 PMCID: PMC10125130 DOI: 10.1161/atvbaha.122.318802] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Marfan Syndrome (MFS) is an inherited connective tissue disorder caused by mutations in the FBN1 (fibrillin-1) gene. Lung abnormalities are common in MFS, but their pathogenesis is poorly understood. IL11 (interleukin-11) causes aortic disease in a mouse model of MFS and was studied here in the lung. METHODS We examined histological and molecular phenotypes in the lungs of Fbn1C1041G/+ mice (mouse model of Marfan Syndrome [mMFS]), an established mouse model of MFS. To identify IL11-expressing cells, we used immunohistochemistry on lungs of 4- and 16-week-old Fbn1C1041G/+:Il11EGFP/+ reporter mice. We studied the effects of IL11 inhibition by RT-qPCR, immunoblots and histopathology in lungs from genetic or pharmacologic models: (1) 16-week-old IL11 receptor (IL11RA) knockout mMFS mice (Fbn1C1041G/+:Il11ra1-/- mice) and (2) in mMFS mice administered IgG control or interleukin-11 receptor antibodies twice weekly from 4 to 24 weeks of age. RESULTS mMFS lungs showed progressive loss and enlargement of distal airspaces associated with increased proinflammatory and profibrotic gene expression as well as matrix metalloproteinases 2, 9, and 12. IL11 was increased in mMFS lungs and localized to smooth muscle and endothelial cells in young mMFS mice in the Fbn1C1041G/+:Il11EGFP/+ reporter strain and in fibroblasts, in older mice. In mMFS mice, genetic (Fbn1C1041G/+:Il11ra1-/-) or pharmacologic (anti-interleukin-11 receptor) inhibition of IL11 signaling reduced lung emphysema, fibrosis, and inflammation. This protective effect was associated with reduced pathogenic ERK1/2 signaling and lower metalloproteinase 2, 9, and 12 expression. CONCLUSIONS IL11 causes lung disease in mMFS. This reveals a shared IL11-driven disease mechanism in lung and aorta in MFS and suggests inhibition of IL11 signaling as a holistic approach for treating multiorgan morbidity in MFS.
Collapse
Affiliation(s)
- Benjamin Ng
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| | - Chen Xie
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Liping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Fathima F. Kuthubudeen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| | - Xiu-Yi Kwek
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Daryl Yeong
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Stuart A. Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
- MRC-London Institute of Medical Sciences, United Kingdom (S.A.C.)
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| |
Collapse
|
12
|
Valenzuela I, Zapletalova K, Greyling M, Regin Y, Gie A, Basurto D, Deprest J, van der Merwe J. Fetal Growth Restriction Impairs Lung Function and Neurodevelopment in an Early Preterm Rabbit Model. Biomedicines 2023; 11:biomedicines11010139. [PMID: 36672647 PMCID: PMC9855731 DOI: 10.3390/biomedicines11010139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/08/2023] Open
Abstract
We previously reported the multi-system sequelae of fetal growth restriction, induced by placental underperfusion, in near-term born rabbits, in the immediate neonatal period and up to pre-adolescence. Herein, we describe the pulmonary and neurodevelopmental consequences of FGR in rabbits born preterm. We hypothesize that FGR has an additional detrimental effect on prematurity in both pulmonary function and neurodevelopment. FGR was induced at gestational day (GD) 25 by placental underperfusion, accomplished by partial uteroplacental vessel ligation in one uterine horn. Rabbits were delivered by cesarean section at GD 29, and placentas were harvested for histology. Neonates underwent neurobehavioral or pulmonary functional assessment at postnatal day 1, followed by brain or lung harvesting, respectively. The neurodevelopmental assessment included neurobehavioral testing and multiregional quantification of cell density and apoptosis in the brain. Lung assessment included functional testing, alveolar morphometry, and airway histology. FGR was associated with higher perinatal mortality, lower birth and placental weight, and a similar brain-to-body weight ratio compared to controls. Placental underperfusion decreased labyrinth and junction zone volumes in FGR placentas. FGR impaired pulmonary function, depicted by higher parenchymal resistance, damping, and elastance. Alveolar morphometry and airway smooth muscle content were comparable between groups. Neurobehavioral tests showed motoric and sensorial impairment in FGR rabbits. In FGR brains, cell density was globally reduced, with higher apoptosis in selected areas. In conclusion, in preterm-born rabbits, placental underperfusion leads to higher mortality, FGR, and impaired lung and brain development in early assessment. This study complements previous findings of placental, pulmonary, and neurodevelopmental impairment in near-term born rabbits in this model.
Collapse
Affiliation(s)
- Ignacio Valenzuela
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
| | - Katerina Zapletalova
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
- Third Faculty of Medicine, Institute for the Care of Mother and Child, Charles University, 147 10 Prague, Czech Republic
| | - Marnel Greyling
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
| | - Yannick Regin
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
| | - Andre Gie
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town 7505, South Africa
| | - David Basurto
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
| | - Jan Deprest
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Johannes van der Merwe
- Cluster Woman and Child, Department of Development and Regeneration, Group Biomedical Sciences, KU Leuven Herestraat 49, 3000 Leuven, Belgium
- Department of Obstetrics and Gynecology, Division Woman and Child, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
13
|
Sengupta S, Abhinav N, Singh S, Dutta J, Mabalirajan U, Kaliyamurthy K, Mukherjee PK, Jaisankar P, Bandyopadhyay A. Standardised Sonneratia apetala Buch.-Ham. fruit extract inhibits human neutrophil elastase and attenuates elastase-induced lung injury in mice. Front Pharmacol 2022; 13:1011216. [PMID: 36569308 PMCID: PMC9768866 DOI: 10.3389/fphar.2022.1011216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) along with asthma is a major and increasing global health problem. Smoking contributes to about 80%-90% of total COPD cases in the world. COPD leads to the narrowing of small airways and destruction of lung tissue leading to emphysema primarily caused by neutrophil elastase. Neutrophil elastase plays an important role in disease progression in COPD patients and has emerged as an important target for drug discovery. Sonneratia apetala Buch.-Ham. is a mangrove plant belonging to family Sonneratiaceae. It is widely found in the Sundarban regions of India. While the fruits of this plant have antibacterial, antifungal, antioxidant and astringent activities, fruit and leaf extracts have been shown to reduce the symptoms of asthma and cough. The aim of this study is to find whether hydro alcoholic fruit extracts of S. apetala inhibit neutrophil elastase and thus prevent the progression of neutrophil elastase-driven lung emphysema. The hydroalcoholic extract, ethanol: water (90:10), of the S. apetala Buch.-Ham. fresh fruits (SAM) were used for neutrophil elastase enzyme kinetic assay and IC50 of the extract was determined. The novel HPLC method has been developed and the extract was standardized with gallic acid and ellagic acid as standards. The extract was further subjected to LC-MS2 profiling to identify key phytochemicals. The standardized SAM extract contains 53 μg/mg of gallic acid and 95 μg/mg of ellagic acid, based on the HPLC calibration curve. SAM also reversed the elastase-induced morphological change of human epithelial cells and prevented the release of ICAM-1 in vitro and an MTT assay was conducted to assess the viability. Further, 10 mg/kg SAM had reduced alveolar collapse induced by neutrophil elastase in the mice model. Thus, in this study, we reported for the first time that S. apetala fruit extract has the potential to inhibit human neutrophil elastase in vitro and in vivo.
Collapse
Affiliation(s)
- Sayantan Sengupta
- Cardiovascular Disease and Respiratory Disorders Laboratory, Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nipun Abhinav
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Sabita Singh
- Molecular Pathobiology of Respiratory Diseases Laboratory, Cell Biology and Physiology Department, CSIR-Indian Institute of Chemical Biology, Kolkata, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Joytri Dutta
- Molecular Pathobiology of Respiratory Diseases Laboratory, Cell Biology and Physiology Department, CSIR-Indian Institute of Chemical Biology, Kolkata, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ulaganathan Mabalirajan
- Molecular Pathobiology of Respiratory Diseases Laboratory, Cell Biology and Physiology Department, CSIR-Indian Institute of Chemical Biology, Kolkata, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Karthigeyan Kaliyamurthy
- Central National Herbarium, Botanical Survey of India, A.J.C.B. Indian Botanic Garden, Howrah, India
| | | | - Parasuraman Jaisankar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,Laboratory of Catalysis and Chemical Biology, Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, Kolkata, India,*Correspondence: Parasuraman Jaisankar, ; Arun Bandyopadhyay,
| | - Arun Bandyopadhyay
- Cardiovascular Disease and Respiratory Disorders Laboratory, Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,*Correspondence: Parasuraman Jaisankar, ; Arun Bandyopadhyay,
| |
Collapse
|
14
|
Placental vascular alterations are associated with early neurodevelopmental and pulmonary impairment in the rabbit fetal growth restriction model. Sci Rep 2022; 12:19720. [PMID: 36385147 PMCID: PMC9668827 DOI: 10.1038/s41598-022-22895-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
Abstract
Fetal growth restriction is one of the leading causes of perinatal mortality and morbidity and has consequences that extend well beyond the neonatal period. Current management relies on timely delivery rather than improving placental function. Several prenatal strategies have failed to show benefit in clinical trials after promising results in animal models. Most of these animal models have important developmental and structural differences compared to the human and/or are insufficiently characterized. We aimed to describe placental function and structure in an FGR rabbit model, and to characterize the early brain and lung developmental morbidity using a multimodal approach. FGR was induced in time-mated rabbits at gestational day 25 by partial uteroplacental vessel ligation in one horn. Umbilical artery Doppler was measured before caesarean delivery at gestational day 30, and placentas were harvested for computed microtomography and histology. Neonates underwent neurobehavioral or pulmonary functional assessment the day after delivery, followed by brain or lung harvesting, respectively. Neuropathological assessment included multiregional quantification of neuron density, apoptosis, astrogliosis, cellular proliferation, and oligodendrocyte progenitors. Brain region volumes and diffusion metrics were obtained from ex-vivo brain magnetic resonance imaging. Lung assessment included biomechanical tests and pulmonary histology. Fetal growth restriction was associated with labyrinth alterations in the placenta, driven by fetal capillary reduction, and overall reduced vessels volume. FGR caused altered neurobehavior paralleled by regional neuropathological deficits and reduced fractional anisotropy in the cortex, white matter, and hippocampus. In addition, FGR kittens presented functional alterations in the peripheral lung and structurally underdeveloped alveoli. In conclusion, in a uteroplacental insufficiency FGR rabbit model, placental vascular alterations coincide with neurodevelopmental and pulmonary disruption.
Collapse
|
15
|
Sengupta S, Reddy JR, Rajesh N, Jaiswal A, Mabalirajan U, Palakodety RK, Mukherjee P, Bandyopadhyay A. Novel benzoxazinone derivative as potent human neutrophil elastase inhibitor: Potential implications in lung injury. Eur J Pharmacol 2022; 931:175187. [PMID: 35952844 DOI: 10.1016/j.ejphar.2022.175187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022]
Abstract
Neutrophil elastase, a powerful physiological defence tool, may serve as drug target for diverse diseases due to its bystander effect on host cells like chronic obstructive pulmonary disease (COPD). Here, we synthesised seven novel benzoxazinone derivatives and identified that these synthetic compounds are human neutrophil elastase inhibitor that was demonstrated by enzyme substrate kinetic assay. One such compound, PD05, emerged as the most potent inhibitor with lower IC50 as compared to control drug sivelestat. While this inhibition is competitive based on substrate dilution assay, PD05 showed a high binding affinity for human neutrophil elastase (Kd = 1.63 nM) with faster association and dissociation rate compared to notable elastase inhibitors like ONO 6818 and AZD9668, and its interaction with human neutrophil elastase was fully reversible.Preclinical pharmacokinetic studies were performed in vitro where protein binding was found to be 72% with a high recovery rate, aqueous solubility of 194.7 μM, low permeability along with a favourable hERG. Experiments with cell line revealed that the molecule successfully prevented elastase induced rounding and retracted cell morphology and cell cytotoxicity. In mouse model PD05 is able to reduce the alveolar collapse induced by neutrophil elastase. In summary, we demonstrate the in situ, in vitro and in vivo anti-elastase potential of the newly synthesised benzoxazinone derivative PD05 and thus this could be promising candidate for further investigation as a drug for the treatment of COPD.
Collapse
Affiliation(s)
- Sayantan Sengupta
- Cardiovascular Disease & Respiratory Disorders Laboratory, Department of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Jala Ranjith Reddy
- Division of Organic and Biomolecular Medicine, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Nomula Rajesh
- Division of Organic and Biomolecular Medicine, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Ashish Jaiswal
- Molecular Pathobiology of Respiratory Diseases, Department of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Ulaganathan Mabalirajan
- Molecular Pathobiology of Respiratory Diseases, Department of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Radha Krishna Palakodety
- Division of Organic and Biomolecular Medicine, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Pulok Mukherjee
- Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
| | - Arun Bandyopadhyay
- Cardiovascular Disease & Respiratory Disorders Laboratory, Department of Cell Biology & Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India.
| |
Collapse
|
16
|
De Bie FR, Halline CG, Kotzur T, Hayes K, Rouse CC, Chang J, Larson AC, Khan SA, Spina A, Tilden S, Russo FM, Hedrick HL, Deprest J, Partridge EA. Prenatal treprostinil reduces the pulmonary hypertension phenotype in the rat model of congenital diaphragmatic hernia. EBioMedicine 2022; 81:104106. [PMID: 35779494 PMCID: PMC9244734 DOI: 10.1016/j.ebiom.2022.104106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
Background Persistent pulmonary hypertension (PH) causes significant mortality and morbidity in infants with congenital diaphragmatic hernia (CDH). Since pulmonary vascular abnormalities in CDH develop early during foetal development, we hypothesized that prenatal maternal administration of treprostinil, through its anti-remodelling effect, would improve the PH-phenotype in the nitrofen rat model of CDH. Methods In a dose-finding study in normal, healthy pregnant rats, we demonstrated target-range foetal plasma treprostinil concentrations without signs of toxicity. Next, an efficacy study was performed assessing the effects of treprostinil administration at 900 and 1500ng/kg/min from gestational day (GD) 16 until term (GD 21) in CDH and control pups. Pulmonary vascular and airway morphometry, lung mechanics, and expression patterns of genes implicated in the prostaglandin vasoactive pathway were studied. Findings In rats maternal administration of 1500ng/kg/min treprostinil reached target foetal concentrations, with no detrimental maternal or foetal side-effects. Prenatal exposure to 900 and 1500 ng/kg/min treprostinil reduced the medial wall thickness (%MWT) (CDH·900, 38.5± 8·4%; CDH.1500, 40·2±9·7%; CDH, 46·6±8·2%; both p < 0·0001) in rat pups with CDH, however increased the %MWT in normal foetuses (C.T.900, 36·6±11·1%; C.T.1500, 36·9±9·3%; C.P., 26·9±6·2%; both p < 0·001). Pulmonary airway development, lung hypoplasia and pulmonary function were unaffected by drug exposure. Interpretation In pregnant rats maternally administered treprostinil crosses the placenta, attains foetal target concentrations, and is well tolerated by both mother and foetuses. This report shows a significant reduction of pulmonary arteriole muscularization with prenatal treprostinil in a nitrofen rat model, supporting the promise of this treatment approach for PH of CDH. Funding United Therapeutics Corporation provided treprostinil and financial support (ISS-2020-10879).
Collapse
|
17
|
Moraes L, Trentini MM, Fousteris D, Eto SF, Chudzinski-Tavassi AM, Leite LCDC, Kanno AI. CRISPR/Cas9 Approach to Generate an Auxotrophic BCG Strain for Unmarked Expression of LTAK63 Adjuvant: A Tuberculosis Vaccine Candidate. Front Immunol 2022; 13:867195. [PMID: 35432328 PMCID: PMC9005855 DOI: 10.3389/fimmu.2022.867195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/09/2022] [Indexed: 12/30/2022] Open
Abstract
Tuberculosis is one of the deadliest infectious diseases and a huge healthcare burden in many countries. New vaccines, including recombinant BCG-based candidates, are currently under evaluation in clinical trials. Our group previously showed that a recombinant BCG expressing LTAK63 (rBCG-LTAK63), a genetically detoxified subunit A of heat-labile toxin (LT) from Escherichia coli, induces improved protection against Mycobacterium tuberculosis (Mtb) in mouse models. This construct uses a traditional antibiotic resistance marker to enable heterologous expression. In order to avoid the use of these markers, not appropriate for human vaccines, we used CRISPR/Cas9 to generate unmarked mutations in the lysA gene, thus obtaining a lysine auxotrophic BCG strain. A mycobacterial vector carrying lysA and ltak63 gene was used to complement the auxotrophic BCG which co-expressed the LTAK63 antigen (rBCGΔ-LTAK63) at comparable levels to the original construct. The intranasal challenge with Mtb confirmed the superior protection induced by rBCGΔ-LTAK63 compared to wild-type BCG. Furthermore, mice immunized with rBCGΔ-LTAK63 showed improved lung function. In this work we showed the practical application of CRISPR/Cas9 in the tuberculosis vaccine development field.
Collapse
Affiliation(s)
- Luana Moraes
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa de Pós-Graduação Interunidades em Biotecnologia Universidade de São Paulo - Instituto de Pesquisas Tecnológicas - Instituto Butantan (USP-IPT-IB), São Paulo, Brazil
| | | | - Dimitrios Fousteris
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,UnivLyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Silas Fernandes Eto
- Development and Innovation Laboratory, Instituto Butantan, São Paulo, Brazil
| | - Ana Marisa Chudzinski-Tavassi
- Development and Innovation Laboratory, Instituto Butantan, São Paulo, Brazil.,Center of Excellence in New Target Discovery (CENTD) Special Laboratory, Instituto Butantan, São Paulo, Brazil
| | | | - Alex Issamu Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
18
|
Toth A, Steinmeyer S, Kannan P, Gray J, Jackson CM, Mukherjee S, Demmert M, Sheak JR, Benson D, Kitzmiller J, Wayman JA, Presicce P, Cates C, Rubin R, Chetal K, Du Y, Miao Y, Gu M, Guo M, Kalinichenko VV, Kallapur SG, Miraldi ER, Xu Y, Swarr D, Lewkowich I, Salomonis N, Miller L, Sucre JS, Whitsett JA, Chougnet CA, Jobe AH, Deshmukh H, Zacharias WJ. Inflammatory blockade prevents injury to the developing pulmonary gas exchange surface in preterm primates. Sci Transl Med 2022; 14:eabl8574. [PMID: 35353543 PMCID: PMC9082785 DOI: 10.1126/scitranslmed.abl8574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1β and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.
Collapse
Affiliation(s)
- Andrea Toth
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Shelby Steinmeyer
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Paranthaman Kannan
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Jerilyn Gray
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester, Rochester, NY USA
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Martin Demmert
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, Institute for Systemic Inflammation Research, University of Lϋbeck, Lϋbeck, Germany
| | - Joshua R. Sheak
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Daniel Benson
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Joseph Kitzmiller
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Joseph A. Wayman
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA USA
| | - Christopher Cates
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Rhea Rubin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Yina Du
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Yifei Miao
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Mingxia Gu
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Minzhe Guo
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Vladimir V. Kalinichenko
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA USA
| | - Emily R. Miraldi
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Yan Xu
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Daniel Swarr
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Ian Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Nathan Salomonis
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Lisa Miller
- California National Primate Research Center, University of California Davis, Davis, CA USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA USA
| | - Jennifer S. Sucre
- Division of Neonatology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN USA
| | - Jeffrey A. Whitsett
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Alan H. Jobe
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Hitesh Deshmukh
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - William J. Zacharias
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|