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
|
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
|
3
|
Wang L, Xiao J, Zhang B, Hou A. Epigenetic modifications in the development of bronchopulmonary dysplasia: a review. Pediatr Res 2024; 96:632-642. [PMID: 38570557 DOI: 10.1038/s41390-024-03167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
While perinatal medicine advancements have bolstered survival outcomes for premature infants, bronchopulmonary dysplasia (BPD) continues to threaten their long-term health. Gene-environment interactions, mediated by epigenetic modifications such as DNA methylation, histone modification, and non-coding RNA regulation, take center stage in BPD pathogenesis. Recent discoveries link methylation variations across biological pathways with BPD. Also, the potential reversibility of histone modifications fuels new treatment avenues. The review also highlights the promise of utilizing mesenchymal stem cells and their exosomes as BPD therapies, given their ability to modulate non-coding RNA, opening novel research and intervention possibilities. IMPACT: The complexity and universality of epigenetic modifications in the occurrence and development of bronchopulmonary dysplasia were thoroughly discussed. Both molecular and cellular mechanisms contribute to the diverse nature of epigenetic changes, suggesting the need for deeper biochemical techniques to explore these molecular alterations. The utilization of innovative cell-specific drug delivery methods like exosomes and extracellular vesicles holds promise in achieving precise epigenetic regulation.
Collapse
Affiliation(s)
- Lichuan Wang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Jun Xiao
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Bohan Zhang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Chen Y, Jiang W, Wang J, Ma X, Wu D, Liu L, Ji M, Qu X, Liu C, Liu H, Qin X, Xiang Y. Conditional knockout of ITGB4 in bronchial epithelial cells directs bronchopulmonary dysplasia. J Cell Mol Med 2023; 27:3760-3772. [PMID: 37698050 PMCID: PMC10718146 DOI: 10.1111/jcmm.17948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023] Open
Abstract
Neonatal respiratory system disease is closely associated with embryonic lung development. Our group found that integrin β4 (ITGB4) is downregulated in the airway epithelium of asthma patients. Asthma is the most common chronic respiratory illness in childhood. Therefore, we suspect whether the deletion of ITGB4 would affect fetal lung development. In this study, we characterized the role of ITGB4 deficiency in bronchopulmonary dysplasia (BPD). ITGB4 was conditionally knocked out in CCSP-rtTA, Tet-O-Cre and ITGB4f/f triple transgenic mice. Lung tissues at different developmental stages were collected for experimental detection and transcriptome sequencing. The effects of ITGB4 deficiency on lung branching morphogenesis were observed by fetal mouse lung explant culture. Deleting ITGB4 from the airway epithelial cells results in enlargement of alveolar airspaces, inhibition of branching, the abnormal structure of epithelium cells and the impairment of cilia growth during lung development. Scanning electron microscopy showed that the airway epithelial cilia of the β4ccsp.cre group appear to be sparse, shortened and lodging. Lung-development-relevant factors such as SftpC and SOX2 significantly decreased both mRNA and protein levels. KEGG pathway analysis indicated that multiple ontogenesis-regulating-relevant pathways converge to FAK. Accordingly, ITGB4 deletion decreased phospho-FAK, phospho-GSK3β and SOX2 levels, and the correspondingly contrary consequence was detected after treatment with GSK3β agonist (wortmannin). Airway branching defect of β4ccsp.cre mice lung explants was also partly recovered after wortmannin treatment. Airway epithelial-specific deletion of ITGB4 contributes to lung developmental defect, which could be achieved through the FAK/GSK3β/SOX2 signal pathway.
Collapse
Affiliation(s)
- Yu Chen
- School of Basic MedicineCentral South UniversityChangshaChina
- Department of Medical Laboratory, School of MedicineHunan Normal UniversityChangshaChina
| | - Wang Jiang
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Jin‐Mei Wang
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Xiao‐Di Ma
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Di Wu
- School of Basic MedicineCentral South UniversityChangshaChina
- School of MedicineFoshan UniversityFoshanChina
| | - Le‐Xin Liu
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Ming Ji
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Xiang‐Ping Qu
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Chi Liu
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Hui‐Jun Liu
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Xiao‐Qun Qin
- School of Basic MedicineCentral South UniversityChangshaChina
| | - Yang Xiang
- School of Basic MedicineCentral South UniversityChangshaChina
| |
Collapse
|
5
|
Sucre JM, Bock F, Negretti NM, Benjamin JT, Gulleman PM, Dong X, Ferguson KT, Jetter CS, Han W, Liu Y, Kook S, Gokey JJ, Guttentag SH, Kropski JA, Blackwell TS, Zent R, Plosa EJ. Alveolar repair following LPS-induced injury requires cell-ECM interactions. JCI Insight 2023; 8:e167211. [PMID: 37279065 PMCID: PMC10443799 DOI: 10.1172/jci.insight.167211] [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: 11/16/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if β1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of β1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, β1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in β1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that β1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires β1-containing integrins.
Collapse
Affiliation(s)
- Jennifer M.S. Sucre
- Department of Pediatrics, Division of Neonatology
- Department of Cell and Developmental Biology
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | | | - Xinyu Dong
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | - Wei Han
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yang Liu
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jonathan A. Kropski
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Nephrology and Hypertension; and
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
6
|
Liu S, Sun D, Butler R, Rawlins EL. RTK signalling promotes epithelial columnar cell shape and apical junction maintenance in human lung progenitor cells. Development 2023; 150:dev201284. [PMID: 37260147 PMCID: PMC10281517 DOI: 10.1242/dev.201284] [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: 09/09/2022] [Accepted: 05/10/2023] [Indexed: 05/25/2023]
Abstract
Multipotent epithelial progenitor cells can be expanded from human embryonic lungs as organoids and maintained in a self-renewing state using a defined medium. The organoid cells are columnar, resembling the cell morphology of the developing lung tip epithelium in vivo. Cell shape dynamics and fate are tightly coordinated during development. We therefore used the organoid system to identify signalling pathways that maintain the columnar shape of human lung tip progenitors. We found that EGF, FGF7 and FGF10 have distinct functions in lung tip progenitors. FGF7 activates MAPK/ERK and PI3K/AKT signalling, and is sufficient to promote columnar cell shape in primary tip progenitors. Inhibitor experiments show that MAPK/ERK and PI3K/AKT signalling are key downstream pathways, regulating cell proliferation, columnar cell shape and cell junctions. We identified integrin signalling as a key pathway downstream of MAPK/ERK in the tip progenitors; disrupting integrin alters polarity, cell adhesion and tight junction assembly. By contrast, stimulation with FGF10 or EGF alone is not sufficient to maintain organoid columnar cell shape. This study employs organoids to provide insight into the cellular mechanisms regulating human lung development.
Collapse
Affiliation(s)
- Shuyu Liu
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Dawei Sun
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Richard Butler
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Emma L. Rawlins
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
7
|
McCoy AM, Lakhdari O, Shome S, Caoili K, Hernandez GE, Aghaeepour N, Butcher LD, Fisch K, Prince LS. Sp3 is essential for normal lung morphogenesis and cell cycle progression during mouse embryonic development. Development 2023; 150:dev200839. [PMID: 36762637 PMCID: PMC10110423 DOI: 10.1242/dev.200839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023]
Abstract
Members of the Sp family of transcription factors regulate gene expression via binding GC boxes within promoter regions. Unlike Sp1, which stimulates transcription, the closely related Sp3 can either repress or activate gene expression and is required for perinatal survival in mice. Here, we use RNA-seq and cellular phenotyping to show how Sp3 regulates murine fetal cell differentiation and proliferation. Homozygous Sp3-/- mice were smaller than wild-type and Sp+/- littermates, died soon after birth and had abnormal lung morphogenesis. RNA-seq of Sp3-/- fetal lung mesenchymal cells identified alterations in extracellular matrix production, developmental signaling pathways and myofibroblast/lipofibroblast differentiation. The lungs of Sp3-/- mice contained multiple structural defects, with abnormal endothelial cell morphology, lack of elastic fiber formation, and accumulation of lipid droplets within mesenchymal lipofibroblasts. Sp3-/- cells and mice also displayed cell cycle arrest, with accumulation in G0/G1 and reduced expression of numerous cell cycle regulators including Ccne1. These data detail the global impact of Sp3 on in vivo mouse gene expression and development.
Collapse
Affiliation(s)
- Alyssa M. McCoy
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Omar Lakhdari
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sayane Shome
- Department of Pediatrics, Stanford University, Palo Alto, CA 94304, USA
- Department of Anesthesiology, Perioperative and Pain Management, Stanford University, Palo Alto, CA 94305, USA
| | - Kaitlin Caoili
- Department of Pediatrics, Stanford University, Palo Alto, CA 94304, USA
| | - Gilberto E. Hernandez
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Stanford University, Palo Alto, CA 94304, USA
- Department of Anesthesiology, Perioperative and Pain Management, Stanford University, Palo Alto, CA 94305, USA
| | | | - Kathleen Fisch
- Department of Obstetrics, Gynecology, and Reproductive Services, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
8
|
Tang F, Brune JE, Chang MY, Reeves SR, Altemeier WA, Frevert CW. Defining the versican interactome in lung health and disease. Am J Physiol Cell Physiol 2022; 323:C249-C276. [PMID: 35649251 PMCID: PMC9291419 DOI: 10.1152/ajpcell.00162.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM) imparts critical mechanical and biochemical information to cells in the lungs. Proteoglycans are essential constituents of the ECM and play a crucial role in controlling numerous biological processes, including regulating cellular phenotype and function. Versican, a chondroitin sulfate proteoglycan required for embryonic development, is almost absent from mature, healthy lungs and is reexpressed and accumulates in acute and chronic lung disease. Studies using genetically engineered mice show that the versican-enriched matrix can be pro- or anti-inflammatory depending on the cellular source or disease process studied. The mechanisms whereby versican develops a contextual ECM remain largely unknown. The primary goal of this review is to provide an overview of the interaction of versican with its many binding partners, the "versican interactome," and how through these interactions, versican is an integrator of complex extracellular information. Hopefully, the information provided in this review will be used to develop future studies to determine how versican and its binding partners can develop contextual ECMs that control select biological processes. Although this review focuses on versican and the lungs, what is described can be extended to other proteoglycans, tissues, and organs.
Collapse
Affiliation(s)
- Fengying Tang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Jourdan E Brune
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Mary Y Chang
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | - Stephen R Reeves
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| | - William A Altemeier
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Charles W Frevert
- Center for Lung Biology, The University of Washington at South Lake Union, Seattle, Washington
- Department of Comparative Medicine, University of Washington, Seattle, Washington
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
9
|
Yazlovitskaya EM, Plosa E, Bock F, Viquez OM, Mernaugh G, Gewin LS, De Arcangelis A, Georges-Labouesse E, Sonnenberg A, Blackwell TS, Pozzi A, Zent R. The laminin-binding integrins regulate nuclear factor κB-dependent epithelial cell polarity and inflammation. J Cell Sci 2021; 134:jcs259161. [PMID: 34841431 PMCID: PMC8729780 DOI: 10.1242/jcs.259161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
The main laminin-binding integrins α3β1, α6β1 and α6β4 are co-expressed in the developing kidney collecting duct system. We previously showed that deleting the integrin α3 or α6 subunit in the ureteric bud, which gives rise to the kidney collecting system, caused either a mild or no branching morphogenesis phenotype, respectively. To determine whether these two integrin subunits cooperate in kidney collecting duct development, we deleted α3 and α6 in the developing ureteric bud. The collecting system of the double knockout phenocopied the α3 integrin conditional knockout. However, with age, the mice developed severe inflammation and fibrosis around the collecting ducts, resulting in kidney failure. Integrin α3α6-null collecting duct epithelial cells showed increased secretion of pro-inflammatory cytokines and displayed mesenchymal characteristics, causing loss of barrier function. These features resulted from increased nuclear factor kappa-B (NF-κB) activity, which regulated the Snail and Slug (also known as Snai1 and Snai2, respectively) transcription factors and their downstream targets. These data suggest that laminin-binding integrins play a key role in the maintenance of kidney tubule epithelial cell polarity and decrease pro-inflammatory cytokine secretion by regulating NF-κB-dependent signaling.
Collapse
Affiliation(s)
- Eugenia M. Yazlovitskaya
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Erin Plosa
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Olga M. Viquez
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Glenda Mernaugh
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Leslie S. Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Veterans Affairs Hospital, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Adele De Arcangelis
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964/ULP, F-67404 Illkirch, France
| | - Elisabeth Georges-Labouesse
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964/ULP, F-67404 Illkirch, France
| | - Arnoud Sonnenberg
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Timothy S. Blackwell
- Veterans Affairs Hospital, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Veterans Affairs Hospital, Nashville, TN 37232, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Veterans Affairs Hospital, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
10
|
Gut-Lung Dysbiosis Accompanied by Diabetes Mellitus Leads to Pulmonary Fibrotic Change through the NF-κB Signaling Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:838-856. [PMID: 33705752 DOI: 10.1016/j.ajpath.2021.02.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Growing evidence shows that the lungs are an unavoidable target organ of diabetic complications. However, the pathologic mechanisms of diabetic lung injury are still controversial. This study demonstrated the dysbiosis of the gut and lung microbiome, pulmonary alveolar wall thickening, and fibrotic change in streptozotocin-induced diabetic mice and antibiotic-induced gut dysbiosis mice compared with controls. In both animal models, the NF-κB signaling pathway was activated in the lungs. Enhanced pulmonary alveolar well thickening and fibrotic change appeared in the lungs of transgenic mice expressing a constitutively active NF-κB mutant compared with wild type. When lincomycin hydrochloride-induced gut dysbiosis was ameliorated by fecal microbiota transplant, enhanced inflammatory response in the intestine and pulmonary fibrotic change in the lungs were significantly decreased compared with lincomycin hydrochloride-treated mice. Furthermore, the application of fecal microbiota transplant and baicalin could also redress the microbial dysbiosis of the gut and lungs in streptozotocin-induced diabetic mice. Taken together, these data suggest that multiple as yet undefined factors related to microbial dysbiosis of gut and lungs cause pulmonary fibrogenesis associated with diabetes mellitus through an NF-κB signaling pathway.
Collapse
|
11
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
12
|
Vila Ellis L, Chen J. A cell-centric view of lung alveologenesis. Dev Dyn 2020; 250:482-496. [PMID: 33169483 PMCID: PMC8140604 DOI: 10.1002/dvdy.271] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/30/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Lung alveologenesis, formation of the alveolar region, allows sufficient gas exchange surface to be packed inside the chest cavity yet with orderly connection to the trachea. The real-life alveolar region, however, bears little resemblance to idealized cartoons owing to its three-dimensional nature, nonuniform shape, and mostly air-filled void. This morphological complexity is matched by its cellular complexity-comprised of intermixed and often tangled cells of the epithelial, mesenchymal, endothelial, and immune lineages. Modern imaging, genetics, and genomics are shedding light on and updating traditional views of alveologenesis. Accordingly, this review describes a cell-centric 3-phase definition of alveologenesis and discusses its failure in diseases and possible reactivation during regeneration.
Collapse
Affiliation(s)
- Lisandra Vila Ellis
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
13
|
Abstract
Abstract
Background
Organisms show an incredibly diverse array of body and organ shapes that are both unique to their taxon and important for adapting to their environment. Achieving these specific shapes involves coordinating the many processes that transform single cells into complex organs, and regulating their growth so that they can function within a fully-formed body.
Main text
Conceptually, body and organ shape can be separated in two categories, although in practice these categories need not be mutually exclusive. Body shape results from the extent to which organs, or parts of organs, grow relative to each other. The patterns of relative organ size are characterized using allometry. Organ shape, on the other hand, is defined as the geometric features of an organ’s component parts excluding its size. Characterization of organ shape is frequently described by the relative position of homologous features, known as landmarks, distributed throughout the organ. These descriptions fall into the domain of geometric morphometrics.
Conclusion
In this review, we discuss the methods of characterizing body and organ shape, the developmental programs thought to underlie each, highlight when and how the mechanisms regulating body and organ shape might overlap, and provide our perspective on future avenues of research.
Collapse
|
14
|
Sucre JMS, Vickers KC, Benjamin JT, Plosa EJ, Jetter CS, Cutrone A, Ransom M, Anderson Z, Sheng Q, Fensterheim BA, Ambalavanan N, Millis B, Lee E, Zijlstra A, Königshoff M, Blackwell TS, Guttentag SH. Hyperoxia Injury in the Developing Lung Is Mediated by Mesenchymal Expression of Wnt5A. Am J Respir Crit Care Med 2020; 201:1249-1262. [PMID: 32023086 DOI: 10.1164/rccm.201908-1513oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rationale: Bronchopulmonary dysplasia (BPD) is a leading complication of preterm birth that affects infants born in the saccular stage of lung development at <32 weeks of gestation. Although the mechanisms driving BPD remain uncertain, exposure to hyperoxia is thought to contribute to disease pathogenesis.Objectives: To determine the effects of hyperoxia on epithelial-mesenchymal interactions and to define the mediators of activated Wnt/β-catenin signaling after hyperoxia injury.Methods: Three hyperoxia models were used: A three-dimensional organotypic coculture using primary human lung cells, precision-cut lung slices (PCLS), and a murine in vivo hyperoxia model. Comparisons of normoxia- and hyperoxia-exposed samples were made by real-time quantitative PCR, RNA in situ hybridization, quantitative confocal microscopy, and lung morphometry.Measurements and Main Results: Examination of an array of Wnt ligands in the three-dimensional organotypic coculture revealed increased mesenchymal expression of WNT5A. Inhibition of Wnt5A abrogated the BPD transcriptomic phenotype induced by hyperoxia. In the PCLS model, Wnt5A inhibition improved alveolarization following hyperoxia exposure, and treatment with recombinant Wnt5a reproduced features of the BPD phenotype in PCLS cultured in normoxic conditions. Chemical inhibition of NF-κB with BAY11-7082 reduced Wnt5a expression in the PCLS hyperoxia model and in vivo mouse hyperoxia model, with improved alveolarization in the PCLS model.Conclusions: Increased mesenchymal Wnt5A during saccular-stage hyperoxia injury contributes to the impaired alveolarization and septal thickening observed in BPD. Precise targeting of Wnt5A may represent a potential therapeutic strategy for the treatment of BPD.
Collapse
Affiliation(s)
- Jennifer M S Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics.,Department of Cell and Developmental Biology, and
| | | | - John T Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Erin J Plosa
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | | | - Alissa Cutrone
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | - Benjamin A Fensterheim
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan Millis
- Department of Cell and Developmental Biology, and.,Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Ethan Lee
- Department of Cell and Developmental Biology, and
| | | | - Melanie Königshoff
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado; and
| | - Timothy S Blackwell
- Department of Cell and Developmental Biology, and.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | | |
Collapse
|
15
|
Sun Y, Xia Y, Liu X, Liu J, He W, Ye H, Yuan X. Dexmedetomidine alleviates LPS‑induced acute lung injury via regulation of the p38/HO‑1 pathway. Mol Med Rep 2020; 22:2442-2450. [PMID: 32705282 PMCID: PMC7411448 DOI: 10.3892/mmr.2020.11330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is a common critical illness in clinical anesthesia and the intensive care unit that can cause acute hypoxic respiratory insufficiency. Despite various therapeutic regimes having been investigated, there is currently no effective pharmacotherapy available to treat ALI. Previous studies have reported that the NOD-like receptor protein 3 (NLRP3) signaling pathway plays an important role in the inflammatory response and is involved in the pathogenesis of ALI. Moreover, dexmedetomidine (Dex), an α2-adrenergic receptor activating agent, has been routinely used as an adjuvant therapy in treating inflammatory diseases, including ALI. However, the precise pathological mechanisms of Dex in ALI remain to be elucidated. Thus, the present study aimed to investigate the effects of the p38/heme oxygenase 1 (HO-1) signaling pathways in the pathological mechanisms of Dex in ALI. Newborn male Sprague-Dawley rats (n=48) were randomly divided into four groups (n=12 each), and an intravenous injection of lipopolysaccharide (LPS) was used to successfully induce the ALI model, with increased pulmonary damage, cell apoptosis, interleukin-1β (IL-1β) secretion and edema fluid in lungs. Moreover, the mRNA and protein expression levels of NLRP3 were significantly upregulated, while that of HO-1 were downregulated by LPS treatment. Furthermore, the levels of phosphorylated p38 were also upregulated in ALI rats. It was demonstrated that Dex administration significantly alleviated LPS-induced ALI, downregulated the secretion of IL-1β, decreased the expression of NLRP3, inhibited the phospho-activation of p38 and increased HO-1 expression. In addition, pharmacological inhibition of p38 using the inhibitor SB20380 further enhanced the effect of Dex. Collectively, these preliminarily results identified the effects of Dex intervention on the pathogenesis of ALI via the regulation of p38/HO-1 signaling pathways, which impacted the inflammatory effects, thus providing a theoretical basis and novel evidence for the development of new targets for clinical treatment of ALI.
Collapse
Affiliation(s)
- Yingying Sun
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Yin Xia
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xinghui Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Junxia Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Weitian He
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Hongwu Ye
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xianren Yuan
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| |
Collapse
|
16
|
Bridges JP, Sudha P, Lipps D, Wagner A, Guo M, Du Y, Brown K, Filuta A, Kitzmiller J, Stockman C, Chen X, Weirauch MT, Jobe AH, Whitsett JA, Xu Y. Glucocorticoid regulates mesenchymal cell differentiation required for perinatal lung morphogenesis and function. Am J Physiol Lung Cell Mol Physiol 2020; 319:L239-L255. [PMID: 32460513 DOI: 10.1152/ajplung.00459.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
While antenatal glucocorticoids are widely used to enhance lung function in preterm infants, cellular and molecular mechanisms by which glucocorticoid receptor (GR) signaling influences lung maturation remain poorly understood. Deletion of the glucocorticoid receptor gene (Nr3c1) from fetal pulmonary mesenchymal cells phenocopied defects caused by global Nr3c1 deletion, while lung epithelial- or endothelial-specific Nr3c1 deletion did not impair lung function at birth. We integrated genome-wide gene expression profiling, ATAC-seq, and single cell RNA-seq data in mice in which GR was deleted or activated to identify the cellular and molecular mechanisms by which glucocorticoids control prenatal lung maturation. GR enhanced differentiation of a newly defined proliferative mesenchymal progenitor cell (PMP) into matrix fibroblasts (MFBs), in part by directly activating extracellular matrix-associated target genes, including Fn1, Col16a4, and Eln and by modulating VEGF, JAK-STAT, and WNT signaling. Loss of mesenchymal GR signaling blocked fibroblast progenitor differentiation into mature MFBs, which in turn increased proliferation of SOX9+ alveolar epithelial progenitor cells and inhibited differentiation of mature alveolar type II (AT2) and AT1 cells. GR signaling controls genes required for differentiation of a subset of proliferative mesenchymal progenitors into matrix fibroblasts, in turn, regulating signals controlling AT2/AT1 progenitor cell proliferation and differentiation and identifying cells and processes by which glucocorticoid signaling regulates fetal lung maturation.
Collapse
Affiliation(s)
- James P Bridges
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Parvathi Sudha
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dakota Lipps
- College of Engineering and Applied Science, University of Cincinnati, Cincinnati, Ohio
| | - Andrew Wagner
- College of Engineering and Applied Science, University of Cincinnati, Cincinnati, Ohio
| | - Minzhe Guo
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Yina Du
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kari Brown
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alyssa Filuta
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph Kitzmiller
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Courtney Stockman
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Matthew T Weirauch
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alan H Jobe
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Jeffrey A Whitsett
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
17
|
Mereness JA, Bhattacharya S, Ren Y, Wang Q, Anderson CS, Donlon K, Dylag AM, Haak J, Angelin A, Bonaldo P, Mariani TJ. Collagen VI Deficiency Results in Structural Abnormalities in the Mouse Lung. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:426-441. [PMID: 31837950 PMCID: PMC7013274 DOI: 10.1016/j.ajpath.2019.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/16/2019] [Accepted: 10/11/2019] [Indexed: 01/14/2023]
Abstract
Collagen VI (COL6) is known for its role in a spectrum of congenital muscular dystrophies, which are often accompanied by respiratory dysfunction. However, little is known regarding the function of COL6 in the lung. We confirmed the presence of COL6 throughout the basement membrane region of mouse lung tissue. Lung structure and organization were studied in a previously described Col6a1-/- mouse, which does not produce detectable COL6 in the lung. The Col6a1-/- mouse displayed histopathologic alveolar and airway abnormalities. The airspaces of Col6a1-/- lungs appeared simplified, with larger (29%; P < 0.01) and fewer (31%; P < 0.001) alveoli. These airspace abnormalities included reduced isolectin B4+ alveolar capillaries and surfactant protein C-positive alveolar epithelial type-II cells. Alterations in lung function consistent with these histopathologic changes were evident. Col6a1-/- mice also displayed multiple airway changes, including increased branching (59%; P < 0.001), increased mucosal thickness (34%; P < 0.001), and increased epithelial cell density (13%; P < 0.001). Comprehensive transcriptome analysis revealed that the loss of COL6 is associated with reductions in integrin-paxillin-phosphatidylinositol 3-kinase signaling in vivo. In vitro, COL6 promoted steady-state phosphorylated paxillin levels and reduced cell density (16% to 28%; P < 0.05) at confluence. Inhibition of phosphatidylinositol 3-kinase, or its downstream effectors, resulted in increased cell density to a level similar to that seen on matrices lacking COL6.
Collapse
Affiliation(s)
- Jared A Mereness
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Yue Ren
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Qian Wang
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Christopher S Anderson
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Kathy Donlon
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Andrew M Dylag
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Jeannie Haak
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York
| | - Alessia Angelin
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York.
| |
Collapse
|
18
|
Plosa EJ, Benjamin JT, Sucre JM, Gulleman PM, Gleaves LA, Han W, Kook S, Polosukhin VV, Haake SM, Guttentag SH, Young LR, Pozzi A, Blackwell TS, Zent R. β1 Integrin regulates adult lung alveolar epithelial cell inflammation. JCI Insight 2020; 5:129259. [PMID: 31873073 PMCID: PMC7098727 DOI: 10.1172/jci.insight.129259] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/12/2019] [Indexed: 01/04/2023] Open
Abstract
Integrins, the extracellular matrix receptors that facilitate cell adhesion and migration, are necessary for organ morphogenesis; however, their role in maintaining adult tissue homeostasis is poorly understood. To define the functional importance of β1 integrin in adult mouse lung, we deleted it after completion of development in type 2 alveolar epithelial cells (AECs). Aged β1 integrin-deficient mice exhibited chronic obstructive pulmonary disease-like (COPD-like) pathology characterized by emphysema, lymphoid aggregates, and increased macrophage infiltration. These histopathological abnormalities were preceded by β1 integrin-deficient AEC dysfunction such as excessive ROS production and upregulation of NF-κB-dependent chemokines, including CCL2. Genetic deletion of the CCL2 receptor, Ccr2, in mice with β1 integrin-deficient type 2 AECs impaired recruitment of monocyte-derived macrophages and resulted in accelerated inflammation and severe premature emphysematous destruction. The lungs exhibited reduced AEC efferocytosis and excessive numbers of inflamed type 2 AECs, demonstrating the requirement for recruited monocytes/macrophages in limiting lung injury and remodeling in the setting of a chronically inflamed epithelium. These studies support a critical role for β1 integrin in alveolar homeostasis in the adult lung.
Collapse
Affiliation(s)
| | | | | | | | - Linda A. Gleaves
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | - Wei Han
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | | | - Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | - Scott M. Haake
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | | - Lisa R. Young
- Division of Pulmonary Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ambra Pozzi
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
- Division of Nephrology and Hypertension, Department of Medicine
- Department of Molecular Physiology and Biophysics, and
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
- Division of Nephrology and Hypertension, Department of Medicine
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Evren E, Ringqvist E, Willinger T. Origin and ontogeny of lung macrophages: from mice to humans. Immunology 2019; 160:126-138. [PMID: 31715003 DOI: 10.1111/imm.13154] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Macrophages are tissue-resident myeloid cells with essential roles in host defense, tissue repair, and organ homeostasis. The lung harbors a large number of macrophages that reside in alveoli. As a result of their strategic location, alveolar macrophages are critical sentinels of healthy lung function and barrier immunity. They phagocytose inhaled material and initiate protective immune responses to pathogens, while preventing excessive inflammatory responses and tissue damage. Apart from alveolar macrophages, other macrophage populations are found in the lung and recent single-cell RNA-sequencing studies indicate that lung macrophage heterogeneity is greater than previously appreciated. The cellular origin and development of mouse lung macrophages has been extensively studied, but little is known about the ontogeny of their human counterparts, despite the importance of macrophages for lung health. In this context, humanized mice (mice with a human immune system) can give new insights into the biology of human lung macrophages by allowing in vivo studies that are not possible in humans. In particular, we have created humanized mouse models that support the development of human lung macrophages in vivo. In this review, we will discuss the heterogeneity, development, and homeostasis of lung macrophages. Moreover, we will highlight the impact of age, the microbiota, and pathogen exposure on lung macrophage function. Altered macrophage function has been implicated in respiratory infections as well as in common allergic and inflammatory lung diseases. Therefore, understanding the functional heterogeneity and ontogeny of lung macrophages should help to develop future macrophage-based therapies for important lung diseases in humans.
Collapse
Affiliation(s)
- Elza Evren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emma Ringqvist
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tim Willinger
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
McCarthy R, Martin-Fairey C, Sojka DK, Herzog ED, Jungheim ES, Stout MJ, Fay JC, Mahendroo M, Reese J, Herington JL, Plosa EJ, Shelton EL, England SK. Mouse models of preterm birth: suggested assessment and reporting guidelines. Biol Reprod 2019; 99:922-937. [PMID: 29733339 PMCID: PMC6297318 DOI: 10.1093/biolre/ioy109] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/30/2018] [Indexed: 02/03/2023] Open
Abstract
Preterm birth affects approximately 1 out of every 10 births in the United States, leading to high rates of mortality and long-term negative health consequences. To investigate the mechanisms leading to preterm birth so as to develop prevention strategies, researchers have developed numerous mouse models of preterm birth. However, the lack of standard definitions for preterm birth in mice limits our field's ability to compare models and make inferences about preterm birth in humans. In this review, we discuss numerous mouse preterm birth models, propose guidelines for experiments and reporting, and suggest markers that can be used to assess whether pups are premature or mature. We argue that adoption of these recommendations will enhance the utility of mice as models for preterm birth.
Collapse
Affiliation(s)
- Ronald McCarthy
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carmel Martin-Fairey
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dorothy K Sojka
- Rheumatology Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Erik D Herzog
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Emily S Jungheim
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Molly J Stout
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Justin C Fay
- Department of Biology, University of Rochester, Rochester, New York, USA
| | - Mala Mahendroo
- Department of Obstetrics and Gynecology University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jennifer L Herington
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Erin J Plosa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sarah K England
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
21
|
Liu J, Zhou Y, Liu Y, Li L, Chen Y, Liu Y, Feng Y, Yosypiv IV, Song R, Peng H. (Pro)renin receptor regulates lung development via the Wnt/β-catenin signaling pathway. Am J Physiol Lung Cell Mol Physiol 2019; 317:L202-L211. [PMID: 31042081 PMCID: PMC6734386 DOI: 10.1152/ajplung.00295.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/01/2019] [Accepted: 04/28/2019] [Indexed: 11/22/2022] Open
Abstract
The (pro)renin receptor [(P)RR] binds to prorenin to activate the renin-angiotensin system and is essential for the development of many different organ systems. Whether the (P)RR also plays a role in lung development is unknown. Immunostaining was used to determine the spatial-temporal distribution of (P)RR in the embryonic, postnatal, and adult lungs. We created a lung-specific (P)RR knockout mouse [Foxd1cre/+-(P)RRflox/flox] and assessed changes in lung morphology, cell proliferation, and apoptosis using immunohistochemistry and TUNEL staining. (P)RR function was confirmed by using siRNA to knock down (P)RR in human bronchial epithelial cells (HBECs) and then using the CCK-8 assay and flow cytometry to assess cell proliferation and apoptosis. Gene expression changes after knockdown were assessed by RT-PCR and Western blotting. (P)RR is expressed in the club cells of the bronchial epithelium, and expression increases throughout development. Lung-specific (P)RR knockout disrupted branching morphogenesis, leading to lung hypoplasia and neonatal mortality. These defects were associated with increased apoptosis and decreased proliferation of the pulmonary epithelial and mesenchymal cells and may be mediated by downregulation of Wnt11, β-catenin, and Axin2. (P)RR regulates lung development through canonical Wnt/β-catenin signaling and may present a new target for strategies to treat lung hypoplasia.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafan Zhou
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yalan Liu
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Chen
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Liu
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumei Feng
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, Nevada
| | - Ihor V Yosypiv
- Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Renfang Song
- Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Hua Peng
- Department of Pediatrics, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Thotakura S, Basova L, Makarenkova HP. FGF Gradient Controls Boundary Position Between Proliferating and Differentiating Cells and Regulates Lacrimal Gland Growth Dynamics. Front Genet 2019; 10:362. [PMID: 31191595 PMCID: PMC6546953 DOI: 10.3389/fgene.2019.00362] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling plays an important role in controlling cell proliferation, survival, and cell movements during branching morphogenesis of many organs. In mammals branching morphogenesis is primarily regulated by members of the FGF7-subfamily (FGF7 and FGF10), which are expressed in the mesenchyme, and signal to the epithelial cells through the “b” isoform of fibroblast growth factor receptor-2 (FGFR2). Our previous work demonstrated that FGF7 and FGF10 form different gradients in the extracellular matrix (ECM) and induce distinct cellular responses and gene expression profiles in the lacrimal and submandibular glands. The last finding was the most surprising since both FGF7 and FGF10 bind signal most strongly through the same fibroblast growth factor receptor-2b isoform (FGFR2b). Here we revisit this question to gain an explanation of how the different FGFs regulate gene expression. For this purpose, we employed our ex vivo epithelial explant migration assay in which isolated epithelial explants are grown near the FGF loaded beads. We demonstrate that the graded distribution of FGF induces activation of ERK1/2 MAP kinases that define the position of the boundary between proliferating “bud” and differentiating “stalk” cells of growing lacrimal gland epithelium. Moreover, we showed that gene expression profiles of the epithelial explants exposed to distinct FGFs strictly depend on the ratio between “bud” and “stalk” area. Our data also suggests that differentiation of “stalk” and “bud” regions within the epithelial explants is necessary for directional and persistent epithelial migration. Gaining a better understanding of FGF functions is important for development of new approaches to enhance tissue regeneration.
Collapse
Affiliation(s)
- Suharika Thotakura
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| | - Liana Basova
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, CA, United States
| |
Collapse
|
23
|
Kemmotsu T, Yokoyama U, Saito J, Ito S, Uozumi A, Nishimaki S, Iwasaki S, Seki K, Ito S, Ishikawa Y. Antenatal Administration of Betamethasone Contributes to Intimal Thickening of the Rat Ductus Arteriosus. Circ J 2019; 83:654-661. [PMID: 30726804 DOI: 10.1253/circj.cj-18-1033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Antenatal betamethasone (BMZ) is a standard therapy for reducing respiratory distress syndrome in preterm infants. Recently, some reports have indicated that BMZ promotes ductus arteriosus (DA) closure. DA closure requires morphological remodeling; that is, intimal thickening (IT) formation; however, the role of BMZ in IT formation has not yet been reported. METHODS AND RESULTS First, DNA microarray analysis using smooth muscle cells (SMCs) of rat preterm DA on gestational day 20 (pDASMCs) stimulated with BMZ was performed. Among 58,717 probe sets, ADP-ribosyltransferase 3 (Art3) was markedly increased by BMZ stimulation. Quantitative reverse transcription polymerase chain reaction (RT-PCR) confirmed the BMZ-induced increase of Art3 in pDASMCs, but not in aortic SMCs. Immunocytochemistry showed that BMZ stimulation increased lamellipodia formation. BMZ significantly increased total paxillin protein expression and the ratio of phosphorylated to total paxillin. A scratch assay demonstrated that BMZ stimulation promoted pDASMC migration, which was attenuated byArt3-targeted siRNAs transfection. pDASMC proliferation was not promoted by BMZ, which was analyzed by a 5'-bromo-2'-deoxyuridine (BrdU) assay. Whether BMZ increased IT formation in vivo was examined. BMZ or saline was administered intravenously to maternal rats on gestational days 18 and 19, and DA tissues were obtained on gestational day 20. The ratio of IT to tunica media was significantly higher in the BMZ-treated group. CONCLUSIONS These data suggest that antenatal BMZ administration promotes DA IT through Art3-mediated DASMC migration.
Collapse
Affiliation(s)
- Takahiro Kemmotsu
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University
- Cardiovascular Research Institute, Yokohama City University
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University
| | - Junichi Saito
- Cardiovascular Research Institute, Yokohama City University
| | - Satoko Ito
- Cardiovascular Research Institute, Yokohama City University
| | - Azusa Uozumi
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University
| | - Shigeru Nishimaki
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University
| | - Shiho Iwasaki
- Perinatal Center, Yokohama City University Medical Center
| | - Kazuo Seki
- Perinatal Center, Yokohama City University Medical Center
| | - Shuichi Ito
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University
| | | |
Collapse
|
24
|
Volckaert T, Yuan T, Yuan J, Boateng E, Hopkins S, Zhang JS, Thannickal VJ, Fässler R, De Langhe SP. Hippo signaling promotes lung epithelial lineage commitment by curbing Fgf10 and β-catenin signaling. Development 2019; 146:146/2/dev166454. [PMID: 30651296 DOI: 10.1242/dev.166454] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/07/2018] [Indexed: 12/25/2022]
Abstract
Organ growth and tissue homeostasis rely on the proliferation and differentiation of progenitor cell populations. In the developing lung, localized Fgf10 expression maintains distal Sox9-expressing epithelial progenitors and promotes basal cell differentiation in the cartilaginous airways. Mesenchymal Fgf10 expression is induced by Wnt signaling but inhibited by Shh signaling, and epithelial Fgf10 signaling activates β-catenin signaling. The Hippo pathway is a well-conserved signaling cascade that regulates organ size and stem/progenitor cell behavior. Here, we show that Hippo signaling promotes lineage commitment of lung epithelial progenitors by curbing Fgf10 and β-catenin signaling. Our findings show that both inactivation of the Hippo pathway (nuclear Yap) or ablation of Yap result in increased β-catenin and Fgf10 signaling, suggesting a cytoplasmic role for Yap in epithelial lineage commitment. We further demonstrate redundant and non-redundant functions for the two nuclear effectors of the Hippo pathway, Yap and Taz, during lung development.
Collapse
Affiliation(s)
- Thomas Volckaert
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Tingting Yuan
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Jie Yuan
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Eistine Boateng
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Seantel Hopkins
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Jin-San Zhang
- School of Pharmaceutical Sciences and the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.,Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Victor J Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Stijn P De Langhe
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, AL 35294, USA
| |
Collapse
|
25
|
Hewlett JC, Kropski JA, Blackwell TS. Idiopathic pulmonary fibrosis: Epithelial-mesenchymal interactions and emerging therapeutic targets. Matrix Biol 2018; 71-72:112-127. [PMID: 29625182 PMCID: PMC6146058 DOI: 10.1016/j.matbio.2018.03.021] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic disease of the lung that is marked by progressive decline in pulmonary function and ultimately respiratory failure. Genetic and environmental risk factors have been identified that indicate injury to, and dysfunction of the lung epithelium is central to initiating the pathogenic process. Following injury to the lung epithelium, growth factors, matrikines and extracellular matrix driven signaling together activate a variety of repair pathways that lead to inflammatory cell recruitment, fibroblast proliferation and expansion of the extracellular matrix, culminating in tissue fibrosis. This tissue fibrosis then leads to changes in the biochemical and biomechanical properties of the extracellular matrix, which potentiate profibrotic mechanisms through a "feed-forward cycle." This review provides an overview of the interactions of the pathogenic mechanisms of IPF with a focus on epithelial-mesenchymal crosstalk and the extracellular matrix as a therapeutic target for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin C Hewlett
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States.
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
26
|
Shelton EL, Waleh N, Plosa EJ, Benjamin JT, Milne GL, Hooper CW, Ehinger NJ, Poole S, Brown N, Seidner S, McCurnin D, Reese J, Clyman RI. Effects of antenatal betamethasone on preterm human and mouse ductus arteriosus: comparison with baboon data. Pediatr Res 2018; 84:458-465. [PMID: 29976969 PMCID: PMC6258329 DOI: 10.1038/s41390-018-0006-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/21/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although studies involving preterm infants ≤34 weeks gestation report a decreased incidence of patent ductus arteriosus after antenatal betamethasone, studies involving younger gestation infants report conflicting results. METHODS We used preterm baboons, mice, and humans (≤276/7 weeks gestation) to examine betamethasone's effects on ductus gene expression and constriction both in vitro and in vivo. RESULTS In mice, betamethasone increased the sensitivity of the premature ductus to the contractile effects of oxygen without altering the effects of other contractile or vasodilatory stimuli. Betamethasone's effects on oxygen sensitivity could be eliminated by inhibiting endogenous prostaglandin/nitric oxide signaling. In mice and baboons, betamethasone increased the expression of several developmentally regulated genes that mediate oxygen-induced constriction (K+ channels) and inhibit vasodilator signaling (phosphodiesterases). In human infants, betamethasone increased the rate of ductus constriction at all gestational ages. However, in infants born ≤256/7 weeks gestation, betamethasone's contractile effects were only apparent when prostaglandin signaling was inhibited, whereas at 26-27 weeks gestation, betamethasone's contractile effects were apparent even in the absence of prostaglandin inhibitors. CONCLUSIONS We speculate that betamethasone's contractile effects may be mediated through genes that are developmentally regulated. This could explain why betamethasone's effects vary according to the infant's developmental age at birth.
Collapse
Affiliation(s)
- Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nahid Waleh
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Erin J Plosa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John T Benjamin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ginger L Milne
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher W Hooper
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Noah J Ehinger
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stanley Poole
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naoko Brown
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Steven Seidner
- Department of Pediatrics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Donald McCurnin
- Department of Pediatrics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ronald I Clyman
- Departments of Pediatrics and Cardiovascular Research Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
27
|
Long Y, Wang G, Li K, Zhang Z, Zhang P, Zhang J, Zhang X, Bao Y, Yang X, Wang P. Oxidative stress and NF-κB signaling are involved in LPS induced pulmonary dysplasia in chick embryos. Cell Cycle 2018; 17:1757-1771. [PMID: 30010471 DOI: 10.1080/15384101.2018.1496743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inflammation or dysbacteriosis-derived lipopolysaccharides (LPS) adversely influence the embryonic development of respiratory system. However, the precise pathological mechanisms still remain to be elucidated. In this study, we demonstrated that LPS exposure caused lung maldevelopment in chick embryos, including higher embryo mortality, increased thickness of alveolar gas exchange zone, and accumulation of PAS+ immature pulmonary cells, accompanied with reduced expression of alveolar epithelial cell markers and lamellar body count. Upon LPS exposure, pulmonary cell proliferation was significantly altered and cell apoptosis was inhibited as well, indicating a delayed progress of pulmonary development. LPS treatment also resulted in reduced CAV-1 expression and up-regulation of Collagen I, suggesting increased lung fibrosis, which was verified by Masson staining. Moreover, LPS induced enhanced Nrf2 expression in E18 lungs, and the increased reactive oxygen species (ROS) production was confirmed in MLE-12 cells in vitro. Antioxidant vitamin C restored the LPS induced down-regulation of ABCA3, SP-C and GATA-6 in MLE-12 cells. Furthermore, LPS induced activation of NF-κB signaling in MLE-12 cells, and the LPS-induced decrease in SP-C expression was partially abrogated by blocking NF-κB signaling with Bay-11-7082. Bay-11-7082 also inhibited LPS-induced increases of ROS and Nrf2 expression. Taken together, we have demonstrated that oxidative stress and NF-κB signaling are involved in LPS induced disruption of pulmonary cell development in chick embryos.
Collapse
Affiliation(s)
- Yun Long
- a Department of Microbiology and Immunology, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Guang Wang
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Ke Li
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Zongyi Zhang
- a Department of Microbiology and Immunology, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Ping Zhang
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Jing Zhang
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Xiaotan Zhang
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Yongping Bao
- c Norwich Medical School , University of East Anglia , Norwich , Norfolk , UK
| | - Xuesong Yang
- b Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| | - Pengcheng Wang
- a Department of Microbiology and Immunology, School of Basic Medical Sciences , Jinan University , Guangzhou , China
| |
Collapse
|
28
|
Liu C, Yuan L, Zou Y, Yang M, Chen Y, Qu X, Liu H, Jiang J, Xiang Y, Qin X. ITGB4 is essential for containing HDM-induced airway inflammation and airway hyperresponsiveness. J Leukoc Biol 2018; 103:897-908. [PMID: 29393977 DOI: 10.1002/jlb.3a1017-411rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Airway epithelial cells play a significant role in the pathogenesis of asthma. Although the structural and functional defects of airway epithelial cells have been postulated to increase asthma susceptibility and exacerbate asthma severity, the mechanism and implication of these defects remain uncertain. Integrin β4 (ITGB4) is a structural adhesion molecule that is downregulated in the airway epithelium of asthma patients. In this study, we demonstrated that ITGB4 deficiency leads to severe allergy-induced airway inflammation and airway hyper-responsiveness (AHR) in mice. After house dust mite (HDM) challenge, epithelial cell-specific ITGB4-deleted mice showed increased lymphocyte, eosinophil, and neutrophil infiltration into lung compared with that of the wild-type mice. ITGB4 deficiency also resulted in increased expression of the Th2 cytokine IL-4, IL-13, and the Th17 cytokine IL-17A in the lung tissue and in the T cells after HDM challenge. The aggravated inflammation in ITGB4 defect mice was partly caused by enhanced disrupted epithelial barrier integrity after HDM stress, which induced the increased thymic stromal lymphopoietin secretion from airway epithelial cells. This study therefore demonstrates that ITGB4 plays a pivotal role in containing allergen-mediated lung inflammation and airway hyper-responsiveness in allergic asthma.
Collapse
Affiliation(s)
- Chi Liu
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yizhou Zou
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Yu Chen
- Department of Examination, Medical College of Hunan Normal University, Changsha, Hunan, China
| | - Xiangping Qu
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Huijun Liu
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Surgery Research, Third Military Medical University, Chongqing, China
| | - Yang Xiang
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoqun Qin
- Departments of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
29
|
He MY, Wang G, Han SS, Li K, Jin Y, Liu M, Si ZP, Wang J, Liu GS, Yang X. Negative impact of hyperglycaemia on mouse alveolar development. Cell Cycle 2017; 17:80-91. [PMID: 29143549 DOI: 10.1080/15384101.2017.1403683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus in pregnancy has been known to affect the embryonic development of various systems, including cardiovascular and nervous systems. However, whether this disease could have a negative impact on embryonic respiratory system remains controversial. In this study, we demonstrated that pregestational diabetes mellitus (PGDM)-induced defects in lung development in mice are mainly characterized by the changes in the morphological structure of the lung. Immunostaining and Western blotting showed that proliferation increased and apoptosis decreased in PGDM. Hyperglycaemia caused pulmonary tissue fibrationas manifested by an increase in Masson staining and decorin expression in PGDM lungs, and the immunofluorescent pro-SPC+ type II pulmonary epithelial cell number was decreased. The alteration of pulmonary epithelial cell differentiation might be due to hyperglycaemia-activated Wnt signalling and suppressed GATA6 expression in PGDM mouse lung tissues and MLE-12 cells. The treatment of MLE-12 cells with high glucose in the presence/absence of XAV939 or su5402 further proved that hyperglycaemia suppressed the expression of GATA6 and pro-SPC by activating Wnt signalling and induced the expression of decorin, α-SMA and TGF-β by activating Fgf signalling. Therefore, in this study, we revealed that hyperglycemia induced dysfunctional pulmonary cell apoptosis and proliferation, as well as pulmonary myofibroblast hyperplasia, which contributed to the formation of aberrant structure of alveolar walls. Furthermore, the hyperglycaemia also inhibited the differentiation of pulmonary epithelial cells through the canonical Wnt and Fgf signalling, and the alteration of Fgf and Wnt signalling activated TGF-β, which would promote the AECII EMT process.
Collapse
Affiliation(s)
- Mei-Yao He
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Guang Wang
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Sha-Sha Han
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Ke Li
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Ya Jin
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Meng Liu
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| | - Zhen-Peng Si
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Ju Wang
- c Institute of Biomedicine , National Engineering Research Center of Genetic Medicine , Jinan University , Guangzhou 510632 , China
| | - Guo-Sheng Liu
- a Department of Pediatrics and Neonatology , Institute of Fetal-Preterm Labor Medicine ; The First Affiliated Hospital , Jinan University , Guangzhou 510630 , China
| | - Xuesong Yang
- b Division of Histology & Embryology , Key Laboratory for Regenerative Medicine of the Ministry of Education , Medical College , Jinan University , Guangzhou 510632 , China
| |
Collapse
|
30
|
Desgrange A, Heliot C, Skovorodkin I, Akram SU, Heikkilä J, Ronkainen VP, Miinalainen I, Vainio SJ, Cereghini S. HNF1B controls epithelial organization and cell polarity during ureteric bud branching and collecting duct morphogenesis. Development 2017; 144:4704-4719. [PMID: 29158444 DOI: 10.1242/dev.154336] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022]
Abstract
Kidney development depends crucially on proper ureteric bud branching giving rise to the entire collecting duct system. The transcription factor HNF1B is required for the early steps of ureteric bud branching, yet the molecular and cellular events regulated by HNF1B are poorly understood. We report that specific removal of Hnf1b from the ureteric bud leads to defective cell-cell contacts and apicobasal polarity during the early branching events. High-resolution ex vivo imaging combined with a membranous fluorescent reporter strategy show decreased mutant cell rearrangements during mitosis-associated cell dispersal and severe epithelial disorganization. Molecular analysis reveals downregulation of Gdnf-Ret pathway components and suggests that HNF1B acts both upstream and downstream of Ret signaling by directly regulating Gfra1 and Etv5 Subsequently, Hnf1b deletion leads to massively mispatterned ureteric tree network, defective collecting duct differentiation and disrupted tissue architecture, which leads to cystogenesis. Consistently, mRNA-seq analysis shows that the most impacted genes encode intrinsic cell-membrane components with transporter activity. Our study uncovers a fundamental and recurring role of HNF1B in epithelial organization during early ureteric bud branching and in further patterning and differentiation of the collecting duct system in mouse.
Collapse
Affiliation(s)
- Audrey Desgrange
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Claire Heliot
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France.,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Ilya Skovorodkin
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Saad U Akram
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | - Janne Heikkilä
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | | | | | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Silvia Cereghini
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| |
Collapse
|
31
|
He H, Huang M, Sun S, Wu Y, Lin X. Epithelial heparan sulfate regulates Sonic Hedgehog signaling in lung development. PLoS Genet 2017; 13:e1006992. [PMID: 28859094 PMCID: PMC5597256 DOI: 10.1371/journal.pgen.1006992] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 09/13/2017] [Accepted: 08/21/2017] [Indexed: 12/23/2022] Open
Abstract
The tree-like structure of the mammalian lung is generated from branching morphogenesis, a reiterative process that is precisely regulated by numerous factors. How the cell surface and extra cellular matrix (ECM) molecules regulate this process is still poorly understood. Herein, we show that epithelial deletion of Heparan Sulfate (HS) synthetase Ext1 resulted in expanded branching tips and reduced branching number, associated with several mesenchymal developmental defects. We further demonstrate an expanded Fgf10 expression and increased FGF signaling activity in Ext1 mutant lungs, suggesting a cell non-autonomous mechanism. Consistent with this, we observed reduced levels of SHH signaling which is responsible for suppressing Fgf10 expression. Moreover, reactivating SHH signaling in mutant lungs rescued the tip dilation phenotype and attenuated FGF signaling. Importantly, the reduced SHH signaling activity did not appear to be caused by decreased Shh expression or protein stability; instead, biologically active form of SHH proteins were reduced in both the Ext1 mutant epithelium and surrounding wild type mesenchymal cells. Together, our study highlights the epithelial HS as a key player for dictating SHH signaling critical for lung morphogenesis.
Collapse
Affiliation(s)
- Hua He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meina Huang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shenfei Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yihui Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- * E-mail: ,
| |
Collapse
|
32
|
Chen CM, Liu YC, Chen YJ, Chou HC. Genome-Wide Analysis of DNA Methylation in Hyperoxia-Exposed Newborn Rat Lung. Lung 2017; 195:661-669. [DOI: 10.1007/s00408-017-0036-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 07/05/2017] [Indexed: 01/23/2023]
|
33
|
Legan SK, Lee DD, Schwarz MA. α5β1 integrin mediates pulmonary epithelial cyst formation. Dev Dyn 2017; 246:475-484. [PMID: 28370666 DOI: 10.1002/dvdy.24502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Formation of the epithelial cyst involves the establishment of apical-basolateral polarity through a series of cellular interactions that are in part mediated by the extracellular matrix (ECM). We report that in a three-dimensional multi-cellular self-assembly model of lung development, α5 integrin regulates epithelial cyst formation through organization of soluble fibronectin matrix into insoluble fibrils through a process called fibrillogenesis. RESULTS Dissociated murine embryonic lung cells self-assemble into three-dimensional pulmonary bodies that are dependent on α5β1 integrin mediated fibrillogenesis for cell-cell mediated self-assembly: compaction and epithelial cyst formation. Knockdown of α5 integrin resulted in a significant increase in another mediator of fibrillogenesis, αV integrin. Compensatory increased expression of another mediator of fibrillogenesis, αV integrin, was not sufficient to normalize epithelial cyst formation. Loss of α5 integrin-mediated fibrillogenesis perturbed the ability of clustered epithelial cells to establish clear polarity, loss of epithelial cell pyramidal shape, and disrupted apical F-actin-rich deposition. Lack of rich central epithelial localization of F-actin cytoskeleton and Podocalyxin suggests that loss of α5 integrin-mediated fibrillogenesis interferes with the normal cytoskeleton organization that facilitates epithelial cysts polarization. CONCLUSIONS We conclude that lung epithelial cyst formation in development is mediated in part by α5β1 integrin dependent fibrillogenesis. Developmental Dynamics 246:475-484, 2016. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Susan K Legan
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - Daniel D Lee
- Department of Pediatrics, Indiana University, South Bend, Indiana
| | | |
Collapse
|
34
|
Epithelial cell integrin β1 is required for developmental angiogenesis in the pituitary gland. Proc Natl Acad Sci U S A 2016; 113:13408-13413. [PMID: 27810956 DOI: 10.1073/pnas.1614970113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
As a key component of the vertebrate neuroendocrine system, the pituitary gland relies on the progressive and coordinated development of distinct hormone-producing cell types and an invading vascular network. The molecular mechanisms that drive formation of the pituitary vasculature, which is necessary for regulated synthesis and secretion of hormones that maintain homeostasis, metabolism, and endocrine function, remain poorly understood. Here, we report that expression of integrin β1 in embryonic pituitary epithelial cells is required for angiogenesis in the developing mouse pituitary gland. Deletion of pituitary epithelial integrin β1 before the onset of angiogenesis resulted in failure of invading endothelial cells to recruit pericytes efficiently, whereas deletion later in embryogenesis led to decreased vascular density and lumen formation. In both cases, lack of epithelial integrin β1 was associated with a complete absence of vasculature in the pituitary gland at birth. Within pituitary epithelial cells, integrin β1 directs a large transcriptional program that includes components of the extracellular matrix and associated signaling factors that are linked to the observed non-cell-autonomous effects on angiogenesis. We conclude that epithelial integrin β1 functions as a critical and canonical regulator of developmental angiogenesis in the pituitary gland, thus providing insight into the long-standing systems biology conundrum of how vascular invasion is coordinated with tissue development.
Collapse
|
35
|
Well-organized spheroids as a new platform to examine cell interaction and behaviour during organ development. Cell Tissue Res 2016; 366:601-615. [PMID: 27599480 DOI: 10.1007/s00441-016-2487-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
We present an experimental method allowing the production of three-dimensional organ-like structures, namely microtissues (MTs), in vitro without the need for exogenous extracellular matrix (ECM) or growth factors. Submandibular salivary glands (embryonic day ED14), kidneys (ED13) and lungs (ED13) were harvested from mouse embryos and dissociated into single cells by enzyme treatment. Single cells were seeded into special hanging drop culture plates (InSphero) and cultured for up to 14 days to obtain MTs. This strategy permitted full control of the quantity of seeded cells. The development of the MTs into organs was followed histologically and immunohistochemically. Well-organized epithelial structures surrounded by a basal lamina were formed, as confirmed by transmission electron microscopy. Expression of E-cadherin, vimentin, fibronectin and α-SMA was compared in organs and corresponding MTs by real-time quantitative polymerase chain reaction. Branching morphogenesis was induced in MTs (as shown by histology and immunostaining for fibronectin and perlecan) and was conserved even after 14 days of culture. MTs continued their development and their epithelial structures were comparable with those of the physiological organ at postnatal day 2 (PN2). Expression of aquaporins was investigated to obtain better support for the functional differentiation of epithelial cells. Histogenesis proceeded and led to the start of organogenesis. This experimental model might improve our knowledge of epithelial-mesenchymal histogenesis and can be employed to study development or cellular organization during the embryonic formation of organs.
Collapse
|
36
|
Corbin JM, Overcash RF, Wren JD, Coburn A, Tipton GJ, Ezzell JA, McNaughton KK, Fung KM, Kosanke SD, Ruiz-Echevarria MJ. Analysis of TMEFF2 allografts and transgenic mouse models reveals roles in prostate regeneration and cancer. Prostate 2016; 76:97-113. [PMID: 26417683 PMCID: PMC4722803 DOI: 10.1002/pros.23103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous results from our lab indicate a tumor suppressor role for the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) in prostate cancer (PCa). Here, we further characterize this role and uncover new functions for TMEFF2 in cancer and adult prostate regeneration. METHODS The role of TMEFF2 was examined in PCa cells using Matrigel(TM) cultures and allograft models of PCa cells. In addition, we developed a transgenic mouse model that expresses TMEFF2 from a prostate specific promoter. Anatomical, histological, and metabolic characterizations of the transgenic mouse prostate were conducted. The effect of TMEFF2 in prostate regeneration was studied by analyzing branching morphogenesis in the TMEFF2-expressing mouse lobes and alterations in branching morphogenesis were correlated with the metabolomic profiles of the mouse lobes. The role of TMEFF2 in prostate tumorigenesis in whole animals was investigated by crossing the TMEFF2 transgenic mice with the TRAMP mouse model of PCa and analyzing the histopathological changes in the progeny. RESULTS Ectopic expression of TMEFF2 impairs growth of PCa cells in Matrigel or allograft models. Surprisingly, while TMEFF2 expression in the TRAMP mouse did not have a significant effect on the glandular prostate epithelial lesions, the double TRAMP/TMEFF2 transgenic mice displayed an increased incidence of neuroendocrine type tumors. In addition, TMEFF2 promoted increased branching specifically in the dorsal lobe of the prostate suggesting a potential role in developmental processes. These results correlated with data indicating an alteration in the metabolic profile of the dorsal lobe of the transgenic TMEFF2 mice. CONCLUSIONS Collectively, our results confirm the tumor suppressor role of TMEFF2 and suggest that ectopic expression of TMEFF2 in mouse prostate leads to additional lobe-specific effects in prostate regeneration and tumorigenesis. This points to a complex and multifunctional role for TMEFF2 during PCa progression.
Collapse
Affiliation(s)
- JM. Corbin
- Department of Pathology, Oklahoma University Health Sciences Center. Oklahoma City, OK, USA
| | - RF. Overcash
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - JD. Wren
- Arthritis and Clinical Immunology Research Program. Oklahoma Medical Research Foundation. Oklahoma City, OK, USA
| | - A. Coburn
- Department of Comparative Medicine. East Carolina University. Greenville, NC, USA
| | - GJ. Tipton
- Bowles Center for Alcohol Studies. University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - JA. Ezzell
- Department of Cell Biology and Physiology. University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - KK. McNaughton
- Department of Cell Biology and Physiology. University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - KM Fung
- Department of Pathology, Oklahoma University Health Sciences Center. Oklahoma City, OK, USA
- Department of Pathology, Oklahoma City Veterans Affairs Medical Center. Oklahoma City, OK, USA
| | - SD. Kosanke
- Department of Pathology, Oklahoma University Health Sciences Center. Oklahoma City, OK, USA
| | - MJ Ruiz-Echevarria
- Department of Pathology, Oklahoma University Health Sciences Center. Oklahoma City, OK, USA
- Stephenson Cancer Center. Oklahoma City, OK, USA
- Correspondence to: MJ. Ruiz-Echevarria, Associate Professor of Pathology, University of Oklahoma Health Sciences Center, Stanton L. Young Biomedical Research Center, 975 N.E. 10th Street, Room 1368A, Oklahoma City, Oklahoma 73104. Phone: (405) 271.1871; Fax: (405) 271.2141.
| |
Collapse
|
37
|
Silva DMG, Nardiello C, Pozarska A, Morty RE. Recent advances in the mechanisms of lung alveolarization and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1239-72. [PMID: 26361876 DOI: 10.1152/ajplung.00268.2015] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/09/2015] [Indexed: 02/08/2023] Open
Abstract
Alveolarization is the process by which the alveoli, the principal gas exchange units of the lung, are formed. Along with the maturation of the pulmonary vasculature, alveolarization is the objective of late lung development. The terminal airspaces that were formed during early lung development are divided by the process of secondary septation, progressively generating an increasing number of alveoli that are of smaller size, which substantially increases the surface area over which gas exchange can take place. Disturbances to alveolarization occur in bronchopulmonary dysplasia (BPD), which can be complicated by perturbations to the pulmonary vasculature that are associated with the development of pulmonary hypertension. Disturbances to lung development may also occur in persistent pulmonary hypertension of the newborn in term newborn infants, as well as in patients with congenital diaphragmatic hernia. These disturbances can lead to the formation of lungs with fewer and larger alveoli and a dysmorphic pulmonary vasculature. Consequently, affected lungs exhibit a reduced capacity for gas exchange, with important implications for morbidity and mortality in the immediate postnatal period and respiratory health consequences that may persist into adulthood. It is the objective of this Perspectives article to update the reader about recent developments in our understanding of the molecular mechanisms of alveolarization and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Diogo M G Silva
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Claudio Nardiello
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Agnieszka Pozarska
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rory E Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
38
|
Abstract
Insulin-producing β cells within the vertebrate fetal pancreas acquire their fate in a step-wise manner. Whereas the intrinsic factors dictating the transcriptional or epigenetic status of pancreatic lineages have been intensely examined, less is known about cell-cell interactions that might constitute a niche for the developing β cell lineage. It is becoming increasingly clear that understanding and recapitulating these steps may instruct in vitro differentiation of embryonic stem cells and/or therapeutic regeneration. Indeed, directed differentiation techniques have improved since transitioning from 2D to 3D cultures, suggesting that the 3D microenvironment in which β cells are born is critical. However, to date, it remains unknown whether the changing architecture of the pancreatic epithelium impacts the fate of cells therein. An emerging challenge in the field is to elucidate how progenitors are allocated during key events, such as the stratification and subsequent resolution of the pre-pancreatic epithelium, as well as the formation of lumens and branches. Here, we assess the progenitor epithelium and examine how it might influence the emergence of pancreatic multipotent progenitors (MPCs), which give rise to β cells and other pancreatic lineages.
Collapse
Affiliation(s)
- Leilani Marty-Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas (LMS,OC)
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas (LMS,OC)
| |
Collapse
|
39
|
Plosa EJ, Young LR, Gulleman PM, Polosukhin VV, Zaynagetdinov R, Benjamin JT, Im AM, van der Meer R, Gleaves LA, Bulus N, Han W, Prince LS, Blackwell TS, Zent R. Epithelial β1 integrin is required for lung branching morphogenesis and alveolarization. J Cell Sci 2015. [DOI: 10.1242/jcs.167338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|