1
|
Quach H, Farrell S, Wu MJM, Kanagarajah K, Leung JWH, Xu X, Kallurkar P, Turinsky AL, Bear CE, Ratjen F, Kalish B, Goyal S, Moraes TJ, Wong AP. Early human fetal lung atlas reveals the temporal dynamics of epithelial cell plasticity. Nat Commun 2024; 15:5898. [PMID: 39003323 PMCID: PMC11246468 DOI: 10.1038/s41467-024-50281-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 07/05/2024] [Indexed: 07/15/2024] Open
Abstract
Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.
Collapse
Affiliation(s)
- Henry Quach
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Spencer Farrell
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Ming Jia Michael Wu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kayshani Kanagarajah
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Wai-Hin Leung
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqiao Xu
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Prajkta Kallurkar
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrei L Turinsky
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christine E Bear
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian Kalish
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sidhartha Goyal
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Wei Y, Han S, Wen J, Liao J, Liang J, Yu J, Chen X, Xiang S, Huang Z, Zhang B. E26 transformation-specific transcription variant 5 in development and cancer: modification, regulation and function. J Biomed Sci 2023; 30:17. [PMID: 36872348 PMCID: PMC9987099 DOI: 10.1186/s12929-023-00909-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
E26 transformation-specific (ETS) transcription variant 5 (ETV5), also known as ETS-related molecule (ERM), exerts versatile functions in normal physiological processes, including branching morphogenesis, neural system development, fertility, embryonic development, immune regulation, and cell metabolism. In addition, ETV5 is repeatedly found to be overexpressed in multiple malignant tumors, where it is involved in cancer progression as an oncogenic transcription factor. Its roles in cancer metastasis, proliferation, oxidative stress response and drug resistance indicate that it is a potential prognostic biomarker, as well as a therapeutic target for cancer treatment. Post-translational modifications, gene fusion events, sophisticated cellular signaling crosstalk and non-coding RNAs contribute to the dysregulation and abnormal activities of ETV5. However, few studies to date systematically summarized the role and molecular mechanisms of ETV5 in benign diseases and in oncogenic progression. In this review, we specify the molecular structure and post-translational modifications of ETV5. In addition, its critical roles in benign and malignant diseases are summarized to draw a panorama for specialists and clinicians. The updated molecular mechanisms of ETV5 in cancer biology and tumor progression are delineated. Finally, we prospect the further direction of ETV5 research in oncology and its potential translational applications in the clinic.
Collapse
Affiliation(s)
- Yi Wei
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenqi Han
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyuan Wen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shuai Xiang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
3
|
Ikonomou L, Yampolskaya M, Mehta P. Multipotent Embryonic Lung Progenitors: Foundational Units of In Vitro and In Vivo Lung Organogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:49-70. [PMID: 37195526 PMCID: PMC10351616 DOI: 10.1007/978-3-031-26625-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Transient, tissue-specific, embryonic progenitors are important cell populations in vertebrate development. In the course of respiratory system development, multipotent mesenchymal and epithelial progenitors drive the diversification of fates that results to the plethora of cell types that compose the airways and alveolar space of the adult lungs. Use of mouse genetic models, including lineage tracing and loss-of-function studies, has elucidated signaling pathways that guide proliferation and differentiation of embryonic lung progenitors as well as transcription factors that underlie lung progenitor identity. Furthermore, pluripotent stem cell-derived and ex vivo expanded respiratory progenitors offer novel, tractable, high-fidelity systems that allow for mechanistic studies of cell fate decisions and developmental processes. As our understanding of embryonic progenitor biology deepens, we move closer to the goal of in vitro lung organogenesis and resulting applications in developmental biology and medicine.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Department of Oral Biology, University at Buffalo, The State University of New York, Buffalo, NY, USA.
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, The State University of New York, Buffalo, NY, USA.
- Cell, Gene and Tissue Engineering Center, University at Buffalo, The State University of New York, Buffalo, NY, USA.
| | | | - Pankaj Mehta
- Department of Physics, Boston University, Boston, MA, USA
- Faculty of Computing and Data Science, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| |
Collapse
|
4
|
Ali M, LaCanna R, Lian Z, Huang J, Tan Y, Shao W, Yu X, Tian Y. Transcriptional responses to injury of regenerative lung alveolar epithelium. iScience 2022; 25:104843. [PMID: 35996586 PMCID: PMC9391595 DOI: 10.1016/j.isci.2022.104843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/01/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
The significance of alveolar epithelial type 2 (AT2) cell proliferation for lung alveolar epithelial homeostasis and regeneration after injury has been widely accepted. However, the heterogeneity of AT2 cell population for cell proliferation capacity remains disputed. By single-cell RNA sequencing and genetic lineage labeling using the Ki67 knock-in mouse model, we map all proliferative AT2 cells in homeostatic and regenerating murine lungs after injury induced by Streptococcus pneumoniae infection. The proliferative AT2 cell population displays a unique transcriptional program, which is regulated by activating transcription factor 3 (ATF3) and thyroid hormone receptor alpha (THRA) transcription factors. Overexpression of these two transcription factors in AT2 cells promoted AT2 cell proliferation and improved lung function after injury. These results indicate that increased expression of ATF3 and THRA at the onset of lung epithelial regeneration is required to permit rapid AT2 cell proliferation and hence progression through the recovery of lung epithelium.
Collapse
Affiliation(s)
- Mir Ali
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ryan LaCanna
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Zhaorui Lian
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Yinfei Tan
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Wenna Shao
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiang Yu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
5
|
Orstad G, Fort G, Parnell TJ, Jones A, Stubben C, Lohman B, Gillis KL, Orellana W, Tariq R, Klingbeil O, Kaestner K, Vakoc CR, Spike BT, Snyder EL. FoxA1 and FoxA2 control growth and cellular identity in NKX2-1-positive lung adenocarcinoma. Dev Cell 2022; 57:1866-1882.e10. [PMID: 35835117 PMCID: PMC9378547 DOI: 10.1016/j.devcel.2022.06.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/11/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Changes in cellular identity (also known as histologic transformation or lineage plasticity) can drive malignant progression and resistance to therapy in many cancers, including lung adenocarcinoma (LUAD). The lineage-specifying transcription factors FoxA1 and FoxA2 (FoxA1/2) control identity in NKX2-1/TTF1-negative LUAD. However, their role in NKX2-1-positive LUAD has not been systematically investigated. We find that Foxa1/2 knockout severely impairs tumorigenesis in KRAS-driven genetically engineered mouse models and human cell lines. Loss of FoxA1/2 leads to the collapse of a dual-identity state, marked by co-expression of pulmonary and gastrointestinal transcriptional programs, which has been implicated in LUAD progression. Mechanistically, FoxA1/2 loss leads to aberrant NKX2-1 activity and genomic localization, which in turn actively inhibits tumorigenesis and drives alternative cellular identity programs that are associated with non-proliferative states. This work demonstrates that FoxA1/2 expression is a lineage-specific vulnerability in NKX2-1-positive LUAD and identifies mechanisms of response and resistance to targeting FoxA1/2 in this disease.
Collapse
Affiliation(s)
- Grace Orstad
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Gabriela Fort
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Timothy J Parnell
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alex Jones
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Chris Stubben
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Brian Lohman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Katherine L Gillis
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Walter Orellana
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Rushmeen Tariq
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Olaf Klingbeil
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Klaus Kaestner
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Benjamin T Spike
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Eric L Snyder
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA; Department of Pathology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
6
|
Lin Y, Wang D, Zeng Y. A Maverick Review of Common Stem/Progenitor Markers in Lung Development. Stem Cell Rev Rep 2022; 18:2629-2645. [DOI: 10.1007/s12015-022-10422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 10/16/2022]
|
7
|
Okada D, Okamoto Y, Io T, Oka M, Kobayashi D, Ito S, Yamada R, Ishii K, Ono K. Comparative Study of Transcriptome in the Hearts Isolated from Mice, Rats, and Humans. Biomolecules 2022; 12:biom12060859. [PMID: 35740984 PMCID: PMC9221511 DOI: 10.3390/biom12060859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023] Open
Abstract
The heart is a significant organ in mammalian life, and the heartbeat mechanism has been an essential focus of science. However, few studies have focused on species differences. Accordingly, challenges remain in studying genes that have universal functions across species and genes that determine species differences. Here, we analyzed transcriptome data in mouse, rat, and human atria, ventricles, and sinoatrial nodes (SA) obtained from different platforms and compared them by calculating specificity measure (SPM) values in consideration of species differences. Among the three heart regions, the species differences in SA were the greatest, and we searched for genes that determined the essential characteristics of SA, which was SHOX2 in our criteria. The SPM value of SHOX2 was prominently high across species. Similarly, by calculating SPM values, we identified 3 atrial-specific, 11 ventricular-specific, and 17 SA-specific markers. Ontology analysis identified 70 cardiac region- and species-specific ontologies. These results suggest that reanalyzing existing data by calculating SPM values may identify novel tissue-specific genes and species-dependent gene expression. This study identified the importance of SHOX2 as an SA-specific transcription factor, a novel cardiac regional marker, and species-dependent ontologies.
Collapse
Affiliation(s)
- Daigo Okada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Shogoinkawahara-cho, Kyoto 606-8507, Japan; (D.O.); (R.Y.)
| | - Yosuke Okamoto
- Department of Cell Physiology, Akita Graduate School of Medicine, Hondo, Akita 010-8543, Japan; (D.K.); (S.I.); (K.O.)
- Correspondence:
| | - Toshiro Io
- Research Department, Ono Pharmaceutical Co., Ltd., Kyutaromachi, Osaka 618-8585, Japan; (T.I.); (M.O.)
| | - Miho Oka
- Research Department, Ono Pharmaceutical Co., Ltd., Kyutaromachi, Osaka 618-8585, Japan; (T.I.); (M.O.)
| | - Daiki Kobayashi
- Department of Cell Physiology, Akita Graduate School of Medicine, Hondo, Akita 010-8543, Japan; (D.K.); (S.I.); (K.O.)
| | - Suzuka Ito
- Department of Cell Physiology, Akita Graduate School of Medicine, Hondo, Akita 010-8543, Japan; (D.K.); (S.I.); (K.O.)
| | - Ryo Yamada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Shogoinkawahara-cho, Kyoto 606-8507, Japan; (D.O.); (R.Y.)
| | - Kuniaki Ishii
- Department of Pharmacology, Faculty of medicine, Yamagata University, Iida-Nishi, Yamagata 990-9585, Japan;
| | - Kyoichi Ono
- Department of Cell Physiology, Akita Graduate School of Medicine, Hondo, Akita 010-8543, Japan; (D.K.); (S.I.); (K.O.)
| |
Collapse
|
8
|
Age-dependent alveolar epithelial plasticity orchestrates lung homeostasis and regeneration. Cell Stem Cell 2021; 28:1775-1789.e5. [PMID: 33974915 PMCID: PMC8500919 DOI: 10.1016/j.stem.2021.04.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/11/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023]
Abstract
Regeneration of the architecturally complex alveolar niche of the lung requires precise temporal and spatial control of epithelial cell behavior. Injury can lead to a permanent reduction in gas exchange surface area and respiratory function. Using mouse models, we show that alveolar type 1 (AT1) cell plasticity is a major and unappreciated mechanism that drives regeneration, beginning in the early postnatal period during alveolar maturation. Upon acute neonatal lung injury, AT1 cells reprogram into alveolar type 2 (AT2) cells, promoting alveolar regeneration. In contrast, the ability of AT2 cells to regenerate AT1 cells is restricted to the mature lung. Unbiased genomic assessment reveals that this previously unappreciated level of plasticity is governed by the preferential activity of Hippo signaling in the AT1 cell lineage. Thus, cellular plasticity is a temporally acquired trait of the alveolar epithelium and presents an alternative mode of tissue regeneration in the postnatal lung.
Collapse
|
9
|
Chu X, Taghizadeh S, Vazquez-Armendariz AI, Herold S, Chong L, Chen C, Zhang JS, El Agha E, Bellusci S. Validation of a Novel Fgf10 Cre-ERT2 Knock-in Mouse Line Targeting FGF10 Pos Cells Postnatally. Front Cell Dev Biol 2021; 9:671841. [PMID: 34055804 PMCID: PMC8155496 DOI: 10.3389/fcell.2021.671841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 01/14/2023] Open
Abstract
Fgf10 is a key gene during development, homeostasis and repair after injury. We previously reported a knock-in Fgf10 Cre-ERT2 line (with the Cre-ERT2 cassette inserted in frame with the start codon of exon 1), called thereafter Fgf10 Ki-v1, to target FGF10Pos cells. While this line allowed fairly efficient and specific labeling of FGF10Pos cells during the embryonic stage, it failed to target these cells after birth, particularly in the postnatal lung, which has been the focus of our research. We report here the generation and validation of a new knock-in Fgf10 Cre-ERT2 line (called thereafter Fgf10 Ki-v2) with the insertion of the expression cassette in frame with the stop codon of exon 3. Fgf10 Ki-v2/+ heterozygous mice exhibited comparable Fgf10 expression levels to wild type animals. However, a mismatch between Fgf10 and Cre expression levels was observed in Fgf10 Ki-v2/+ lungs. In addition, lung and limb agenesis were observed in homozygous embryos suggesting a loss of Fgf10 functional allele in Fgf10 Ki-v2 mice. Bioinformatic analysis shows that the 3'UTR, where the Cre-ERT2 cassette is inserted, contains numerous putative transcription factor binding sites. By crossing this line with tdTomato reporter line, we demonstrated that tdTomato expression faithfully recapitulated Fgf10 expression during development. Importantly, Fgf10 Ki-v2 mouse is capable of significantly targeting FGF10Pos cells in the adult lung. Therefore, despite the aforementioned limitations, this new Fgf10 Ki-v2 line opens the way for future mechanistic experiments involving the postnatal lung.
Collapse
Affiliation(s)
- Xuran Chu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Sara Taghizadeh
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Ana Ivonne Vazquez-Armendariz
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Lei Chong
- National Key Clinical Specialty of Pediatric Respiratory Medicine, Discipline of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jin-San Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Elie El Agha
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Saverio Bellusci
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
10
|
TP63 basal cells are indispensable during endoderm differentiation into proximal airway cells on acellular lung scaffolds. NPJ Regen Med 2021; 6:12. [PMID: 33674599 PMCID: PMC7935966 DOI: 10.1038/s41536-021-00124-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/01/2021] [Indexed: 12/24/2022] Open
Abstract
The use of decellularized whole-organ scaffolds for bioengineering of organs is a promising avenue to circumvent the shortage of donor organs for transplantation. However, recellularization of acellular scaffolds from multicellular organs like the lung with a variety of different cell types remains a challenge. Multipotent cells could be an ideal cell source for recellularization. Here we investigated the hierarchical differentiation process of multipotent ES-derived endoderm cells into proximal airway epithelial cells on acellular lung scaffolds. The first cells to emerge on the scaffolds were TP63+ cells, followed by TP63+/KRT5+ basal cells, and finally multi-ciliated and secretory airway epithelial cells. TP63+/KRT5+ basal cells on the scaffolds simultaneously expressed KRT14, like basal cells involved in airway repair after injury. Removal of TP63 by CRISPR/Cas9 in the ES cells halted basal and airway cell differentiation on the scaffolds. These findings suggest that differentiation of ES-derived endoderm cells into airway cells on decellularized lung scaffolds proceeds via TP63+ basal cell progenitors and tracks a regenerative repair pathway. Understanding the process of differentiation is key for choosing the cell source for repopulation of a decellularized organ scaffold. Our data support the use of airway basal cells for repopulating the airway side of an acellular lung scaffold.
Collapse
|
11
|
Kuwahara A, Lewis AE, Coombes C, Leung FS, Percharde M, Bush JO. Delineating the early transcriptional specification of the mammalian trachea and esophagus. eLife 2020; 9:e55526. [PMID: 32515350 PMCID: PMC7282815 DOI: 10.7554/elife.55526] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
The genome-scale transcriptional programs that specify the mammalian trachea and esophagus are unknown. Though NKX2-1 and SOX2 are hypothesized to be co-repressive master regulators of tracheoesophageal fates, this is untested at a whole transcriptomic scale and their downstream networks remain unidentified. By combining single-cell RNA-sequencing with bulk RNA-sequencing of Nkx2-1 mutants and NKX2-1 ChIP-sequencing in mouse embryos, we delineate the NKX2-1 transcriptional program in tracheoesophageal specification, and discover that the majority of the tracheal and esophageal transcriptome is NKX2-1 independent. To decouple the NKX2-1 transcriptional program from regulation by SOX2, we interrogate the expression of newly-identified tracheal and esophageal markers in Sox2/Nkx2-1 compound mutants. Finally, we discover that NKX2-1 binds directly to Shh and Wnt7b and regulates their expression to control mesenchymal specification to cartilage and smooth muscle, coupling epithelial identity with mesenchymal specification. These findings create a new framework for understanding early tracheoesophageal fate specification at the genome-wide level.
Collapse
Affiliation(s)
- Akela Kuwahara
- Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
- Department of Cell and Tissue Biology, University of California San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
- Developmental and Stem Cell Biology Graduate Program, University of California San FranciscoSan FranciscoUnited States
| | - Ace E Lewis
- Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
- Department of Cell and Tissue Biology, University of California San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
| | - Coohleen Coombes
- Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
- Department of Cell and Tissue Biology, University of California San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
- Department of Biology, San Francisco State UniversitySan FranciscoUnited States
| | - Fang-Shiuan Leung
- Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
- Department of Cell and Tissue Biology, University of California San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
| | - Michelle Percharde
- MRC London Institute of Medical Sciences (LMS)LondonUnited Kingdom
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College LondonLondonUnited Kingdom
| | - Jeffrey O Bush
- Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
- Department of Cell and Tissue Biology, University of California San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
12
|
Presence of N-acetylglucosamine residues on the surface coating of bronchioloalveolar cells during rat postnatal development: What is their purpose? Acta Histochem 2019; 121:119-124. [PMID: 30448021 DOI: 10.1016/j.acthis.2018.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/26/2018] [Accepted: 10/24/2018] [Indexed: 01/03/2023]
Abstract
Mammalian lung development is a complex process that is partially accomplished during the postnatal period. Surface carbohydrates are crucial in many biological and pathological phenomena and are key partners during development. The outer surface of lung epithelial cells, which is rich in carbohydrate components, plays a pivotal role throughout the developmental process. However, systematic studies on the sugar residue content of the cell surface coating during postnatal rat lung development are scarce. The aim of the present study was to identify and determine the localization of N-acetylglucosamine residues on the bronchioloalveolar cell surface during rat lung development using light and pre-embedding transmission electron microscopy methodologies, and to associate these data with the components underlying postnatal lung growth. Strong binding sites for the lectin Triticum vulgare (common name Wheat Germ, WGA) are present on the luminal surface of adult rat bronchioloalveolar cells throughout the entire postnatal period and have been identified as N-acetylglucosamine residues. The consistent positive reaction observed on the surface coating of bronchioloalveolar lining cells before and after neuraminidase treatment suggests that aside from possible terminal sialic acids, the lectin specificity for N-acetylglucosamine residues is still evident. Our results also suggest a stronger positive reaction on the bronchioloalveolar cell surface when compared with endothelial cell surface. N-acetylglucosamine residues for lectin binding can be present in glycoproteins in the membrane and also within heparin sulfate chains of glycosaminoglycans, which are crucial for lung development. The work described here has sought to highlight the presence and possible importance of N-acetylglucosamine residues on the glycocalyx of bronchioloalveolar cells, during postnatal lung development.
Collapse
|
13
|
Jones MR, Dilai S, Lingampally A, Chao CM, Danopoulos S, Carraro G, Mukhametshina R, Wilhelm J, Baumgart-Vogt E, Al Alam D, Chen C, Minoo P, Zhang JS, Bellusci S. A Comprehensive Analysis of Fibroblast Growth Factor Receptor 2b Signaling on Epithelial Tip Progenitor Cells During Early Mouse Lung Branching Morphogenesis. Front Genet 2019; 9:746. [PMID: 30728831 PMCID: PMC6351499 DOI: 10.3389/fgene.2018.00746] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/27/2018] [Indexed: 01/10/2023] Open
Abstract
This study demonstrates that FGF10/FGFR2b signaling on distal epithelial progenitor cells, via ß-catenin/EP300, controls, through a comprehensive set of developmental genes, morphogenesis, and differentiation. Fibroblast growth factor (FGF) 10 signaling through FGF receptor 2b (FGFR2b) is mandatory during early lung development as the deletion of either the ligand or the receptor leads to lung agenesis. However, this drastic phenotype previously hampered characterization of the primary biological activities, immediate downstream targets and mechanisms of action. Through the use of a dominant negative transgenic mouse model (Rosa26rtTA; tet(o)sFgfr2b), we conditionally inhibited FGF10 signaling in vivo in E12.5 embryonic lungs via doxycycline IP injection to pregnant females, and in vitro by culturing control and experimental lungs with doxycycline. The impact on branching morphogenesis 9 h after doxycycline administration was analyzed by morphometry, fluorescence and electron microscopy. Gene arrays at 6 and 9 h following doxycycline administration were carried out. The relationship between FGF10 and ß-catenin signaling was also analyzed through in vitro experiments using IQ1, a pharmacological inhibitor of ß-catenin/EP300 transcriptional activity. Loss of FGF10 signaling did not impact proliferation or survival, but affected both adherens junctions (up-regulation of E-cadherin), and basement membrane organization (increased laminin). Gene arrays identified multiple direct targets of FGF10, including main transcription factors. Immunofluorescence showed a down-regulation of the distal epithelial marker SOX9 and mis-expression distally of the proximal marker SOX2. Staining for the transcriptionally-active form of ß-catenin showed a reduction in experimental vs. control lungs. In vitro experiments using IQ1 phenocopied the impacts of blocking FGF10. This study demonstrates that FGF10/FGFR2b signaling on distal epithelial progenitor cells via ß-catenin/EP300 controls, through a comprehensive set of developmental genes, cell adhesion, and differentiation.
Collapse
Affiliation(s)
- Matthew R Jones
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Salma Dilai
- Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Arun Lingampally
- Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Cho-Ming Chao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Soula Danopoulos
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles and University of Southern California, Los Angeles, CA, United States
| | - Gianni Carraro
- Department of Medicine, Cedars-Sinai Medical Center, Lung and Regenerative Medicine Institutes, Los Angeles, CA, United States
| | - Regina Mukhametshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Jochen Wilhelm
- Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany
| | - Denise Al Alam
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles and University of Southern California, Los Angeles, CA, United States
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Parviz Minoo
- Division of Newborn Medicine, Department of Pediatrics, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Jin San Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Life Sciences, Wenzhou University, Zhejiang, China.,International Collaborative Research Center on Growth Factors, Wenzhou Medical University, Zhejiang, China
| | - Saverio Bellusci
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Internal Medicine II, Member of the German Lung Center, Excellence Cluster Cardio-Pulmonary Systems, University of Giessen Lung Center, Giessen, Germany.,Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles and University of Southern California, Los Angeles, CA, United States.,Institute of Life Sciences, Wenzhou University, Zhejiang, China.,International Collaborative Research Center on Growth Factors, Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
14
|
Li Q, Jiao J, Li H, Wan H, Zheng C, Cai J, Bao S. Histone arginine methylation by Prmt5 is required for lung branching morphogenesis through repression of BMP signaling. J Cell Sci 2018; 131:jcs.217406. [PMID: 29950483 DOI: 10.1242/jcs.217406] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/19/2018] [Indexed: 12/17/2022] Open
Abstract
Branching morphogenesis is essential for the successful development of a functional lung to accomplish its gas exchange function. Although many studies have highlighted requirements for the bone morphogenetic protein (BMP) signaling pathway during branching morphogenesis, little is known about how BMP signaling is regulated. Here, we report that the protein arginine methyltransferase 5 (Prmt5) and symmetric dimethylation at histone H4 arginine 3 (H4R3sme2) directly associate with chromatin of Bmp4 to suppress its transcription. Inactivation of Prmt5 in the lung epithelium results in halted branching morphogenesis, altered epithelial cell differentiation and neonatal lethality. These defects are accompanied by increased apoptosis and reduced proliferation of lung epithelium, as a consequence of elevated canonical BMP-Smad1/5/9 signaling. Inhibition of BMP signaling by Noggin rescues the lung branching defects of Prmt5 mutant in vitro Taken together, our results identify a novel mechanism through which Prmt5-mediated histone arginine methylation represses canonical BMP signaling to regulate lung branching morphogenesis.
Collapse
Affiliation(s)
- Qiuling Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Jie Jiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huijun Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.,School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huajing Wan
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of the Ministry of Education, West China Institute of Women and Children's Health, and Department of Pediatrics, Huaxi Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Caihong Zheng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Jun Cai
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Shilai Bao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China .,School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
15
|
He Y, Zuo Q, Edwards J, Zhao K, Lei J, Cai W, Nie Q, Li B, Song J. DNA Methylation and Regulatory Elements during Chicken Germline Stem Cell Differentiation. Stem Cell Reports 2018; 10:1793-1806. [PMID: 29681542 PMCID: PMC5989647 DOI: 10.1016/j.stemcr.2018.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
The production of germ cells in vitro would open important new avenues for stem biology and human medicine, but the mechanisms of germ cell differentiation are not well understood. The chicken, as a great model for embryology and development, was used in this study to help us explore its regulatory mechanisms. In this study, we reported a comprehensive genome-wide DNA methylation landscape in chicken germ cells, and transcriptomic dynamics was also presented. By uncovering DNA methylation patterns on individual genes, some genes accurately modulated by DNA methylation were found to be associated with cancers and virus infection, e.g., AKT1 and CTNNB1. Chicken-unique markers were also discovered for identifying male germ cells. Importantly, integrated epigenetic mechanisms were explored during male germ cell differentiation, which provides deep insight into the epigenetic processes associated with male germ cell differentiation and possibly improves treatment options to male infertility in animals and humans. The mechanisms of stem cell differentiation were explored using the chick embryo model The orchestrated stem cell differentiation involves multiple epigenetic events The unique markers in chick embryo were discovered for identifying male germ cells
Collapse
Affiliation(s)
- Yanghua He
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Qisheng Zuo
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009, People's Republic of China
| | - John Edwards
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8220, St. Louis, MO 63110, USA
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinzhi Lei
- Zhou Peiyuan Center for Applied Mathematics, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Wentao Cai
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA; Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, CA 92697-3875, USA
| | - Bichun Li
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009, People's Republic of China.
| | - Jiuzhou Song
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
16
|
Mao Z, Shi Y, Cao Q, Chen Y, Sun Y, Liu Z, Zhang Q, Huang M. Transcriptional regulation on the gene expression signature in combined allergic rhinitis and asthma syndrome. Epigenomics 2018; 10:119-131. [PMID: 29334241 DOI: 10.2217/epi-2017-0072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM This study was intended to evaluate transcriptional regulation of gene expression signatures in combined allergic rhinitis and asthma syndrome (CARAS). MATERIALS & METHODS The blood samples of three patients with CARAS, three patients with allergic rhinitis and three normal controls were obtained. The cuffdiff, miRDeep2 and DEGseq were used to quantify the expression of genes and miRNAs, respectively. And p-value < 0.01 and false discovery rate < 0.001 were considered as significant differences of genes and miRNAs, respectively. Gene ontology and the Kyoto Encyclopedia of Genes and Genomes were used to analyze the biological function. And the cut-off value for significance was p < 0.05. RESULTS SLC14A1, SNCA, TNS1, KAT2B and PARP1 were regulated by hsa-miR-93-5p, hsa-miR-92a-3p and hsa-miR-21-5p. Additionally, phagosome (p = 0.00627769839083361) was the only significantly enriched signal pathway involving HLA-DOA, TUBB2A and MRC2. CONCLUSION Disordered expression of genes under the regulation of miRNAs may play an important role in CARAS.
Collapse
Affiliation(s)
- Zhengdao Mao
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China.,Departmentof Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yujia Shi
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Qi Cao
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Yi Chen
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Yun Sun
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Zhiguang Liu
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory Medicine, Affiliated Changzhou No. 2 People's Hospital, Nanjing Medical University, Changzhou, China
| | - Mao Huang
- Departmentof Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
17
|
Landry-Truchon K, Houde N, Boucherat O, Joncas FH, Dasen JS, Philippidou P, Mansfield JH, Jeannotte L. HOXA5 plays tissue-specific roles in the developing respiratory system. Development 2017; 144:3547-3561. [PMID: 28827394 DOI: 10.1242/dev.152686] [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] [Received: 03/26/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022]
Abstract
Hoxa5 is essential for development of several organs and tissues. In the respiratory system, loss of Hoxa5 function causes neonatal death due to respiratory distress. Expression of HOXA5 protein in mesenchyme of the respiratory tract and in phrenic motor neurons of the central nervous system led us to address the individual contribution of these Hoxa5 expression domains using a conditional gene targeting approach. Hoxa5 does not play a cell-autonomous role in lung epithelium, consistent with lack of HOXA5 expression in this cell layer. In contrast, ablation of Hoxa5 in mesenchyme perturbed trachea development, lung epithelial cell differentiation and lung growth. Further, deletion of Hoxa5 in motor neurons resulted in abnormal diaphragm innervation and musculature, and lung hypoplasia. It also reproduced the neonatal lethality observed in null mutants, indicating that the defective diaphragm is the main cause of impaired survival at birth. Thus, Hoxa5 possesses tissue-specific functions that differentially contribute to the morphogenesis of the respiratory tract.
Collapse
Affiliation(s)
- Kim Landry-Truchon
- Centre de Recherche sur le Cancer de l'Université Laval, CRCHU de Québec, L'Hôtel-Dieu de Québec, Québec G1R 3S3, Canada
| | - Nicolas Houde
- Centre de Recherche sur le Cancer de l'Université Laval, CRCHU de Québec, L'Hôtel-Dieu de Québec, Québec G1R 3S3, Canada
| | - Olivier Boucherat
- Centre de Recherche sur le Cancer de l'Université Laval, CRCHU de Québec, L'Hôtel-Dieu de Québec, Québec G1R 3S3, Canada
| | - France-Hélène Joncas
- Centre de Recherche sur le Cancer de l'Université Laval, CRCHU de Québec, L'Hôtel-Dieu de Québec, Québec G1R 3S3, Canada
| | - Jeremy S Dasen
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10036, USA
| | - Polyxeni Philippidou
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10036, USA
| | - Jennifer H Mansfield
- Department of Biology, Barnard College-Columbia University, New York, NY 10027, USA
| | - Lucie Jeannotte
- Centre de Recherche sur le Cancer de l'Université Laval, CRCHU de Québec, L'Hôtel-Dieu de Québec, Québec G1R 3S3, Canada
| |
Collapse
|
18
|
Nikolić MZ, Caritg O, Jeng Q, Johnson JA, Sun D, Howell KJ, Brady JL, Laresgoiti U, Allen G, Butler R, Zilbauer M, Giangreco A, Rawlins EL. Human embryonic lung epithelial tips are multipotent progenitors that can be expanded in vitro as long-term self-renewing organoids. eLife 2017; 6. [PMID: 28665271 PMCID: PMC5555721 DOI: 10.7554/elife.26575] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
The embryonic mouse lung is a widely used substitute for human lung development. For example, attempts to differentiate human pluripotent stem cells to lung epithelium rely on passing through progenitor states that have only been described in mouse. The tip epithelium of the branching mouse lung is a multipotent progenitor pool that self-renews and produces differentiating descendants. We hypothesized that the human distal tip epithelium is an analogous progenitor population and tested this by examining morphology, gene expression and in vitro self-renewal and differentiation capacity of human tips. These experiments confirm that human and mouse tips are analogous and identify signalling pathways that are sufficient for long-term self-renewal of human tips as differentiation-competent organoids. Moreover, we identify mouse-human differences, including markers that define progenitor states and signalling requirements for long-term self-renewal. Our organoid system provides a genetically-tractable tool that will allow these human-specific features of lung development to be investigated. DOI:http://dx.doi.org/10.7554/eLife.26575.001 Degenerative lung disease occurs when the structure of the lungs breaks down, which makes it harder to get enough oxygen into the bloodstream. Most, but not all, cases occur in smokers and ex-smokers or people who have been exposed to a lot of air pollution. Currently, there is no way to reverse the damage, and even slowing the progress of the disease is extremely difficult. Some researchers are looking for ways to treat patients with degenerative lung diseases by regenerating the surface of their lungs. However, it is still not clear what the most effective route towards this long-term goal will be. One approach to lung regeneration is to use findings from developmental biology to understand how embryos normally build the gas exchange surfaces in the lungs. This knowledge may allow scientists to trigger a similar process in an adult lung to renew or replace any diseased tissue. Alternatively, cells could be collected from patients, reprogrammed and then coaxed into becoming a gas exchange surface in the laboratory. Such a “lung-in-a-dish” could be used to understand how degenerative diseases develop, to discover and test new drugs, or even to treat the patient directly via a transplant. To date, the embryonic development of lungs has mostly been studied using mouse lungs as a model system. However, it was not clear if human lungs actually develop in similar ways to mouse lungs, and whether using mice is a valid research strategy. Nikolić et al. compared embryonic lungs from humans and mice and showed that they are indeed very similar in terms of the cell types that they contain and how they mature. However, some key differences were identified that can only be explored in human cells and tissue. Nikolić et al. went on to identify conditions that allowed them to grow cells from human embryonic lungs indefinitely in a dish. These cells can now be used to investigate the aspects of lung development that are specific to humans. Together these findings provide a useful guide to allow scientists to coax human cells growing in a laboratory to become lung cells. Further improvements to this process will make the lungs-in-a-dish more true to the real organs, meaning that they could be used to better understand lung disease and identify new medicines. In the longer term, Nikolić et al. hope to gain enough insight from the human lung-in-a-dish model to eventually be able to regenerate the lungs of patients with degenerative lung disease. However, this possibility is still many years away. DOI:http://dx.doi.org/10.7554/eLife.26575.002
Collapse
Affiliation(s)
- Marko Z Nikolić
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Oriol Caritg
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Quitz Jeng
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jo-Anne Johnson
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Dawei Sun
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kate J Howell
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Jane L Brady
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Usua Laresgoiti
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - George Allen
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Richard Butler
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Matthias Zilbauer
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom.,Department of Paediatric Gastroenterology, University of Cambridge and Addenbrookes Hospital, Cambridge, United Kingdom
| | - Adam Giangreco
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Emma L Rawlins
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust/MRC Stem Cell Institute, Cambridge, United Kingdom
| |
Collapse
|
19
|
Hawkins F, Kramer P, Jacob A, Driver I, Thomas DC, McCauley KB, Skvir N, Crane AM, Kurmann AA, Hollenberg AN, Nguyen S, Wong BG, Khalil AS, Huang SX, Guttentag S, Rock JR, Shannon JM, Davis BR, Kotton DN. Prospective isolation of NKX2-1-expressing human lung progenitors derived from pluripotent stem cells. J Clin Invest 2017; 127:2277-2294. [PMID: 28463226 DOI: 10.1172/jci89950] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1-expressing (NKX2-1+) precursor cells. However, this hypothesis has not been formally tested owing to an inability to purify or track these progenitors for detailed characterization. Here we have engineered and developmentally differentiated NKX2-1GFP reporter pluripotent stem cells (PSCs) in vitro to generate and isolate human primordial lung progenitors that express NKX2-1 but are initially devoid of differentiated lung lineage markers. After sorting to purity, these primordial lung progenitors exhibited lung epithelial maturation. In the absence of mesenchymal coculture support, this NKX2-1+ population was able to generate epithelial-only spheroids in defined 3D cultures. Alternatively, when recombined with fetal mouse lung mesenchyme, the cells recapitulated epithelial-mesenchymal developing lung interactions. We imaged these progenitors in real time and performed time-series global transcriptomic profiling and single-cell RNA sequencing as they moved through the earliest moments of lung lineage specification. The profiles indicated that evolutionarily conserved, stage-dependent gene signatures of early lung development are expressed in primordial human lung progenitors and revealed a CD47hiCD26lo cell surface phenotype that allows their prospective isolation from untargeted, patient-specific PSCs for further in vitro differentiation and future applications in regenerative medicine.
Collapse
Affiliation(s)
- Finn Hawkins
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Philipp Kramer
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Anjali Jacob
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ian Driver
- Department of Anatomy, UCSF, San Francisco, California, USA
| | | | - Katherine B McCauley
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Ana M Crane
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Anita A Kurmann
- Center for Regenerative Medicine, and.,Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Brandon G Wong
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA
| | - Ahmad S Khalil
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Sarah Xl Huang
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA.,Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA
| | - Susan Guttentag
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital, Vanderbilt University, Nashville, Tennessee, USA
| | - Jason R Rock
- Department of Anatomy, UCSF, San Francisco, California, USA
| | - John M Shannon
- Division of Pulmonary Biology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Brian R Davis
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Transcription factor Etv5 is essential for the maintenance of alveolar type II cells. Proc Natl Acad Sci U S A 2017; 114:3903-3908. [PMID: 28351980 DOI: 10.1073/pnas.1621177114] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Alveolar type II (AT2) cell dysfunction contributes to a number of significant human pathologies including respiratory distress syndrome, lung adenocarcinoma, and debilitating fibrotic diseases, but the critical transcription factors that maintain AT2 cell identity are unknown. Here we show that the E26 transformation-specific (ETS) family transcription factor Etv5 is essential to maintain AT2 cell identity. Deletion of Etv5 from AT2 cells produced gene and protein signatures characteristic of differentiated alveolar type I (AT1) cells. Consistent with a defect in the AT2 stem cell population, Etv5 deficiency markedly reduced recovery following bleomycin-induced lung injury. Lung tumorigenesis driven by mutant KrasG12D was also compromised by Etv5 deficiency. ERK activation downstream of Ras was found to stabilize Etv5 through inactivation of the cullin-RING ubiquitin ligase CRL4COP1/DET1 that targets Etv5 for proteasomal degradation. These findings identify Etv5 as a critical output of Ras signaling in AT2 cells, contributing to both lung homeostasis and tumor initiation.
Collapse
|
21
|
Danopoulos S, Krainock M, Toubat O, Thornton M, Grubbs B, Al Alam D. Rac1 modulates mammalian lung branching morphogenesis in part through canonical Wnt signaling. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1036-L1049. [PMID: 27765763 DOI: 10.1152/ajplung.00274.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/03/2016] [Indexed: 01/06/2023] Open
Abstract
Lung branching morphogenesis relies on a number of factors, including proper epithelial cell proliferation and differentiation, cell polarity, and migration. Rac1, a small Rho GTPase, orchestrates a number of these cellular processes, including cell proliferation and differentiation, cellular alignment, and polarization. Furthermore, Rac1 modulates both noncanonical and canonical Wnt signaling, important pathways in lung branching morphogenesis. Culture of embryonic mouse lung explants in the presence of the Rac1 inhibitor (NSC23766) resulted in a dose-dependent decrease in branching. Increased cell death and BrdU uptake were notably seen in the mesenchyme, while no direct effect on the epithelium was observed. Moreover, vasculogenesis was impaired following Rac1 inhibition as shown by decreased Vegfa expression and impaired LacZ staining in Flk1-Lacz reporter mice. Rac1 inhibition decreased Fgf10 expression in conjunction with many of its associated factors. Moreover, using the reporter lines TOPGAL and Axin2-LacZ, there was an evident decrease in canonical Wnt signaling in the explants treated with the Rac1 inhibitor. Activation of canonical Wnt pathway using WNT3a or WNT7b only partially rescued the branching inhibition. Moreover, these results were validated on human explants, where Rac1 inhibition resulted in impaired branching and decreased AXIN2 and FGFR2b expression. We therefore conclude that Rac1 regulates lung branching morphogenesis, in part through canonical Wnt signaling. However, the exact mechanisms by which Rac1 interacts with canonical Wnt in human and mouse lung requires further investigation.
Collapse
Affiliation(s)
- Soula Danopoulos
- Developmental Biology and Regenerative Medicine Program, Department of Pediatric Surgery, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Michael Krainock
- Developmental Biology and Regenerative Medicine Program, Department of Pediatric Surgery, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Omar Toubat
- Developmental Biology and Regenerative Medicine Program, Department of Pediatric Surgery, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Matthew Thornton
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Denise Al Alam
- Developmental Biology and Regenerative Medicine Program, Department of Pediatric Surgery, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; .,Keck School of Medicine, University of Southern California, Los Angeles, California; and
| |
Collapse
|
22
|
Beauchemin KJ, Wells JM, Kho AT, Philip VM, Kamir D, Kohane IS, Graber JH, Bult CJ. Temporal dynamics of the developing lung transcriptome in three common inbred strains of laboratory mice reveals multiple stages of postnatal alveolar development. PeerJ 2016; 4:e2318. [PMID: 27602285 PMCID: PMC4991849 DOI: 10.7717/peerj.2318] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
To characterize temporal patterns of transcriptional activity during normal lung development, we generated genome wide gene expression data for 26 pre- and post-natal time points in three common inbred strains of laboratory mice (C57BL/6J, A/J, and C3H/HeJ). Using Principal Component Analysis and least squares regression modeling, we identified both strain-independent and strain-dependent patterns of gene expression. The 4,683 genes contributing to the strain-independent expression patterns were used to define a murine Developing Lung Characteristic Subtranscriptome (mDLCS). Regression modeling of the Principal Components supported the four canonical stages of mammalian embryonic lung development (embryonic, pseudoglandular, canalicular, saccular) defined previously by morphology and histology. For postnatal alveolar development, the regression model was consistent with four stages of alveolarization characterized by episodic transcriptional activity of genes related to pulmonary vascularization. Genes expressed in a strain-dependent manner were enriched for annotations related to neurogenesis, extracellular matrix organization, and Wnt signaling. Finally, a comparison of mouse and human transcriptomics from pre-natal stages of lung development revealed conservation of pathways associated with cell cycle, axon guidance, immune function, and metabolism as well as organism-specific expression of genes associated with extracellular matrix organization and protein modification. The mouse lung development transcriptome data generated for this study serves as a unique reference set to identify genes and pathways essential for normal mammalian lung development and for investigations into the developmental origins of respiratory disease and cancer. The gene expression data are available from the Gene Expression Omnibus (GEO) archive (GSE74243). Temporal expression patterns of mouse genes can be investigated using a study specific web resource (http://lungdevelopment.jax.org).
Collapse
Affiliation(s)
- Kyle J. Beauchemin
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, The University of Maine, Orono, ME, United States
| | | | - Alvin T. Kho
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | | | - Daniela Kamir
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Isaac S. Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, United States
| | | | - Carol J. Bult
- The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
23
|
E3 ubiquitin ligase RFWD2 controls lung branching through protein-level regulation of ETV transcription factors. Proc Natl Acad Sci U S A 2016; 113:7557-62. [PMID: 27335464 DOI: 10.1073/pnas.1603310113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mammalian lung is an elaborate branching organ, and it forms following a highly stereotypical morphogenesis program. It is well established that precise control at the transcript level is a key genetic underpinning of lung branching. In comparison, little is known about how regulation at the protein level may play a role. Ring finger and WD domain 2 (RFWD2, also termed COP1) is an E3 ubiquitin ligase that modifies specific target proteins, priming their degradation via the ubiquitin proteasome system. RFWD2 is known to function in the adult in pathogenic processes such as tumorigenesis. Here, we show that prenatal inactivation of Rfwd2 gene in the lung epithelium led to a striking halt in branching morphogenesis shortly after secondary branch formation. This defect is accompanied by distalization of the lung epithelium while growth and cellular differentiation still occurred. In the mutant lung, two E26 transformation-specific (ETS) transcription factors essential for normal lung branching, ETS translocation variant 4 (ETV4) and ETV5, were up-regulated at the protein level, but not at the transcript level. Introduction of Etv loss-of-function alleles into the Rfwd2 mutant background attenuated the branching phenotype, suggesting that RFWD2 functions, at least in part, through degrading ETV proteins. Because a number of E3 ligases are known to target factors important for lung development, our findings provide a preview of protein-level regulatory network essential for lung branching morphogenesis.
Collapse
|
24
|
Makanya AN. Membrane mediated development of the vertebrate blood-gas-barrier. ACTA ACUST UNITED AC 2016; 108:85-97. [PMID: 26991887 DOI: 10.1002/bdrc.21120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/22/2016] [Indexed: 01/24/2023]
Abstract
During embryonic lung development, establishment of the gas-exchanging units is guided by epithelial tubes lined by columnar cells. Ultimately, a thin blood-gas barrier (BGB) is established and forms the interface for efficient gas exchange. This thin BGB is achieved through processes, which entail lowering of tight junctions, stretching, and thinning in mammals. In birds the processes are termed peremerecytosis, if they involve cell squeezing and constriction, or secarecytosis, if they entail cutting cells to size. In peremerecytosis, cells constrict at a point below the protruding apical part, resulting in fusion of the opposing membranes and discharge of the aposome, or the cell may be squeezed by the more endowed cognate neighbors. Secarecytosis may entail formation of double membranes below the aposome, subsequent unzipping and discharge of the aposome, or vesicles form below the aposome, fuse in a bilateral manner, and release the aposome. These processes occur within limited developmental windows, and are mediated through cell membranes that appear to be of intracellular in origin. In addition, basement membranes (BM) play pivotal roles in differentiation of the epithelial and endothelial layers of the BGB. Laminins found in the BM are particularly important in the signaling pathways that result in formation of squamous pneumocytes and pulmonary capillaries, the two major components of the BGB. Some information exists on the contribution by BM to BGB formation, but little is known regarding the molecules that drive peremerecytosis, or even the origins and composition of the double and vesicular membranes involved in secarecytosis.
Collapse
Affiliation(s)
- Andrew N Makanya
- Department of Vet Anatomy and Physiology, Riverside Drive, Chiromo Campus, University of , Box 30197-00100, Nairobi
| |
Collapse
|
25
|
Herriges JC, Verheyden JM, Zhang Z, Sui P, Zhang Y, Anderson MJ, Swing DA, Zhang Y, Lewandoski M, Sun X. FGF-Regulated ETV Transcription Factors Control FGF-SHH Feedback Loop in Lung Branching. Dev Cell 2016; 35:322-32. [PMID: 26555052 DOI: 10.1016/j.devcel.2015.10.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/17/2015] [Accepted: 10/09/2015] [Indexed: 01/13/2023]
Abstract
The mammalian lung forms its elaborate tree-like structure following a largely stereotypical branching sequence. While a number of genes have been identified to play essential roles in lung branching, what coordinates the choice between branch growth and new branch formation has not been elucidated. Here we show that loss of FGF-activated transcription factor genes, Etv4 and Etv5 (collectively Etv), led to prolonged branch tip growth and delayed new branch formation. Unexpectedly, this phenotype is more similar to mutants with increased rather than decreased FGF activity. Indeed, an increased Fgf10 expression is observed, and reducing Fgf10 dosage can attenuate the Etv mutant phenotype. Further evidence indicates that ETV inhibits Fgf10 via directly promoting Shh expression. SHH in turn inhibits local Fgf10 expression and redirects growth, thereby initiating new branches. Together, our findings establish ETV as a key node in the FGF-ETV-SHH inhibitory feedback loop that dictates branching periodicity.
Collapse
Affiliation(s)
- John C Herriges
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jamie M Verheyden
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Zhen Zhang
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Physiological Chemistry, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Pengfei Sui
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ying Zhang
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21702, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21702, USA
| | - Deborah A Swing
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD 21702, USA
| | - Yan Zhang
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21702, USA
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
26
|
Matsuda R, Hosono C, Saigo K, Samakovlis C. The intersection of the extrinsic hedgehog and WNT/wingless signals with the intrinsic Hox code underpins branching pattern and tube shape diversity in the drosophila airways. PLoS Genet 2015; 11:e1004929. [PMID: 25615601 PMCID: PMC4304712 DOI: 10.1371/journal.pgen.1004929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/28/2014] [Indexed: 01/04/2023] Open
Abstract
The tubular networks of the Drosophila respiratory system and our vasculature show distinct branching patterns and tube shapes in different body regions. These local variations are crucial for organ function and organismal fitness. Organotypic patterns and tube geometries in branched networks are typically controlled by variations of extrinsic signaling but the impact of intrinsic factors on branch patterns and shapes is not well explored. Here, we show that the intersection of extrinsic hedgehog(hh) and WNT/wingless (wg) signaling with the tube-intrinsic Hox code of distinct segments specifies the tube pattern and shape of the Drosophila airways. In the cephalic part of the airways, hh signaling induces expression of the transcription factor (TF) knirps (kni) in the anterior dorsal trunk (DTa1). kni represses the expression of another TF spalt major (salm), making DTa1 a narrow and long tube. In DTa branches of more posterior metameres, Bithorax Complex (BX-C) Hox genes autonomously divert hh signaling from inducing kni, thereby allowing DTa branches to develop as salm-dependent thick and short tubes. Moreover, the differential expression of BX-C genes is partly responsible for the anterior-to-posterior gradual increase of the DT tube diameter through regulating the expression level of Salm, a transcriptional target of WNT/wg signaling. Thus, our results highlight how tube intrinsic differential competence can diversify tube morphology without changing availabilities of extrinsic factors. Tubes are common structural elements of many internal organs,
facilitating fluid flow and material exchange. To meet the local needs of diverse tissues, the branching patterns and tube shapes vary regionally. Diametric tapering and specialized branch targeting to the brain represent two common examples of variations with organismal benefits in the Drosophila airways and our vascular system. Several extrinsic signals instruct tube diversifications but the impact of intrinsic factors remains underexplored. Here, we show that the local, tube-intrinsic Hox code instructs the pattern and shape of the dorsal trunk (DT), the main Drosophila airway. In the cephalic part (DT1), where Bithorax Complex (BX-C) Hox genes are not expressed, the extrinsic Hedgehog signal is epistatic to WNT/Wingless signals. Hedgehog instructs anterior DT1 cells to take a long and narrow tube fate targeting the brain. In more posterior metameres, BX-C genes make the extrinsic WNT/Wingless signals epistatic over Hedgehog. There, WNT/Wingless instruct all DT cells to take the thick and short tube fate. Moreover, BX-C genes modulate the outputs of WNT/wingless signaling, making the DT tubes thicker in more posterior metameres. We provide a model for how intrinsic factors modify extrinsic signaling to control regional tube morphologies in a network.
Collapse
Affiliation(s)
- Ryo Matsuda
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Chie Hosono
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Kaoru Saigo
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Christos Samakovlis
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- ECCPS, University of Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
27
|
Hogan BLM, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CCW, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014; 15:123-38. [PMID: 25105578 PMCID: PMC4212493 DOI: 10.1016/j.stem.2014.07.012] [Citation(s) in RCA: 632] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Respiratory disease is the third leading cause of death in the industrialized world. Consequently, the trachea, lungs, and cardiopulmonary vasculature have been the focus of extensive investigations. Recent studies have provided new information about the mechanisms driving lung development and differentiation. However, there is still much to learn about the ability of the adult respiratory system to undergo repair and to replace cells lost in response to injury and disease. This Review highlights the multiple stem/progenitor populations in different regions of the adult lung, the plasticity of their behavior in injury models, and molecular pathways that support homeostasis and repair.
Collapse
Affiliation(s)
- Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA.
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke Medicine, Durham, NC 27705, USA
| | - Harold A Chapman
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Niklason
- Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Elizabeth Calle
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Andrew Le
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason Rock
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melinda Snitow
- Perleman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Krummel
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barry R Stripp
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Thiennu Vu
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric S White
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey A Whitsett
- Section of Neonatology, Perinatal and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Edward E Morrisey
- Departments of Medicine and Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Yang J, Chen J. Developmental programs of lung epithelial progenitors: a balanced progenitor model. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:331-47. [PMID: 25124755 DOI: 10.1002/wdev.141] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/17/2014] [Accepted: 05/04/2014] [Indexed: 01/17/2023]
Abstract
UNLABELLED The daunting task of lung epithelium development is to transform a cluster of foregut progenitors into a three-dimensional (3D) tubular network with distinct cell types distributed at their appropriate locations. A complete understanding of lung development needs to address not only how, but also where, different cell types form. We propose that the lung epithelium forms through regulated deployment of three developmental programs: branching morphogenesis to expand progenitors and build a tree-like tubular network, airway differentiation to specify cells for the proximal conducting airways, and alveolar differentiation to specify cells for the peripheral gas exchange region. Each developmental program has its unique morphological features and molecular control mechanisms; their spatiotemporal coordination can be accounted for in a balanced progenitor model where progenitors balance between alternative developmental programs in response to spatiotemporal cues. This model integrates progenitor morphogenesis and differentiation, and provides new insights to lung immaturity in preterm birth and lung evolution. Advanced gene targeting and 3D imaging tools are needed to achieve a comprehensive understanding of lung epithelial progenitors on molecular, cellular, and morphological levels. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The authors have declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Jun Yang
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
29
|
Bérubé-Simard FA, Prudhomme C, Jeannotte L. YY1 acts as a transcriptional activator of Hoxa5 gene expression in mouse organogenesis. PLoS One 2014; 9:e93989. [PMID: 24705708 PMCID: PMC3976385 DOI: 10.1371/journal.pone.0093989] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/11/2014] [Indexed: 12/15/2022] Open
Abstract
The Hox gene family encodes homeodomain-containing transcriptional regulators that confer positional information to axial and paraxial tissues in the developing embryo. The dynamic Hox gene expression pattern requires mechanisms that differentially control Hox transcription in a precise spatio-temporal fashion. This implies an integrated regulation of neighbouring Hox genes achieved through the sharing and the selective use of defined enhancer sequences. The Hoxa5 gene plays a crucial role in lung and gut organogenesis. To position Hoxa5 in the regulatory hierarchy that drives organ morphogenesis, we searched for cis-acting regulatory sequences and associated trans-acting factors required for Hoxa5 expression in the developing lung and gut. Using mouse transgenesis, we identified two DNA regions included in a 1.5-kb XbaI-XbaI fragment located in the Hoxa4-Hoxa5 intergenic domain and known to control Hoxa4 organ expression. The multifunctional YY1 transcription factor binds the two regulatory sequences in vitro and in vivo. Moreover, the mesenchymal deletion of the Yy1 gene function in mice results in a Hoxa5-like lung phenotype with decreased Hoxa5 and Hoxa4 gene expression. Thus, YY1 acts as a positive regulator of Hoxa5 expression in the developing lung and gut. Our data also support a role for YY1 in the coordinated expression of Hox genes for correct organogenesis.
Collapse
Affiliation(s)
- Félix-Antoine Bérubé-Simard
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l′Université Laval, Québec, Canada
- Centre de recherche du Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Québec, Canada
| | - Christelle Prudhomme
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l′Université Laval, Québec, Canada
- Centre de recherche du Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Québec, Canada
| | - Lucie Jeannotte
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
- Centre de recherche sur le cancer de l′Université Laval, Québec, Canada
- Centre de recherche du Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Québec, Canada
| |
Collapse
|
30
|
Sox9 plays multiple roles in the lung epithelium during branching morphogenesis. Proc Natl Acad Sci U S A 2013; 110:E4456-64. [PMID: 24191021 DOI: 10.1073/pnas.1311847110] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Lung branching morphogenesis is a highly orchestrated process that gives rise to the complex network of gas-exchanging units in the adult lung. Intricate regulation of signaling pathways, transcription factors, and epithelial-mesenchymal cross-talk are critical to ensuring branching morphogenesis occurs properly. Here, we describe a role for the transcription factor Sox9 during lung branching morphogenesis. Sox9 is expressed at the distal tips of the branching epithelium in a highly dynamic manner as branching occurs and is down-regulated starting at embryonic day 16.5, concurrent with the onset of terminal differentiation of type 1 and type 2 alveolar cells. Using epithelial-specific genetic loss- and gain-of-function approaches, our results demonstrate that Sox9 controls multiple aspects of lung branching. Fine regulation of Sox9 levels is required to balance proliferation and differentiation of epithelial tip progenitor cells, and loss of Sox9 leads to direct and indirect cellular defects including extracellular matrix defects, cytoskeletal disorganization, and aberrant epithelial movement. Our evidence shows that unlike other endoderm-derived epithelial tissues, such as the intestine, Wnt/β-catenin signaling does not regulate Sox9 expression in the lung. We conclude that Sox9 collectively promotes proper branching morphogenesis by controlling the balance between proliferation and differentiation and regulating the extracellular matrix.
Collapse
|
31
|
Boucherat O, Montaron S, Bérubé-Simard FA, Aubin J, Philippidou P, Wellik DM, Dasen JS, Jeannotte L. Partial functional redundancy between Hoxa5 and Hoxb5 paralog genes during lung morphogenesis. Am J Physiol Lung Cell Mol Physiol 2013; 304:L817-30. [PMID: 23585229 DOI: 10.1152/ajplung.00006.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hox genes encode transcription factors governing complex developmental processes in several organs. A subset of Hox genes are expressed in the developing lung. Except for Hoxa5, the lack of overt lung phenotype in single mutants suggests that Hox genes may not play a predominant role in lung ontogeny or that functional redundancy may mask anomalies. In the Hox5 paralog group, both Hoxa5 and Hoxb5 genes are expressed in the lung mesenchyme whereas Hoxa5 is also expressed in the tracheal mesenchyme. Herein, we generated Hoxa5;Hoxb5 compound mutant mice to evaluate the relative contribution of each gene to lung development. Hoxa5;Hoxb5 mutants carrying the four mutated alleles displayed an aggravated lung phenotype, resulting in the death of the mutant pups at birth. Characterization of the phenotype highlighted the role of Hoxb5 in lung formation, the latter being involved in branching morphogenesis, goblet cell specification, and postnatal air space structure, revealing partial functional redundancy with Hoxa5. However, the Hoxb5 lung phenotypes were less severe than those seen in Hoxa5 mutants, likely because of Hoxa5 compensation. New specific roles for Hoxa5 were also unveiled, demonstrating the extensive contribution of Hoxa5 to the developing respiratory system. The exclusive expression of Hoxa5 in the trachea and the phrenic motor column likely underlies the Hoxa5-specific trachea and diaphragm phenotypes. Altogether, our observations establish that the Hoxa5 and Hoxb5 paralog genes shared some functions during lung morphogenesis, Hoxa5 playing a predominant role.
Collapse
Affiliation(s)
- Olivier Boucherat
- Centre de recherche en cancérologie de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Hirasawa T, Kuratani S. A new scenario of the evolutionary derivation of the mammalian diaphragm from shoulder muscles. J Anat 2013; 222:504-17. [PMID: 23448284 PMCID: PMC3633340 DOI: 10.1111/joa.12037] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2013] [Indexed: 12/30/2022] Open
Abstract
The evolutionary origin of the diaphragm remains unclear, due to the lack of a comparable structure in other extant taxa. However, recent researches into the developmental mechanism of this structure have yielded new insights into its origin. Here we summarize current understanding regarding the development of the diaphragm, and present a possible scenario for the evolutionary acquisition of this uniquely mammalian structure. Recent developmental analyses indicate that the diaphragm and forelimb muscles are derived from a shared cell population during embryonic development. Therefore, the embryonic positions of forelimb muscle progenitors, which correspond to the position of the brachial plexus, likely played an important role in the evolution of the diaphragm. We surveyed the literature to reexamine the position of the brachial plexus among living amniotes and confirmed that the cervico-thoracic transition in ribs reflects the brachial plexus position. Using this osteological correlate, we concluded that the anterior borders of the brachial plexuses in the stem synapsids were positioned at the level of the fourth spinal nerve, suggesting that the forelimb buds were laid in close proximity of the infrahyoid muscles. The topology of the phrenic and suprascapular nerves of mammals is similar to that of subscapular and supracoracoid nerves, respectively, of the other amniotes, suggesting that the diaphragm evolved from a muscle positioned medial to the pectoral girdle (cf. subscapular muscle). We hypothesize that the diaphragm was acquired in two steps: first, forelimb muscle cells were incorporated into tissues to form a primitive diaphragm in the stem synapsid grade, and second, the diaphragm in cynodonts became entrapped in the region controlled by pulmonary development.
Collapse
Affiliation(s)
- Tatsuya Hirasawa
- Laboratory for Evolutionary Morphology, RIKEN Center for Developmental Biology, Kobe, Japan.
| | | |
Collapse
|
33
|
Development and remodeling of the vertebrate blood-gas barrier. BIOMED RESEARCH INTERNATIONAL 2012; 2013:101597. [PMID: 23484070 PMCID: PMC3591247 DOI: 10.1155/2013/101597] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/24/2012] [Indexed: 11/18/2022]
Abstract
During vertebrate development, the lung inaugurates as an endodermal bud from the primitive foregut. Dichotomous subdivision of the bud results in arborizing airways that form the prospective gas exchanging chambers, where a thin blood-gas barrier (BGB) is established. In the mammalian lung, this proceeds through conversion of type II cells to type I cells, thinning, and elongation of the cells as well as extrusion of the lamellar bodies. Subsequent diminution of interstitial tissue and apposition of capillaries to the alveolar epithelium establish a thin BGB. In the noncompliant avian lung, attenuation proceeds through cell-cutting processes that result in remarkable thinning of the epithelial layer. A host of morphoregulatory molecules, including transcription factors such as Nkx2.1, GATA, HNF-3, and WNT5a; signaling molecules including FGF, BMP-4, Shh, and TFG- β and extracellular proteins and their receptors have been implicated. During normal physiological function, the BGB may be remodeled in response to alterations in transmural pressures in both blood capillaries and airspaces. Such changes are mitigated through rapid expression of the relevant genes for extracellular matrix proteins and growth factors. While an appreciable amount of information regarding molecular control has been documented in the mammalian lung, very little is available on the avian lung.
Collapse
|