151
|
Li L, Feng J, Zhao S, Rong Z, Lin Y. SOX9 inactivation affects the proliferation and differentiation of human lung organoids. Stem Cell Res Ther 2021; 12:343. [PMID: 34112251 PMCID: PMC8194236 DOI: 10.1186/s13287-021-02422-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
Background The regulation of the transcription factor sex-determining region Y-box transcription factor 9 (SOX9) in lung development has been described in mouse, but the same principles apply to human lung development is unknown due to a lack of appropriate experimental approaches and models. Methods Here, we used gene editing technology to inactivate SOX9 in human embryonic stem cells that were then induced to differentiate into lung organoids to investigate the role of SOX9 in human lung epithelium development. Results Complete knockout of the transactivation domain of SOX9 by gene editing resulted in indels in both alleles of SOX9. SOX9−/− hESCs could be induced to differentiate into lung progenitor organoids. In vitro long-term expansion showed that SOX9 inactivation did not affect the differentiation of pulmonary epithelial cells, but promoted apoptosis and reduced proliferative capacity in the organoids. When lung progenitor organoids were transplanted under the kidney capsule of immunodeficient mice, expression of the club cell marker secretoglobin family 1A member 1 (SCGB1A1) was detected in SOX9−/− transplants but was absent in wild-type (WT) transplants. The maturation of goblet cells was also affected by SOX9 inactivation, as evidenced by the presence of mucin 5 AC (MUC5AC) in the cytoplasm of SOX9−/− grafts as compared to WT grafts in which most MUC5AC was secreted into the lumen. In vivo lung orthotopic transplantations showed that SOX9 inactivation had a limited effect on the differentiation of alveolar cells and lung regeneration in injured mice. Conclusions SOX9 modulates the proliferative capacity of lung epithelium but is not an indispensable transcription factor in the regulation of human lung epithelium development. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02422-6.
Collapse
Affiliation(s)
- Lian Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jianqi Feng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shanshan Zhao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhili Rong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
| | - Ying Lin
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
152
|
Lang C, Conrad L, Iber D. Organ-Specific Branching Morphogenesis. Front Cell Dev Biol 2021; 9:671402. [PMID: 34150767 PMCID: PMC8212048 DOI: 10.3389/fcell.2021.671402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 01/09/2023] Open
Abstract
A common developmental process, called branching morphogenesis, generates the epithelial trees in a variety of organs, including the lungs, kidneys, and glands. How branching morphogenesis can create epithelial architectures of very different shapes and functions remains elusive. In this review, we compare branching morphogenesis and its regulation in lungs and kidneys and discuss the role of signaling pathways, the mesenchyme, the extracellular matrix, and the cytoskeleton as potential organ-specific determinants of branch position, orientation, and shape. Identifying the determinants of branch and organ shape and their adaptation in different organs may reveal how a highly conserved developmental process can be adapted to different structural and functional frameworks and should provide important insights into epithelial morphogenesis and developmental disorders.
Collapse
Affiliation(s)
- Christine Lang
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lisa Conrad
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
153
|
Aros CJ, Pantoja CJ, Gomperts BN. Wnt signaling in lung development, regeneration, and disease progression. Commun Biol 2021; 4:601. [PMID: 34017045 PMCID: PMC8138018 DOI: 10.1038/s42003-021-02118-w] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
The respiratory tract is a vital, intricate system for several important biological processes including mucociliary clearance, airway conductance, and gas exchange. The Wnt signaling pathway plays several crucial and indispensable roles across lung biology in multiple contexts. This review highlights the progress made in characterizing the role of Wnt signaling across several disciplines in lung biology, including development, homeostasis, regeneration following injury, in vitro directed differentiation efforts, and disease progression. We further note uncharted directions in the field that may illuminate important biology. The discoveries made collectively advance our understanding of Wnt signaling in lung biology and have the potential to inform therapeutic advancements for lung diseases. Cody Aros, Carla Pantoja, and Brigitte Gomperts review the key role of Wnt signaling in all aspects of lung development, repair, and disease progression. They provide an overview of recent research findings and highlight where research is needed to further elucidate mechanisms of action, with the aim of improving disease treatments.
Collapse
Affiliation(s)
- Cody J Aros
- UCLA Department of Molecular Biology Interdepartmental Program, UCLA, Los Angeles, CA, USA.,UCLA Medical Scientist Training Program, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.,UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Carla J Pantoja
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Brigitte N Gomperts
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA. .,Division of Pulmonary and Critical Care MedicineDavid Geffen School of Medicine, UCLA, Los Angeles, CA, USA. .,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA. .,Eli and Edythe Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
154
|
Lee H, Ko HW. Cell cycle-related kinase is a crucial regulator for ciliogenesis and Hedgehog signaling in embryonic mouse lung development. BMB Rep 2021. [PMID: 32317081 PMCID: PMC7396919 DOI: 10.5483/bmbrep.2020.53.7.295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell cycle-related kinase (CCRK) has a conserved role in ciliogenesis, and Ccrk defects in mice lead to developmental defects, including exencephaly, preaxial polydactyly, skeletal abnormalities, retinal degeneration, and polycystic kidney. Here, we found that Ccrk is highly expressed in mouse trachea and bronchioles. Ccrk mutants exhibited pulmonary hypoplasia and abnormal branching morphogenesis in respiratory organ development. Furthermore, we demonstrated that Ccrk mutant lungs exhibit not only impaired branching morphogenesis but also a significant sacculation deficiency in alveoli associated with reduced epithelial progenitor cell proliferation. In pseudoglandular stages, Ccrk mutant lungs showed a downregulation of Hedgehog (Hh) signaling and defects in cilia morphology and frequency during progenitor-cell proliferation. Interestingly, we observed that activation of the Hh signaling pathway by small-molecule smoothened agonist (SAG) partially rescued bud morphology during branch bifurcation in explants from Ccrk mutant lungs. Therefore, CCRK properly regulates respiratory airway architecture in part through Hh-signal transduction and ciliogenesis.
Collapse
Affiliation(s)
- Hankyu Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
155
|
Basil MC, Katzen J, Engler AE, Guo M, Herriges MJ, Kathiriya JJ, Windmueller R, Ysasi AB, Zacharias WJ, Chapman HA, Kotton DN, Rock JR, Snoeck HW, Vunjak-Novakovic G, Whitsett JA, Morrisey EE. The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future. Cell Stem Cell 2021; 26:482-502. [PMID: 32243808 PMCID: PMC7128675 DOI: 10.1016/j.stem.2020.03.009] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The respiratory system, which includes the trachea, airways, and distal alveoli, is a complex multi-cellular organ that intimately links with the cardiovascular system to accomplish gas exchange. In this review and as members of the NIH/NHLBI-supported Progenitor Cell Translational Consortium, we discuss key aspects of lung repair and regeneration. We focus on the cellular compositions within functional niches, cell-cell signaling in homeostatic health, the responses to injury, and new methods to study lung repair and regeneration. We also provide future directions for an improved understanding of the cell biology of the respiratory system, as well as new therapeutic avenues.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeremy Katzen
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anna E Engler
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Minzhe Guo
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael J Herriges
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jaymin J Kathiriya
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Rebecca Windmueller
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra B Ysasi
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - William J Zacharias
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Hal A Chapman
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jason R Rock
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Hans-Willem Snoeck
- Center for Human Development, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Gordana Vunjak-Novakovic
- Departments of Biomedical Engineering and Medicine, Columbia University, New York, NY 10027, USA
| | - Jeffrey A Whitsett
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Edward E Morrisey
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
156
|
Blitz MJ, Ghorayeb SR, Solmonovich R, Glykos S, Jauhari A, Rochelson B, Bracero LA. Fetal Lung Echo Texture in Pregnancies at Risk for Pulmonary Hypoplasia. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2021; 40:805-810. [PMID: 32865280 DOI: 10.1002/jum.15454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/14/2020] [Accepted: 07/25/2020] [Indexed: 06/11/2023]
Abstract
Pulmonary hypoplasia is associated with severe respiratory distress immediately after birth and frequently leads to neonatal death. In this study, we compared the fetal lung echo texture in pregnancies at high and low risk for pulmonary hypoplasia. Ultrasonic tissue heterogeneity was determined by a dynamic range calculation. This quantification uses a dithering technique based on the Floyd-Steinberg algorithm, in which the pixels are transformed into a binary map. Pregnancies at high risk for pulmonary hypoplasia showed decreased fetal lung heterogeneity on ultrasound imaging. This image-processing technique may allow improved risk stratification, patient counseling, and treatment approaches for pulmonary hypoplasia.
Collapse
Affiliation(s)
- Matthew J Blitz
- Division of Maternal-Fetal Medicine, Southside Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Bay Shore, New York, USA
| | - Sleiman R Ghorayeb
- School of Engineering and Applied Sciences, Ultrasound Research Laboratory, Hofstra University, Hempstead, New York, USA
- Departments of Radiology and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Rachel Solmonovich
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Stella Glykos
- School of Engineering and Applied Sciences, Ultrasound Research Laboratory, Hofstra University, Hempstead, New York, USA
| | - Arushi Jauhari
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Burton Rochelson
- Division of Maternal-Fetal Medicine, North Shore University Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Luis A Bracero
- Division of Maternal-Fetal Medicine, Southside Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Bay Shore, New York, USA
| |
Collapse
|
157
|
Cuevas-Gómez I, McGee M, Sánchez JM, O'Riordan E, Byrne N, McDaneld T, Earley B. Association between clinical respiratory signs, lung lesions detected by thoracic ultrasonography and growth performance in pre-weaned dairy calves. Ir Vet J 2021; 74:7. [PMID: 33766106 PMCID: PMC7992334 DOI: 10.1186/s13620-021-00187-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/11/2021] [Indexed: 11/10/2022] Open
Abstract
Background Bovine respiratory disease (BRD) is the main cause of mortality among 1-to-5 month old calves in Ireland, accounting for approximately one-third of deaths. Despite widespread use of clinical respiratory signs for diagnosing BRD, lung lesions are detected, using thoracic ultrasonography (TUS) or following post-mortem, in calves showing no clinical signs. This highlights the limitation of clinical respiratory signs as a method of detecting sub-clinical BRD. Using 53 purchased artificially-reared male dairy calves, the objectives of this study were to: (i) characterise the BRD incidence detected by clinical respiratory signs and/or TUS, (ii) investigate the association between clinical respiratory signs and lung lesions detected by TUS, and (iii) assess the effect of BRD on pre-weaning growth. Results Clinical BRD (based on Wisconsin clinical respiratory score and/or rectal temperature > 39.6 ºC) was detected in 43 % and sonographic changes (lung lesions) were detected in 64 % of calves from purchase (23 (SD; 6.2) days of age) until weaning, 53 days post-arrival. Calves with clinical BRD were treated. Sixty-one per cent calves affected with clinical BRD had lung lesions 10.5 days (median) before detection of clinical signs. Moderate correlations (rsp 0.70; P < 0.05) were found between cough and severe lung lesions on arrival day, and between rectal temperature > 39.6 ºC and lung lesions ≥ 2 cm2 on day 7 (rsp 0.40; P < 0.05) post-arrival. Mean average daily live weight gain (ADG) of calves from purchase to weaning was 0.75 (SD; 0.10) kg; calves with or without clinical BRD did not differ in ADG (P > 0.05), whereas ADG of those with severe lung lesions (lung lobe completely consolidated or pulmonary emphysema) was 0.12 kg/d less (P < 0.05) than calves without lung lesions. Conclusions Thoracic ultrasonography detected lung consolidation in calves that did not show signs of respiratory disease. The presence of severe lung lesions was associated with reduced pre-weaning growth. These findings emphasise the importance of using TUS in addition to clinical respiratory scoring of calves for an early and accurate detection of clinical and sub-clinical BRD.
Collapse
Affiliation(s)
- Inmaculada Cuevas-Gómez
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland
| | - Mark McGee
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland
| | - José María Sánchez
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland
| | - Edward O'Riordan
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland
| | - Nicky Byrne
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland
| | - Tara McDaneld
- Meat Animal Research Center, USDA, ARS, Nebraska, Clay Center, USA
| | - Bernadette Earley
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Co. Meath, Dunsany, Ireland.
| |
Collapse
|
158
|
Abstract
Branching morphogenesis generates epithelial trees which facilitate gas exchange, filtering, as well as secretion processes with their large surface to volume ratio. In this review, we focus on the developmental mechanisms that control the early stages of lung branching morphogenesis. Lung branching morphogenesis involves the stereotypic, recurrent definition of new branch points, subsequent epithelial budding, and lung tube elongation. We discuss current models and experimental evidence for each of these steps. Finally, we discuss the role of the mesenchyme in determining the organ-specific shape.
Collapse
Affiliation(s)
- Dagmar Iber
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), Basel, Switzerland.
| |
Collapse
|
159
|
Browne JA, NandyMazumdar M, Paranjapye A, Leir SH, Harris A. The Bromodomain Containing 8 (BRD8) transcriptional network in human lung epithelial cells. Mol Cell Endocrinol 2021; 524:111169. [PMID: 33476703 PMCID: PMC8035426 DOI: 10.1016/j.mce.2021.111169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Mechanisms regulating gene expression in the airway epithelium underlie its response to the environment. A network of transcription factors (TFs) and architectural proteins, modulate chromatin accessibility and recruit activating or repressive signals. Bromodomain-containing proteins function as TFs or by engaging methyltransferase or acetyltransferase activity to induce chromatin modifications. Here we investigate the role of Bromodomain Containing 8 (BRD8) in coordinating lung epithelial function. Sites of BRD8 occupancy genome-wide were mapped in human lung epithelial cell lines (Calu-3 and 16HBE14o-). CCCTC-Binding Factor (CTCF) was identified as a predicted co-factor of BRD8, based upon motif over-representation under BRD8 ChIP-seq peaks. Following siRNA-mediated depletion of BRD8, differentially expressed genes with nearby peaks of BRD8 occupancy were subject to gene ontology process enrichment analysis. BRD8 targets are enriched for genes involved in the innate immune response and the cell cycle. Depletion of BRD8 increased the secretion of the antimicrobial peptide beta-defensin 1 and multiple chemokines, and reduced cell proliferation.
Collapse
Affiliation(s)
- James A Browne
- Department of Genetics and Genome Sciences, Cleveland, OH, USA
| | | | | | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
160
|
Abstract
The transforming growth factor β (TGFβ) signaling family is evolutionarily conserved in metazoans. The signal transduction mechanisms of TGFβ family members have been expansively investigated and are well understood. During development and homeostasis, numerous TGFβ family members are expressed in various cell types with temporally changing levels, playing diverse roles in embryonic development, adult tissue homeostasis and human diseases by regulating cell proliferation, differentiation, adhesion, migration and apoptosis. Here, we discuss the molecular mechanisms underlying signal transduction and regulation of the TGFβ subfamily pathways, and then highlight their key functions in mesendoderm induction, dorsoventral patterning and laterality development, as well as in the formation of several representative tissues/organs.
Collapse
Affiliation(s)
- Shunji Jia
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Anming Meng
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
161
|
Zahmatkesh E, Khoshdel-Rad N, Mirzaei H, Shpichka A, Timashev P, Mahmoudi T, Vosough M. Evolution of organoid technology: Lessons learnt in Co-Culture systems from developmental biology. Dev Biol 2021; 475:37-53. [PMID: 33684433 DOI: 10.1016/j.ydbio.2021.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
In recent years, the development of 3D organoids has opened new avenues of investigation into development, physiology, and regenerative medicine. Organoid formation and the process of organogenesis share common developmental pathways; thus, our knowledge of developmental biology can help model the complexity of different organs to refine organoids into a more sophisticated platform. The developmental process is strongly dependent on complex networks and communication of cell-cell and cell-matrix interactions among different cell populations and their microenvironment, during embryogenesis. These interactions affect cell behaviors such as proliferation, survival, migration, and differentiation. Co-culture systems within the organoid technology were recently developed and provided the highly physiologically relevant systems. Supportive cells including various types of endothelial and stromal cells provide the proper microenvironment, facilitate organoid assembly, and improve vascularization and maturation of organoids. This review discusses the role of the co-culture systems in organoid generation, with a focus on how knowledge of developmental biology has directed and continues to shape the development of more evolved 3D co-culture system-derived organoids.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia; Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, N.N.Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
162
|
Widowski H, Reynaert NL, Ophelders DRMG, Hütten MC, Nikkels PGJ, Severens-Rijvers CAH, Cleutjens JPM, Kemp MW, Newnham JP, Saito M, Usuda H, Payne MS, Jobe AH, Kramer BW, Delhaas T, Wolfs TGAM. Sequential Exposure to Antenatal Microbial Triggers Attenuates Alveolar Growth and Pulmonary Vascular Development and Impacts Pulmonary Epithelial Stem/Progenitor Cells. Front Med (Lausanne) 2021; 8:614239. [PMID: 33693012 PMCID: PMC7937719 DOI: 10.3389/fmed.2021.614239] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/02/2021] [Indexed: 01/01/2023] Open
Abstract
Perinatal inflammatory stress is strongly associated with adverse pulmonary outcomes after preterm birth. Antenatal infections are an essential perinatal stress factor and contribute to preterm delivery, induction of lung inflammation and injury, pre-disposing preterm infants to bronchopulmonary dysplasia. Considering the polymicrobial nature of antenatal infection, which was reported to result in diverse effects and outcomes in preterm lungs, the aim was to examine the consequences of sequential inflammatory stimuli on endogenous epithelial stem/progenitor cells and vascular maturation, which are crucial drivers of lung development. Therefore, a translational ovine model of antenatal infection/inflammation with consecutive exposures to chronic and acute stimuli was used. Ovine fetuses were exposed intra-amniotically to Ureaplasma parvum 42 days (chronic stimulus) and/or to lipopolysaccharide 2 or 7 days (acute stimulus) prior to preterm delivery at 125 days of gestation. Pulmonary inflammation, endogenous epithelial stem cell populations, vascular modulators and morphology were investigated in preterm lungs. Pre-exposure to UP attenuated neutrophil infiltration in 7d LPS-exposed lungs and prevented reduction of SOX-9 expression and increased SP-B expression, which could indicate protective responses induced by re-exposure. Sequential exposures did not markedly impact stem/progenitors of the proximal airways (P63+ basal cells) compared to single exposure to LPS. In contrast, the alveolar size was increased solely in the UP+7d LPS group. In line, the most pronounced reduction of AEC2 and proliferating cells (Ki67+) was detected in these sequentially UP + 7d LPS-exposed lambs. A similar sensitization effect of UP pre-exposure was reflected by the vessel density and expression of vascular markers VEGFR-2 and Ang-1 that were significantly reduced after UP exposure prior to 2d LPS, when compared to UP and LPS exposure alone. Strikingly, while morphological changes of alveoli and vessels were seen after sequential microbial exposure, improved lung function was observed in UP, 7d LPS, and UP+7d LPS-exposed lambs. In conclusion, although sequential exposures did not markedly further impact epithelial stem/progenitor cell populations, re-exposure to an inflammatory stimulus resulted in disturbed alveolarization and abnormal pulmonary vascular development. Whether these negative effects on lung development can be rescued by the potentially protective responses observed, should be examined at later time points.
Collapse
Affiliation(s)
- Helene Widowski
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,Department of BioMedical Engineering, Maastricht University Medical Center, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University, Maastricht, Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Daan R M G Ophelders
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Matthias C Hütten
- Neonatology, Pediatrics Department, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center, Maastricht, Netherlands.,University Children's Hospital Würzburg, University of Würzburg, Würzburg, Germany
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Jack P M Cleutjens
- Department of Pathology, Maastricht University Medical Center, Maastricht, Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, Netherlands
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia
| | - John P Newnham
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia
| | - Masatoshi Saito
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia.,Tohoku University Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia.,Tohoku University Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Matthew S Payne
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynecology, The University of Western Australia, Crawley, WA, Australia.,Perinatal Institute Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Tammo Delhaas
- Department of BioMedical Engineering, Maastricht University Medical Center, Maastricht, Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, Netherlands
| | - Tim G A M Wolfs
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
163
|
Cuevas-Gómez I, McGee M, McCabe M, Cormican P, O'Riordan E, McDaneld T, Earley B. Growth performance and hematological changes of weaned beef calves diagnosed with respiratory disease using respiratory scoring and thoracic ultrasonography. J Anim Sci 2021; 98:5936636. [PMID: 33095858 DOI: 10.1093/jas/skaa345] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
This study investigated 1) the effect of clinical bovine respiratory disease (BRD) and associated lung consolidations on growth performance and hematological profiles of recently weaned beef calves and 2) the relationship between clinical respiratory signs and lung consolidation detected by thoracic ultrasonography (TUS). One hundred and fifty-three weaned beef calves (209 days old [SD: 35.8] and 306 kg [SD: 26.3], at arrival) purchased and transported from auction markets were accommodated indoors in concrete slatted floor pens. Calves were weighed weekly from arrival until day 28 and on day 65 post-arrival. Assessment of BRD and blood sample collection for hematological profiles were performed on scheduled days (at arrival, on days 7, 14, and 28) and on other days upon BRD diagnosis. Animals were assessed for BRD using a total clinical respiratory score (CRS) of five clinical signs (rectal temperature, ear position, cough, nasal secretion, and eye secretion with each ranging from normal [0] to abnormal [3]) and TUS scores (normal [0] to lung consolidation ≥ 1 cm2 [2]). Based on CRS, 35% of calves were CRS+ (CRS ≥ 5) and 65% were CRS- (CRS < 5). Although no lung consolidations (TUS-) were detected at arrival, 34% of calves developed lung consolidation (≥1 cm2) (TUS+) during the first 28 d post-arrival. Only fever (>39.6 °C) and nasal discharge were weakly associated (r = 0.19, P <0.05) with lung consolidation. On the day of BRD detection, neutrophil number and neutrophil:lymphocyte ratio were 58% and 73% greater, respectively, in BRD calves with lung consolidation compared with healthy calves. From day 0 to 65, calf average daily gain (ADG) did not differ (P >0.05) between CRS+ and CRS- calves but was 0.09 kg/d lower (P < 0.05) for TUS+ compared with TUS- calves. Calves classified as BRD (CRS + TUS ≥ 5) with lung consolidation had lower (P < 0.05) ADG from arrival until day 28 than healthy calves and BRD calves without lung consolidation (0.11 ± 0.10 vs. 0.53 ± 0.07 vs. 0.57 ± 0.10 kg/d, respectively); however, no differences in ADG were observed from day 0 to 65. Conventional methods to diagnose BRD failed to detect calves with lung lesions. TUS is a useful tool to detect lung lesions and its implementation in combination with CRS should provide a more accurate and early diagnosis of BRD, which is fundamental to successful treatment, animal welfare, and growth performance.
Collapse
Affiliation(s)
- Inmaculada Cuevas-Gómez
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| | - Mark McGee
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| | - Matthew McCabe
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| | - Paul Cormican
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| | - Edward O'Riordan
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| | - Tara McDaneld
- US Meat Animal Research Center, USDA, ARS, Clay Center, NE
| | - Bernadette Earley
- Teagasc, Animal & Grassland Research and Innovation Centre (AGRIC), Grange, Dunsany, Co. Meath, Ireland
| |
Collapse
|
164
|
Takahashi T, Friedmacher F, Zimmer J, Puri P. Pbx1, Meis1, and Runx1 Expression Is Decreased in the Diaphragmatic and Pulmonary Mesenchyme of Rats with Nitrofen-Induced Congenital Diaphragmatic Hernia. Eur J Pediatr Surg 2021; 31:120-125. [PMID: 32862424 DOI: 10.1055/s-0040-1714736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Congenital diaphragmatic hernia (CDH) and associated pulmonary hypoplasia (PH) are thought to originate from mesenchymal defects in pleuroperitoneal folds (PPFs) and primordial lungs. Pre-B-cell leukemia homeobox 1 (Pbx1), its binding partner myeloid ecotropic integration site 1 (Meis1), and runt-related transcription factor 1 (Runx1) are expressed in diaphragmatic and lung mesenchyme, functioning as transcription cofactors that modulate mesenchymal cell proliferation. Furthermore, Pbx1 -/- mice develop diaphragmatic defects and PH similar to human CDH. We hypothesized that diaphragmatic and pulmonary Pbx1, Meis1, and Runx1 expression is decreased in the nitrofen-induced CDH model. MATERIALS AND METHODS Time-mated rats were exposed to nitrofen or vehicle on gestational day 9 (D9). Fetal diaphragms (n = 72) and lungs (n = 48) were microdissected on D13, D15, and D18, and were divided into control and nitrofen-exposed specimens. Diaphragmatic and pulmonary gene expression levels of Pbx1, Meis1, and Runx1 were analyzed by quantitative real-time polymerase chain reaction. Immunofluorescence-double-staining for Pbx1, Meis1, and Runx1 was combined with mesenchymal/myogenic markers Gata4 and myogenin to evaluate protein expression. RESULTS Relative mRNA expression of Pbx1, Meis1, and Runx1 was significantly decreased in PPFs (D13), developing diaphragms/lungs (D15), and muscularized diaphragms/differentiated lungs (D18) of nitrofen-exposed fetuses compared with controls. Confocal-laser-scanning-microscopy revealed markedly diminished Pbx1, Meis1, and Runx1 immunofluorescence in diaphragmatic and pulmonary mesenchyme, associated with less proliferating mesenchymal cells in nitrofen-exposed fetuses on D13, D15, and D18 compared with controls. CONCLUSION Decreased Pbx1, Meis1, and Runx1 expression during diaphragmatic development and lung branching morphogenesis may reduce mesenchymal cell proliferation, causing malformed PPFs and disrupted airway branching, thus leading to diaphragmatic defects and PH in the nitrofen-induced CDH model.
Collapse
Affiliation(s)
- Toshiaki Takahashi
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.,Department of Pediatric Surgery, Kansai Medical University, Osaka, Japan
| | - Florian Friedmacher
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.,Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Julia Zimmer
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.,Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Prem Puri
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.,Beacon Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
165
|
Inactivation of Lats1 and Lats2 highlights the role of hippo pathway effector YAP in larynx and vocal fold epithelium morphogenesis. Dev Biol 2021; 473:33-49. [PMID: 33515576 DOI: 10.1016/j.ydbio.2021.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 01/03/2021] [Accepted: 01/20/2021] [Indexed: 11/22/2022]
Abstract
Proliferation and differentiation of vocal fold epithelial cells during embryonic development is poorly understood. We examined the role of Hippo signaling, a vital pathway known for regulating organ size, in murine laryngeal development. Conditional inactivation of the Hippo kinase genes Lats1 and Lats2, specifically in vocal fold epithelial cells, resulted in severe morphogenetic defects. Deletion of Lats1 and Lats2 caused abnormalities in epithelial differentiation, epithelial lamina separation, cellular adhesion, basement membrane organization with secondary failed cartilage, and laryngeal muscle development. Further, Lats1 and Lats2 inactivation led to failure in differentiation of p63+ basal progenitors. Our results reveal novel roles of Hippo-Lats-YAP signaling in proper regulation of VF epithelial fate and larynx morphogenesis.
Collapse
|
166
|
Karolak JA, Gambin T, Szafranski P, Stankiewicz P. Potential interactions between the TBX4-FGF10 and SHH-FOXF1 signaling during human lung development revealed using ChIP-seq. Respir Res 2021; 22:26. [PMID: 33478486 PMCID: PMC7818749 DOI: 10.1186/s12931-021-01617-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
Background The epithelial-mesenchymal signaling involving SHH-FOXF1, TBX4-FGF10, and TBX2 pathways is an essential transcriptional network operating during early lung organogenesis. However, precise regulatory interactions between different genes and proteins in this pathway are incompletely understood. Methods To identify TBX2 and TBX4 genome-wide binding sites, we performed chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq) in human fetal lung fibroblasts IMR-90. Results We identified 14,322 and 1,862 sites strongly-enriched for binding of TBX2 and TBX4, respectively, 43.95% and 18.79% of which are located in the gene promoter regions. Gene Ontology, pathway enrichment, and DNA binding motif analyses revealed a number of overrepresented cues and transcription factor binding motifs relevant for lung branching that can be transcriptionally regulated by TBX2 and/or TBX4. In addition, TBX2 and TBX4 binding sites were found enriched around and within FOXF1 and its antisense long noncoding RNA FENDRR, indicating that the TBX4-FGF10 cascade may directly interact with the SHH-FOXF1 signaling. Conclusions We highlight the complexity of transcriptional network driven by TBX2 and TBX4 and show that disruption of this crosstalk during morphogenesis can play a substantial role in etiology of lung developmental disorders.
Collapse
Affiliation(s)
- Justyna A Karolak
- Department of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-781, Poznan, Poland
| | - Tomasz Gambin
- Department of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Institute of Computer Science, Warsaw University of Technology, 00-665, Warsaw, Poland
| | - Przemyslaw Szafranski
- Department of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Paweł Stankiewicz
- Department of Molecular & Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.
| |
Collapse
|
167
|
Ameis D, Liu F, Kirby E, Patel D, Keijzer R. The RNA-binding protein Quaking regulates multiciliated and basal cell abundance in the developing lung. Am J Physiol Lung Cell Mol Physiol 2021; 320:L557-L567. [PMID: 33438508 DOI: 10.1152/ajplung.00481.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RNA-binding proteins (RBPs) form complexes with RNA, changing how the RNA is processed and thereby regulating gene expression. RBPs are important sources of gene regulation during organogenesis, including the development of lungs. The RBP called Quaking (QK) is critical for embryogenesis, yet it has not been studied in the developing lung. Here, we show that QK is widely expressed during rat lung development and into adulthood. The QK isoforms QK5 and QK7 colocalize to the nuclei of nearly all lung cells. QK6 is present in the nuclei and cytoplasm of mesenchymal cells and is only present in the epithelium during branching morphogenesis. QK knockdown in embryonic lung explants caused a greater number of multiciliated cells to appear in the airways, at the expense of basal cells. The mRNA of multiciliated cell genes and the abundance of FOXJ1/SOX2+ cells increased after knockdown, whereas P63/SOX2+ cells decreased. The cytokine IL-6, a known regulator of multiciliated cell differentiation, had increased mRNA levels after QK knockdown, although protein levels remained unchanged. Further studies are necessary to confirm whether QK acts as a blocker for the IL-6-induced differentiation of basal cells into multiciliated cells, and a conditional QK knockout would likely lead to additional discoveries on QK's role during lung development.
Collapse
Affiliation(s)
- Dustin Ameis
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Franklin Liu
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Eimear Kirby
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Daywin Patel
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
168
|
Robbins ME, Cho HY, Hansen JM, Luchsinger JR, Locy ML, Velten M, Kleeberger SR, Rogers LK, Tipple TE. Glutathione reductase deficiency alters lung development and hyperoxic responses in neonatal mice. Redox Biol 2021; 38:101797. [PMID: 33254076 PMCID: PMC7708869 DOI: 10.1016/j.redox.2020.101797] [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: 10/06/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular antioxidants protect against hyperoxic lung injury. The role of the glutathione (GSH) system in lung development and bronchopulmonary dysplasia (BPD) pathogenesis has not been systematically investigated. The current study utilized GSH reductase-deficient (Gsr-KO) neonatal mice to test the hypothesis that early disruption of the GSH system negatively impacts lung development and hyperoxic responses. Lungs from wild-type (Gsr-WT) and Gsr-KO mice were analyzed for histopathology, developmental markers, redox indices, and transcriptome profiling at different developmental stages following exposure to room air or hyperoxia (85% O2) for up to 14 d. Lungs from Gsr-KO mice exhibited alveolar epithelial dysplasia in the embryonic and neonatal periods with relatively normal lung architecture in adulthood. GSH and its oxidized form (GSSG) were 50-70% lower at E19-PND14 in Gsr-KO lungs than in age-matched Gsr-WT. Differential gene expression between Gsr-WT and Gsr-KO lungs was analyzed at discrete developmental stages. Gsr-KO lungs exhibited downregulated cell cycle and DNA damage checkpoint genes at E19, as well as lung lipid metabolism and surfactant genes at PND5. In addition to abnormal baseline lung morphometry, Gsr-KO mice displayed a blunted response to hyperoxia. Hyperoxia caused a more robust upregulation of the lung thioredoxin system in Gsr-KO compared to Gsr-WT. Gsr-dependent, hyperoxia-responsive genes were highly associated with abnormal cytoskeleton, skeletal-muscular function, and tissue morphology at PND5. Overall, our data in Gsr-KO mice implicate the GSH system as a key regulator of lung development, cellular differentiation, and hyperoxic responses in neonatal mice.
Collapse
Affiliation(s)
- Mary E Robbins
- Division of Neonatology, Department of Pediatrics, Northwestern University, Chicago, IL, USA.
| | - Hye-Youn Cho
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jason M Hansen
- Physiology & Developmental Biology, Brigham Young University, Provo, UT, USA
| | - Joseph R Luchsinger
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Morgan L Locy
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich- Wilhelms University, University Medical Center, Bonn, Germany
| | - Steven R Kleeberger
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Lynette K Rogers
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Trent E Tipple
- Center for Pregnancy and Newborn Research, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
169
|
Hu Y, Ciminieri C, Hu Q, Lehmann M, Königshoff M, Gosens R. WNT Signalling in Lung Physiology and Pathology. Handb Exp Pharmacol 2021; 269:305-336. [PMID: 34463851 DOI: 10.1007/164_2021_521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany. .,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
170
|
Yeung AK, Villacorta-Martin C, Hon S, Rock JR, Murphy GJ. Lung megakaryocytes display distinct transcriptional and phenotypic properties. Blood Adv 2020; 4:6204-6217. [PMID: 33351116 PMCID: PMC7757004 DOI: 10.1182/bloodadvances.2020002843] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Megakaryocytes (MKs) are responsible for platelet biogenesis, which is believed to occur canonically in adult bone marrow (BM) and in the fetal liver during development. However, emerging evidence highlights the lung as a previously underappreciated residence for MKs that may contribute significantly to circulating platelet mass. Although a diversity of cells specific to the BM is known to promote the maturation and trafficking of MKs, little investigation into the impact of the lung niche on the development and function of MKs has been done. Here, we describe the application of single-cell RNA sequencing, coupled with histological, ploidy, and flow cytometric analyses, to profile primary MKs derived from syngeneic mouse lung and hematopoietic tissues. Transcriptional profiling demonstrated that lung MKs have a unique signature distinct from their hematopoietic counterparts, with lung MKs displaying enrichment for maturation markers, potentially indicating a propensity for more efficient platelet production. Reciprocally, fetal lung MKs also showed the robust expression of cytokines and growth factors that are known to promote lung development. Lastly, lung MKs possess an enrichment profile skewed toward roles in immunity and inflammation. These findings highlight the existence of a lung-specific MK phenotype and support the notion that the lung plays an independent role in the development and functional maturation of MKs. The immune phenotype displayed by lung MKs also introduces their potential role in microbial surveillance and antigen presentation.
Collapse
Affiliation(s)
- Anthony K Yeung
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; and
- Section of Hematology and Medical Oncology and
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; and
| | - Stephanie Hon
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; and
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Jason R Rock
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; and
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - George J Murphy
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA; and
- Section of Hematology and Medical Oncology and
| |
Collapse
|
171
|
de Fátima Martins M, Honório-Ferreira A, S Reis M, Cortez-Vaz C, Gonçalves CA. Sialic acids expression in newborn rat lungs: implications for pulmonary developmental biology. Acta Histochem 2020; 122:151626. [PMID: 33068965 DOI: 10.1016/j.acthis.2020.151626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 08/15/2020] [Accepted: 09/02/2020] [Indexed: 11/30/2022]
Abstract
Mammalian lung development proceeds during the postnatal period and continues throughout life. Intricate tubular systems of airways and vessels lined by epithelial cells are developed during this process. All cells, and particularly epithelial cells, carry an array of glycans on their surfaces. N-acetylneuraminic (Neu5Ac) and N-glycolylneuraminic (Neu5Gc) acids, two most frequently-occurring sialic acid residues, are essential determinants during development and in the homeostasis of cells and organisms. However, systematic data about the presence of cell surface sialic acids in the postnatal lung and their content is still scarce. In the present study, we addressed the histochemical localization of Neu5Ac > Neu5Gc in 0-day-old rat lungs. Furthermore, both residues were separated, identified and quantified in lung membranes isolated from 0-day-old rat lungs using high-performance liquid chromatography (HPLC) methodologies. Finally, we compared these results with those previously reported by us for adult rat lungs. The Neu5Ac > Neu5Gc residues were located on the surface of ciliated and non-ciliated cells and the median values for both residues in the purified lung membranes of newborn rats were 5.365 and 1.935 μg/mg prot., respectively. Comparing these results with those reported for the adults, it was possible to observe a significant difference between the levels of Neu5Ac and Neu5Gc (p < 0.001). A more substantial change was found for the case of Neu5Ac. The preponderance of Neu5Ac and its expressive increase during the postnatal development points towards a more prominent role of this residue. Bearing in mind that sialic acids are negatively charged molecules, the high content of Neu5Ac could contribute to the formation of an anion "shield" and have a role in pulmonary development and physiology.
Collapse
Affiliation(s)
- Maria de Fátima Martins
- Instituto de Histologia e Embriologia, Faculdade de Medicina, Universidade de Coimbra, Polo I Rua Larga, 3004-504, Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal.
| | - Ana Honório-Ferreira
- Instituto de Histologia e Embriologia, Faculdade de Medicina, Universidade de Coimbra, Polo I Rua Larga, 3004-504, Coimbra, Portugal
| | - Marco S Reis
- CIEPQPF, Departamento de Engenharia Química, Universidade de Coimbra, Pólo II, Rua Sílvio Lima, 3030-790 Coimbra, Portugal
| | - Catarina Cortez-Vaz
- Instituto de Histologia e Embriologia, Faculdade de Medicina, Universidade de Coimbra, Polo I Rua Larga, 3004-504, Coimbra, Portugal
| | - Carlos Alberto Gonçalves
- Instituto de Histologia e Embriologia, Faculdade de Medicina, Universidade de Coimbra, Polo I Rua Larga, 3004-504, Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| |
Collapse
|
172
|
Mižíková I, Thébaud B. Looking at the developing lung in single-cell resolution. Am J Physiol Lung Cell Mol Physiol 2020; 320:L680-L687. [PMID: 33205990 DOI: 10.1152/ajplung.00385.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lung development is a complicated and delicate process, facilitated by spatially and temporarily coordinated cross talk of up to 40 cell types. Developmental origin and heterogeneity of lung cell lineages in context of lung development have been a focus of research efforts for decades. Bulk RNA and protein measurements, RNA and protein labeling, and lineage tracing techniques have been traditionally employed. However, the complex and heterogeneous nature of lung tissue presents a particular challenge when identifying subtle changes in gene expression in individual cell types. Rapidly developing single-cell RNA sequencing (scRNA-seq) techniques allow for unbiased and robust assessment of complex cellular dynamics during biological processes in unprecedented ways. Discovered a decade ago, scRNA-seq has been applied in respiratory research to understand lung cellular composition and to identify novel cell types. Still, very few studies to date have addressed the single-cell transcriptome in healthy or aberrantly developing lung. In this review, we discuss principal discoveries with scRNA-seq in the field of prenatal and postnatal lung development. In addition, we examine challenges and expectations, and propose future steps associated with the use of scRNA-seq to study developmental lung diseases.
Collapse
Affiliation(s)
- I Mižíková
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - B Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
173
|
Dyamenahalli K, Garg G, Shupp JW, Kuprys PV, Choudhry MA, Kovacs EJ. Inhalation Injury: Unmet Clinical Needs and Future Research. J Burn Care Res 2020; 40:570-584. [PMID: 31214710 DOI: 10.1093/jbcr/irz055] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary and systemic insults from inhalation injury can complicate the care of burn patients and contribute to significant morbidity and mortality. However, recent progress in diagnosis and treatment of inhalation injury has not kept pace with the care of cutaneous thermal injury. There are many challenges unique to inhalation injury that have slowed advancement, including deficiencies in our understanding of its pathophysiology, the relative difficulty and subjectivity of bronchoscopic diagnosis, the lack of diagnostic biomarkers, the necessarily urgent manner in which decisions are made about intubation, and the lack of universal recommendations for the application of mucolytics, anticoagulants, bronchodilators, modified ventilator strategies, and other measures. This review represents a summary of critical shortcomings in our understanding and management of inhalation injury identified by the American Burn Association's working group on Cutaneous Thermal Injury and Inhalation Injury in 2018. It addresses our current understanding of the diagnosis, pathophysiology, and treatment of inhalation injury and highlights topics in need of additional research, including 1) airway repair mechanisms; 2) the airway microbiome in health and after injury; and 3) candidate biomarkers of inhalation injury.
Collapse
Affiliation(s)
- Kiran Dyamenahalli
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado School of Medicine, Aurora
| | - Gaurav Garg
- Department of Surgery, Georgetown University School of Medicine, Washington, District of Columbia
| | - Jeffrey W Shupp
- Department of Surgery, Georgetown University School of Medicine, Washington, District of Columbia
| | - Paulius V Kuprys
- Department of Surgery, Burn & Shock Trauma Research Institute, Health Sciences Division, Loyola University, Maywood, Illinois
| | - Mashkoor A Choudhry
- Department of Surgery, Burn & Shock Trauma Research Institute, Health Sciences Division, Loyola University, Maywood, Illinois
| | - Elizabeth J Kovacs
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, University of Colorado School of Medicine, Aurora
| |
Collapse
|
174
|
Tian L, Gao J, Garcia IM, Chen HJ, Castaldi A, Chen YW. Human pluripotent stem cell-derived lung organoids: Potential applications in development and disease modeling. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e399. [PMID: 33145915 DOI: 10.1002/wdev.399] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 09/09/2020] [Accepted: 10/07/2020] [Indexed: 01/01/2023]
Abstract
The pulmonary system is comprised of two main compartments, airways and alveolar space. Their tissue and cellular complexity ensure lung function and protection from external agents, for example, virus. Two-dimensional (2D) in vitro systems and animal models have been largely employed to elucidate the molecular mechanisms underlying human lung development, physiology, and pathogenesis. However, neither of these models accurately recapitulate the human lung environment and cellular crosstalk. More recently, human-derived three-dimensional (3D) models have been generated allowing for a deeper understanding of cell-to-cell communication. However, the availability and accessibility of primary human cell sources from which generate the 2D and 3D models may be limited. In the past few years, protocols have been developed to successfully employ human pluripotent stem cells (hPSCs) and differentiate them toward pulmonary fate in vitro. In the present review, we discuss the advantages and pitfalls of hPSC-derived lung 2D and 3D models, including the main characteristics and potentials for these models and their current and future applications for modeling development and diseases. Lung organoids currently represent the closest model to the human pulmonary system. We further focus on the applications of lung organoids for the study of human diseases such as pulmonary fibrosis, infectious diseases, and lung cancer. Finally, we discuss the present limitations and potential future applications of 3D lung organoids. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion.
Collapse
Affiliation(s)
- Lu Tian
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jinghui Gao
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Irving M Garcia
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Huanhuan Joyce Chen
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA.,Ben May department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Alessandra Castaldi
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ya-Wen Chen
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
175
|
Sah RK, Ma J, Bah FB, Xing Z, Adlat S, Oo ZM, Wang Y, Bahadar N, Bohio AA, Nagi FH, Feng X, Zhang L, Zheng Y. Targeted Disruption of Mouse Dip2B Leads to Abnormal Lung Development and Prenatal Lethality. Int J Mol Sci 2020; 21:E8223. [PMID: 33153107 PMCID: PMC7663123 DOI: 10.3390/ijms21218223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/21/2022] Open
Abstract
Molecular and anatomical functions of mammalian Dip2 family members (Dip2A, Dip2B and Dip2C) during organogenesis are largely unknown. Here, we explored the indispensable role of Dip2B in mouse lung development. Using a LacZ reporter, we explored Dip2B expression during embryogenesis. This study shows that Dip2B expression is widely distributed in various neuronal, myocardial, endothelial, and epithelial cell types during embryogenesis. Target disruption of Dip2b leads to intrauterine growth restriction, defective lung formation and perinatal mortality. Dip2B is crucial for late lung maturation rather than early-branching morphogenesis. The morphological analysis shows that Dip2b loss leads to disrupted air sac formation, interstitium septation and increased cellularity. In BrdU incorporation assay, it is shown that Dip2b loss results in increased cell proliferation at the saccular stage of lung development. RNA-seq analysis reveals that 1431 genes are affected in Dip2b deficient lungs at E18.5 gestation age. Gene ontology analysis indicates cell cycle-related genes are upregulated and immune system related genes are downregulated. KEGG analysis identifies oxidative phosphorylation as the most overrepresented pathways along with the G2/M phase transition pathway. Loss of Dip2b de-represses the expression of alveolar type I and type II molecular markers. Altogether, the study demonstrates an important role of Dip2B in lung maturation and survival.
Collapse
Affiliation(s)
- Rajiv Kumar Sah
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Jun Ma
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China;
| | - Fatoumata Binta Bah
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Zhenkai Xing
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Salah Adlat
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Zin Ma Oo
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Yajun Wang
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Noor Bahadar
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Ameer Ali Bohio
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Farooq Hayel Nagi
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Xuechao Feng
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| | - Luqing Zhang
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Yaowu Zheng
- Key Laboratory of Molecular Epigenetics, Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China; (R.K.S.); (F.B.B.); (Z.X.); (S.A.); (Z.M.O.); (Y.W.); (N.B.); (A.A.B.); (F.H.N.); (L.Z.)
| |
Collapse
|
176
|
Vazquez-Armendariz AI, Heiner M, El Agha E, Salwig I, Hoek A, Hessler MC, Shalashova I, Shrestha A, Carraro G, Mengel JP, Günther A, Morty RE, Vadász I, Schwemmle M, Kummer W, Hain T, Goesmann A, Bellusci S, Seeger W, Braun T, Herold S. Multilineage murine stem cells generate complex organoids to model distal lung development and disease. EMBO J 2020; 39:e103476. [PMID: 32985719 PMCID: PMC7604576 DOI: 10.15252/embj.2019103476] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/23/2022] Open
Abstract
Organoids derived from mouse and human stem cells have recently emerged as a powerful tool to study organ development and disease. We here established a three‐dimensional (3D) murine bronchioalveolar lung organoid (BALO) model that allows clonal expansion and self‐organization of FACS‐sorted bronchioalveolar stem cells (BASCs) upon co‐culture with lung‐resident mesenchymal cells. BALOs yield a highly branched 3D structure within 21 days of culture, mimicking the cellular composition of the bronchioalveolar compartment as defined by single‐cell RNA sequencing and fluorescence as well as electron microscopic phenotyping. Additionally, BALOs support engraftment and maintenance of the cellular phenotype of injected tissue‐resident macrophages. We also demonstrate that BALOs recapitulate lung developmental defects after knockdown of a critical regulatory gene, and permit modeling of viral infection. We conclude that the BALO model enables reconstruction of the epithelial–mesenchymal‐myeloid unit of the distal lung, thereby opening numerous new avenues to study lung development, infection, and regenerative processes in vitro.
Collapse
Affiliation(s)
- Ana Ivonne Vazquez-Armendariz
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Monika Heiner
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Elie El Agha
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Isabelle Salwig
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andreas Hoek
- Bioinformatics and Systems Biology, Justus-Liebig-University, Giessen, Germany
| | - Marie Christin Hessler
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Irina Shalashova
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Amit Shrestha
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Gianni Carraro
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jan Philip Mengel
- Institute for Medical Microbiology, German Center for Infection Research (DZIF), Justus-Liebig-University Giessen, Partner Site Giessen-Marburg-Langen, Giessen, Germany
| | - Andreas Günther
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Rory Edward Morty
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany.,Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - István Vadász
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Kummer
- Institute for Anatomy and Cell Biology, Justus-Liebig-University, UGMLC, DZL, Giessen, Germany
| | - Torsten Hain
- Institute for Medical Microbiology, German Center for Infection Research (DZIF), Justus-Liebig-University Giessen, Partner Site Giessen-Marburg-Langen, Giessen, Germany
| | - Alexander Goesmann
- Bioinformatics and Systems Biology, Justus-Liebig-University, Giessen, Germany
| | - Saverio Bellusci
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany.,Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Susanne Herold
- Department of Internal Medicine II and Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) and The Institute of Lung Health (ILH), Giessen, Germany
| |
Collapse
|
177
|
Lekovic MH, Drekovic NE, Granica ND, Mahmutovic EH, Djordjevic NZ. Extremely low-frequency electromagnetic field induces a change in proliferative capacity and redox homeostasis of human lung fibroblast cell line MRC-5. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:39466-39473. [PMID: 32651790 DOI: 10.1007/s11356-020-10039-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/06/2020] [Indexed: 06/11/2023]
Abstract
Numerous studies have shown that extremely low-frequency electromagnetic field (ELF-EMF) by modulating oxidative-antioxidative balance in the cells achieved beneficial and harmful effects on living organisms. The aim of this study was to research changes of both proliferative capacity and redox homeostasis of human lung fibroblast cell line MRC-5 during exposure to ELF-EMF (50 Hz). The human lung fibroblast cell line MRC-5 were exposed to ELF-EMF once a day in duration of 1 h during 24 h (1 treatment 1 h/day), 48 h (2 treatments 1 h/day), 72 h (3 treatments 1 h/day), and 7 days (7 treatments 1 h/day). After 24 h of the last treatment, the proliferative capacity of the cells and the concentrations and activities of the components of the oxidative/antioxidative system were determined: superoxide anion (O2.-), hydrogen peroxide (H2O2), nitric oxide (NO), peroxynitrite (ONOO-), reduced glutathione (GSH), oxidized glutathione (GSSG), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), glutathione reductase (GR), and glutathione-S-transferase (GST). The results of this study show that ELF-EMF may affect a cell cycle regulation of human lung fibroblast cell line MRC-5 through modulation of oxidative/antioxidative defense system. The effects of ELF-EMF on proliferation and redox balance of human lung fibroblast cell line MRC-5 depend on exposure time.
Collapse
Affiliation(s)
- Maida H Lekovic
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica bb, Novi Pazar, 36300, Serbia
| | - Nerkesa E Drekovic
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica bb, Novi Pazar, 36300, Serbia
| | - Nihat Dz Granica
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica bb, Novi Pazar, 36300, Serbia
| | - Elvis H Mahmutovic
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica bb, Novi Pazar, 36300, Serbia
| | - Natasa Z Djordjevic
- Department of Biomedical Sciences, State University of Novi Pazar, Vuka Karadzica bb, Novi Pazar, 36300, Serbia.
| |
Collapse
|
178
|
Jiang M, Roth MG, Chun-On P, Sullivan DI, Alder JK. Phenotypic Diversity Caused by Differential Expression of SFTPC-Cre-Transgenic Alleles. Am J Respir Cell Mol Biol 2020; 62:692-698. [PMID: 32208105 DOI: 10.1165/rcmb.2019-0416ma] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Type II alveolar epithelial cells (AEC2s) play an essential role in the function and maintenance of the pulmonary epithelium. Several transgenic mice have been developed to study the function of these cells in vivo by using the human SFTPC promoter to drive expression of Cre recombinase. The precise activity of each of these transgenic alleles has not been studied, and previous reports suggest that their activity can depend on breeding strategies. We bred mice with a conditional allele of the essential telomere capping protein TRF2 with two different SFTPC-Cre-transgenic strains and observed opposite phenotypes (100% lethality vs. 100% viability). We characterized the Cre recombinase activity in these two transgenic lines and found that the contrasting phenotypes were driven by difference in embryonic expression of the two transgenes, likely due to position effects or differences in the transgenic constructs. We also tested if SFTPC-Cre activity was dependent on maternal or paternal inheritance. When paternally inherited, both SFTPC-Cre alleles produced offspring with constitutive reporter activity independent of the inheritance of the Cre allele, suggesting that Cre recombinase was expressed in the male germline before meiosis. Immunohistochemical analysis of the testis showed reporter activity during spermatogenesis. Analysis of single-cell RNA sequencing data from murine and human testis demonstrated SFTPC expression uniquely during human spermatogenesis, suggesting that use of the human promoter in these constructs is responsible for male germline activity. Our data highlight the importance of careful analysis of transgenic allele activity and identify an SFTPC-Cre allele that is useful for panepithelial targeting in the mouse.
Collapse
Affiliation(s)
- Mao Jiang
- The Third Xiangya Hospital and Xiangya Hospital, Central South University, Changsha, China.,Division of Pulmonary, Allergy, and Critical Care Medicine, Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, Pennsylvania
| | - Mark G Roth
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, Pennsylvania
| | - Pattra Chun-On
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, Pennsylvania.,Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand; and.,Environmental and Occupational Health Department, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel I Sullivan
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, Pennsylvania
| | - Jonathan K Alder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Pittsburgh, Pennsylvania
| |
Collapse
|
179
|
Cadiz L, Jonz MG. A comparative perspective on lung and gill regeneration. ACTA ACUST UNITED AC 2020; 223:223/19/jeb226076. [PMID: 33037099 DOI: 10.1242/jeb.226076] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The ability to continuously grow and regenerate the gills throughout life is a remarkable property of fish and amphibians. Considering that gill regeneration was first described over one century ago, it is surprising that the underlying mechanisms of cell and tissue replacement in the gills remain poorly understood. By contrast, the mammalian lung is a largely quiescent organ in adults but is capable of facultative regeneration following injury. In the course of the past decade, it has been recognized that lungs contain a population of stem or progenitor cells with an extensive ability to restore tissue; however, despite recent advances in regenerative biology of the lung, the signaling pathways that underlie regeneration are poorly understood. In this Review, we discuss the common evolutionary and embryological origins shared by gills and mammalian lungs. These are evident in homologies in tissue structure, cell populations, cellular function and genetic pathways. An integration of the literature on gill and lung regeneration in vertebrates is presented using a comparative approach in order to outline the challenges that remain in these areas, and to highlight the importance of using aquatic vertebrates as model organisms. The study of gill regeneration in fish and amphibians, which have a high regenerative potential and for which genetic tools are widely available, represents a unique opportunity to uncover common signaling mechanisms that may be important for regeneration of respiratory organs in all vertebrates. This may lead to new advances in tissue repair following lung disease.
Collapse
Affiliation(s)
- Laura Cadiz
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, Canada, K1N 6N5
| | - Michael G Jonz
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, Canada, K1N 6N5
| |
Collapse
|
180
|
Improved detection of tumor suppressor events in single-cell RNA-Seq data. NPJ Genom Med 2020; 5:43. [PMID: 33083012 PMCID: PMC7541488 DOI: 10.1038/s41525-020-00151-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Tissue-specific transcription factors are frequently inactivated in cancer. To fully dissect the heterogeneity of such tumor suppressor events requires single-cell resolution, yet this is challenging because of the high dropout rate. Here we propose a simple yet effective computational strategy called SCIRA to infer regulatory activity of tissue-specific transcription factors at single-cell resolution and use this tool to identify tumor suppressor events in single-cell RNA-Seq cancer studies. We demonstrate that tissue-specific transcription factors are preferentially inactivated in the corresponding cancer cells, suggesting that these are driver events. For many known or suspected tumor suppressors, SCIRA predicts inactivation in single cancer cells where differential expression does not, indicating that SCIRA improves the sensitivity to detect changes in regulatory activity. We identify NKX2-1 and TBX4 inactivation as early tumor suppressor events in normal non-ciliated lung epithelial cells from smokers. In summary, SCIRA can help chart the heterogeneity of tumor suppressor events at single-cell resolution.
Collapse
|
181
|
Lungova V, Thibeault SL. Mechanisms of larynx and vocal fold development and pathogenesis. Cell Mol Life Sci 2020; 77:3781-3795. [PMID: 32253462 PMCID: PMC7511430 DOI: 10.1007/s00018-020-03506-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
The larynx and vocal folds sit at the crossroad between digestive and respiratory tracts and fulfill multiple functions related to breathing, protection and phonation. They develop at the head and trunk interface through a sequence of morphogenetic events that require precise temporo-spatial coordination. We are beginning to understand some of the molecular and cellular mechanisms that underlie critical processes such as specification of the laryngeal field, epithelial lamina formation and recanalization as well as the development and differentiation of mesenchymal cell populations. Nevertheless, many gaps remain in our knowledge, the filling of which is essential for understanding congenital laryngeal disorders and the evaluation and treatment approaches in human patients. This review highlights recent advances in our understanding of the laryngeal embryogenesis. Proposed genes and signaling pathways that are critical for the laryngeal development have a potential to be harnessed in the field of regenerative medicine.
Collapse
Affiliation(s)
- Vlasta Lungova
- Department of Surgery, University of Wisconsin Madison, 5103 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Susan L Thibeault
- Department of Surgery, University of Wisconsin Madison, 5103 WIMR, 1111 Highland Ave, Madison, WI, 53705, USA.
| |
Collapse
|
182
|
Miao B, Fu S, Lyu C, Gontarz P, Wang T, Zhang B. Tissue-specific usage of transposable element-derived promoters in mouse development. Genome Biol 2020; 21:255. [PMID: 32988383 PMCID: PMC7520981 DOI: 10.1186/s13059-020-02164-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Transposable elements (TEs) are a significant component of eukaryotic genomes and play essential roles in genome evolution. Mounting evidence indicates that TEs are highly transcribed in early embryo development and contribute to distinct biological functions and tissue morphology. RESULTS We examine the epigenetic dynamics of mouse TEs during the development of five tissues: intestine, liver, lung, stomach, and kidney. We found that TEs are associated with over 20% of open chromatin regions during development. Close to half of these accessible TEs are only activated in a single tissue and a specific developmental stage. Most accessible TEs are rodent-specific. Across these five tissues, 453 accessible TEs are found to create the transcription start sites of downstream genes in mouse, including 117 protein-coding genes and 144 lincRNA genes, 93.7% of which are mouse-specific. Species-specific TE-derived transcription start sites are found to drive the expression of tissue-specific genes and change their tissue-specific expression patterns during evolution. CONCLUSION Our results suggest that TE insertions increase the regulatory potential of the genome, and some TEs have been domesticated to become a crucial component of gene and regulate tissue-specific expression during mouse tissue development.
Collapse
Affiliation(s)
- Benpeng Miao
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63108, USA
- Department of Genetics, Edison Family Center for Genomic Sciences and Systems Biology, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Shuhua Fu
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Cheng Lyu
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Paul Gontarz
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63108, USA
| | - Ting Wang
- Department of Genetics, Edison Family Center for Genomic Sciences and Systems Biology, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63108, USA.
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63108, USA.
| |
Collapse
|
183
|
Scudieri P, Musante I, Venturini A, Guidone D, Genovese M, Cresta F, Caci E, Palleschi A, Poeta M, Santamaria F, Ciciriello F, Lucidi V, Galietta LJV. Ionocytes and CFTR Chloride Channel Expression in Normal and Cystic Fibrosis Nasal and Bronchial Epithelial Cells. Cells 2020; 9:cells9092090. [PMID: 32933106 PMCID: PMC7565890 DOI: 10.3390/cells9092090] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022] Open
Abstract
The airway epithelium contains ionocytes, a rare cell type with high expression of Forkhead Box I1 (FOXI1) transcription factor and Cystic Fibrosis Transmembrane conductance Regulator (CFTR), a chloride channel that is defective in cystic fibrosis (CF). Our aim was to verify if ionocyte development is altered in CF and to investigate the relationship between ionocytes and CFTR-dependent chloride secretion. We collected nasal cells by brushing to determine ionocyte abundance. Nasal and bronchial cells were also expanded in vitro and reprogrammed to differentiated epithelia for morphological and functional studies. We found a relatively high (~3%) ionocyte abundance in ex vivo nasal samples, with no difference between CF and control individuals. In bronchi, ionocytes instead appeared very rarely as previously reported, thus suggesting a possible proximal-distal gradient in human airways. The difference between nasal and bronchial epithelial cells was maintained in culture, which suggests an epigenetic control of ionocyte development. In the differentiation phase of the culture procedure, we used two media that resulted in a different pattern of CFTR expression: confined to ionocytes or more broadly expressed. CFTR function was similar in both conditions, thus indicating that chloride secretion equally occurs irrespective of CFTR expression pattern.
Collapse
Affiliation(s)
- Paolo Scudieri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genova, 16147 Genova, Italy; (P.S.); (I.M.)
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Ilaria Musante
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genova, 16147 Genova, Italy; (P.S.); (I.M.)
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Arianna Venturini
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Federico Cresta
- Centro Fibrosi Cistica, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Emanuela Caci
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Alessandro Palleschi
- Thoracic Surgery and Lung Transplantation Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Marco Poeta
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
| | - Francesca Santamaria
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
| | - Fabiana Ciciriello
- Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, 00165 Roma, Italy; (F.C.); (V.L.)
| | - Vincenzina Lucidi
- Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, 00165 Roma, Italy; (F.C.); (V.L.)
| | - Luis J. V. Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
- Correspondence:
| |
Collapse
|
184
|
Mullen DJ, Yan C, Kang DS, Zhou B, Borok Z, Marconett CN, Farnham PJ, Offringa IA, Rhie SK. TENET 2.0: Identification of key transcriptional regulators and enhancers in lung adenocarcinoma. PLoS Genet 2020; 16:e1009023. [PMID: 32925947 PMCID: PMC7515200 DOI: 10.1371/journal.pgen.1009023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 09/24/2020] [Accepted: 08/02/2020] [Indexed: 01/09/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death and lung adenocarcinoma is its most common subtype. Although genetic alterations have been identified as drivers in subsets of lung adenocarcinoma, they do not fully explain tumor development. Epigenetic alterations have been implicated in the pathogenesis of tumors. To identify epigenetic alterations driving lung adenocarcinoma, we used an improved version of the Tracing Enhancer Networks using Epigenetic Traits method (TENET 2.0) in primary normal lung and lung adenocarcinoma cells. We found over 32,000 enhancers that appear differentially activated between normal lung and lung adenocarcinoma. Among the identified transcriptional regulators inactivated in lung adenocarcinoma vs. normal lung, NKX2-1 was linked to a large number of silenced enhancers. Among the activated transcriptional regulators identified, CENPA, FOXM1, and MYBL2 were linked to numerous cancer-specific enhancers. High expression of CENPA, FOXM1, and MYBL2 is particularly observed in a subgroup of lung adenocarcinomas and is associated with poor patient survival. Notably, CENPA, FOXM1, and MYBL2 are also key regulators of cancer-specific enhancers in breast adenocarcinoma of the basal subtype, but they are associated with distinct sets of activated enhancers. We identified individual lung adenocarcinoma enhancers linked to CENPA, FOXM1, or MYBL2 that were associated with poor patient survival. Knockdown experiments of FOXM1 and MYBL2 suggest that these factors regulate genes involved in controlling cell cycle progression and cell division. For example, we found that expression of TK1, a potential target gene of a MYBL2-linked enhancer, is associated with poor patient survival. Identification and characterization of key transcriptional regulators and associated enhancers in lung adenocarcinoma provides important insights into the deregulation of lung adenocarcinoma epigenomes, highlighting novel potential targets for clinical intervention.
Collapse
Affiliation(s)
- Daniel J. Mullen
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Chunli Yan
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Diane S. Kang
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Beiyun Zhou
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Zea Borok
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Crystal N. Marconett
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Peggy J. Farnham
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Ite A. Offringa
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, CA, United States of America
| | - Suhn Kyong Rhie
- Department of Biochemistry and Molecular Medicine and the Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, CA, United States of America
| |
Collapse
|
185
|
Sucre JMS, Vickers KC, Benjamin JT, Plosa EJ, Jetter CS, Cutrone A, Ransom M, Anderson Z, Sheng Q, Fensterheim BA, Ambalavanan N, Millis B, Lee E, Zijlstra A, Königshoff M, Blackwell TS, Guttentag SH. Hyperoxia Injury in the Developing Lung Is Mediated by Mesenchymal Expression of Wnt5A. Am J Respir Crit Care Med 2020; 201:1249-1262. [PMID: 32023086 DOI: 10.1164/rccm.201908-1513oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rationale: Bronchopulmonary dysplasia (BPD) is a leading complication of preterm birth that affects infants born in the saccular stage of lung development at <32 weeks of gestation. Although the mechanisms driving BPD remain uncertain, exposure to hyperoxia is thought to contribute to disease pathogenesis.Objectives: To determine the effects of hyperoxia on epithelial-mesenchymal interactions and to define the mediators of activated Wnt/β-catenin signaling after hyperoxia injury.Methods: Three hyperoxia models were used: A three-dimensional organotypic coculture using primary human lung cells, precision-cut lung slices (PCLS), and a murine in vivo hyperoxia model. Comparisons of normoxia- and hyperoxia-exposed samples were made by real-time quantitative PCR, RNA in situ hybridization, quantitative confocal microscopy, and lung morphometry.Measurements and Main Results: Examination of an array of Wnt ligands in the three-dimensional organotypic coculture revealed increased mesenchymal expression of WNT5A. Inhibition of Wnt5A abrogated the BPD transcriptomic phenotype induced by hyperoxia. In the PCLS model, Wnt5A inhibition improved alveolarization following hyperoxia exposure, and treatment with recombinant Wnt5a reproduced features of the BPD phenotype in PCLS cultured in normoxic conditions. Chemical inhibition of NF-κB with BAY11-7082 reduced Wnt5a expression in the PCLS hyperoxia model and in vivo mouse hyperoxia model, with improved alveolarization in the PCLS model.Conclusions: Increased mesenchymal Wnt5A during saccular-stage hyperoxia injury contributes to the impaired alveolarization and septal thickening observed in BPD. Precise targeting of Wnt5A may represent a potential therapeutic strategy for the treatment of BPD.
Collapse
Affiliation(s)
- Jennifer M S Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics.,Department of Cell and Developmental Biology, and
| | | | - John T Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Erin J Plosa
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | | | - Alissa Cutrone
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | - Benjamin A Fensterheim
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan Millis
- Department of Cell and Developmental Biology, and.,Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Ethan Lee
- Department of Cell and Developmental Biology, and
| | | | - Melanie Königshoff
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado; and
| | - Timothy S Blackwell
- Department of Cell and Developmental Biology, and.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | | |
Collapse
|
186
|
Körner RW, Majjouti M, Alcazar MAA, Mahabir E. Of Mice and Men: The Coronavirus MHV and Mouse Models as a Translational Approach to Understand SARS-CoV-2. Viruses 2020; 12:E880. [PMID: 32806708 PMCID: PMC7471983 DOI: 10.3390/v12080880] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
The fatal acute respiratory coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since COVID-19 was declared a pandemic by the World Health Organization in March 2020, infection and mortality rates have been rising steadily worldwide. The lack of a vaccine, as well as preventive and therapeutic strategies, emphasize the need to develop new strategies to mitigate SARS-CoV-2 transmission and pathogenesis. Since mouse hepatitis virus (MHV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2 share a common genus, lessons learnt from MHV and SARS-CoV could offer mechanistic insights into SARS-CoV-2. This review provides a comprehensive review of MHV in mice and SARS-CoV-2 in humans, thereby highlighting further translational avenues in the development of innovative strategies in controlling the detrimental course of SARS-CoV-2. Specifically, we have focused on various aspects, including host species, organotropism, transmission, clinical disease, pathogenesis, control and therapy, MHV as a model for SARS-CoV and SARS-CoV-2 as well as mouse models for infection with SARS-CoV and SARS-CoV-2. While MHV in mice and SARS-CoV-2 in humans share various similarities, there are also differences that need to be addressed when studying murine models. Translational approaches, such as humanized mouse models are pivotal in studying the clinical course and pathology observed in COVID-19 patients. Lessons from prior murine studies on coronavirus, coupled with novel murine models could offer new promising avenues for treatment of COVID-19.
Collapse
Affiliation(s)
- Robert W. Körner
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Mohamed Majjouti
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany;
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Member of the German Center for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Center (UGMLC), 50937 Cologne, Germany
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany;
| |
Collapse
|
187
|
Notch Transduction in Non-Small Cell Lung Cancer. Int J Mol Sci 2020; 21:ijms21165691. [PMID: 32784481 PMCID: PMC7461113 DOI: 10.3390/ijms21165691] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily-conserved Notch signaling pathway plays critical roles in cell communication, function and homeostasis equilibrium. The pathway serves as a cell-to-cell juxtaposed molecular transducer and is crucial in a number of cell processes including cell fate specification, asymmetric cell division and lateral inhibition. Notch also plays critical roles in organismal development, homeostasis, and regeneration, including somitogenesis, left-right asymmetry, neurogenesis, tissue repair, self-renewal and stemness, and its dysregulation has causative roles in a number of congenital and acquired pathologies, including cancer. In the lung, Notch activity is necessary for cell fate specification and expansion, and its aberrant activity is markedly linked to various defects in club cell formation, alveologenesis, and non-small cell lung cancer (NSCLC) development. In this review, we focus on the role this intercellular signaling device plays during lung development and on its functional relevance in proximo-distal cell fate specification, branching morphogenesis, and alveolar cell determination and maturation, then revise its involvement in NSCLC formation, progression and treatment refractoriness, particularly in the context of various mutational statuses associated with NSCLC, and, lastly, conclude by providing a succinct outlook of the therapeutic perspectives of Notch targeting in NSCLC therapy, including an overview on prospective synthetic lethality approaches.
Collapse
|
188
|
Ren Y, Lyu Y, Mereness JA, Wang S, Pang J, Mariani TJ. Rare Pulmonary Connective Tissue Type Mast Cells Regulate Lung Endothelial Cell Angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1763-1773. [PMID: 32450152 PMCID: PMC9808505 DOI: 10.1016/j.ajpath.2020.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/24/2020] [Accepted: 04/27/2020] [Indexed: 01/07/2023]
Abstract
Within the human lung, mast cells typically reside adjacent to the conducting airway and assume a mucosal phenotype (MCT). In rare pathologic conditions, connective tissue phenotype mast cells (MCTCs) can be found in the lung parenchyma. MCTCs accumulate in the lungs of infants with severe bronchopulmonary dysplasia, a chronic lung disease associated with preterm birth, which is characterized by pulmonary vascular dysmorphia. The human mast cell line (LUVA) was used to model MCTCs or MCTs. The ability of MCTCs to affect vascular organization during fetal lung development was tested in mouse lung explant cultures. The effect of MCTCs on in vitro tube formation and barrier function was studied using primary fetal human pulmonary microvascular endothelial cells. The mechanistic role of MCTC proteases was tested using inhibitors. MCTCLUVA but not MCTLUVA was associated with vascular dysmorphia in lung explants. In vitro, the addition of MCTCLUVA potentiated fetal human pulmonary microvascular endothelial cell interactions, inhibited tube stability, and disrupted endothelial cell junctions. Protease inhibitors ameliorated the ability of MCTCLUVA to alter endothelial cell angiogenic activities in vitro and ex vivo. These data indicate that MCTCs may directly contribute to disrupted angiogenesis in bronchopulmonary dysplasia. A better understanding of factors that regulate mast cell subtype and their different effector functions is essential.
Collapse
Affiliation(s)
- Yue Ren
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester, Rochester, New York; Department of Biology, University of Rochester, Rochester, New York
| | - Yuyan Lyu
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Jared A Mereness
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York
| | - Shumin Wang
- Department of Pediatrics, University of Rochester, Rochester, New York; Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York
| | - Jinjiang Pang
- Department of Pediatrics, University of Rochester, Rochester, New York; Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester, Rochester, New York; Department of Biomedical Genetics, University of Rochester, Rochester, New York.
| |
Collapse
|
189
|
Bartman CM, Matveyenko A, Prakash YS. It's about time: clocks in the developing lung. J Clin Invest 2020; 130:39-50. [PMID: 31895049 DOI: 10.1172/jci130143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The discovery of peripheral intracellular clocks revealed circadian oscillations of clock genes and their targets in all cell types, including those in the lung, sparking exploration of clocks in lung disease pathophysiology. While the focus has been on the role of these clocks in adult airway diseases, clock biology is also likely to be important in perinatal lung development, where it has received far less attention. Historically, fetal circadian rhythms have been considered irrelevant owing to lack of external light exposure, but more recent insights into peripheral clock biology raise questions of clock emergence, its concordance with tissue-specific structure/function, the interdependence of clock synchrony and functionality in perinatal lung development, and the possibility of lung clocks in priming the fetus for postnatal life. Understanding the perinatal molecular clock may unravel mechanistic targets for chronic airway disease across the lifespan. With current research providing more questions than answers, it is about time to investigate clocks in the developing lung.
Collapse
Affiliation(s)
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine and.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
190
|
Strunz M, Simon LM, Ansari M, Kathiriya JJ, Angelidis I, Mayr CH, Tsidiridis G, Lange M, Mattner LF, Yee M, Ogar P, Sengupta A, Kukhtevich I, Schneider R, Zhao Z, Voss C, Stoeger T, Neumann JHL, Hilgendorff A, Behr J, O'Reilly M, Lehmann M, Burgstaller G, Königshoff M, Chapman HA, Theis FJ, Schiller HB. Alveolar regeneration through a Krt8+ transitional stem cell state that persists in human lung fibrosis. Nat Commun 2020; 11:3559. [PMID: 32678092 PMCID: PMC7366678 DOI: 10.1038/s41467-020-17358-3] [Citation(s) in RCA: 416] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022] Open
Abstract
The cell type specific sequences of transcriptional programs during lung regeneration have remained elusive. Using time-series single cell RNA-seq of the bleomycin lung injury model, we resolved transcriptional dynamics for 28 cell types. Trajectory modeling together with lineage tracing revealed that airway and alveolar stem cells converge on a unique Krt8 + transitional stem cell state during alveolar regeneration. These cells have squamous morphology, feature p53 and NFkB activation and display transcriptional features of cellular senescence. The Krt8+ state appears in several independent models of lung injury and persists in human lung fibrosis, creating a distinct cell-cell communication network with mesenchyme and macrophages during repair. We generated a model of gene regulatory programs leading to Krt8+ transitional cells and their terminal differentiation to alveolar type-1 cells. We propose that in lung fibrosis, perturbed molecular checkpoints on the way to terminal differentiation can cause aberrant persistence of regenerative intermediate stem cell states.
Collapse
Affiliation(s)
- Maximilian Strunz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Lukas M Simon
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - Meshal Ansari
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Jaymin J Kathiriya
- Biomedical Center, University of California San Francisco, San Francisco, CA, USA
| | - Ilias Angelidis
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christoph H Mayr
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - George Tsidiridis
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Marius Lange
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Laura F Mattner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Min Yee
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Paulina Ogar
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Arunima Sengupta
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Igor Kukhtevich
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Munich, Germany
| | - Robert Schneider
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Munich, Germany
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - Carola Voss
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Tobias Stoeger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jens H L Neumann
- Institute of Pathology, Ludwig Maximilians University Hospital Munich, Munich, Germany
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Member of the German Center for Lung Research (DZL), Center for Comprehensive Developmental Care (CDeCLMU), Department of Neonatology, Perinatal Center Grosshadern, Hospital of the Ludwig-Maximilians University (LMU), Munich, Germany
| | - Jürgen Behr
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Member of the German Center for Lung Research (DZL), Department of Internal Medicine V, Ludwig Maximilians University Hospital (LMU) Munich, Munich, Germany
- Asklepios Fachkliniken in Munich-Gauting, Munich, Germany
| | - Michael O'Reilly
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Mareike Lehmann
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- University of Colorado, Department of Pulmonary Sciences and Critical Care Medicine, Denver, CO, USA
| | - Harold A Chapman
- Biomedical Center, University of California San Francisco, San Francisco, CA, USA
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Department of Mathematics, Technische Universität München, Munich, Germany.
| | - Herbert B Schiller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
191
|
Okolo F, Zhang G, Rhodes J, Gittes GK, Potoka DA. Intra-Amniotic Sildenafil Treatment Promotes Lung Growth and Attenuates Vascular Remodeling in an Experimental Model of Congenital Diaphragmatic Hernia. Fetal Diagn Ther 2020; 47:787-799. [PMID: 32663823 DOI: 10.1159/000508986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/26/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Defective lung development resulting in lung hypoplasia and an attenuated and hypermuscularized pulmonary vasculature contributes to significant postnatal mortality in congenital diaphragmatic hernia (CDH). We hypothesize that deficient embryonic pulmonary blood flow contributes to defective lung development in CDH, which may therefore be ameliorated via enhancement of embryonic pulmonary blood flow. METHODS The mouse nitrofen model of CDH was utilized to measure embryonic pulmonary blood flow by in utero intracardiac injection of FITC-labeled tomato lectin and color-flow Doppler ultrasound. The effect of prenatal intra-amniotic treatment with sildenafil on survival, lung growth, and vascular morphology in the nitrofen model was determined. RESULTS Nitrofen-treated embryos exhibited decreased blood flow in the lung periphery compared to controls, and intra-amniotic sildenafil significantly improved embryonic pulmonary blood flow. Similar to nitrofen alone, pups delivered after nitrofen treatment and intra-amniotic injection of dextrose control exhibited respiratory distress and never survived beyond 6 h. Intra-amniotic sildenafil ameliorated respiratory distress in nitrofen-treated pups and improved postnatal survival to 82%. Following intra-amniotic sildenafil treatment at embryonic day (E)10.5, nitrofen-treated P0 lungs were larger with increased left lobe weight, reduced small pulmonary arterial wall muscularization, and increased airway branching complexity compared to controls. Intra-amniotic sildenafil treatment later at E15.5 also resulted in improved survival, lung growth, and attenuation of vascular remodeling in nitrofen-treated embryos. CONCLUSIONS Defective embryonic pulmonary blood flow may contribute to lung maldevelopment in CDH. Enhancement of embryonic pulmonary blood flow via intra-amniotic sildenafil results in lung growth and attenuation of pulmonary vascular remodeling and may have therapeutic potential for CDH.
Collapse
Affiliation(s)
- Frances Okolo
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - GuangFeng Zhang
- Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Julie Rhodes
- Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA,
| | - Douglas A Potoka
- Department of Surgery, University of Pittsburgh School of Medicine and Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
192
|
Sun Y, Xia Y, Liu X, Liu J, He W, Ye H, Yuan X. Dexmedetomidine alleviates LPS‑induced acute lung injury via regulation of the p38/HO‑1 pathway. Mol Med Rep 2020; 22:2442-2450. [PMID: 32705282 PMCID: PMC7411448 DOI: 10.3892/mmr.2020.11330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is a common critical illness in clinical anesthesia and the intensive care unit that can cause acute hypoxic respiratory insufficiency. Despite various therapeutic regimes having been investigated, there is currently no effective pharmacotherapy available to treat ALI. Previous studies have reported that the NOD-like receptor protein 3 (NLRP3) signaling pathway plays an important role in the inflammatory response and is involved in the pathogenesis of ALI. Moreover, dexmedetomidine (Dex), an α2-adrenergic receptor activating agent, has been routinely used as an adjuvant therapy in treating inflammatory diseases, including ALI. However, the precise pathological mechanisms of Dex in ALI remain to be elucidated. Thus, the present study aimed to investigate the effects of the p38/heme oxygenase 1 (HO-1) signaling pathways in the pathological mechanisms of Dex in ALI. Newborn male Sprague-Dawley rats (n=48) were randomly divided into four groups (n=12 each), and an intravenous injection of lipopolysaccharide (LPS) was used to successfully induce the ALI model, with increased pulmonary damage, cell apoptosis, interleukin-1β (IL-1β) secretion and edema fluid in lungs. Moreover, the mRNA and protein expression levels of NLRP3 were significantly upregulated, while that of HO-1 were downregulated by LPS treatment. Furthermore, the levels of phosphorylated p38 were also upregulated in ALI rats. It was demonstrated that Dex administration significantly alleviated LPS-induced ALI, downregulated the secretion of IL-1β, decreased the expression of NLRP3, inhibited the phospho-activation of p38 and increased HO-1 expression. In addition, pharmacological inhibition of p38 using the inhibitor SB20380 further enhanced the effect of Dex. Collectively, these preliminarily results identified the effects of Dex intervention on the pathogenesis of ALI via the regulation of p38/HO-1 signaling pathways, which impacted the inflammatory effects, thus providing a theoretical basis and novel evidence for the development of new targets for clinical treatment of ALI.
Collapse
Affiliation(s)
- Yingying Sun
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Yin Xia
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xinghui Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Junxia Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Weitian He
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Hongwu Ye
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Xianren Yuan
- Department of Anesthesiology, Anhui Provincial Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| |
Collapse
|
193
|
Affiliation(s)
- David N Cornfield
- Department of Pediatricsand
- Center for Excellence in Pulmonary BiologyStanford University School of MedicineStanford, California
| |
Collapse
|
194
|
Zhang L, Valizadeh H, Alipourfard I, Bidares R, Aghebati-Maleki L, Ahmadi M. Epigenetic Modifications and Therapy in Chronic Obstructive Pulmonary Disease (COPD): An Update Review. COPD 2020; 17:333-342. [PMID: 32558592 DOI: 10.1080/15412555.2020.1780576] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) that is one of the most prevalent chronic adult diseases and the third leading cause of fatality until 2020. Elastase/anti-elastase hypothesis, chronic inflammation, apoptosis, oxidant-antioxidant balance and infective repair cause pathogenesis of COPD are among the factors at play. Epigenetic changes are post-translational modifications in histone proteins and DNA such as methylation and acetylation as well as dysregulation of miRNAs expression. In this update review, we have examined recent studies on the upregulation or downregulation of methylation in different genes associated with COPD. Dysregulation of HDAC activity which is caused by some factors and miRNAs plays a key role in the suppression and reduction of COPD development. Also, some therapeutic approaches are proposed against COPD by targeting HDAC2 and miRNAs, which have therapeutic effects.
Collapse
Affiliation(s)
- Lingzhi Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hamed Valizadeh
- Department of Internal Medicine and Pulmonology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Iraj Alipourfard
- Faculty of Life Sciences, Center of pharmaceutical sciences, University of Vienna, Vienna, Austria.,Faculty of Sciences, School of Pharmacy, University of Rome Tor Vergata, Roma, Italy
| | - Ramtin Bidares
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
195
|
Dylag AM, Haak J, Yee M, O’Reilly MA. Pulmonary mechanics and structural lung development after neonatal hyperoxia in mice. Pediatr Res 2020; 87:1201-1210. [PMID: 31835269 PMCID: PMC7255955 DOI: 10.1038/s41390-019-0723-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/22/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Supplemental oxygen exposure administered to premature infants is associated with chronic lung disease and abnormal pulmonary function. This study used mild (40%), moderate (60%), and severe (80%) oxygen to determine how hyperoxia-induced changes in lung structure impact pulmonary mechanics in mice. METHODS C57BL/6J mice were exposed to room air or hyperoxia from birth through postnatal day 8. Baseline pulmonary function and methacholine challenge was assessed at 4 and 8 weeks of age, accompanied by immunohistochemical assessments of both airway (smooth muscle, tethering) and alveolar (simplification, elastin deposition) structure. RESULTS Mild/moderate hyperoxia increased baseline airway resistance (40% only) and airway hyperreactivity (40 and 60%) at 4 weeks accompanied by increased airway smooth muscle deposition, which resolved at 8 weeks. Severe hyperoxia increased baseline compliance, baseline resistance, and total elastin/surface area ratio without increasing airway hyperreactivity, and was accompanied by increased alveolar simplification, decreased airway tethering, and changes in elastin distribution at both time points. CONCLUSIONS Mild to moderate hyperoxia causes changes in airway function and airway hyperreactivity with minimal parenchymal response. Severe hyperoxia drives its functional changes through alveolar simplification, airway tethering, and elastin redistribution. These differential responses can be leveraged to further develop hyperoxia mouse models.
Collapse
Affiliation(s)
- Andrew M. Dylag
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Jeannie Haak
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Min Yee
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael A. O’Reilly
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
196
|
Abstract
ABSTRACT
Over the past 5 years, several studies have begun to uncover the links between the classical signal transduction pathways and the physical mechanisms that are used to sculpt branched tissues. These advances have been made, in part, thanks to innovations in live imaging and reporter animals. With modern research tools, our conceptual models of branching morphogenesis are rapidly evolving, and the differences in branching mechanisms between each organ are becoming increasingly apparent. Here, we highlight four branched epithelia that develop at different spatial scales, within different surrounding tissues and via divergent physical mechanisms. Each of these organs has evolved to employ unique branching strategies to achieve a specialized final architecture.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M. Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
197
|
Evidence for Overlapping and Distinct Biological Activities and Transcriptional Targets Triggered by Fibroblast Growth Factor Receptor 2b Signaling between Mid- and Early Pseudoglandular Stages of Mouse Lung Development. Cells 2020; 9:cells9051274. [PMID: 32455591 PMCID: PMC7290466 DOI: 10.3390/cells9051274] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/12/2020] [Accepted: 04/20/2020] [Indexed: 12/17/2022] Open
Abstract
Branching morphogenesis is the basic developmental mode common to organs such as the lungs that undergo a process of ramification from a rudimentary tree. However, the precise molecular and cellular bases underlying the formation of branching organs are still unclear. As inactivation of fibroblast growth factor receptor 2b (Fgfr2b) signaling during early development leads to lung agenesis, thereby preventing the analysis of this pathway at later developmental stages, we used transgenic mice to induce expression of a soluble form of Fgfr2b to inactivate Fgfr2b ligands at embryonic day (E) 14.5, corresponding to the mid-pseudoglandular stage of lung development. We identified an Fgfr2b signaling signature comprised of 46 genes enriched in the epithelium, some of which were common to, but most of them distinct from, the previously identified Fgfr2b signaling signature at E12.5. Our results indicate that Fgfr2b signaling at E14.5 controls mostly proliferation and alveolar type 2 cell (AT2) differentiation. In addition, inhibition of Fgfr2b signaling at E14.5 leads to morphological and cellular impairment at E18.5, with defective alveolar lineage formation. Further studies will have to be conducted to elucidate the role of Fgfr2b signaling at successive stages (canalicular/saccular/alveolar) of lung development as well as during homeostasis and regeneration and repair after injury.
Collapse
|
198
|
Paranjapye A, Mutolo MJ, Ebron JS, Leir SH, Harris A. The FOXA1 transcriptional network coordinates key functions of primary human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2020; 319:L126-L136. [PMID: 32432922 DOI: 10.1152/ajplung.00023.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The differentiated functions of the human airway epithelium are coordinated by a complex network of transcription factors. These include the pioneer factors Forkhead box A1 and A2 (FOXA1 and FOXA2), which are well studied in several tissues, but their role in airway epithelial cells is poorly characterized. Here, we define the cistrome of FOXA1 and FOXA2 in primary human bronchial epithelial (HBE) cells by chromatin immunoprecipitation with deep-sequencing (ChIP-seq). Next, siRNA-mediated depletion of each factor is used to investigate their transcriptome by RNA-seq. We found that, as predicted from their DNA-binding motifs, genome-wide occupancy of the two factors showed substantial overlap; however, their global impact on gene expression differed. FOXA1 is an abundant transcript in HBE cells, while FOXA2 is expressed at low levels, and both these factors likely exhibit autoregulation and cross-regulation. FOXA1 regulated loci are involved in cell adhesion and the maintenance of epithelial cell identity, particularly through repression of genes associated with epithelial to mesenchymal transition (EMT). FOXA1 also directly targets other transcription factors with a known role in the airway epithelium such as SAM-pointed domain-containing Ets-like factor (SPDEF). The intersection of the cistrome and transcriptome for FOXA1 revealed enrichment of genes involved in epithelial development and tissue morphogenesis. Moreover, depletion of FOXA1 was shown to reduce the transepithelial resistance of HBE cells, confirming the role of this factor in maintaining epithelial barrier integrity.
Collapse
Affiliation(s)
- Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Michael J Mutolo
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Jey Sabith Ebron
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
199
|
Kennedy JC, Khabibullin D, Hougard T, Nijmeh J, Shi W, Henske EP. Loss of FLCN inhibits canonical WNT signaling via TFE3. Hum Mol Genet 2020; 28:3270-3281. [PMID: 31272105 DOI: 10.1093/hmg/ddz158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/10/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022] Open
Abstract
Lower lobe predominant pulmonary cysts occur in up to 90% of patients with Birt-Hogg-Dubé (BHD) syndrome, but the key pathologic cell type and signaling events driving this distinct phenotype remain elusive. Through examination of the LungMAP database, we found that folliculin (FLCN) is highly expressed in neonatal lung mesenchymal cells. Using RNA-Seq, we found that inactivation of Flcn in mouse embryonic fibroblasts leads to changes in multiple Wnt ligands, including a 2.8-fold decrease in Wnt2. This was associated with decreased TCF/LEF activity, a readout of canonical WNT activity, after treatment with a GSK3-α/β inhibitor. Similarly, FLCN deficiency in HEK293T cells decreased WNT pathway activity by 76% post-GSK3-α/β inhibition. Inactivation of FLCN in human fetal lung fibroblasts (MRC-5) led to ~ 100-fold decrease in Wnt2 expression and a 33-fold decrease in Wnt7b expression-two ligands known to be necessary for lung development. Furthermore, canonical WNT activity was decreased by 60%. Classic WNT targets such as AXIN2 and BMP4, and WNT enhanceosome members including TCF4, LEF1 and BCL9 were also decreased after GSK3-α/β inhibition. FLCN-deficient MRC-5 cells failed to upregulate LEF1 in response to GSK3-α/β inhibition. Finally, we found that a constitutively active β-catenin could only partially rescue the decreased WNT activity phenotype seen in FLCN-deficient cells, whereas silencing the transcription factor TFE3 completely reversed this phenotype. In summary, our data establish FLCN as a critical regulator of the WNT pathway via TFE3 and suggest that FLCN-dependent defects in WNT pathway developmental cues may contribute to lung cyst pathogenesis in BHD.
Collapse
Affiliation(s)
- John C Kennedy
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.,Division of Pulmonary and Respiratory Diseases, Boston Children's Hospital, Boston, MA 02115, USA
| | - Damir Khabibullin
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Hougard
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Julie Nijmeh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Wei Shi
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Elizabeth P Henske
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
200
|
Yue H, Ji X, Ku T, Li G, Sang N. Sex difference in bronchopulmonary dysplasia of offspring in response to maternal PM 2.5 exposure. JOURNAL OF HAZARDOUS MATERIALS 2020; 389:122033. [PMID: 32004849 DOI: 10.1016/j.jhazmat.2020.122033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 01/05/2020] [Indexed: 06/10/2023]
Abstract
The adverse effects of fine particulate matters (PM2.5) on respiratory diseases start in utero. In order to investigate whether maternal PM2.5 exposure could lead to bronchopulmonary dysplasia (BPD) in offspring, PM2.5 was collected in Taiyuan, Shanxi, China during the annual heating period. Mice were mated and gestation day 0 (GD0) was considered the day on which a vaginal plug was observed. The plug-positive mice received 3 mg/kg b.w. PM2.5 by oropharyngeal aspiration every other day starting on GD0 and throughout the gestation period. Offspring were sacrificed at postnatal days (PNDs) 1, 7, 14 and 21. We assessed some typical BPD-like symptoms in offspring. The results showed that maternal PM2.5 exposure caused low birth weight, hypoalveolarization, decreased angiogenesis, suppressed production of secretory and surfactant proteins, and increased inflammation in the lungs of male offspring. However, maternal PM2.5 exposure induced only hypoalveolarization and inflammation in the lungs of female offspring. Furthermore, these alterations were reversed during postnatal development. Our results demonstrated that maternal exposure to PM2.5 caused reversible BPD-related consequences in offspring, and male offspring were more sensitive than females. However, these alterations were reversed during postnatal development.
Collapse
Affiliation(s)
- Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|