1
|
Shirazi SP, Negretti NM, Jetter CS, Sharkey AL, Garg S, Kapp ME, Wilkins D, Fortier G, Mallapragada S, Banovich NE, Wright CVE, Frank DB, Kropski JA, Sucre JMS. Bronchopulmonary Dysplasia with Pulmonary Hypertension Associates with Loss of Semaphorin Signaling and Functional Decrease in FOXF1 Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609955. [PMID: 39253423 PMCID: PMC11383282 DOI: 10.1101/2024.08.27.609955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Lung injury in preterm infants leads to structural and functional respiratory deficits, with a risk for bronchopulmonary dysplasia (BPD) that in its most severe form is accompanied by pulmonary hypertension (PH). To examine cellular and molecular dynamics driving evolving BPD in humans, we performed single-cell RNA sequencing of preterm infant lungs in early stages of BPD and BPD+PH compared to term infants. Analysis of the endothelium revealed a unique aberrant capillary cell-state primarily in BPD+PH marked by ANKRD1 expression. Predictive signaling analysis identified deficits in the semaphorin guidance-cue signaling pathway and decreased expression of pro-angiogenic transcription factor FOXF1 within the alveolar parenchyma in neonatal lung samples with BPD/BPD+PH. Loss of semaphorin signaling was replicated in a murine BPD model and in humans with alveolar capillary dysplasia (ACDMPV), suggesting a mechanistic link between the developmental programs underlying BPD and ACDMPV and a critical role for semaphorin signaling in normal lung development.
Collapse
|
2
|
Taylor J, Dubois F, Bergot E, Levallet G. Targeting the Hippo pathway to prevent radioresistance brain metastases from the lung (Review). Int J Oncol 2024; 65:68. [PMID: 38785155 PMCID: PMC11155713 DOI: 10.3892/ijo.2024.5656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/04/2024] [Indexed: 05/25/2024] Open
Abstract
The prognosis for patients with non‑small cell lung cancer (NSCLC), a cancer type which represents 85% of all lung cancers, is poor with a 5‑year survival rate of 19%, mainly because NSCLC is diagnosed at an advanced and metastatic stage. Despite recent therapeutic advancements, ~50% of patients with NSCLC will develop brain metastases (BMs). Either surgical BM treatment alone for symptomatic patients and patients with single cerebral metastases, or in combination with stereotactic radiotherapy (RT) for patients who are not suitable for surgery or presenting with fewer than four cerebral lesions with a diameter range of 5‑30 mm, or whole‑brain RT for numerous or large BMs can be administered. However, radioresistance (RR) invariably prevents the action of RT. Several mechanisms of RR have been described including hypoxia, cellular stress, presence of cancer stem cells, dysregulation of apoptosis and/or autophagy, dysregulation of the cell cycle, changes in cellular metabolism, epithelial‑to‑mesenchymal transition, overexpression of programmed cell death‑ligand 1 and activation several signaling pathways; however, the role of the Hippo signaling pathway in RR is unclear. Dysregulation of the Hippo pathway in NSCLC confers metastatic properties, and inhibitors targeting this pathway are currently in development. It is therefore essential to evaluate the effect of inhibiting the Hippo pathway, particularly the effector yes‑associated protein‑1, on cerebral metastases originating from lung cancer.
Collapse
Affiliation(s)
- Jasmine Taylor
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
| | - Fatéméh Dubois
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pathology, and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| | - Emmanuel Bergot
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pneumology and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| | - Guénaëlle Levallet
- University of Caen Normandy, National Center for Scientific Research, Normandy University, Unit of Imaging and Therapeutic Strategies for Cancers and Cerebral Tissues (ISTCT)-UMR6030, GIP CYCERON, F-14074 Caen, France
- Departments of Pathology, and Thoracic Oncology, Caen University Hospital, F-14033 Caen, France
| |
Collapse
|
3
|
Govorova IA, Nikitochkina SY, Vorotelyak EA. Influence of intersignaling crosstalk on the intracellular localization of YAP/TAZ in lung cells. Cell Commun Signal 2024; 22:289. [PMID: 38802925 PMCID: PMC11129370 DOI: 10.1186/s12964-024-01662-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/11/2024] [Indexed: 05/29/2024] Open
Abstract
A cell is a dynamic system in which various processes occur simultaneously. In particular, intra- and intercellular signaling pathway crosstalk has a significant impact on a cell's life cycle, differentiation, proliferation, growth, regeneration, and, consequently, on the normal functioning of an entire organ. Hippo signaling and YAP/TAZ nucleocytoplasmic shuttling play a pivotal role in normal development, homeostasis, and tissue regeneration, particularly in lung cells. Intersignaling communication has a significant impact on the core components of the Hippo pathway and on YAP/TAZ localization. This review describes the crosstalk between Hippo signaling and key lung signaling pathways (WNT, SHH, TGFβ, Notch, Rho, and mTOR) using lung cells as an example and highlights the remaining unanswered questions.
Collapse
Affiliation(s)
- I A Govorova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia.
| | - S Y Nikitochkina
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia
| | - E A Vorotelyak
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia
| |
Collapse
|
4
|
Liang H, Xu Y, Zhao J, Chen M, Wang M. Hippo pathway in non-small cell lung cancer: mechanisms, potential targets, and biomarkers. Cancer Gene Ther 2024; 31:652-666. [PMID: 38499647 PMCID: PMC11101353 DOI: 10.1038/s41417-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Lung cancer is the primary contributor to cancer-related deaths globally, and non-small cell lung cancer (NSCLC) constitutes around 85% of all lung cancer cases. Recently, the emergence of targeted therapy and immunotherapy revolutionized the treatment of NSCLC and greatly improved patients' survival. However, drug resistance is inevitable, and extensive research has demonstrated that the Hippo pathway plays a crucial role in the development of drug resistance in NSCLC. The Hippo pathway is a highly conserved signaling pathway that is essential for various biological processes, including organ development, maintenance of epithelial balance, tissue regeneration, wound healing, and immune regulation. This pathway exerts its effects through two key transcription factors, namely Yes-associated protein (YAP) and transcriptional co-activator PDZ-binding motif (TAZ). They regulate gene expression by interacting with the transcriptional-enhanced associate domain (TEAD) family. In recent years, this pathway has been extensively studied in NSCLC. The review summarizes a comprehensive overview of the involvement of this pathway in NSCLC, and discusses the mechanisms of drug resistance, potential targets, and biomarkers associated with this pathway in NSCLC.
Collapse
Affiliation(s)
- Hongge Liang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Ahmed DW, Eiken MK, DePalma SJ, Helms AS, Zemans RL, Spence JR, Baker BM, Loebel C. Integrating mechanical cues with engineered platforms to explore cardiopulmonary development and disease. iScience 2023; 26:108472. [PMID: 38077130 PMCID: PMC10698280 DOI: 10.1016/j.isci.2023.108472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024] Open
Abstract
Mechanical forces provide critical biological signals to cells during healthy and aberrant organ development as well as during disease processes in adults. Within the cardiopulmonary system, mechanical forces, such as shear, compressive, and tensile forces, act across various length scales, and dysregulated forces are often a leading cause of disease initiation and progression such as in bronchopulmonary dysplasia and cardiomyopathies. Engineered in vitro models have supported studies of mechanical forces in a number of tissue and disease-specific contexts, thus enabling new mechanistic insights into cardiopulmonary development and disease. This review first provides fundamental examples where mechanical forces operate at multiple length scales to ensure precise lung and heart function. Next, we survey recent engineering platforms and tools that have provided new means to probe and modulate mechanical forces across in vitro and in vivo settings. Finally, the potential for interdisciplinary collaborations to inform novel therapeutic approaches for a number of cardiopulmonary diseases are discussed.
Collapse
Affiliation(s)
- Donia W. Ahmed
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Madeline K. Eiken
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, Division of Pulmonary Sciences and Critical Care Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Lee KY, Yang CC, Shueng PW, Wu SM, Chen CH, Chao YC, Chang YC, Han CL, Chuang HC, Lee CC, Lin CW. Downregulation of TAZ elicits a mitochondrial redox imbalance and ferroptosis in lung epithelial cells exposed to diesel exhaust particles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115555. [PMID: 37832483 DOI: 10.1016/j.ecoenv.2023.115555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
Mitochondrial dysfunction was reported to be involved in the development of lung diseases including chronic obstructive pulmonary disease (COPD). However, molecular regulation underlying metabolic disorders in the airway epithelia exposed to air pollution remains unclear. In the present study, lung bronchial epithelial BEAS-2B and alveolar epithelial A549 cells were treated with diesel exhaust particles (DEPs), the primary representative of ambient particle matter. This treatment elicited cell death accompanied by induction of lipid reactive oxygen species (ROS) production and ferroptosis. Lipidomics analyses revealed that DEPs increased glycerophospholipid contents. Accordingly, DEPs upregulated expression of the electron transport chain (ETC) complex and induced mitochondrial ROS production. Mechanistically, DEP exposure downregulated the Hippo transducer transcriptional co-activator with PDZ-binding motif (TAZ), which was further identified to be crucial for the ferroptosis-associated antioxidant system, including glutathione peroxidase 4 (GPX4), the glutamate-cysteine ligase catalytic subunit (GCLC), and glutathione-disulfide reductase (GSR). Moreover, immunohistochemistry confirmed downregulation of GPX4 and upregulation of lipid peroxidation in the bronchial epithelium of COPD patients and Sprague-Dawley rats exposed to air pollution. Finally, proteomics analyses confirmed alterations of ETC-related proteins in bronchoalveolar lavage from COPD patients compared to healthy subjects. Together, our study discovered that involvement of mitochondrial redox dysregulation plays a vital role in pulmonary epithelial cell destruction after exposure to air pollution.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Ching-Chieh Yang
- Division of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sheng-Min Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsuan Chen
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chun Chao
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chu Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Li Han
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
7
|
Kizawa R, Araya J, Fujita Y. Divergent roles of the Hippo pathway in the pathogenesis of idiopathic pulmonary fibrosis: tissue homeostasis and fibrosis. Inflamm Regen 2023; 43:45. [PMID: 37735707 PMCID: PMC10512581 DOI: 10.1186/s41232-023-00295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/10/2023] [Indexed: 09/23/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive aging-related lung disease with a poor prognosis. Despite extensive research, the cause of IPF remains largely unknown and treatment strategies are limited. Proposed mechanisms of the pathogenesis of IPF are a combination of excessive accumulation of the extracellular matrix and dysfunctional lung tissue regeneration. Epithelial cell dysfunction, in addition to fibroblast activation, is considered a key process in the progression of IPF. Epithelial cells normally maintain homeostasis of the lung tissue through regulated proliferation, differentiation, cell death, and cellular senescence. However, various stresses can cause repetitive damage to lung epithelial cells, leading to dysfunctional regeneration and acquisition of profibrotic functions. The Hippo pathway is a central signaling pathway that maintains tissue homeostasis and plays an essential role in fundamental biological processes. Dysregulation of the Hippo pathway has been implicated in various diseases, including IPF. However, the role of the Hippo pathway in the pathogenesis of IPF remains unclear, particularly given the pathway's opposing effects on the 2 key pathogenic mechanisms of IPF: epithelial cell dysfunction and fibroblast activation. A deeper understanding of the relationship between the Hippo pathway and the pathogenesis of IPF will pave the way for novel Hippo-targeted therapies.
Collapse
Affiliation(s)
- Ryusuke Kizawa
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
- Division of Next-Generation Drug Development, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yu Fujita
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
- Division of Next-Generation Drug Development, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
8
|
Laiman V, Hsiao TC, Fang YT, Chen YY, Lo YC, Lee KY, Chen TT, Chen KY, Ho SC, Wu SM, Chen JK, Heriyanto DS, Chung KF, Ho KF, Chuang KJ, Chang JH, Chuang HC. Hippo signaling pathway contributes to air pollution exposure-induced emphysema in ageing rats. JOURNAL OF HAZARDOUS MATERIALS 2023; 452:131188. [PMID: 36963197 DOI: 10.1016/j.jhazmat.2023.131188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/07/2023] [Accepted: 03/08/2023] [Indexed: 05/03/2023]
Affiliation(s)
- Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Fang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - You-Yin Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Industrial Ph.D. Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chun Lo
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kin-Fai Ho
- School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong, China
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Departments of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; National Heart and Lung Institute, Imperial College London, London, UK; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
10
|
da Rosa NN, Appel JM, Irioda AC, Mogharbel BF, de Oliveira NB, Perussolo MC, Stricker PEF, Rosa-Fernandes L, Marinho CRF, de Carvalho KAT. Three-Dimensional Bioprinting of an In Vitro Lung Model. Int J Mol Sci 2023; 24:5852. [PMID: 36982923 PMCID: PMC10059924 DOI: 10.3390/ijms24065852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
In December 2019, COVID-19 emerged in China, and in January 2020, the World Health Organization declared a state of international emergency. Within this context, there is a significant search for new drugs to fight the disease and a need for in vitro models for preclinical drug tests. This study aims to develop a 3D lung model. For the execution, Wharton's jelly mesenchymal stem cells (WJ-MSC) were isolated and characterized through flow cytometry and trilineage differentiation. For pulmonary differentiation, the cells were seeded in plates coated with natural functional biopolymer matrix as membrane until spheroid formation, and then the spheroids were cultured with differentiation inductors. The differentiated cells were characterized using immunocytochemistry and RT-PCR, confirming the presence of alveolar type I and II, ciliated, and goblet cells. Then, 3D bioprinting was performed with a sodium alginate and gelatin bioink in an extrusion-based 3D printer. The 3D structure was analyzed, confirming cell viability with a live/dead assay and the expression of lung markers with immunocytochemistry. The results showed that the differentiation of WJ-MSC into lung cells was successful, as well as the bioprinting of these cells in a 3D structure, a promising alternative for in vitro drug testing.
Collapse
Affiliation(s)
- Nádia Nascimento da Rosa
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Julia Maurer Appel
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Ana Carolina Irioda
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Bassam Felipe Mogharbel
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Nathalia Barth de Oliveira
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Maiara Carolina Perussolo
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Priscila Elias Ferreira Stricker
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| | - Lívia Rosa-Fernandes
- Experimental Immunoparasitology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (L.R.-F.); (C.R.F.M.)
| | - Cláudio Romero Farias Marinho
- Experimental Immunoparasitology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (L.R.-F.); (C.R.F.M.)
| | - Katherine Athayde Teixeira de Carvalho
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Pelé Pequeno Príncipe Research Institute & Pequeno Príncipe Faculties, Curitiba 80240-020, Brazil; (N.N.d.R.); (J.M.A.); (A.C.I.); (B.F.M.); (N.B.d.O.); (M.C.P.); (P.E.F.S.)
| |
Collapse
|
11
|
Molecular insights of Hippo signaling in the chick developing lung. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194904. [PMID: 36572276 DOI: 10.1016/j.bbagrm.2022.194904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Hippo signaling pathway and its effector YAP have been recognized as an essential growth regulator during embryonic development. Hippo has been studied in different contexts; nevertheless, its role during chick lung branching morphogenesis remains unknown. Therefore, this work aims to determine Hippo role during early pulmonary organogenesis in the avian animal model. The current study describes the spatial distribution of Hippo signaling members in the embryonic chick lung by in situ hybridization. Overall, their expression is comparable to their mammalian counterparts. Moreover, the expression levels of phosphorylated-YAP (pYAP) and total YAP revealed that Hippo signaling is active in the embryonic chick lung. Furthermore, the presence of pYAP in the cytoplasm demonstrated that the Hippo machinery distribution is maintained in this tissue. In vitro studies were performed to assess the role of the Hippo signaling pathway in lung branching. Lung explants treated with a YAP/TEAD complex inhibitor (verteporfin) displayed a significant reduction in lung size and branching and decreased expression of ctgf (Hippo target gene) compared to the control. This approach also revealed that Hippo seems to modulate the expression of key molecular players involved in lung branching morphogenesis (sox2, sox9, axin2, and gli1). Conversely, when treated with dobutamine, an upstream regulator that promotes YAP phosphorylation, explant morphology was not severely affected. Overall, our data indicate that Hippo machinery is present and active in the early stages of avian pulmonary branching and that YAP is likely involved in the regulation of lung growth.
Collapse
|
12
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
13
|
Chung YL, Laiman V, Tsao PN, Chen CM, Heriyanto DS, Chung KF, Chuang KJ, Chuang HC. Diesel exhaust particles inhibit lung branching morphogenesis via the YAP/TAZ pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 861:160682. [PMID: 36481141 DOI: 10.1016/j.scitotenv.2022.160682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
Prenatal exposure to air pollution may associated with inhibition of lung development in the child, however the possible mechanism is unclear. We investigated the effects of traffic-related diesel exhaust particle (DEP) exposure on fetal lung branching morphogenesis and elucidate the possible mechanism. Ex vivo fetal lungs collected from ICR mice at an age of 11.5 embryonic (E) days were exposed to DEPs at 0 (control), 10, and 50 μg/mL and branching morphogenesis was measured for 3 days. Normal IMR-90 human fetal lung fibroblast cells were exposed to DEPs at 0 (control), 10, and 50 μg/mL for 24 h. We observed that DEP exposure significantly inhibited lung branching morphogenesis with reduced lung branching ratios and surface areas on day 3. RNA sequencing (RNA-Seq) showed that DEP increased the inflammatory response and impaired lung development-related gene expressions. DEPs significantly decreased Yes-associated protein (YAP), phosphorylated (p)-YAP, transcriptional coactivator with a PDZ-binding motif (TAZ), and p-TAZ in IMR-90 cells at 10 and 50 μg/mL. Treatment of fetal lungs with the YAP inhibitor, PFI-2, also demonstrated restricted lung branching development similar to that of DEP exposure, with a significantly decreased lung branching ratio on day 3. DEP exposure significantly decreased the lung branching modulators fibroblast growth factor receptor 2 (FGFR2), sex-determining region Y-box 2 (SOX2), and SOX9 in IMR-90 cells at 10 and 50 μg/mL. Fetal lung immunofluorescence staining showed that DEP decreased SOX2 expression in fibronectin+ fibroblasts. DEP exposure decreased the cellular senescence regulator, p-sirtuin 1 (SIRT1)/SIRT1 in IMR-90 cells, with RNA-Seq showing impaired telomere maintenance. DEP exposure impaired fetal lung growth during the pseudoglandular stage through dysregulating the Hippo signaling pathway, causing fibroblast lung branching restriction and early senescence. Prenatal exposure to traffic-related air pollution has adverse effects on fetal lung development.
Collapse
Affiliation(s)
- Yu-Ling Chung
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; The Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Noël A, Yilmaz S, Farrow T, Schexnayder M, Eickelberg O, Jelesijevic T. Sex-Specific Alterations of the Lung Transcriptome at Birth in Mouse Offspring Prenatally Exposed to Vanilla-Flavored E-Cigarette Aerosols and Enhanced Susceptibility to Asthma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3710. [PMID: 36834405 PMCID: PMC9967225 DOI: 10.3390/ijerph20043710] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Currently, approximately 8 million adult Americans use electronic cigarettes (e-cigs) daily, including women of childbearing age. It is known that more than 10% of women smoke during their pregnancy, and recent surveys show that rates of maternal vaping are similar to rates of maternal cigarette smoking. However, the effects of inhaling e-cig aerosol on the health of fetuses remain unknown. The objective of the present study was to increase our understanding of the molecular effects caused by in utero exposures to e-cig aerosols on developing mouse lungs and, later in life, on the offspring's susceptibility to developing asthma. METHODS Pregnant mice were exposed throughout gestation to either filtered air or vanilla-flavored e-cig aerosols containing 18 mg/mL of nicotine. Male and female exposed mouse offspring were sacrificed at birth, and then the lung transcriptome was evaluated. Additionally, once sub-groups of male offspring mice reached 4 weeks of age, they were challenged with house dust mites (HDMs) for 3 weeks to assess asthmatic responses. RESULTS The lung transcriptomic responses of the mouse offspring at birth showed that in utero vanilla-flavored e-cig aerosol exposure significantly regulated 88 genes in males (62 genes were up-regulated and 26 genes were down-regulated), and 65 genes were significantly regulated in females (17 genes were up-regulated and 48 genes were down-regulated). Gene network analyses revealed that in utero e-cig aerosol exposure affected canonical pathways associated with CD28 signaling in T helper cells, the role of NFAT in the regulation of immune responses, and phospholipase C signaling in males, whereas the dysregulated genes in the female offspring were associated with NRF2-mediated oxidative stress responses. Moreover, we found that in utero exposures to vanilla-flavored e-cig aerosol exacerbated HDM-induced asthma in 7-week-old male mouse offspring compared to respective in utero air + HDM controls. CONCLUSIONS Overall, these data demonstrate that in utero e-cig aerosol exposure alters the developing mouse lung transcriptome at birth in a sex-specific manner and provide evidence that the inhalation of e-cig aerosols is detrimental to the respiratory health of offspring by increasing the offspring' susceptibility to developing lung diseases later in life.
Collapse
Affiliation(s)
- Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Sultan Yilmaz
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Tori Farrow
- Department of Environmental Toxicology, Southern University and A & M College, Baton Rouge, LA 70813, USA
| | | | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tomislav Jelesijevic
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
15
|
Ko HS, Laiman V, Tsao PN, Chen CM, Chuang HC. Alteration in branching morphogenesis via YAP/TAZ in fibroblasts of fetal lungs in an LPS-induced inflammation model. Mol Med 2023; 29:16. [PMID: 36717779 PMCID: PMC9887856 DOI: 10.1186/s10020-023-00613-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Chorioamnionitis is a common cause of preterm birth and leads to serious complications in newborns. The objective of this study was to investigate the role of the Hippo signaling pathway in lung branching morphogenesis under a lipopolysaccharide (LPS)-induced inflammation model. MATERIALS AND METHODS IMR-90 cells and ex vivo fetal lungs were treated with 0, 10, 30, or 50 μg/ml LPS for 24 and 72 h. Supernatant levels of lactate dehydrogenase (LDH), interleukin (IL)-6, IL-8, Chemokine (C-X-C motif) ligand 1(CXCL1), branching and the surface area ratio, Yes-associated protein (YAP), transcription coactivator with PDZ-binding motif (TAZ), fibroblast growth factor 10 (FGF10), fibroblast growth factor receptor II (FGFR2), SRY-box transcription factor 2 (SOX2), SOX9, and sirtuin 1 (SIRT1) levels were examined. Differentially expressed genes in fetal lungs after LPS treatment were identified by RNA-sequencing. RESULTS LPS at 50 μg/ml increased IL-6 and IL-8 in IMR-90 cells and increased IL-6, CXCL1 and LDH in fetal lungs. The branching ratio significantly increased by LPS at 30 μg/ml compared to the control but the increased level had decreased by 50 μg/ml LPS exposure. Exposure to 50 μg/ml LPS increased phosphorylated (p)-YAP, p-YAP/YAP, and p-TAZ/TAZ in IMR-90 cells, whereas 50 μg/ml LPS decreased FGF10 and SOX2. Consistently, p-YAP/YAP and p-TAZ/TAZ were increased in fibronectin+ cells of fetal lungs. Moreover, results of RNA-sequencing in fetal lungs showed that SMAD, FGF, IκB phosphorylation, tissue remodeling and homeostasis was involved in branching morphogenesis following exposure to 50 μg/ml LPS. The p-SIRT1/SIRT1 ratio increased in IMR-90 cells by LPS treatment. CONCLUSIONS This study showed that regulation of the Hippo pathway in fibroblasts of fetal lungs was involved in branching morphogenesis under an inflammatory disease such as chorioamnionitis.
Collapse
Affiliation(s)
- Hung-Shuo Ko
- grid.412896.00000 0000 9337 0481School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Vincent Laiman
- grid.412896.00000 0000 9337 0481International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan ,grid.8570.a0000 0001 2152 4506Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Dr. Sardjito Hospital, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Po-Nien Tsao
- grid.412094.a0000 0004 0572 7815Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Ming Chen
- grid.412897.10000 0004 0639 0994Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan ,grid.412896.00000 0000 9337 0481Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- grid.412896.00000 0000 9337 0481School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031 Taiwan ,grid.412896.00000 0000 9337 0481Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan ,grid.412896.00000 0000 9337 0481Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan ,grid.412896.00000 0000 9337 0481Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan ,grid.7445.20000 0001 2113 8111National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
16
|
Ortiz-Zapater E, Bagley DC, Hernandez VL, Roberts LB, Maguire TJA, Voss F, Mertins P, Kirchner M, Peset-Martin I, Woszczek G, Rosenblatt J, Gotthardt M, Santis G, Parsons M. Epithelial coxsackievirus adenovirus receptor promotes house dust mite-induced lung inflammation. Nat Commun 2022; 13:6407. [PMID: 36302767 PMCID: PMC9613683 DOI: 10.1038/s41467-022-33882-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 10/06/2022] [Indexed: 12/25/2022] Open
Abstract
Airway inflammation and remodelling are important pathophysiologic features in asthma and other respiratory conditions. An intact epithelial cell layer is crucial to maintain lung homoeostasis, and this depends on intercellular adhesion, whilst damaged respiratory epithelium is the primary instigator of airway inflammation. The Coxsackievirus Adenovirus Receptor (CAR) is highly expressed in the epithelium where it modulates cell-cell adhesion stability and facilitates immune cell transepithelial migration. However, the contribution of CAR to lung inflammation remains unclear. Here we investigate the mechanistic contribution of CAR in mediating responses to the common aeroallergen, House Dust Mite (HDM). We demonstrate that administration of HDM in mice lacking CAR in the respiratory epithelium leads to loss of peri-bronchial inflammatory cell infiltration, fewer goblet-cells and decreased pro-inflammatory cytokine release. In vitro analysis in human lung epithelial cells confirms that loss of CAR leads to reduced HDM-dependent inflammatory cytokine release and neutrophil migration. Epithelial CAR depletion also promoted smooth muscle cell proliferation mediated by GSK3β and TGF-β, basal matrix production and airway hyperresponsiveness. Our data demonstrate that CAR coordinates lung inflammation through a dual function in leucocyte recruitment and tissue remodelling and may represent an important target for future therapeutic development in inflammatory lung diseases.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
| | - Dustin C Bagley
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | | | - Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Thomas J A Maguire
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Felizia Voss
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- DZHK Partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Marieluise Kirchner
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Grzegorz Woszczek
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jody Rosenblatt
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Michael Gotthardt
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - George Santis
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
- Department of Respiratory Medicine, Guy's & St Thomas NHS Trust, London, UK
| | - Maddy Parsons
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
17
|
YAP and TAZ: Monocorial and bicorial transcriptional co-activators in human cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188756. [PMID: 35777600 DOI: 10.1016/j.bbcan.2022.188756] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/09/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
The transcriptional regulators YAP and TAZ are involved in numerous physiological processes including organ development, growth, immunity and tissue regeneration. YAP and TAZ dysregulation also contribute to tumorigenesis, thereby making them attractive cancer therapeutic targets. Arbitrarily, YAP and TAZ are often considered as a single protein, and are referred to as YAP/TAZ in most studies. However, increasing experimental evidences documented that YAP and TAZ perform both overlapping and distinct functions in several physiological and pathological processes. In addition to regulating distinct processes, YAP and TAZ are also regulated by distinct upstream cues. The aim of the review is to describe the distinct roles of YAP and TAZ focusing particularly on cancer. Therapeutic strategies targeting either YAP and TAZ proteins or only one of them should be carefully evaluated. Selective targeting of YAP or TAZ may in fact impair different pathways and determine diverse clinical outputs.
Collapse
|
18
|
Lin CR, Bahmed K, Kosmider B. Impaired Alveolar Re-Epithelialization in Pulmonary Emphysema. Cells 2022; 11:2055. [PMID: 35805139 PMCID: PMC9265977 DOI: 10.3390/cells11132055] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/24/2023] Open
Abstract
Alveolar type II (ATII) cells are progenitors in alveoli and can repair the alveolar epithelium after injury. They are intertwined with the microenvironment for alveolar epithelial cell homeostasis and re-epithelialization. A variety of ATII cell niches, transcription factors, mediators, and signaling pathways constitute a specific environment to regulate ATII cell function. Particularly, WNT/β-catenin, YAP/TAZ, NOTCH, TGF-β, and P53 signaling pathways are dynamically involved in ATII cell proliferation and differentiation, although there are still plenty of unknowns regarding the mechanism. However, an imbalance of alveolar cell death and proliferation was observed in patients with pulmonary emphysema, contributing to alveolar wall destruction and impaired gas exchange. Cigarette smoking causes oxidative stress and is the primary cause of this disease development. Aberrant inflammatory and oxidative stress responses result in loss of cell homeostasis and ATII cell dysfunction in emphysema. Here, we discuss the current understanding of alveolar re-epithelialization and altered reparative responses in the pathophysiology of this disease. Current therapeutics and emerging treatments, including cell therapies in clinical trials, are addressed as well.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
19
|
Fallah S, Beaulieu JF. Differential influence of YAP1 and TAZ on differentiation of intestinal epithelial cell: A review. Anat Rec (Hoboken) 2022; 306:1054-1061. [PMID: 35648375 DOI: 10.1002/ar.24996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 11/06/2022]
Abstract
Intestinal cell stemness, proliferation and differentiation are complex processes all occurring in distinct compartments of the crypt that need to be closely regulated to ensure proper epithelial renewal. The involvement of the Hippo pathway in intestinal epithelial proliferation and regeneration after injury via the regulation of its effectors YAP1 and TAZ has been well-documented over the last decade. The implication of YAP1 and TAZ on intestinal epithelial cell differentiation is less clear. Using intestinal cell models in which the expression of YAP1 and TAZ can be modulated, our group showed that YAP1 inhibits differentiation of the two main intestinal epithelial cell types, goblet and absorptive cells through a specific mechanism involving the repression of prodifferentiation transcription factor CDX2 expression. Further analysis provided evidence that the repressive effect of YAP1 on intestinal differentiation is mediated by regulation of the Hippo pathway by Src family kinase activity. Interestingly, the TAZ paralog does not seem to be involved in this process, which provides another example of the lack of perfect complementarity of the two main Hippo effectors.
Collapse
Affiliation(s)
- Sepideh Fallah
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
20
|
Duong TE, Wu Y, Sos BC, Dong W, Limaye S, Rivier LH, Myers G, Hagood JS, Zhang K. A single-cell regulatory map of postnatal lung alveologenesis in humans and mice. CELL GENOMICS 2022; 2:100108. [PMID: 35434692 PMCID: PMC9012447 DOI: 10.1016/j.xgen.2022.100108] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 05/05/2021] [Accepted: 02/02/2022] [Indexed: 04/14/2023]
Abstract
Ex-utero regulation of the lungs' responses to breathing air and continued alveolar development shape adult respiratory health. Applying single-cell transposome hypersensitive site sequencing (scTHS-seq) to over 80,000 cells, we assembled the first regulatory atlas of postnatal human and mouse lung alveolar development. We defined regulatory modules and elucidated new mechanistic insights directing alveolar septation, including alveolar type 1 and myofibroblast cell signaling and differentiation, and a unique human matrix fibroblast population. Incorporating GWAS, we mapped lung function causal variants to myofibroblasts and identified a pathogenic regulatory unit linked to lineage marker FGF18, demonstrating the utility of chromatin accessibility data to uncover disease mechanism targets. Our regulatory map and analysis model provide valuable new resources to investigate age-dependent and species-specific control of critical developmental processes. Furthermore, these resources complement existing atlas efforts to advance our understanding of lung health and disease across the human lifespan.
Collapse
Affiliation(s)
- Thu Elizabeth Duong
- Department of Pediatrics, Division of Respiratory Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Yan Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Brandon Chin Sos
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Weixiu Dong
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Siddharth Limaye
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Lauraine H. Rivier
- Department of Pediatrics, Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Greg Myers
- Department of Pediatrics, Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - James S. Hagood
- Department of Pediatrics, Division of Pediatric Pulmonology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
21
|
Zinc Oxide Nanoparticles Promote YAP/TAZ Nuclear Localization in Alveolar Epithelial Type II Cells. ATMOSPHERE 2022. [DOI: 10.3390/atmos13020334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated roles of Hippo signaling pathway components in alveolar type II cells (AECII) after zinc oxide nanoparticle (ZnONP) exposure. ZnONPs physicochemistry was characterized using field emission-scanning electron microscopy (FE-SEM) and energy-dispersive X-ray (EDX) microanalysis. ZnONP deposition in human respiratory tract was estimated using multiple-path particle dosimetry (MPPD) model. MLE-12 AECII were cultured and exposed to 0, 1, and 5 μg/mL of ZnONPs for 24 h. Western blots were used to investigate signaling pathways associated with Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ), cell adherens junctions, differentiation, and senescence. ZnONPs morphology was irregular, with Zn and O identified. Approximately 72% of inhaled ZnONPs were deposited in lungs, with 26% being deposited in alveolar regions. ZnONP exposure increased nuclear YAP expression and decreased cytoplasmic YAP expression by AECII. Adherens junction proteins, E-cadherin, α-catenin, and β-catenin, on AECII decreased after ZnONP exposure. ZnONP exposure of AECII increased alveolar type I (AECI) transition protein, LGALS3, and the AECI protein, T1α, while decreasing AECII SPC expression. ZnONP exposure induced Sirt1 and p53 senescence proteins by AECII. Our findings showed that inhalable ZnONPs can deposit in alveoli, which promotes YAP nuclear localization in AECII, resulting in decrease tight junctions, cell differentiation, and cell senescence.
Collapse
|
22
|
Jaslove JM, Goodwin K, Sundarakrishnan A, Spurlin JW, Mao S, Košmrlj A, Nelson CM. Transmural pressure signals through retinoic acid to regulate lung branching. Development 2022; 149:274047. [PMID: 35051272 PMCID: PMC8917413 DOI: 10.1242/dev.199726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 12/10/2021] [Indexed: 01/22/2023]
Abstract
During development, the mammalian lung undergoes several rounds of branching, the rate of which is tuned by the relative pressure of the fluid within the lumen of the lung. We carried out bioinformatics analysis of RNA-sequencing of embryonic mouse lungs cultured under physiologic or sub-physiologic transmural pressure and identified transcription factor-binding motifs near genes whose expression changes in response to pressure. Surprisingly, we found retinoic acid (RA) receptor binding sites significantly overrepresented in the promoters and enhancers of pressure-responsive genes. Consistently, increasing transmural pressure activates RA signaling, and pharmacologically inhibiting RA signaling decreases airway epithelial branching and smooth muscle wrapping. We found that pressure activates RA signaling through the mechanosensor Yap. A computational model predicts that mechanical signaling through Yap and RA affects lung branching by altering the balance between epithelial proliferation and smooth muscle wrapping, which we test experimentally. Our results reveal that transmural pressure signals through RA to balance the relative rates of epithelial growth and smooth muscle differentiation in the developing mouse lung and identify RA as a previously unreported component in the mechanotransduction machinery of embryonic tissues.
Collapse
Affiliation(s)
- Jacob M. Jaslove
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA,Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Aswin Sundarakrishnan
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - James W. Spurlin
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA,Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Sheng Mao
- Department of Mechanics and Engineering Science, BIC-ESAT, College of Engineering, Peking University, Beijing 100871, People's Republic of China,Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Andrej Košmrlj
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA,Princeton Institute for the Science & Technology of Materials, Princeton, NJ 08544, USA
| | - Celeste M. Nelson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA,Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA,Author for correspondence ()
| |
Collapse
|
23
|
Stanton AE, Goodwin K, Sundarakrishnan A, Jaslove JM, Gleghorn JP, Pavlovich AL, Nelson CM. Negative Transpulmonary Pressure Disrupts Airway Morphogenesis by Suppressing Fgf10. Front Cell Dev Biol 2021; 9:725785. [PMID: 34926440 PMCID: PMC8673560 DOI: 10.3389/fcell.2021.725785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanical forces are increasingly recognized as important determinants of cell and tissue phenotype and also appear to play a critical role in organ development. During the fetal stages of lung morphogenesis, the pressure of the fluid within the lumen of the airways is higher than that within the chest cavity, resulting in a positive transpulmonary pressure. Several congenital defects decrease or reverse transpulmonary pressure across the developing airways and are associated with a reduced number of branches and a correspondingly underdeveloped lung that is insufficient for gas exchange after birth. The small size of the early pseudoglandular stage lung and its relative inaccessibility in utero have precluded experimental investigation of the effects of transpulmonary pressure on early branching morphogenesis. Here, we present a simple culture model to explore the effects of negative transpulmonary pressure on development of the embryonic airways. We found that negative transpulmonary pressure decreases branching, and that it does so in part by altering the expression of fibroblast growth factor 10 (Fgf10). The morphogenesis of lungs maintained under negative transpulmonary pressure can be rescued by supplementing the culture medium with exogenous FGF10. These data suggest that Fgf10 expression is regulated by mechanical stress in the developing airways. Understanding the mechanical signaling pathways that connect transpulmonary pressure to FGF10 can lead to the establishment of novel non-surgical approaches for ameliorating congenital lung defects.
Collapse
Affiliation(s)
- Alice E Stanton
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Katharine Goodwin
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Aswin Sundarakrishnan
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Jacob M Jaslove
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Jason P Gleghorn
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Amira L Pavlovich
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Celeste M Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States.,Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
24
|
Nasri A, Foisset F, Ahmed E, Lahmar Z, Vachier I, Jorgensen C, Assou S, Bourdin A, De Vos J. Roles of Mesenchymal Cells in the Lung: From Lung Development to Chronic Obstructive Pulmonary Disease. Cells 2021; 10:3467. [PMID: 34943975 PMCID: PMC8700565 DOI: 10.3390/cells10123467] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal cells are an essential cell type because of their role in tissue support, their multilineage differentiation capacities and their potential clinical applications. They play a crucial role during lung development by interacting with airway epithelium, and also during lung regeneration and remodeling after injury. However, much less is known about their function in lung disease. In this review, we discuss the origins of mesenchymal cells during lung development, their crosstalk with the epithelium, and their role in lung diseases, particularly in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Amel Nasri
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France; (A.N.); (F.F.); (C.J.); (S.A.)
| | - Florent Foisset
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France; (A.N.); (F.F.); (C.J.); (S.A.)
| | - Engi Ahmed
- Department of Respiratory Diseases, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34090 Montpellier, France; (E.A.); (Z.L.); (I.V.); (A.B.)
- PhyMedExp, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34295 Montpellier, France
| | - Zakaria Lahmar
- Department of Respiratory Diseases, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34090 Montpellier, France; (E.A.); (Z.L.); (I.V.); (A.B.)
- PhyMedExp, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34295 Montpellier, France
| | - Isabelle Vachier
- Department of Respiratory Diseases, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34090 Montpellier, France; (E.A.); (Z.L.); (I.V.); (A.B.)
| | - Christian Jorgensen
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France; (A.N.); (F.F.); (C.J.); (S.A.)
| | - Said Assou
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France; (A.N.); (F.F.); (C.J.); (S.A.)
| | - Arnaud Bourdin
- Department of Respiratory Diseases, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34090 Montpellier, France; (E.A.); (Z.L.); (I.V.); (A.B.)
- PhyMedExp, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34295 Montpellier, France
| | - John De Vos
- Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, INSERM, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France; (A.N.); (F.F.); (C.J.); (S.A.)
- Department of Cell and Tissue Engineering, Université de Montpellier, Centre Hospitalier Universitaire de Montpellier, 34000 Montpellier, France
| |
Collapse
|
25
|
Rippa AL, Alpeeva EV, Vasiliev AV, Vorotelyak EA. Alveologenesis: What Governs Secondary Septa Formation. Int J Mol Sci 2021; 22:ijms222212107. [PMID: 34829987 PMCID: PMC8618598 DOI: 10.3390/ijms222212107] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/30/2022] Open
Abstract
The simplification of alveoli leads to various lung pathologies such as bronchopulmonary dysplasia and emphysema. Deep insight into the process of emergence of the secondary septa during development and regeneration after pneumonectomy, and into the contribution of the drivers of alveologenesis and neo-alveolarization is required in an efficient search for therapeutic approaches. In this review, we describe the formation of the gas exchange units of the lung as a multifactorial process, which includes changes in the actomyosin cytoskeleton of alveocytes and myofibroblasts, elastogenesis, retinoic acid signaling, and the contribution of alveolar mesenchymal cells in secondary septation. Knowledge of the mechanistic context of alveologenesis remains incomplete. The characterization of the mechanisms that govern the emergence and depletion of αSMA will allow for an understanding of how the niche of fibroblasts is changing. Taking into account the intense studies that have been performed on the pool of lung mesenchymal cells, we present data on the typing of interstitial fibroblasts and their role in the formation and maintenance of alveoli. On the whole, when identifying cell subpopulations in lung mesenchyme, one has to consider the developmental context, the changing cellular functions, and the lability of gene signatures.
Collapse
|
26
|
Gokey JJ, Snowball J, Sridharan A, Sudha P, Kitzmiller JA, Xu Y, Whitsett JA. YAP regulates alveolar epithelial cell differentiation and AGER via NFIB/KLF5/NKX2-1. iScience 2021; 24:102967. [PMID: 34466790 PMCID: PMC8383002 DOI: 10.1016/j.isci.2021.102967] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/26/2021] [Accepted: 08/06/2021] [Indexed: 01/04/2023] Open
Abstract
Ventilation is dependent upon pulmonary alveoli lined by two major epithelial cell types, alveolar type-1 (AT1) and 2 (AT2) cells. AT1 cells mediate gas exchange while AT2 cells synthesize and secrete pulmonary surfactants and serve as progenitor cells which repair the alveoli. We developed transgenic mice in which YAP was activated or deleted to determine its roles in alveolar epithelial cell differentiation. Postnatal YAP activation increased epithelial cell proliferation, increased AT1 cell numbers, and caused indeterminate differentiation of subsets of alveolar cells expressing atypical genes normally restricted to airway epithelial cells. YAP deletion increased expression of genes associated with mature AT2 cells. YAP activation enhanced DNA accessibility in promoters of transcription factors and motif enrichment analysis predicted target genes associated with alveolar cell differentiation. YAP participated with KLF5, NFIB, and NKX2-1 to regulate AGER. YAP plays a central role in a transcriptional network that regulates alveolar epithelial differentiation.
Collapse
Affiliation(s)
- Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John Snowball
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Anusha Sridharan
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Parvathi Sudha
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joseph A. Kitzmiller
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Yan Xu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jeffrey A. Whitsett
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
27
|
Xu X, Nie Y, Wang W, Ullah I, Tung WT, Ma N, Lendlein A. Generation of 2.5D lung bud organoids from human induced pluripotent stem cells. Clin Hemorheol Microcirc 2021; 79:217-230. [PMID: 34487028 DOI: 10.3233/ch-219111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are a promising cell source to generate the patient-specific lung organoid given their superior differentiation potential. However, the current 3D cell culture approach is tedious and time-consuming with a low success rate and high batch-to-batch variability. Here, we explored the establishment of lung bud organoids by systematically adjusting the initial confluence levels and homogeneity of cell distribution. The efficiency of single cell seeding and clump seeding was compared. Instead of the traditional 3D culture, we established a 2.5D organoid culture to enable the direct monitoring of the internal structure via microscopy. It was found that the cell confluence and distribution prior to induction were two key parameters, which strongly affected hiPSC differentiation trajectories. Lung bud organoids with positive expression of NKX 2.1, in a single-cell seeding group with homogeneously distributed hiPSCs at 70% confluence (SC_70%_hom) or a clump seeding group with heterogeneously distributed cells at 90% confluence (CL_90%_het), can be observed as early as 9 days post induction. These results suggest that a successful lung bud organoid formation with single-cell seeding of hiPSCs requires a moderate confluence and homogeneous distribution of cells, while high confluence would be a prominent factor to promote the lung organoid formation when seeding hiPSCs as clumps. 2.5D organoids generated with defined culture conditions could become a simple, efficient, and valuable tool facilitating drug screening, disease modeling and personalized medicine.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Yan Nie
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Weiwei Wang
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Imran Ullah
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Wing Tai Tung
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Nan Ma
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| |
Collapse
|
28
|
Forno E, Abman SH, Singh J, Robbins ME, Selvadurai H, Schumacker PT, Robinson PD. Update in Pediatrics 2020. Am J Respir Crit Care Med 2021; 204:274-284. [PMID: 34126039 DOI: 10.1164/rccm.202103-0605up] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Erick Forno
- Division of Pediatric Pulmonary Medicine, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Steven H Abman
- Department of Pediatrics, Children's Hospital Colorado, Denver, Colorado.,University of Colorado Anschutz School of Medicine, Denver, Colorado
| | - Jagdev Singh
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mary E Robbins
- Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois; and.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hiran Selvadurai
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| | - Paul T Schumacker
- Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois; and.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Paul D Robinson
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Hicks-Berthet J, Ning B, Federico A, Tilston-Lunel A, Matschulat A, Ai X, Lenburg ME, Beane J, Monti S, Varelas X. Yap/Taz inhibit goblet cell fate to maintain lung epithelial homeostasis. Cell Rep 2021; 36:109347. [PMID: 34260916 PMCID: PMC8346236 DOI: 10.1016/j.celrep.2021.109347] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/22/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Proper lung function relies on the precise balance of specialized epithelial cells that coordinate to maintain homeostasis. Herein, we describe essential roles for the transcriptional regulators YAP/TAZ in maintaining lung epithelial homeostasis, reporting that conditional deletion of Yap and Wwtr1/Taz in the lung epithelium of adult mice results in severe defects, including alveolar disorganization and the development of airway mucin hypersecretion. Through in vivo lineage tracing and in vitro molecular experiments, we reveal that reduced YAP/TAZ activity promotes intrinsic goblet transdifferentiation of secretory airway epithelial cells. Global gene expression and chromatin immunoprecipitation sequencing (ChIP-seq) analyses suggest that YAP/TAZ act cooperatively with TEA domain (TEAD) transcription factors and the NuRD complex to suppress the goblet cell fate program, directly repressing the SPDEF gene. Collectively, our study identifies YAP/TAZ as critical factors in lung epithelial homeostasis and offers molecular insight into the mechanisms promoting goblet cell differentiation, which is a hallmark of many lung diseases.
Collapse
Affiliation(s)
- Julia Hicks-Berthet
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Boting Ning
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony Federico
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Adeline Matschulat
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marc E Lenburg
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jennifer Beane
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Stefano Monti
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
30
|
Takeda T, Tsubaki M, Genno S, Matsuda T, Yamamoto Y, Kimura A, Shimizu N, Nishida S. Inhibition of yes-associated protein suppresses migration, invasion, and metastasis in non-small cell lung cancer in vitro and in vivo. Clin Exp Med 2021; 22:221-228. [PMID: 34196881 DOI: 10.1007/s10238-021-00738-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a highly aggressive cancer with one of the most prevalent malignant tumors. Metastasis in NSCLC is the major cause of treatment failure and cancer-related deaths. Yes-associated protein (YAP) is a transcriptional coactivator regulated by the evolutionarily conserved Hippo signaling pathway that regulates organ size, growth, and regeneration. YAP is highly expressed in several malignant tumor types. Furthermore, YAP promotes tumor initiation and/or progression in various types of cancer. However, it is unclear whether YAP contributes to the metastasis in NSCLC and serves as a useful therapeutic target. Here, we investigated whether levels of YAP correlate with metastatic phenotype in NSCLC cells and serve as a useful therapeutic target. We found that high levels of YAP associate with high cell migration, invasion, and metastasis in NSCLC cell lines. Furthermore, YAP siRNA decreased the migration and invasion in NSCLC cells. Additionally, verteporfin, an agent used for the treatment of symptomatic polypoidal choroidal vasculopathy, decreased the expression of YAP and inhibited migration, invasion, and metastasis in NSCLC cells. Thus, the study suggests that targeting YAP may present a new avenue to develop therapeutics against metastasis in NSCLC and that verteporfin has potential molecular therapeutic strategy for the treatment of metastatic NSCLC.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shuji Genno
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yuuta Yamamoto
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Akihiro Kimura
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Nao Shimizu
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
31
|
Warburton D. Conserved Mechanisms in the Formation of the Airways and Alveoli of the Lung. Front Cell Dev Biol 2021; 9:662059. [PMID: 34211971 PMCID: PMC8239290 DOI: 10.3389/fcell.2021.662059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/12/2021] [Indexed: 11/15/2022] Open
Abstract
Branching is an intrinsic property of respiratory epithelium that can be induced and modified by signals emerging from the mesenchyme. However, during stereotypic branching morphogenesis of the airway, the relatively thick upper respiratory epithelium extrudes through a mesenchymal orifice to form a new branch, whereas during alveologenesis the relatively thin lower respiratory epithelium extrudes to form sacs or bubbles. Thus, both branching morphogenesis of the upper airway and alveolarization in the lower airway seem to rely on the same fundamental physical process: epithelial extrusion through an orifice. Here I propose that it is the orientation and relative stiffness of the orifice boundary that determines the stereotypy of upper airway branching as well as the orientation of individual alveolar components of the gas exchange surface. The previously accepted dogma of the process of alveologenesis, largely based on 2D microscopy, is that alveoli arise by erection of finger-like interalveolar septae to form septal clefts that subdivide pre-existing saccules, a process for which the contractile properties of specialized alveolar myofibroblasts are necessary. Here I suggest that airway tip splitting and stereotypical side domain branching are actually conserved processes, but modified somewhat by evolution to achieve both airway tip splitting and side branching of the upper airway epithelium, as well as alveologenesis. Viewed in 3D it is clear that alveolar “septal tips” are in fact ring or purse string structures containing elastin and collagen that only appear as finger like projections in cross section. Therefore, I propose that airway branch orifices as well as alveolar mouth rings serve to delineate and stabilize the budding of both airway and alveolar epithelium, from the tips and sides of upper airways as well as from the sides and tips of alveolar ducts. Certainly, in the case of alveoli arising laterally and with radial symmetry from the sides of alveolar ducts, the mouth of each alveolus remains within the plane of the side of the ductal lumen. This suggests that the thin epithelium lining these lateral alveolar duct buds may extrude or “pop out” from the duct lumen through rings rather like soap or gum bubbles, whereas the thicker upper airway epithelium extrudes through a ring like toothpaste from a tube to form a new branch.
Collapse
Affiliation(s)
- David Warburton
- The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
32
|
Isago H, Mitani A, Kohno S, Nagoshi S, Ishimori T, Saito M, Tamiya H, Miyashita N, Ishii T, Matsuzaki H, Yatomi Y, Nagase T, Jo T. The Japanese Herbal (Kampo) Medicine Hochuekkito Attenuates Lung Inflammation in Lung Emphysema. Biol Pharm Bull 2021; 44:39-45. [PMID: 33390548 DOI: 10.1248/bpb.b20-00556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a systemic inflammatory disorder. It often causes weight loss, which is considered a poor prognostic factor. A Japanese herbal Kampo medicine, Hochuekkito (TJ-41), has been reported to prevent systemic inflammation and weight loss in COPD patients, but the underlying biological mechanisms remain unknown. In the present study, we investigated the role of TJ-41 in vivo using a mouse model of lung emphysema. We used lung epithelium-specific Taz conditional knockout mice (Taz CKO mice) as the lung emphysema model mimicking the chronic pulmonary inflammation in COPD. Acute inflammation was induced by intratracheal lipopolysaccharide administration, simulating COPD exacerbation. Mice were fed a diet containing 2% TJ-41 or a control diet. Taz CKO mice showed increased numbers of inflammatory cells in the bronchoalveolar lavage fluid compared to control mice. This effect was reduced by TJ-41 treatment. In the acute exacerbation model, TJ-41 mitigated the increased numbers of inflammatory cells in the bronchoalveolar lavage fluid and attenuated lung inflammation in histopathological studies. Additional in vitro experiments using the human macrophage cell line U-937 demonstrated that lipopolysaccharide-induced tumor necrosis factor-alpha expression was significantly downregulated by TJ-41. These results suggest that TJ-41 has anti-inflammatory effects in lung emphysema both in the chronic phase and during an acute exacerbation. In conclusion, our study sheds light on the anti-inflammatory effects of TJ-41 in lung emphysema. This establishes its potential as a new anti-inflammatory therapy and a preventive medicine for exacerbations during the long-time maintenance of COPD patients.
Collapse
Affiliation(s)
- Hideaki Isago
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo Hospital.,Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Akihisa Mitani
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Shiho Kohno
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Saki Nagoshi
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Taro Ishimori
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Minako Saito
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Hiroyuki Tamiya
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Naoya Miyashita
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Takashi Ishii
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Hirotaka Matsuzaki
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo Hospital
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo
| | - Taisuke Jo
- Department of Health Services Research, Graduate School of Medicine, the University of Tokyo
| |
Collapse
|
33
|
Zeng C, Motta-Ribeiro GC, Hinoshita T, Lessa MA, Winkler T, Grogg K, Kingston NM, Hutchinson JN, Sholl LM, Fang X, Varelas X, Layne MD, Baron RM, Vidal Melo MF. Lung Atelectasis Promotes Immune and Barrier Dysfunction as Revealed by Transcriptome Sequencing in Female Sheep. Anesthesiology 2020; 133:1060-1076. [PMID: 32796202 PMCID: PMC7572680 DOI: 10.1097/aln.0000000000003491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Pulmonary atelectasis is frequent in clinical settings. Yet there is limited mechanistic understanding and substantial clinical and biologic controversy on its consequences. The authors hypothesize that atelectasis produces local transcriptomic changes related to immunity and alveolar-capillary barrier function conducive to lung injury and further exacerbated by systemic inflammation. METHODS Female sheep underwent unilateral lung atelectasis using a left bronchial blocker and thoracotomy while the right lung was ventilated, with (n = 6) or without (n = 6) systemic lipopolysaccharide infusion. Computed tomography guided samples were harvested for NextGen RNA sequencing from atelectatic and aerated lung regions. The Wald test was used to detect differential gene expression as an absolute fold change greater than 1.5 and adjusted P value (Benjamini-Hochberg) less than 0.05. Functional analysis was performed by gene set enrichment analysis. RESULTS Lipopolysaccharide-unexposed atelectatic versus aerated regions presented 2,363 differentially expressed genes. Lipopolysaccharide exposure induced 3,767 differentially expressed genes in atelectatic lungs but only 1,197 genes in aerated lungs relative to the corresponding lipopolysaccharide-unexposed tissues. Gene set enrichment for immune response in atelectasis versus aerated tissues yielded negative normalized enrichment scores without lipopolysaccharide (less than -1.23, adjusted P value less than 0.05) but positive scores with lipopolysaccharide (greater than 1.33, adjusted P value less than 0.05). Leukocyte-related processes (e.g., leukocyte migration, activation, and mediated immunity) were enhanced in lipopolysaccharide-exposed atelectasis partly through interferon-stimulated genes. Furthermore, atelectasis was associated with negatively enriched gene sets involving alveolar-capillary barrier function irrespective of lipopolysaccharide (normalized enrichment scores less than -1.35, adjusted P value less than 0.05). Yes-associated protein signaling was dysregulated with lower nuclear distribution in atelectatic versus aerated lung (lipopolysaccharide-unexposed: 10.0 ± 4.2 versus 13.4 ± 4.2 arbitrary units, lipopolysaccharide-exposed: 8.1 ± 2.0 versus 11.3 ± 2.4 arbitrary units, effect of lung aeration, P = 0.003). CONCLUSIONS Atelectasis dysregulates the local pulmonary transcriptome with negatively enriched immune response and alveolar-capillary barrier function. Systemic lipopolysaccharide converts the transcriptomic immune response into positive enrichment but does not affect local barrier function transcriptomics. Interferon-stimulated genes and Yes-associated protein might be novel candidate targets for atelectasis-associated injury. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Congli Zeng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, United States
| | - Gabriel C. Motta-Ribeiro
- Biomedical Engineering Program, Alberto Luiz Coimbra Institute of Post-Graduation and Engineering Research, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Takuga Hinoshita
- Department of Intensive Care Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Marcos Adriano Lessa
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Tilo Winkler
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, United States
| | - Kira Grogg
- Department of Radiology, Massachusetts General Hospital, Boston, United States
| | - Nathan M Kingston
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - John N. Hutchinson
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, United States
| | - Lynette Marie Sholl
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Matthew D. Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, United States
| | - Rebecca M. Baron
- Department of Medicine (Pulmonary and Critical Care), Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Marcos F. Vidal Melo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, United States
| |
Collapse
|
34
|
Cheong SS, Akram KM, Matellan C, Kim SY, Gaboriau DCA, Hind M, del Río Hernández AE, Griffiths M, Dean CH. The Planar Polarity Component VANGL2 Is a Key Regulator of Mechanosignaling. Front Cell Dev Biol 2020; 8:577201. [PMID: 33195213 PMCID: PMC7658195 DOI: 10.3389/fcell.2020.577201] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/08/2020] [Indexed: 12/02/2022] Open
Abstract
VANGL2 is a component of the planar cell polarity (PCP) pathway, which regulates tissue polarity and patterning. The Vangl2 Lp mutation causes lung branching defects due to dysfunctional actomyosin-driven morphogenesis. Since the actomyosin network regulates cell mechanics, we speculated that mechanosignaling could be impaired when VANGL2 is disrupted. Here, we used live-imaging of precision-cut lung slices (PCLS) from Vangl2 Lp/+ mice to determine that alveologenesis is attenuated as a result of impaired epithelial cell migration. Vangl2 Lp/+ tracheal epithelial cells (TECs) and alveolar epithelial cells (AECs) exhibited highly disrupted actomyosin networks and focal adhesions (FAs). Functional assessment of cellular forces confirmed impaired traction force generation in Vangl2 Lp/+ TECs. YAP signaling in Vangl2 Lp airway epithelium was reduced, consistent with a role for VANGL2 in mechanotransduction. Furthermore, activation of RhoA signaling restored actomyosin organization in Vangl2 Lp/+ , confirming RhoA as an effector of VANGL2. This study identifies a pivotal role for VANGL2 in mechanosignaling, which underlies the key role of the PCP pathway in tissue morphogenesis.
Collapse
Affiliation(s)
- Sek-Shir Cheong
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Khondoker M. Akram
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Sally Yunsun Kim
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David C. A. Gaboriau
- Facility for Imaging by Light Microscopy, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- National Institute for Health Research, Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mark Griffiths
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Peri-Operative Medicine Department, St Bartholomew’s Hospital, London, United Kingdom
| | - Charlotte H. Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| |
Collapse
|
35
|
Reggiani F, Gobbi G, Ciarrocchi A, Sancisi V. YAP and TAZ Are Not Identical Twins. Trends Biochem Sci 2020; 46:154-168. [PMID: 32981815 DOI: 10.1016/j.tibs.2020.08.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
Yes-associated protein (YAP) and TAZ (WW domain containing transcription regulator 1, or WWTR1) are paralog transcriptional regulators, able to integrate mechanical, metabolic, and signaling inputs to regulate cell growth and differentiation during development and neoplastic progression. YAP and TAZ hold common and distinctive structural features, reflecting only partially overlapping regulatory mechanisms. The two paralogs interact with both shared and specific transcriptional partners and control nonidentical transcriptional programs. Although most of the available literature considers YAP and TAZ as functionally redundant, they play distinctive or even contrasting roles in different contexts. The issue of their divergent roles is currently underexplored but holds fundamental implications for mechanistic and translational studies. Here, we aim to review the available literature on the biological functions of YAP and TAZ, highlighting differential roles that distinguish these two paralogues.
Collapse
Affiliation(s)
- Francesca Reggiani
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Gobbi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Sancisi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
36
|
Conway RF, Frum T, Conchola AS, Spence JR. Understanding Human Lung Development through In Vitro Model Systems. Bioessays 2020; 42:e2000006. [PMID: 32310312 PMCID: PMC7433239 DOI: 10.1002/bies.202000006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/28/2020] [Indexed: 12/19/2022]
Abstract
An abundance of information about lung development in animal models exists; however, comparatively little is known about lung development in humans. Recent advances using primary human lung tissue combined with the use of human in vitro model systems, such as human pluripotent stem cell-derived tissue, have led to a growing understanding of the mechanisms governing human lung development. They have illuminated key differences between animal models and humans, underscoring the need for continued advancements in modeling human lung development and utilizing human tissue. This review discusses the use of human tissue and the use of human in vitro model systems that have been leveraged to better understand key regulators of human lung development and that have identified uniquely human features of development. This review also examines the implementation and challenges of human model systems and discusses how they can be applied to address knowledge gaps.
Collapse
Affiliation(s)
- Renee F Conway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Tristan Frum
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Ansley S Conchola
- Cell and Molecular Biology (CMB) Training Program, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Cell and Molecular Biology (CMB) Training Program, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, 48104, USA
| |
Collapse
|
37
|
Hsu PC, Yang CT, Jablons DM, You L. The Crosstalk between Src and Hippo/YAP Signaling Pathways in Non-Small Cell Lung Cancer (NSCLC). Cancers (Basel) 2020; 12:1361. [PMID: 32466572 PMCID: PMC7352956 DOI: 10.3390/cancers12061361] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 12/22/2022] Open
Abstract
The advancement of new therapies, including targeted therapies and immunotherapies, has improved the survival of non-small-cell lung cancer (NSCLC) patients in the last decade. Some NSCLC patients still do not benefit from therapies or encounter progressive disease during the course of treatment because they have intrinsic resistance, acquired resistance, or lack a targetable driver mutation. More investigations on the molecular biology of NSCLC are needed to find useful biomarkers for current therapies and to develop novel therapeutic strategies. Src is a non-receptor tyrosine kinase protein that interacts with cell surface growth factor receptors and the intracellular signaling pathway to maintain cell survival tumorigenesis in NSCLC. The Yes-associated protein (YAP) is one of the main effectors of the Hippo pathway and has been identified as a promoter of drug resistance, cancer progression, and metastasis in NSCLC. Here, we review studies that have investigated the activation of YAP as mediated by Src kinases and demonstrate that Src regulates YAP through three main mechanisms: (1) direct phosphorylation; (2) the activation of pathways repressing Hippo kinases; and (3) Hippo-independent mechanisms. Further work should focus on the efficacy of Src inhibitors in inhibiting YAP activity in NSCLC. In addition, future efforts toward developing potentially reasonable combinations of therapy targeting the Src-YAP axis using other therapies, including targeted therapies and/or immunotherapies, are warranted.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
- Division of Thoracic Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan;
| | - Cheng-Ta Yang
- Division of Thoracic Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan;
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - David M. Jablons
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
| | - Liang You
- Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA; (P.-C.H.); (D.M.J.)
| |
Collapse
|
38
|
Affiliation(s)
- David Warburton
- The Saban Research InstituteChildren's Hospital Los AngelesLos Angeles, California
| |
Collapse
|