1
|
Song JY, Wehbe F, Wong AK, Hall BM, Vander Heiden JA, Brightbill HD, Arron JR, Garfield DA, Dey A, Rock JR. YAP/TAZ activity in PDGFRα-expressing alveolar fibroblasts modulates AT2 proliferation through Wnt4. Cell Rep 2025; 44:115645. [PMID: 40333185 DOI: 10.1016/j.celrep.2025.115645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/06/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
The Hippo pathway, mediated by its transcriptional effectors Yes-associated protein 1 (YAP) and WW-domain-containing transcription regulator 1 (TAZ), is crucial in maintaining lung homeostasis and facilitating injury repair. While its roles in epithelial cells are well established, its regulatory effects on lung fibroblasts remain less understood. We engineered a mouse model for the inducible knockdown of YAP/TAZ and showed that fibroblast-specific knockdown enhances PDGFRα+ alveolar fibroblasts' support for alveolar-epithelial-stem-cell-derived organoids in vitro. Single-cell profiling revealed changes in fibroblast subpopulations, including the emergence of a Wnt4+ enriched subpopulation. Epigenomic analyses revealed shifts in transcription factor motif enrichment in both fibroblasts and epithelial cells due to fibroblast YAP/TAZ suppression. Further computational and in vivo analyses confirmed increased Wnt signaling and Wnt4 expression in PDGFRα-lineage+ fibroblasts, which enhanced SPC+ alveolar type 2 (AT2) cell proliferation. These findings highlight a mechanistic role of YAP/TAZ in PDGFRα+ alveolar fibroblasts in supporting AT2 cell maintenance and proliferation via Wnt4 secretion.
Collapse
Affiliation(s)
- Jane Y Song
- Department of Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Fabien Wehbe
- Data & Analytics Chapter-Computational Science, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Aaron K Wong
- Department of Immunology and Infectious Diseases, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ben M Hall
- Department of Immunology and Infectious Diseases, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A Vander Heiden
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hans D Brightbill
- Department of Immunology and Infectious Diseases, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph R Arron
- Department of Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - David A Garfield
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason R Rock
- Department of Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
2
|
Walke P, Price JDW, Vizeacoumar FS, Joseph N, Maranda V, Chowdhury B, Patel J, Zhang Y, Dong H, New L, Ganapathysamy A, Gong LH, Lazell-Wright M, Elhasasna H, Bhanumathy KK, Wu Y, Wang Y, Freywald A, Krishnan A, Vizeacoumar FJ. A novel role for Neurog2 in MYCN driven neuroendocrine plasticity of prostate cancer. Oncogene 2025:10.1038/s41388-025-03413-0. [PMID: 40301542 DOI: 10.1038/s41388-025-03413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) presents a formidable clinical challenge owing to its aggressive progression and resistance to conventional therapies. A key driver of NEPC is the overexpression of MYCN, a well-established oncogene associated with neuroendocrine tumors. However, efforts to directly inhibit the N-Myc protein encoded by this gene have resulted in limited success, thereby hindering therapeutic advancements. To overcome this obstacle, we conducted unbiased genome-wide screening using isogenic prostate cancer cell lines to identify the synthetic vulnerabilities of MYCN. Among the identified candidates, NEUROG2 emerged as a significant candidate. Neurog2 is a proneural transcription factor (PTF) known for its role in developmental processes and trans-differentiation of adult cells. Our findings demonstrate that Neurog2 depletion does not affect non-malignant cells but significantly suppresses the growth of MYCN-overexpressing cells and tumors in orthotopic NEPC models. Furthermore, our observations indicate that Neurog2-driven modulation of PTFs potentially contribute to NEPC development. Thus, targeting Neurog2 holds promise as an effective therapeutic strategy for MYCN-overexpressing NEPC.
Collapse
Affiliation(s)
- Prachi Walke
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Jared D W Price
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Frederick S Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Nickson Joseph
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Vincent Maranda
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Bari Chowdhury
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Jay Patel
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | - Yue Zhang
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - He Dong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Lara New
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada
| | | | - Li Hui Gong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Mary Lazell-Wright
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Hussain Elhasasna
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Kalpana K Bhanumathy
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Yuliang Wu
- Department of Biochemistry Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Yuzhuo Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
- BC Cancer Research Centre, Vancouver, BC, Canada.
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada.
| | - Anand Krishnan
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St, Saskatoon, S7K 0M7, SK, Canada.
| | - Franco J Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada.
- Cancer Research Department, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, S7N 5E5, SK, Canada.
| |
Collapse
|
3
|
Lao Z, Chen X, Pan B, Fang B, Yang W, Qian Y. Pharmacological regulators of Hippo pathway: Advances and challenges of drug development. FASEB J 2025; 39:e70438. [PMID: 40100056 DOI: 10.1096/fj.202401895rr] [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: 09/06/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
The Hippo signaling pathway is crucial in regulating organ size, tumor progression, tissue regeneration, and bone homeostasis. Inactivation of the Hippo pathway results in the nuclear translocation and activation of YAP/TAZ. This activation not only promotes tumor progression but also enhances tissue regeneration, wound healing, and maintenance of bone stability Although its discovery occurred over two decades ago, developing effective inhibitors or activators for the Hippo pathway remains challenging. Recently, however, the pace of advancements in developing Hippo signaling-related agonists and antagonists has accelerated, with some drugs that target TEAD advancing to clinical trials and showing promise for treating related diseases. This review summarizes the progress in research on Hippo signaling-related agonists and inhibitors, offering an in-depth analysis of their regulatory mechanisms, pharmacological properties, and potential in vivo applications.
Collapse
Affiliation(s)
- Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Fang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Peng Y, Yuan Q, Zhou S, Gan J, Shen Z, Xia X, Jiang Y, Chen Q, Yuan Y, He G, Wei Q, Feng X. FAK mediates mechanical signaling to maintain epithelial homeostasis through YAP/TAZ-TEADs. Histochem Cell Biol 2025; 163:31. [PMID: 39918604 DOI: 10.1007/s00418-025-02360-x] [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] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Epithelial homeostasis ensures that the epithelium can perform its normal physiological functions. Mechanical signaling response through integrin-mediated adhesions of the basement membrane (BM) is crucial for maintaining epithelial homeostasis. The essential mechanosensors YAP and the paralog TAZ (YAP/TAZ) have been shown to play a critical role in epithelial homeostasis, but the key regulator that mediates mechanical signaling to YAP/TAZ in maintaining epithelial homeostasis has not been fully understood. In this study, we noticed that mechanical signals correlated with YAP/TAZ activation and basal state maintenance in epithelial stem/progenitor cells through immunohistochemistry. Subsequently, we found that inhibition of focal adhesion kinase (FAK) suppressed YAP/TAZ activation in the human keratinocyte line HaCaT cells. Furthermore, inhibition of the interaction between YAP/TAZ and the transcriptional enhanced associate domains (TEADs) resulted in the differentiation of HaCaT cells. Finally, we used primary mouse epithelial cells to reconstruct the epithelium in vitro and found that FAK inhibition led to both a reduction in YAP/TAZ activity and an increase of differentiation in the basal layer cells. In conclusion, our findings reveal that FAK mediates mechanical signaling to maintain epithelial homeostasis via YAP/TAZ-TEADs.
Collapse
Affiliation(s)
- Yang Peng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiuyun Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jianguo Gan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhengzhong Shen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuchen Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, 310000, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Gu He
- Department of Dermatology and Venerology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China.
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
McCall AS, Gutor S, Tanjore H, Burman A, Sherrill T, Chapman M, Calvi CL, Han D, Camarata J, Hunt RP, Nichols D, Banovich NE, Lawson WE, Gokey JJ, Kropski JA, Blackwell TS. Hypoxia-inducible factor 2 regulates alveolar regeneration after repetitive injury in three-dimensional cellular and in vivo models. Sci Transl Med 2025; 17:eadk8623. [PMID: 39772774 PMCID: PMC12051389 DOI: 10.1126/scitranslmed.adk8623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 09/23/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease in which repetitive epithelial injury and incomplete alveolar repair result in accumulation of profibrotic intermediate/transitional "aberrant" epithelial cell states. The mechanisms leading to the emergence and persistence of aberrant epithelial populations in the distal lung remain incompletely understood. By interrogating single-cell RNA sequencing (scRNA-seq) data from patients with IPF and a mouse model of repeated lung epithelial injury, we identified persistent activation of hypoxia-inducible factor (HIF) signaling in these aberrant epithelial cells. Using mouse genetic lineage-tracing strategies together with scRNA-seq, we found that these disease-emergent aberrant epithelial cells predominantly arose from airway-derived (Scgb1a1-CreER-traced) progenitors and exhibited transcriptional programs of Hif2a activation. In mice treated with repetitive intratracheal bleomycin, deletion of Epas1 (Hif2a) but not Hif1a, from airway-derived progenitors, or administration of the small-molecule HIF2 inhibitor PT-2385, using both prevention and rescue approaches, attenuated experimental lung fibrosis, reduced the appearance of aberrant epithelial cells, and promoted alveolar repair. In mouse alveolar organoids, genetic or pharmacologic inhibition of Hif2 promoted alveolar differentiation of airway-derived epithelial progenitors. In addition, treatment of human distal lung organoids with PT-2385 increased colony-forming efficiency, enhanced protein and transcriptional markers of alveolar type 2 epithelial cell maturation, and prevented the emergence of aberrant epithelial cells. Together, these studies showed that HIF2 activation drives the emergence of aberrant epithelial populations after repetitive injury and that targeted HIF2 inhibition may represent an effective therapeutic strategy to promote functional alveolar repair in IPF and other interstitial lung diseases.
Collapse
Affiliation(s)
- A. Scott McCall
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Sergey Gutor
- Department of Internal Medicine, University of Michigan Medical School, 48109
| | - Hari Tanjore
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ankita Burman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- University Health Network, Toronto Lung Transplant Program, Toronto, Ontario, Canada, M5G 2N2
| | - Taylor Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Micah Chapman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Carla L. Calvi
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jane Camarata
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Raphael P. Hunt
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David Nichols
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | | | - William E. Lawson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Veterans Affairs Medical Center, Nashville, TN, 37212
| | - Jason J. Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Translational Genomics Research Institute, Phoenix, AZ, 85004
- Department of Veterans Affairs Medical Center, Nashville, TN, 37212
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Internal Medicine, University of Michigan Medical School, 48109
| |
Collapse
|
6
|
Bridges JP, Vladar EK, Kurche JS, Krivoi A, Stancil IT, Dobrinskikh E, Hu Y, Sasse SK, Lee JS, Blumhagen RZ, Yang IV, Gerber AN, Peljto AL, Evans CM, Redente EF, Riches DW, Schwartz DA. Progressive lung fibrosis: reprogramming a genetically vulnerable bronchoalveolar epithelium. J Clin Invest 2025; 135:e183836. [PMID: 39744946 PMCID: PMC11684817 DOI: 10.1172/jci183836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is etiologically complex, with well-documented genetic and nongenetic origins. In this Review, we speculate that the development of IPF requires two hits: the first establishes a vulnerable bronchoalveolar epithelium, and the second triggers mechanisms that reprogram distal epithelia to initiate and perpetuate a profibrotic phenotype. While vulnerability of the bronchoalveolar epithelia is most often driven by common or rare genetic variants, subsequent injury of the bronchoalveolar epithelia results in persistent changes in cell biology that disrupt tissue homeostasis and activate fibroblasts. The dynamic biology of IPF can best be contextualized etiologically and temporally, including stages of vulnerability, early disease, and persistent and progressive lung fibrosis. These dimensions of IPF highlight critical mechanisms that adversely disrupt epithelial function, activate fibroblasts, and lead to lung remodeling. Together with better recognition of early disease, this conceptual approach should lead to the development of novel therapeutics directed at the etiologic and temporal drivers of lung fibrosis that will ultimately transform the care of patients with IPF from palliative to curative.
Collapse
Affiliation(s)
- James P. Bridges
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eszter K. Vladar
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jonathan S. Kurche
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Andrei Krivoi
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ian T. Stancil
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, School of Medicine, Stanford, California, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yan Hu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Joyce S. Lee
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel Z. Blumhagen
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Ivana V. Yang
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Anna L. Peljto
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher M. Evans
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - David W.H. Riches
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A. Schwartz
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
7
|
Yang J, Li Y, Huang Y, Chen H, Sui P. Unlocking lung regeneration: insights into progenitor cell dynamics and metabolic control. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:31. [PMID: 39676102 PMCID: PMC11646969 DOI: 10.1186/s13619-024-00212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Regenerative responses are particularly important in the lungs, which are critical for gas exchange and frequently challenged by environmental insults. The lung progenitor cells play a central role in the lung regeneration response, and their dysfunction is associated with various lung diseases. Understanding the mechanisms regulating lung progenitor cell function is essential for developing new therapeutic approaches to promote lung regeneration. This review summarizes recent advancements in the field of lung regeneration, focusing on the metabolic control of lung progenitor cell function. We discuss cell lineage plasticity and cell-cell signaling under different physiological conditions. Additionally, we highlight the connection between progenitor cell dysfunction and lung diseases, emphasizing the need to develop new therapeutic strategies in regenerative medicine to improve lung regenerative capacity.
Collapse
Affiliation(s)
- Jiaying Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yawen Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huaiyong Chen
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China.
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin, China.
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
8
|
Warren R, Klinkhammer K, Lyu H, Knopp J, Yuan T, Yao C, Stripp B, De Langhe SP. Cell competition drives bronchiolization and pulmonary fibrosis. Nat Commun 2024; 15:10624. [PMID: 39639058 PMCID: PMC11621346 DOI: 10.1038/s41467-024-54997-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive respiratory scarring disease arising from the maladaptive differentiation of lung stem cells into bronchial epithelial cells rather than into alveolar type 1 (AT1) cells, which are responsible for gas exchange. Here, we report that healthy lungs maintain their stem cells through tonic Hippo and β-catenin signaling, which promote Yap/Taz degradation and allow for low-level expression of the Wnt target gene Myc. Inactivation of upstream activators of the Hippo pathway in lung stem cells inhibits this tonic β-catenin signaling and Myc expression and promotes their Taz-mediated differentiation into AT1 cells. Vice versa, increased Myc in collaboration with Yap promotes the differentiation of lung stem cells along the basal and myoepithelial-like lineages allowing them to invade and bronchiolize the lung parenchyma in a process reminiscent of submucosal gland development. Our findings indicate that stem cells exhibiting the highest Myc levels become supercompetitors that drive remodeling, whereas loser cells with lower Myc levels terminally differentiate into AT1 cells.
Collapse
Affiliation(s)
- Rachel Warren
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kylie Klinkhammer
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN, USA
| | - Handeng Lyu
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph Knopp
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tingting Yuan
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Changfu Yao
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barry Stripp
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stijn P De Langhe
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Liu J, Luo D, Huang H, Mu R, Yuan J, Jiang M, Lin C, Xiang H, Lin X, Song H, Zhang Y. Hippo cooperates with p53 to regulate lung airway mucous cell metaplasia. Dis Model Mech 2024; 17:dmm052074. [PMID: 39428818 PMCID: PMC11603118 DOI: 10.1242/dmm.052074] [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: 08/21/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024] Open
Abstract
Airway mucous cell metaplasia is a significant feature of many chronic airway diseases, such as chronic obstructive pulmonary disease, cystic fibrosis and asthma. However, the mechanisms underlying this process remain poorly understood. Here, we employed in vivo mouse genetic models to demonstrate that Hippo and p53 (encoded by Trp53) cooperate to modulate the differentiation of club cells into goblet cells. We revealed that ablation of Mst1 (Stk4) and Mst2 (Stk3), encoding the core components of Hippo signaling, significantly reduces mucous metaplasia in the lung airways in a lipopolysaccharide (LPS)-induced lung inflammation murine model while promoting club cell proliferation in a Yap (Yap1)-dependent manner. Additionally, we showed that deleting Mst1/2 is sufficient to suppress p53 deficiency-mediated goblet cell metaplasia. Finally, single-cell RNA-sequencing analysis revealed downregulation of YAP and p53 signaling in goblet cells in human airways. These findings underscore the important role of Hippo and p53 signaling in regulating airway mucous metaplasia.
Collapse
Affiliation(s)
- Jiangying Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Luo
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haidi Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Rongzi Mu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianghong Yuan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ming Jiang
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310030, Zhejiang, China
| | - Chuwen Lin
- Department of Histology and Embryology, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Honggang Xiang
- Department of General Surgery, Pudong New Area People's Hospital, Shanghai 201299, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University Shanghai, Shanghai 200438, China
| | - Haihan Song
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai 201299, China
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC V6B 5A6, Canada
| | - Yongchun Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Inner Mongolia Research Institute, Shenzhen Research Institute, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of General Surgery, Pudong New Area People's Hospital, Shanghai 201299, China
| |
Collapse
|
10
|
Blakely WJ, Hatterschide J, White EA. HPV18 E7 inhibits LATS1 kinase and activates YAP1 by degrading PTPN14. mBio 2024; 15:e0181124. [PMID: 39248565 PMCID: PMC11481495 DOI: 10.1128/mbio.01811-24] [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: 06/25/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024] Open
Abstract
High-risk human papillomavirus (HPV) oncoproteins inactivate cellular tumor suppressors to reprogram host cell signaling pathways. HPV E7 proteins bind and degrade the tumor suppressor PTPN14, thereby promoting the nuclear localization of the YAP1 oncoprotein and inhibiting keratinocyte differentiation. YAP1 is a transcriptional coactivator that drives epithelial cell stemness and self-renewal. YAP1 activity is inhibited by the highly conserved Hippo pathway, which is frequently inactivated in human cancers. MST1/2 and LATS1/2 kinases form the core of the Hippo kinase cascade. Active LATS1 kinase is phosphorylated on threonine 1079 and inhibits YAP1 by phosphorylating it on amino acids including serine 127. Here, we tested the effect of high-risk (carcinogenic) HPV18 E7 on Hippo pathway activity. We found that either PTPN14 knockout or PTPN14 degradation by HPV18 E7 decreased the phosphorylation of LATS1 T1079 and YAP1 S127 in human keratinocytes and inhibited keratinocyte differentiation. Conversely, PTPN14-dependent differentiation required LATS kinases and certain PPxY motifs in PTPN14. Neither MST1/2 kinases nor the putative PTPN14 phosphatase active sites were required for PTPN14 to promote differentiation. Together, these data support that PTPN14 inactivation or degradation of PTPN14 by HPV18 E7 reduce LATS1 activity, promoting active YAP1 and inhibiting keratinocyte differentiation.IMPORTANCEThe Hippo kinase cascade inhibits YAP1, an oncoprotein and driver of cell stemness and self-renewal. There is mounting evidence that the Hippo pathway is targeted by tumor viruses including human papillomavirus. The high-risk HPV E7 oncoprotein promotes YAP1 nuclear localization and the carcinogenic activity of high-risk HPV E7 requires YAP1 activity. Blocking HPV E7-dependent YAP1 activation could inhibit HPV-mediated carcinogenesis, but the mechanism by which HPV E7 activates YAP1 has not been elucidated. Here we report that by degrading the tumor suppressor PTPN14, HPV18 E7 inhibits LATS1 kinase, reducing inhibitory phosphorylation on YAP1. These data support that an HPV oncoprotein can inhibit Hippo signaling to activate YAP1 and strengthen the link between PTPN14 and Hippo signaling in human epithelial cells.
Collapse
Affiliation(s)
- William J. Blakely
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joshua Hatterschide
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth A. White
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Berdnikovs S, Newcomb DC, Hartert TV. How early life respiratory viral infections impact airway epithelial development and may lead to asthma. Front Pediatr 2024; 12:1441293. [PMID: 39156016 PMCID: PMC11327159 DOI: 10.3389/fped.2024.1441293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Childhood asthma is a common chronic disease of the airways that results from host and environment interactions. Most risk factor studies of asthma point to the first year of life as a susceptibility window of mucosal exposure that directly impacts the airway epithelium and airway epithelial cell development. The development of the airway epithelium, which forms a competent barrier resulting from coordinated interactions of different specialized cell subsets, occurs during a critical time frame in normal postnatal development in the first year of life. Understanding the normal and aberrant developmental trajectory of airway epithelial cells is important in identifying pathways that may contribute to barrier dysfunction and asthma pathogenesis. Respiratory viruses make first contact with and infect the airway mucosa. Human rhinovirus (HRV) and respiratory syncytial virus (RSV) are mucosal pathogens that are consistently identified as asthma risk factors. Respiratory viruses represent a unique early life exposure, different from passive irritant exposures which injure the developing airway epithelium. To replicate, respiratory viruses take over the host cell transcriptional and translational processes and exploit host cell energy metabolism. This takeover impacts the development and differentiation processes of airway epithelial cells. Therefore, delineating the mechanisms through which early life respiratory viral infections alter airway epithelial cell development will allow us to understand the maturation and heterogeneity of asthma and develop tools tailored to prevent disease in specific children. This review will summarize what is understood about the impact of early life respiratory viruses on the developing airway epithelium and define critical gaps in our knowledge.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dawn C. Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tina V. Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
12
|
Ji X, Li Y, Liu M, Chen L, Zhang X, Wang M, Tian S, Lu L, Zhang M, Zheng Y, Tang J. Diesel exhaust exposure induced squamous metaplasia of corneal epithelium via yes-associated protein activation. CHEMOSPHERE 2024; 362:142564. [PMID: 38885762 DOI: 10.1016/j.chemosphere.2024.142564] [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: 02/03/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Atmospheric pollution has been demonstrated to be associated with ocular surface diseases characterized by corneal epithelial damage, including impaired barrier function and squamous metaplasia. However, the specific mechanisms underlying the impact of atmospheric pollution on corneal damage are still unknow. To address this gap in knowledge, we conducted a study using a whole-body exposure system to investigate the detrimental effects of traffic-related air pollution, specifically diesel exhaust (DE), on corneal epithelium in C57BL/6 mice over a 28-day period. Following DE exposure, the pathological alterations in corneal epithelium, including significant increase in corneal thickness and epithelial stratification, were observed in mice. Additionally, exposure to DE was also shown to disrupt the barrier functions of corneal epithelium, leading to excessive proliferation of basal cells and even causing squamous metaplasia in corneal epithelium. Further studies have found that the activation of yes-associated protein (YAP), characterized by nuclear translocation, may play a significant role in DE-induced corneal squamous metaplasia. In vitro assays confirmed that DE exposure triggered the YAP/β-catenin pathway, resulting in squamous metaplasia and destruction of barrier functions. These findings provide the preliminary evidence that YAP activation is one of the mechanisms of the damage to corneal epithelium caused by traffic-related air pollution. These findings contribute to the knowledge base for promoting eye health in the context of atmospheric pollution.
Collapse
Affiliation(s)
- Xiaoya Ji
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yanting Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Meike Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Linfei Chen
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xinglin Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Mingyue Wang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Shuhan Tian
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Lin Lu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Mingliang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jinglong Tang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
13
|
Quach H, Farrell S, Wu MJM, Kanagarajah K, Leung JWH, Xu X, Kallurkar P, Turinsky AL, Bear CE, Ratjen F, Kalish B, Goyal S, Moraes TJ, Wong AP. Early human fetal lung atlas reveals the temporal dynamics of epithelial cell plasticity. Nat Commun 2024; 15:5898. [PMID: 39003323 PMCID: PMC11246468 DOI: 10.1038/s41467-024-50281-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 07/05/2024] [Indexed: 07/15/2024] Open
Abstract
Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.
Collapse
Affiliation(s)
- Henry Quach
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Spencer Farrell
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Ming Jia Michael Wu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kayshani Kanagarajah
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Wai-Hin Leung
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqiao Xu
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Prajkta Kallurkar
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrei L Turinsky
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christine E Bear
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian Kalish
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sidhartha Goyal
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Blakely WJ, Hatterschide J, White EA. HPV18 E7 inhibits LATS1 kinase and activates YAP1 by degrading PTPN14. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583953. [PMID: 38496413 PMCID: PMC10942435 DOI: 10.1101/2024.03.07.583953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
High-risk human papillomavirus (HPV) oncoproteins inactivate cellular tumor suppressors to reprogram host cell signaling pathways. HPV E7 proteins bind and degrade the tumor suppressor PTPN14, thereby promoting the nuclear localization of the YAP1 oncoprotein and inhibiting keratinocyte differentiation. YAP1 is a transcriptional coactivator that drives epithelial cell stemness and self-renewal. YAP1 activity is inhibited by the highly conserved Hippo pathway, which is frequently inactivated in human cancers. MST1/2 and LATS1/2 kinases form the core of the Hippo kinase cascade. Active LATS1 kinase is phosphorylated on threonine 1079 and inhibits YAP1 by phosphorylating it on amino acids including serine 127. Here, we tested the effect of high-risk (carcinogenic) HPV18 E7 on Hippo pathway activity. We found that either PTPN14 knockout or PTPN14 degradation by HPV18 E7 decreased phosphorylation of LATS1 T1079 and YAP1 S127 in human keratinocytes and inhibited keratinocyte differentiation. Conversely, PTPN14-dependent differentiation required LATS kinases and certain PPxY motifs in PTPN14. Neither MST1/2 kinases nor the putative PTPN14 phosphatase active site were required for PTPN14 to promote differentiation. Taken together, these data support that PTPN14 inactivation or degradation of PTPN14 by HPV18 E7 reduce LATS1 activity, promoting active YAP1 and inhibiting keratinocyte differentiation.
Collapse
Affiliation(s)
- William J. Blakely
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Joshua Hatterschide
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Current address: Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Elizabeth A. White
- Department of Otorhinolaryngology: Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
15
|
Stuart WD, Ito M, Baldauf IF, Fukazawa T, Yamatsuji T, Tsuchiya T, Watanabe H, Okada M, Snyder EL, Mino-Kenudson M, Guo M, Maeda Y. Patho-transcriptomic analysis of invasive mucinous adenocarcinoma of the lung (IMA): comparison with lung adenocarcinoma with signet ring cell features (SRCC). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598839. [PMID: 38948839 PMCID: PMC11212912 DOI: 10.1101/2024.06.13.598839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Invasive mucinous adenocarcinoma (IMA) comprises ∼5% of lung adenocarcinoma. There is no effective therapy for IMA when surgical resection is not possible. IMA is sometimes confused with adenocarcinoma with signet ring cell features (SRCC) pathologically since both adenocarcinomas feature tumor cells with abundant intracellular mucin. The molecular mechanisms by which such mucin-producing lung adenocarcinomas develop remain unknown. Methods Using a Visium spatial transcriptomics approach, we analyzed IMA and compared it with SRCC patho-transcriptomically. Combining spatial transcriptomics data with in vitro studies using RNA-seq and ChIP-seq, we assessed downstream targets of transcription factors HNF4A and SPDEF that are highly expressed in IMA and/or SRCC. Results Spatial transcriptomics analysis indicated that there are 6 distinct cell clusters in IMA and SRCC. Notably, two clusters (C1 and C3) of mucinous tumor cells exist in both adenocarcinomas albeit at a different ratio. Importantly, a portion of genes (e.g., NKX2-1 , GKN1 , HNF4A and FOXA3 ) are distinctly expressed while some mucous-related genes (e.g., SPDEF and FOXA2 ) are expressed in both adenocarcinomas. We determined that HNF4A induces MUC3A/B and TM4SF4 and that BI 6015, an HNF4A antagonist, suppressed the growth of IMA cells. Using mutant SPDEF that is associated with COVID-19, we also determined that an intact DNA-binding domain of SPDEF is required for SPDEF-mediated induction of mucin genes ( MUC5AC , MUC5B and AGR2 ). Additionally, we found that XMU-MP-1, a SPDEF inhibitor, suppressed the growth of IMA cells. Conclusion These results revealed that IMA and SRCC contain heterogenous tumor cell types, some of which are targetable.
Collapse
|
16
|
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
|
17
|
Warren R, Klinkhammer K, Lyu H, Yao C, Stripp B, De Langhe SP. Cell competition drives bronchiolization and pulmonary fibrosis. RESEARCH SQUARE 2024:rs.3.rs-4177351. [PMID: 38746309 PMCID: PMC11092845 DOI: 10.21203/rs.3.rs-4177351/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive scarring disease arising from the maladaptive differentiation of lung stem cells into bronchial epithelial cells rather than into alveolar type 1 (AT1) cells, which are responsible for gas exchange. Here, we report that healthy lungs maintain their stem cells through tonic Hippo and β-catenin signaling, which promote Yap/Taz degradation and allow for low level expression of the Wnt target gene Myc. Inactivation of upstream activators of the Hippo pathway in lung stem cells inhibits this tonic β-catenin signaling and Myc expression and promotes their Taz mediated differentiation into AT1 cells. Vice versa, increased Myc in collaboration with Yap promotes the differentiation of lung stem cells along the basal and myoepithelial like lineages allowing them to invade and bronchiolize the lung parenchyma in a process reminiscent of submucosal gland development. Our findings indicate that stem cells exhibiting the highest Myc levels become supercompetitors that drive remodeling, whereas loser cells with lower Myc levels terminally differentiate into AT1 cells.
Collapse
Affiliation(s)
- Rachel Warren
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kylie Klinkhammer
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Handeng Lyu
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Changfu Yao
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Barry Stripp
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stijn P. De Langhe
- Department of Medicine, Division of Pulmonary and Critical Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Kim HK, Jeong H, Jeong MG, Won HY, Lee G, Bae SH, Nam M, Lee SH, Hwang GS, Hwang ES. TAZ deficiency impairs the autophagy-lysosomal pathway through NRF2 dysregulation and lysosomal dysfunction. Int J Biol Sci 2024; 20:2592-2606. [PMID: 38725855 PMCID: PMC11077375 DOI: 10.7150/ijbs.88897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Transcriptional coactivator with a PDZ-binding motif (TAZ) plays a key role in normal tissue homeostasis and tumorigenesis through interaction with several transcription factors. In particular, TAZ deficiency causes abnormal alveolarization and emphysema, and persistent TAZ overexpression contributes to lung cancer and pulmonary fibrosis, suggesting the possibility of a complex mechanism of TAZ function. Recent studies suggest that nuclear factor erythroid 2-related factor 2 (NRF2), an antioxidant defense system, induces TAZ expression during tumorigenesis and that TAZ also activates the NRF2-mediated antioxidant pathway. We thus thought to elucidate the cross-regulation of TAZ and NRF2 and the underlying molecular mechanisms and functions. TAZ directly interacted with NRF2 through the N-terminal domain and suppressed the transcriptional activity of NRF2 by preventing NRF2 from binding to DNA. In addition, the return of NRF2 to basal levels after signaling was inhibited in TAZ deficiency, resulting in sustained nuclear NRF2 levels and aberrantly increased expression of NRF2 targets. TAZ deficiency failed to modulate optimal NRF2 signaling and concomitantly impaired lysosomal acidification and lysosomal enzyme function, accumulating the abnormal autophagy vesicles and reactive oxygen species and causing protein oxidation and cellular damage in the lungs. TAZ restoration to TAZ deficiency normalized dysregulated NRF2 signaling and aberrant lysosomal function and triggered the normal autophagy-lysosomal pathway. Therefore, TAZ is indispensable for the optimal regulation of NRF2-mediated autophagy-lysosomal pathways and for preventing pulmonary damage caused by oxidative stress and oxidized proteins.
Collapse
Affiliation(s)
- Hyo Kyeong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hana Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hee Yeon Won
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Gibbeum Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Soo Han Bae
- College of Medicine, Severance Biomedical Science Institute, Yonsei University, Seoul 03722, Korea
| | - Miso Nam
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
19
|
Zhong Z, Jiao Z, Yu FX. The Hippo signaling pathway in development and regeneration. Cell Rep 2024; 43:113926. [PMID: 38457338 DOI: 10.1016/j.celrep.2024.113926] [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: 12/02/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.
Collapse
Affiliation(s)
- Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
20
|
López-Posadas R, Bagley DC, Pardo-Pastor C, Ortiz-Zapater E. The epithelium takes the stage in asthma and inflammatory bowel diseases. Front Cell Dev Biol 2024; 12:1258859. [PMID: 38529406 PMCID: PMC10961468 DOI: 10.3389/fcell.2024.1258859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
The epithelium is a dynamic barrier and the damage to this epithelial layer governs a variety of complex mechanisms involving not only epithelial cells but all resident tissue constituents, including immune and stroma cells. Traditionally, diseases characterized by a damaged epithelium have been considered "immunological diseases," and research efforts aimed at preventing and treating these diseases have primarily focused on immuno-centric therapeutic strategies, that often fail to halt or reverse the natural progression of the disease. In this review, we intend to focus on specific mechanisms driven by the epithelium that ensure barrier function. We will bring asthma and Inflammatory Bowel Diseases into the spotlight, as we believe that these two diseases serve as pertinent examples of epithelium derived pathologies. Finally, we will argue how targeting the epithelium is emerging as a novel therapeutic strategy that holds promise for addressing these chronic diseases.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universtiy Eralngen-Nürnberg, Erlangen, Germany
| | - Dustin C. Bagley
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Carlos Pardo-Pastor
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- Instituto Investigación Hospital Clínico-INCLIVA, Valencia, Spain
| |
Collapse
|
21
|
Hata K, Tsubouchi K, Suzuki K, Eto D, Ando H, Yanagihara T, Kan-O K, Okamoto I. Surfactant protein D prevents mucin overproduction in airway goblet cells via SIRPα. Sci Rep 2024; 14:1799. [PMID: 38245585 PMCID: PMC10799941 DOI: 10.1038/s41598-024-52328-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Abstract
Mucin overproduction is a common feature of chronic airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), and exacerbates their underlying respiratory condition. Surfactant protein D (SP-D) protects against airway diseases through modulation of immune reactions, but whether it also exerts direct effects on airway epithelial cells has remained unclear. Therefore, we sought to investigate the inhibitory role of SP-D on mucin production in airway epithelial cells. We prepared air-liquid interface (ALI) cultures of human primary bronchial epithelial cells (HBECs), which recapitulated a well-differentiated human airway epithelium. Benzo(a)pyrene (BaP), a key toxicant in cigarette smoke, induced mucin 5AC (MUC5AC) production in ALI-cultured HBECs, airway secretory cell lines, and airway epithelia of mice. Then, the protective effects of SP-D against the BaP-induced mucin overproduction were examined. BaP increased MUC5AC production in ALI cultures of HBECs, and this effect was attenuated by SP-D. SP-D also suppressed the BaP-induced phosphorylation of extracellular signal-regulated kinase (ERK) and MUC5AC expression in NCI-H292 goblet-like cells, but not in NCI-H441 club-like cells. Signal regulatory protein α (SIRPα) was found to be expressed in HBECs and NCI-H292 cells but absent in NCI-H441 cells. In NCI-H292 cells, SP-D activated SH2 domain-containing tyrosine phosphatase-1 (SHP-1), downstream of SIRPα, and knockdown of SIRPα abolished the suppressive effects of SP-D on BaP-induced ERK phosphorylation and MUC5AC production. Consistent with these in vitro findings, intratracheal instillation of SP-D prevented the BaP-induced phosphorylation of ERK and Muc5ac expression in airway epithelial cells in a mouse model. SP-D acts directly on airway epithelial cells to inhibit mucin secretion through ligation of SIRPα and SHP-1-mediated dephosphorylation of ERK. Targeting of SIRPα is therefore a potential new therapeutic approach to suppression of mucin hypersecretion in chronic airway diseases such as COPD and asthma.
Collapse
Affiliation(s)
- Kentaro Hata
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kazuya Tsubouchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Kunihiro Suzuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Daisuke Eto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hiroyuki Ando
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Toyoshi Yanagihara
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keiko Kan-O
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
22
|
Gelbard MK, Munger K. Human papillomaviruses: Knowns, mysteries, and unchartered territories. J Med Virol 2023; 95:e29191. [PMID: 37861365 PMCID: PMC10608791 DOI: 10.1002/jmv.29191] [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: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
There has been an explosion in the number of papillomaviruses that have been identified and fully sequenced. Yet only a minute fraction of these has been studied in any detail. Most of our molecular research efforts have focused on the E6 and E7 proteins of "high-risk," cancer-associated human papillomaviruses (HPVs). Interactions of the high-risk HPV E6 and E7 proteins with their respective cellular targets, the p53 and the retinoblastoma tumor suppressors, have been investigated in minute detail. Some have thus questioned if research on papillomaviruses remains an exciting and worthwhile area of investigation. However, fundamentally new insights on the biological activities and cellular targets of the high-risk HPV E6 and E7 proteins have been discovered and previously unstudied HPVs have been newly associated with human diseases. HPV infections continue to be an important cause of human morbidity and mortality and since there are no antivirals to combat HPV infections, research on HPVs should remain attractive to new investigators and biomedical funding agencies, alike.
Collapse
Affiliation(s)
- Maya K. Gelbard
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences
- Department of Developmental, Molecular and Cellular Biology, Tufts University School of Medicine, Boston, MA 02111
| | - Karl Munger
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences
- Department of Developmental, Molecular and Cellular Biology, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Ning B, Tilston-Lunel AM, Simonetti J, Hicks-Berthet J, Matschulat A, Pfefferkorn R, Spira A, Edwards M, Mazzilli S, Lenburg ME, Beane JE, Varelas X. Convergence of YAP/TAZ, TEAD and TP63 activity is associated with bronchial premalignant severity and progression. J Exp Clin Cancer Res 2023; 42:116. [PMID: 37150829 PMCID: PMC10165825 DOI: 10.1186/s13046-023-02674-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Bronchial premalignant lesions (PMLs) are composed primarily of cells resembling basal epithelial cells of the airways, which through poorly understood mechanisms have the potential to progress to lung squamous cell carcinoma (LUSC). Despite ongoing efforts that have mapped gene expression and cell diversity across bronchial PML pathologies, signaling and transcriptional events driving malignancy are poorly understood. Evidence has suggested key roles for the Hippo pathway effectors YAP and TAZ and associated TEAD and TP63 transcription factor families in bronchial basal cell biology and LUSC. In this study we examine the functional association of YAP/TAZ, TEADs and TP63 in bronchial epithelial cells and PMLs. METHODS We performed RNA-seq in primary human bronchial epithelial cells following small interfering RNA (siRNA)-mediated depletion of YAP/TAZ, TEADs or TP63, and combined these data with ChIP-seq analysis of these factors. Directly activated or repressed genes were identified and overlapping genes were profiled across gene expression data obtained from progressive or regressive human PMLs and across lung single cell RNA-seq data sets. RESULTS Analysis of genes regulated by YAP/TAZ, TEADs, and TP63 in human bronchial epithelial cells revealed a converged transcriptional network that is strongly associated with the pathological progression of bronchial PMLs. Our observations suggest that YAP/TAZ-TEAD-TP63 associate to cooperatively promote basal epithelial cell proliferation and repress signals associated with interferon responses and immune cell communication. Directly repressed targets we identified include the MHC Class II transactivator CIITA, which is repressed in progressive PMLs and associates with adaptive immune responses in the lung. Our findings provide molecular insight into the control of gene expression events driving PML progression, including those contributing to immune evasion, offering potential new avenues for lung cancer interception. CONCLUSIONS Our study identifies important gene regulatory functions for YAP/TAZ-TEAD-TP63 in the early stages of lung cancer development, which notably includes immune-suppressive roles, and suggest that an assessment of the activity of this transcriptional complex may offer a means to identify immune evasive bronchial PMLs and serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Boting Ning
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Bioinformatics Program, Boston University, 72 East Concord Street, Boston, MA, 02215, USA
| | - Andrew M Tilston-Lunel
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Room K620, Boston, MA, 02118, USA
| | - Justice Simonetti
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Room K620, Boston, MA, 02118, USA
| | - Julia Hicks-Berthet
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Room K620, Boston, MA, 02118, USA
| | - Adeline Matschulat
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Room K620, Boston, MA, 02118, USA
| | - Roxana Pfefferkorn
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Bioinformatics Program, Boston University, 72 East Concord Street, Boston, MA, 02215, USA
| | - Avrum Spira
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Johnson and Johnson Innovation, Cambridge, MA, 02142, USA
| | | | - Sarah Mazzilli
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Bioinformatics Program, Boston University, 72 East Concord Street, Boston, MA, 02215, USA
| | - Marc E Lenburg
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- Bioinformatics Program, Boston University, 72 East Concord Street, Boston, MA, 02215, USA.
| | - Jennifer E Beane
- Department of Medicine, Computational Biomedicine Section, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- Bioinformatics Program, Boston University, 72 East Concord Street, Boston, MA, 02215, USA.
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Room K620, Boston, MA, 02118, USA.
| |
Collapse
|
26
|
Sveiven SN, Anesko K, Morgan J, Nair MG, Nordgren TM. Lipid-Sensing Receptor FFAR4 Modulates Pulmonary Epithelial Homeostasis following Immunogenic Exposures Independently of the FFAR4 Ligand Docosahexaenoic Acid (DHA). Int J Mol Sci 2023; 24:ijms24087072. [PMID: 37108233 PMCID: PMC10138935 DOI: 10.3390/ijms24087072] [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: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The role of pulmonary free fatty acid receptor 4 (FFAR4) is not fully elucidated and we aimed to clarify the impact of FFAR4 on the pulmonary immune response and return to homeostasis. We employed a known high-risk human pulmonary immunogenic exposure to extracts of dust from swine confinement facilities (DE). WT and Ffar4-null mice were repetitively exposed to DE via intranasal instillation and supplemented with docosahexaenoic acid (DHA) by oral gavage. We sought to understand if previous findings of DHA-mediated attenuation of the DE-induced inflammatory response are FFAR4-dependent. We identified that DHA mediates anti-inflammatory effects independent of FFAR4 expression, and that DE-exposed mice lacking FFAR4 had reduced immune cells in the airways, epithelial dysplasia, and impaired pulmonary barrier integrity. Analysis of transcripts using an immunology gene expression panel revealed a role for FFAR4 in lungs related to innate immune initiation of inflammation, cytoprotection, and immune cell migration. Ultimately, the presence of FFAR4 in the lung may regulate cell survival and repair following immune injury, suggestive of potential therapeutic directions for pulmonary disease.
Collapse
Affiliation(s)
- Stefanie N Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Kyle Anesko
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Joshua Morgan
- Department of Bioengineering, Bourns College of Engineering, University of California-Riverside, Riverside, CA 92521, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Tara M Nordgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
27
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
28
|
Li X, Zhuo S, Cho YS, Liu Y, Yang Y, Zhu J, Jiang J. YAP antagonizes TEAD-mediated AR signaling and prostate cancer growth. EMBO J 2023; 42:e112184. [PMID: 36588499 PMCID: PMC9929633 DOI: 10.15252/embj.2022112184] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/03/2023] Open
Abstract
Hippo signaling restricts tumor growth by inhibiting the oncogenic potential of YAP/TAZ-TEAD transcriptional complex. Here, we uncover a context-dependent tumor suppressor function of YAP in androgen receptor (AR) positive prostate cancer (PCa) and show that YAP impedes AR+ PCa growth by antagonizing TEAD-mediated AR signaling. TEAD forms a complex with AR to enhance its promoter/enhancer occupancy and transcriptional activity. YAP and AR compete for TEAD binding and consequently, elevated YAP in the nucleus disrupts AR-TEAD interaction and prevents TEAD from promoting AR signaling. Pharmacological inhibition of MST1/2 or LATS1/2, or transgenic activation of YAP suppressed the growth of PCa expressing therapy resistant AR splicing variants. Our study uncovers an unanticipated crosstalk between Hippo and AR signaling pathways, reveals an antagonistic relationship between YAP and TEAD in AR+ PCa, and suggests that targeting the Hippo signaling pathway may provide a therapeutical opportunity to treat PCa driven by therapy resistant AR variants.
Collapse
Affiliation(s)
- Xu Li
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Shu Zhuo
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Center for Cancer Targeted Therapies, Signet Therapeutics Inc.Research Institute of Tsinghua University in ShenzhenShenzhenChina
| | - Yong Suk Cho
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Yuchen Liu
- Department of Developmental BiologyHarvard School of Dental MedicineBostonMAUSA
- Harvard Stem Cell InstituteBostonMAUSA
- Dana‐Farber/Harvard Cancer CenterBostonMAUSA
| | - Yingzi Yang
- Department of Developmental BiologyHarvard School of Dental MedicineBostonMAUSA
- Harvard Stem Cell InstituteBostonMAUSA
- Dana‐Farber/Harvard Cancer CenterBostonMAUSA
| | - Jian Zhu
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of General Surgery, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Jin Jiang
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of PharmacologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
29
|
Tilston-Lunel AM, Varelas X. Polarity in respiratory development, homeostasis and disease. Curr Top Dev Biol 2023; 154:285-315. [PMID: 37100521 DOI: 10.1016/bs.ctdb.2023.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
The respiratory system is composed of a multitude of cells that organize to form complex branched airways that end in alveoli, which respectively function to guide air flow and mediate gas exchange with the bloodstream. The organization of the respiratory sytem relies on distinct forms of cell polarity, which guide lung morphogenesis and patterning in development and provide homeostatic barrier protection from microbes and toxins. The stability of lung alveoli, the luminal secretion of surfactants and mucus in the airways, and the coordinated motion of multiciliated cells that generate proximal fluid flow, are all critical functions regulated by cell polarity, with defects in polarity contributing to respiratory disease etiology. Here, we summarize the current knowledge of cell polarity in lung development and homeostasis, highlighting key roles for polarity in alveolar and airway epithelial function and outlining relationships with microbial infections and diseases, such as cancer.
Collapse
|
30
|
Effective Component Compatibility of Bufei Yishen Formula III Which Regulates the Mucus Hypersecretion of COPD Rats via the miR-146a-5p/EGFR/MEK/ERK Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9423435. [PMID: 36619199 PMCID: PMC9812609 DOI: 10.1155/2022/9423435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022]
Abstract
Background The effective-component compatibility of Bufei Yishen formula III (ECC-BYF III) with 5 ingredients (ginsenoside Rh1, astragaloside, icariin, nobiletin, and paeonol) has been shown to protect against chronic obstructive pulmonary disease (COPD). The present study aimed to observe the effects of ECC-BYF III in a COPD rat model and dissect its potential mechanisms in regulating mucus hypersecretion via the miR-146a-5p/epidermal growth factor receptor (EGFR)/MEK/ERK pathway. Methods COPD model rats were treated with normal saline, ECC-BYF III, or N-acetylcysteine (NAC). Pulmonary function, lung tissue histology with H & E and AB-PAS staining, expression levels of interleukin (IL)-4, IL-6, IL-1β, MUC5AC, MUC5B, and FOXA2 in lung tissues and the mRNA and proteins involved in the miR-146a-5p/EGFR/MEK/ERK pathway were evaluated. Results The COPD rats showed a significant decrease in the pulmonary function and serious pathological damage to the lung tissue. ECC-BYF III and NAC significantly improved the ventilation function and small airway pathological damage in the COPD rats. The goblet cells and the expression levels of IL-1β, IL-6, MUC5AC, and MUC5B were increased in the COPD rats and were significantly decreased after ECC-BYF III or NAC intervention. The expression levels of IL-4 and FOXA2 in the COPD rats were markedly decreased and were improved in the ECC-BYF III and NAC groups. ECC-BYF III appeared to have a potent effect in restoring the reduced expression of miR-146a-5p. The increased phosphorylation levels of EGFR, MEK, and ERK1/2 and the protein expression levels of SPDEF in the lungs of COPD rats could be significantly reduced by ECC-BYF III. Conclusions ECC-BYF III has a significant effect in improving the airway mucus hypersecretion in COPD model rats, as well as a protective effect against limited pulmonary function and injured lung histopathology. The protective effect of ECC-BYF III in reducing airway mucus hypersecretion in COPD may involve the miR-146a-5p/EGFR/MEK/ERK pathway.
Collapse
|
31
|
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
|
32
|
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
|
33
|
Lee SN, Yoon SA, Song JM, Kim HC, Cho HJ, Choi AMK, Yoon JH. Cell-Type Specific Expression of Hyaluronan Synthases HAS2 and HAS3 Promotes Goblet Cell Hyperplasia in Allergic Airway Inflammation. Am J Respir Cell Mol Biol 2022; 67:360-374. [PMID: 35679095 DOI: 10.1165/rcmb.2021-0527oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Allergic rhinitis (AR) is a multifactorial airway disease characterized by basal and goblet cell hyperplasia. Hyaluronic acid (HA) is a major component of extracellular matrix and a critical contributor to tissue repair and remodeling after injury. We previously demonstrated that the intermediate progenitor cell (IPC) surface marker CD44v3 is upregulated in the basal and suprabasal layers of well-differentiated primary human nasal epithelial (HNE) cells after stimulation with the Th2 cytokine interleukin (IL)-4, and an antibody blocking the CD44v3-HA interaction suppressed IL-4-induced goblet cell hyperplasia. We now show that the expression of HA and two HA synthases, HAS2 and HAS3, was upregulated in both the nasal surface epithelium of subjects with AR and IL-4-stimulated HNE cells. Inhibition of HA synthesis by 4-methylumbelliferone (4-MU) suppressed IL-4-induced goblet cell hyperplasia. Moreover, HAS2 and HAS3 were expressed in IPCs depending on the differentiation events, as follows: the rapid, transient upregulation of HAS2 induced basal IPC proliferation and basal-to-suprabasal transition, whereas the delayed upregulation of HAS3 promoted the transition of suprabasal IPCs to a goblet cell fate. 4-MU treatment in house dust mite-induced murine AR model attenuated goblet cell metaplasia. Lastly, HA levels in nasal epithelial lining fluids from AR patients positively correlated with the levels of mediators causing allergic inflammation. These data suggest that HA produced following the sequential upregulation of HAS2 and HAS3 contributes to goblet cell hyperplasia in allergic airway inflammation and modulates disease progression.
Collapse
Affiliation(s)
- Sang-Nam Lee
- Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Seol Ah Yoon
- Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Ji Min Song
- Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Hyung Chul Kim
- Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Hyung-Ju Cho
- Yonsei university college of medicine, otorhinolaryngology, Seoul, Korea (the Republic of)
| | - Augustine M K Choi
- Cornell University Joan and Sanford I Weill Medical College, 12295, New York, New York, United States
| | - Joo-Heon Yoon
- Yonsei University College of Medicine, Department of Otorhinolaryngology, Seoul, Korea (the Republic of);
| |
Collapse
|
34
|
Man K, Liu J, Phan KM, Wang K, Lee JY, Sun X, Story M, Saha D, Liao J, Sadat H, Yang Y. Dimensionality-Dependent Mechanical Stretch Regulation of Cell Behavior. ACS APPLIED MATERIALS & INTERFACES 2022; 14:17081-17092. [PMID: 35380801 DOI: 10.1021/acsami.2c01266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A variety of cells are subject to mechanical stretch in vivo, which plays a critical role in the function and homeostasis of cells, tissues, and organs. Deviations from the physiologically relevant mechanical stretch are often associated with organ dysfunction and various diseases. Although mechanical stretch is provided in some in vitro cell culture models, the effects of stretch dimensionality on cells are often overlooked and it remains unclear whether and how stretch dimensionality affects cell behavior. Here we develop cell culture platforms that provide 1-D uniaxial, 2-D circumferential, or 3-D radial mechanical stretches, which recapitulate the three major types of mechanical stretches that cells experience in vivo. We investigate the behavior of human microvascular endothelial cells and human alveolar epithelial cells cultured on these platforms, showing that the mechanical stretch influences cell morphology and cell-cell and cell-substrate interactions in a stretch dimensionality-dependent manner. Furthermore, the endothelial and epithelial cells are sensitive to the physiologically relevant 2-D and 3-D stretches, respectively, which could promote the formation of endothelium and epithelium. This study underscores the importance of recreating the physiologically relevant mechanical stretch in the development of in vitro tissue/organ models.
Collapse
Affiliation(s)
- Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Khang Minh Phan
- Department of Mechanical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jung Yeon Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Michael Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76010, United States
| | - Hamid Sadat
- Department of Mechanical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
35
|
Li H, Wu BK, Kanchwala M, Cai J, Wang L, Xing C, Zheng Y, Pan D. YAP/TAZ drives cell proliferation and tumour growth via a polyamine-eIF5A hypusination-LSD1 axis. Nat Cell Biol 2022; 24:373-383. [PMID: 35177822 PMCID: PMC8930503 DOI: 10.1038/s41556-022-00848-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/12/2022] [Indexed: 12/22/2022]
Abstract
Metabolic reprogramming is central to oncogene-induced tumorigenesis by providing the necessary building blocks and energy sources, but how oncogenic signalling controls metabolites that play regulatory roles in driving cell proliferation and tumour growth is less understood. Here we show that oncogene YAP/TAZ promotes polyamine biosynthesis by activating the transcription of the rate-limiting enzyme ornithine decarboxylase 1. The increased polyamine levels, in turn, promote the hypusination of eukaryotic translation factor 5A (eIF5A) to support efficient translation of histone demethylase LSD1, a transcriptional repressor that mediates a bulk of YAP/TAZ-downregulated genes including tumour suppressors in YAP/TAZ-activated cells. Accentuating the importance of the YAP/TAZ-polyamine-eIF5A hypusination-LSD1 axis, inhibiting polyamine biosynthesis or LSD1 suppressed YAP/TAZ-induced cell proliferation and tumour growth. Given the frequent upregulation of YAP/TAZ activity and polyamine levels in diverse cancers, our identification of YAP/TAZ as an upstream regulator and LSD1 as a downstream effector of the oncometabolite polyamine offers a molecular framework in which oncogene-induced metabolic and epigenetic reprogramming coordinately drives tumorigenesis, and suggests potential therapeutic strategies in YAP/TAZ- or polyamine-dependent human malignancies.
Collapse
Affiliation(s)
- Hongde Li
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo-Kuan Wu
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mohammed Kanchwala
- Eugene McDermott Center for Human Growth and Development/Center for Human Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jing Cai
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Li Wang
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development/Center for Human Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
36
|
Hatterschide J, Castagnino P, Kim HW, Sperry SM, Montone KT, Basu D, White EA. YAP1 activation by human papillomavirus E7 promotes basal cell identity in squamous epithelia. eLife 2022; 11:75466. [PMID: 35170430 PMCID: PMC8959598 DOI: 10.7554/elife.75466] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/15/2022] [Indexed: 11/27/2022] Open
Abstract
Persistent human papillomavirus (HPV) infection of stratified squamous epithelial cells causes nearly 5% of cancer cases worldwide. HPV-positive oropharyngeal cancers harbor few mutations in the Hippo signaling pathway compared to HPV-negative cancers at the same anatomical site, prompting the hypothesis that an HPV-encoded protein inactivates the Hippo pathway and activates the Hippo effector yes-associated protein (YAP1). The HPV E7 oncoprotein is required for HPV infection and for HPV-mediated oncogenic transformation. We investigated the effects of HPV oncoproteins on YAP1 and found that E7 activates YAP1, promoting YAP1 nuclear localization in basal epithelial cells. YAP1 activation by HPV E7 required that E7 binds and degrades the tumor suppressor protein tyrosine phosphatase non-receptor type 14 (PTPN14). E7 required YAP1 transcriptional activity to extend the lifespan of primary keratinocytes, indicating that YAP1 activation contributes to E7 carcinogenic activity. Maintaining infection in basal cells is critical for HPV persistence, and here we demonstrate that YAP1 activation causes HPV E7 expressing cells to be retained in the basal compartment of stratified epithelia. We propose that YAP1 activation resulting from PTPN14 inactivation is an essential, targetable activity of the HPV E7 oncoprotein relevant to HPV infection and carcinogenesis. The ‘epithelial’ cells that cover our bodies are in a constant state of turnover. Every few weeks, the outermost layers die and are replaced by new cells from the layers below. For scientists, this raises a difficult question. Cells infected by human papillomaviruses, often known as HPV, can become cancerous over years or even decades. How do infected cells survive while the healthy cells around them mature and get replaced? One clue could lie in PTPN14, a human protein which many papillomaviruses eliminate using their viral E7 protein; this mechanism could be essential for the virus to replicate and cause cancer. To find out the impact of losing PTPN14, Hatterschide et al. used human epithelial cells to make three-dimensional models of infected tissues. These experiments showed that, when papillomaviruses destroy PTPN14, a human protein called YAP1 turns on in the lowest, most long-lived layer of the tissue. Cells in which YAP1 is activated survive while those that carry the inactive version mature and die. This suggests that papillomaviruses turn on YAP1 to remain in tissues for long periods. Papillomaviruses cause about five percent of all human cancers. Finding ways to stop them from activating YAP1 has the potential to prevent disease. Overall, the research by Hatterschide et al. also sheds light on other epithelial cancers which are not caused by viruses.
Collapse
Affiliation(s)
- Joshua Hatterschide
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| | - Paola Castagnino
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| | - Hee Won Kim
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| | - Steven M Sperry
- Department of Otolaryngology-Head and Neck Surgery, Aurora St. Luke's Medical Center, Milwaukee, United States
| | - Kathleen T Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Devraj Basu
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| | - Elizabeth A White
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
37
|
Xiao H, Zhang QN, Sun QX, Li LD, Xu SY, Li CQ. Transcriptomic Analysis Reveals a Link Between Hippo Signaling Pathway and Macrophages in Lungs of Mice with OVA-Induced Allergic Asthma. J Inflamm Res 2022; 15:423-437. [PMID: 35082511 PMCID: PMC8784274 DOI: 10.2147/jir.s346505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Huan Xiao
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Qian-nan Zhang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Qi-xiang Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Lao-dong Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Si-yue Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Chao-qian Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
- Correspondence: Chao-qian Li, Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Province, People’s Republic of China, Tel +86 13807887867, Fax +86 771-5350031, Email
| |
Collapse
|