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
|
Feng Y, Mao T, Yi J, Zhang N, Gu Y, Shen H, Chen J. Runt-related transcription factors: from pathogenesis to therapeutic targets in multiple-organ fibrosis. Front Cell Dev Biol 2025; 13:1528645. [PMID: 40356603 PMCID: PMC12066561 DOI: 10.3389/fcell.2025.1528645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Fibrosis is a partially manageable process that leads to scarring and tissue hardening by prompting myofibroblasts to deposit significant amounts of extracellular matrix (ECM) following injury. It results in detrimental consequences and pathological characteristics, which hinder the functioning of associated organs and increase mortality rates. Runt-related transcription factors (RUNX) are part of a highly conserved family of heterodimer transcription factors, comprising RUNX1, RUNX2, and RUNX3. They are involved in several biological processes and undergo various forms of post-translational modification. RUNX regulates multiple targets and pathways to impact fibrosis, indicating promise for clinical application. Therefore, its significance in the fibrosis process should not be disregarded. The review begins with an objective description of the structure, transcriptional mechanism, and biological function of RUNX1, RUNX2, and RUNX3. A subsequent analysis is made of their physiological relationship with heart, lung, kidney, and liver, followed by a focus on the signaling mechanism of RUNX in regulating fibrosis of these organs. Furthermore, potential agents or drugs targeting RUNX for treating organ fibrosis are summarized, along with an evaluation of the therapeutic prospects and potential value of RUNX in fibrosis. Further research into RUNX could contribute to the development of novel therapeutic approaches for fibrosis.
Collapse
Affiliation(s)
- Yuan Feng
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Tianshi Mao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jifei Yi
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Na Zhang
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yinying Gu
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Huifen Shen
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jie Chen
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Pezet MG, Torres JA, Thimraj TA, Matkovic I, Schrode N, Murray JW, Saqi A, Beaumont KG, Snoeck HW. Human respiratory airway progenitors derived from pluripotent cells generate alveolar epithelial cells and model pulmonary fibrosis. Nat Biotechnol 2025:10.1038/s41587-025-02569-0. [PMID: 39994483 DOI: 10.1038/s41587-025-02569-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/17/2025] [Indexed: 02/26/2025]
Abstract
Human lungs contain unique cell populations in distal respiratory airways or terminal and respiratory bronchioles (RA/TRBs) that accumulate in persons with lung injury and idiopathic pulmonary fibrosis (IPF), a lethal lung disease. As these populations are absent in rodents, deeper understanding requires a human in vitro model. Here we convert human pluripotent stem cells (hPS cells) into expandable spheres, called induced respiratory airway progenitors (iRAPs), consisting of ~98% RA/TRB-associated cell types. One hPS cell can give rise to 1010 iRAP cells. We differentiate iRAPs through a stage consistent with transitional type 2 alveolar epithelial (AT2) cells into a population corresponding to mature AT1 cells with 95% purity. iRAPs with deletion of Heřmanský-Pudlák Syndrome 1 (HPS1), which causes pulmonary fibrosis in humans, replicate the aberrant differentiation and recruitment of profibrotic fibroblasts observed in IPF, indicating that intrinsic dysfunction of RA/TRB-associated alveolar progenitors contributes to HPS1-related IPF. iRAPs may provide a system suitable for IPF drug discovery and validation.
Collapse
Affiliation(s)
- Mikael G Pezet
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Juan A Torres
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Tania A Thimraj
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ivana Matkovic
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Nadine Schrode
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Center for Advanced Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John W Murray
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Kristin G Beaumont
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Center for Advanced Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hans-Willem Snoeck
- Columbia Center for Stem Cell Therapies/Columbia Center for Human Development, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Division of Pulmonary Medicine, Allergy and Critical Care, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
4
|
Spalding VA, Fellenstein BA, Ahodantin J, Jeyarajan AJ, Wang Y, Khan SK, Xu M, Lin W, Alatrakchi N, Su L, Chung RT, Salloum S. YAP mediates HIV-related liver fibrosis. JHEP Rep 2024; 6:101163. [PMID: 39524207 PMCID: PMC11544392 DOI: 10.1016/j.jhepr.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background & Aims HIV accelerates liver fibrosis attributable to multiple etiologies, including HCV, HBV, and steatotic liver disease. Evidence also suggests that HIV infection itself is associated with liver fibrogenesis. Recent studies have implicated Yes-associated protein 1 (YAP1) and the upstream lysophosphatidic acid (LPA)/PI3K/AKT pathway as critical regulators of hepatic fibrogenesis, and suggest a connection to HIV-related liver fibrosis. However, the relationship between YAP/PI3K/AKT pathway activation and HIV-related liver fibrosis remains uncertain. Methods qPCR, western blot, immunofluorescence, and ELISA (replicates n ≥3) were performed in an unbiased humanized mouse model (NRG-hu HSC mice, n = 6), the precision cut liver slice ex vivo model, and both traditional in vitro models as well as a 3D spheroid system. Results YAP target gene mRNA and protein levels (ANKRD, CTGF, CYR61) were upregulated across all models exposed to HIV. Humanized mice infected with HIV had significant increases in the percentage of YAP-positive nuclei (2.2-fold) and the percentage area of Sirius Red collagen staining (3.3-fold) compared to control mice. Serum concentrations of LPA were increased 5.8-fold in people living with HIV compared to healthy controls. Modulation of LPAR1, PI3K, and AKT by either inhibitors or small-interfering RNAs abrogated the fibrotic effects of HIV exposure and downregulated YAP target genes within cultured liver cells. Conclusions The LPAR/PI3K/AKT axis is vital for the activation of YAP and hepatic fibrogenesis due to HIV infection. This novel mechanistic insight suggests new pharmacologic targets for treatment of liver fibrosis in people living with HIV. Impact and implications There are currently no FDA-approved treatments for cirrhosis, while liver disease is the second leading cause of mortality among people living with HIV after AIDS. Increased lysophosphatidic acid concentrations and AKT activation after HIV infection found in recent work suggest that the Hippo pathway may be a key regulator of HIV-related fibrogenesis. By linking lysophosphatidic acid signaling, YAP activation, and HIV-related fibrogenesis, this mechanism presents a target for future research into therapeutic interventions for not only HIV but also other liver diseases, e.g. metabolic dysfunction- or alcohol-associated liver disease.
Collapse
Affiliation(s)
- Volney A. Spalding
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Brian A. Fellenstein
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - James Ahodantin
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andre J. Jeyarajan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Yongtao Wang
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sanjoy K. Khan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Min Xu
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Nadia Alatrakchi
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Lishan Su
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Shadi Salloum
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
5
|
Wang J, Li K, Hao D, Li X, Zhu Y, Yu H, Chen H. Pulmonary fibrosis: pathogenesis and therapeutic strategies. MedComm (Beijing) 2024; 5:e744. [PMID: 39314887 PMCID: PMC11417429 DOI: 10.1002/mco2.744] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive lung disease characterized by extensive alterations of cellular fate and function and excessive accumulation of extracellular matrix, leading to lung tissue scarring and impaired respiratory function. Although our understanding of its pathogenesis has increased, effective treatments remain scarce, and fibrotic progression is a major cause of mortality. Recent research has identified various etiological factors, including genetic predispositions, environmental exposures, and lifestyle factors, which contribute to the onset and progression of PF. Nonetheless, the precise mechanisms by which these factors interact to drive fibrosis are not yet fully elucidated. This review thoroughly examines the diverse etiological factors, cellular and molecular mechanisms, and key signaling pathways involved in PF, such as TGF-β, WNT/β-catenin, and PI3K/Akt/mTOR. It also discusses current therapeutic strategies, including antifibrotic agents like pirfenidone and nintedanib, and explores emerging treatments targeting fibrosis and cellular senescence. Emphasizing the need for omni-target approaches to overcome the limitations of current therapies, this review integrates recent findings to enhance our understanding of PF and contribute to the development of more effective prevention and management strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Jianhai Wang
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Kuan Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - De Hao
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
| | - Xue Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Yu Zhu
- Department of Clinical LaboratoryNankai University Affiliated Third Central HospitalTianjinChina
- Department of Clinical LaboratoryThe Third Central Hospital of TianjinTianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesArtificial Cell Engineering Technology Research Center of TianjinTianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Hongzhi Yu
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Huaiyong Chen
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| |
Collapse
|
6
|
Raslan AA, Pham TX, Lee J, Kontodimas K, Tilston-Lunel A, Schmottlach J, Hong J, Dinc T, Bujor AM, Caporarello N, Thiriot A, von Andrian UH, Huang SK, Nicosia RF, Trojanowska M, Varelas X, Ligresti G. Lung injury-induced activated endothelial cell states persist in aging-associated progressive fibrosis. Nat Commun 2024; 15:5449. [PMID: 38937456 PMCID: PMC11211333 DOI: 10.1038/s41467-024-49545-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] [Received: 02/08/2023] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
Progressive lung fibrosis is associated with poorly understood aging-related endothelial cell dysfunction. To gain insight into endothelial cell alterations in lung fibrosis we performed single cell RNA-sequencing of bleomycin-injured lungs from young and aged mice. Analysis reveals activated cell states enriched for hypoxia, glycolysis and YAP/TAZ activity in ACKR1+ venous and TrkB+ capillary endothelial cells. Endothelial cell activation is prevalent in lungs of aged mice and can also be detected in human fibrotic lungs. Longitudinal single cell RNA-sequencing combined with lineage tracing demonstrate that endothelial activation resolves in young mouse lungs but persists in aged ones, indicating a failure of the aged vasculature to return to quiescence. Genes associated with activated lung endothelial cells states in vivo can be induced in vitro by activating YAP/TAZ. YAP/TAZ also cooperate with BDNF, a TrkB ligand that is reduced in fibrotic lungs, to promote capillary morphogenesis. These findings offer insights into aging-related lung endothelial cell dysfunction that may contribute to defective lung injury repair and persistent fibrosis.
Collapse
Affiliation(s)
- Ahmed A Raslan
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Tho X Pham
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jisu Lee
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jillian Schmottlach
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Jeongmin Hong
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Taha Dinc
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Andreea M Bujor
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | | | - Aude Thiriot
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Trojanowska
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| | - Giovanni Ligresti
- Arthritis and Autoimmune Diseases Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
7
|
Leong E, Al-Bitar H, Marshall JS, Bezuhly M. Ketotifen directly modifies the fibrotic response of human skin fibroblasts. Sci Rep 2024; 14:7076. [PMID: 38528089 DOI: 10.1038/s41598-024-57776-7] [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: 10/26/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
Fibrosis is a destructive, end-stage disease process. In the skin, it is associated with systemic sclerosis and scarring with considerable health burden. Ketotifen is a clinical antihistamine and mast cell stabilizer. Studies have demonstrated mast cell-dependent anti-fibrotic effects of ketotifen but direct effects on fibroblasts have not been determined. Human dermal fibroblasts were treated with pro-fibrotic transforming growth factor-β1 (TGFβ) followed by ketotifen or control treatments to determine direct effects on fibrotic fibroblasts. Ketotifen impaired TGFβ-induced α-smooth muscle actin gene and protein responses and decreased cytoskeletal- and contractility-associated gene responses associated with fibrosis. Ketotifen reduced Yes-associated protein phosphorylation, transcriptional coactivator with PDZ binding motif transcript and protein levels, and phosphorylation of protein kinase B. In a fibroblast-populated collagen gel contraction assay, ketotifen reduced the contractile activity of TGFβ-activated fibroblasts. In a murine model of bleomycin-induced skin fibrosis, collagen density and dermal thickness were significantly decreased in ketotifen-treated mice supporting in vitro findings. These results support a novel, direct anti-fibrotic activity of ketotifen, reducing pro-fibrotic phenotypic changes in fibroblasts and reducing collagen fibres in fibrotic mouse skin. Together, these findings suggest novel therapeutic potential and a novel mechanism of action for ketotifen in the context of fibrosis.
Collapse
Affiliation(s)
- Edwin Leong
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada
| | - Haya Al-Bitar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada.
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
| | - Michael Bezuhly
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
- Division of Plastic Surgery, Izaak Walton Killam Health Centre, 5850/5980 University Avenue, PO Box 9700, Halifax, NS, B3K 6R8, Canada.
- Department of Surgery, Dalhousie University, Halifax, Canada.
| |
Collapse
|
8
|
Saneh H, Wanczyk H, Walker J, Finck C. Effectiveness of extracellular vesicles derived from hiPSCs in repairing hyperoxia-induced injury in a fetal murine lung explant model. Stem Cell Res Ther 2024; 15:80. [PMID: 38486338 PMCID: PMC10941466 DOI: 10.1186/s13287-024-03687-3] [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: 10/02/2023] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among preterm infants. Human induced pluripotent stem cells (hiPSCs) have shown promise in repairing injury in animal BPD models. Evidence suggests they exert their effects via paracrine mechanisms. We aim herein to assess the effectiveness of extracellular vesicles (EVs) derived from hiPSCs and their alveolar progenies (diPSCs) in attenuating hyperoxic injury in a preterm lung explant model. METHODS Murine lung lobes were harvested on embryonic day 17.5 and maintained in air-liquid interface. Following exposure to 95% O2 for 24 h, media was supplemented with 5 × 106 particles/mL of EVs isolated from hiPSCs or diPSCs by size-exclusion chromatography. On day 3, explants were assessed using Hematoxylin-Eosin staining with mean linear intercept (MLI) measurements, immunohistochemistry, VEGFa and antioxidant gene expression. Statistical analysis was conducted using one-way ANOVA and Multiple Comparison Test. EV proteomic profiling was performed, and annotations focused on alveolarization and angiogenesis signaling pathways, as well as anti-inflammatory, anti-oxidant, and regenerative pathways. RESULTS Exposure of fetal lung explants to hyperoxia induced airspace enlargement, increased MLI, upregulation of anti-oxidants Prdx5 and Nfe2l2 with decreased VEGFa expression. Treatment with hiPSC-EVs improved parenchymal histologic changes. No overt changes in vasculature structure were observed on immunohistochemistry in our in vitro model. However, VEGFa and anti-oxidant genes were upregulated with diPSC-EVs, suggesting a pro-angiogenic and cytoprotective potential. EV proteomic analysis provided new insights in regard to potential pathways influencing lung regeneration. CONCLUSION This proof-of-concept in vitro study reveals a potential role for hiPSC- and diPSC-EVs in attenuating lung changes associated with prematurity and oxygen exposure. Our findings pave the way for a novel cell free approach to prevent and/or treat BPD, and ultimately reduce the global burden of the disease.
Collapse
Affiliation(s)
- Hala Saneh
- Department of Neonatal Medicine, Connecticut Children's Medical Center, Hartford, CT, USA.
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Joanne Walker
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
- Department of Pediatric Surgery, Connecticut Children's Medical Center, Hartford, CT, USA
| |
Collapse
|
9
|
Chen XY, Kao C, Peng SW, Chang JH, Lee YL, Laiman V, Chung KF, Bhavsar PK, Heriyanto DS, Chuang KJ, Chuang HC. Role of DCLK1/Hippo pathway in type II alveolar epithelial cells differentiation in acute respiratory distress syndrome. Mol Med 2023; 29:159. [PMID: 37996782 PMCID: PMC10668445 DOI: 10.1186/s10020-023-00760-0] [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: 05/31/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Delay in type II alveolar epithelial cell (AECII) regeneration has been linked to higher mortality in patients with acute respiratory distress syndrome (ARDS). However, the interaction between Doublecortin-like kinase 1 (DCLK1) and the Hippo signaling pathway in ARDS-associated AECII differentiation remains unclear. Therefore, the objective of this study was to understand the role of the DCLK1/Hippo pathway in mediating AECII differentiation in ARDS. MATERIALS AND METHODS AECII MLE-12 cells were exposed to 0, 0.1, or 1 μg/mL of lipopolysaccharide (LPS) for 6 and 12 h. In the mouse model, C57BL/6JNarl mice were intratracheally (i.t.) injected with 0 (control) or 5 mg/kg LPS and were euthanized for lung collection on days 3 and 7. RESULTS We found that LPS induced AECII markers of differentiation by reducing surfactant protein C (SPC) and p53 while increasing T1α (podoplanin) and E-cadherin at 12 h. Concurrently, nuclear YAP dynamic regulation and increased TAZ levels were observed in LPS-exposed AECII within 12 h. Inhibition of YAP consistently decreased cell levels of SPC, claudin 4 (CLDN-4), galectin 3 (LGALS-3), and p53 while increasing transepithelial electrical resistance (TEER) at 6 h. Furthermore, DCLK1 expression was reduced in isolated human AECII of ARDS, consistent with the results in LPS-exposed AECII at 6 h and mouse SPC-positive (SPC+) cells after 3-day LPS exposure. We observed that downregulated DCLK1 increased p-YAP/YAP, while DCLK1 overexpression slightly reduced p-YAP/YAP, indicating an association between DCLK1 and Hippo-YAP pathway. CONCLUSIONS We conclude that DCLK1-mediated Hippo signaling components of YAP/TAZ regulated markers of AECII-to-AECI differentiation in an LPS-induced ARDS model.
Collapse
Affiliation(s)
- Xiao-Yue Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ching Kao
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Syue-Wei Peng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
| | - Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- National Heart and Lung Institute, Imperial College London, London, UK.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Inhalation Toxicology Research Lab (ITRL), School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan.
| |
Collapse
|
10
|
Blumhagen RZ, Kurche JS, Cool CD, Walts AD, Heinz D, Fingerlin TE, Yang IV, Schwartz DA. Spatially distinct molecular patterns of gene expression in idiopathic pulmonary fibrosis. Respir Res 2023; 24:287. [PMID: 37978501 PMCID: PMC10655274 DOI: 10.1186/s12931-023-02572-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/21/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a heterogeneous disease that is pathologically characterized by areas of normal-appearing lung parenchyma, active fibrosis (transition zones including fibroblastic foci) and dense fibrosis. Defining transcriptional differences between these pathologically heterogeneous regions of the IPF lung is critical to understanding the distribution and extent of fibrotic lung disease and identifying potential therapeutic targets. Application of a spatial transcriptomics platform would provide more detailed spatial resolution of transcriptional signals compared to previous single cell or bulk RNA-Seq studies. METHODS We performed spatial transcriptomics using GeoMx Nanostring Digital Spatial Profiling on formalin-fixed paraffin-embedded (FFPE) tissue from 32 IPF and 12 control subjects and identified 231 regions of interest (ROIs). We compared normal-appearing lung parenchyma and airways between IPF and controls with histologically normal lung tissue, as well as histologically distinct regions within IPF (normal-appearing lung parenchyma, transition zones containing fibroblastic foci, areas of dense fibrosis, and honeycomb epithelium metaplasia). RESULTS We identified 254 differentially expressed genes (DEGs) between IPF and controls in histologically normal-appearing regions of lung parenchyma; pathway analysis identified disease processes such as EIF2 signaling (important for cap-dependent mRNA translation), epithelial adherens junction signaling, HIF1α signaling, and integrin signaling. Within IPF, we identified 173 DEGs between transition and normal-appearing lung parenchyma and 198 DEGs between dense fibrosis and normal lung parenchyma; pathways dysregulated in both transition and dense fibrotic areas include EIF2 signaling pathway activation (upstream of endoplasmic reticulum (ER) stress proteins ATF4 and CHOP) and wound healing signaling pathway deactivation. Through cell deconvolution of transcriptome data and immunofluorescence staining, we confirmed loss of alveolar parenchymal signals (AGER, SFTPB, SFTPC), gain of secretory cell markers (SCGB3A2, MUC5B) as well as dysregulation of the upstream regulator ATF4, in histologically normal-appearing tissue in IPF. CONCLUSIONS Our findings demonstrate that histologically normal-appearing regions from the IPF lung are transcriptionally distinct when compared to similar lung tissue from controls with histologically normal lung tissue, and that transition zones and areas of dense fibrosis within the IPF lung demonstrate activation of ER stress and deactivation of wound healing pathways.
Collapse
Affiliation(s)
- Rachel Z Blumhagen
- Center for Genes, Environment and Health, National Jewish Health, 1400 Jackson St, Office M222D, Denver, CO, 80206, USA.
| | - Jonathan S Kurche
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, CO, 80045, USA
- Medical Service, Rocky Mountain Regional Veterans Administration Medical Center, 1700 N Wheeling St, Aurora, CO, 80045, USA
| | - Carlyne D Cool
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, CO, 80045, USA
- Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO, 80206, USA
| | - Avram D Walts
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, CO, 80045, USA
| | - David Heinz
- Pathology Laboratory, National Jewish Health, 1400 Jackson St., Denver, CO, 80206, USA
| | - Tasha E Fingerlin
- Center for Genes, Environment and Health, National Jewish Health, 1400 Jackson St, Office M222D, Denver, CO, 80206, USA
| | - Ivana V Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, CO, 80045, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Aurora, CO, 80045, USA
- Medical Service, Rocky Mountain Regional Veterans Administration Medical Center, 1700 N Wheeling St, Aurora, CO, 80045, USA
| |
Collapse
|
11
|
李 明, 孙 美, 贾 渊, 任 徽, 刘 含. [Biomechanical properties of epithelial mesenchymal transition in idiopathic pulmonary fibrosis]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2023; 40:632-637. [PMID: 37666752 PMCID: PMC10477379 DOI: 10.7507/1001-5515.202206016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 02/02/2023] [Indexed: 09/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive scar-forming disease with a high mortality rate that has received widespread attention. Epithelial mesenchymal transition (EMT) is an important part of the pulmonary fibrosis process, and changes in the biomechanical properties of lung tissue have an important impact on it. In this paper, we summarize the changes in the biomechanical microenvironment of lung tissue in IPF-EMT in recent years, and provide a systematic review on the effects of alterations in the mechanical microenvironment in pulmonary fibrosis on the process of EMT, the effects of mechanical factors on the behavior of alveolar epithelial cells in EMT and the biomechanical signaling in EMT, in order to provide new references for the research on the prevention and treatment of IPF.
Collapse
Affiliation(s)
- 明艳 李
- 河南中医药大学 中医药科学院 呼吸疾病中医药防治省部共建协同创新中心 河南省中医药防治呼吸病重点实验室(郑州 450016)Henan University of Chinese Medicine, Academy of Chinese Medicine Sciences, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan & Ministry of Education of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Zhengzhou 450016, P.R. China
| | - 美好 孙
- 河南中医药大学 中医药科学院 呼吸疾病中医药防治省部共建协同创新中心 河南省中医药防治呼吸病重点实验室(郑州 450016)Henan University of Chinese Medicine, Academy of Chinese Medicine Sciences, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan & Ministry of Education of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Zhengzhou 450016, P.R. China
| | - 渊博 贾
- 河南中医药大学 中医药科学院 呼吸疾病中医药防治省部共建协同创新中心 河南省中医药防治呼吸病重点实验室(郑州 450016)Henan University of Chinese Medicine, Academy of Chinese Medicine Sciences, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan & Ministry of Education of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Zhengzhou 450016, P.R. China
| | - 徽 任
- 河南中医药大学 中医药科学院 呼吸疾病中医药防治省部共建协同创新中心 河南省中医药防治呼吸病重点实验室(郑州 450016)Henan University of Chinese Medicine, Academy of Chinese Medicine Sciences, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan & Ministry of Education of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Zhengzhou 450016, P.R. China
- 西安交通大学 仿生工程与生物力学中心(西安 710049)Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - 含 刘
- 河南中医药大学 中医药科学院 呼吸疾病中医药防治省部共建协同创新中心 河南省中医药防治呼吸病重点实验室(郑州 450016)Henan University of Chinese Medicine, Academy of Chinese Medicine Sciences, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan & Ministry of Education of PR China, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Zhengzhou 450016, P.R. China
| |
Collapse
|
12
|
Doryab A, Heydarian M, Yildirim AÖ, Hilgendorff A, Behr J, Schmid O. Breathing-induced stretch enhances the efficacy of an inhaled and orally delivered anti-fibrosis drug in vitro. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
13
|
Engelbrecht E, Kooistra T, Knipe RS. The Vasculature in Pulmonary Fibrosis. CURRENT TISSUE MICROENVIRONMENT REPORTS 2022; 3:83-97. [PMID: 36712832 PMCID: PMC9881604 DOI: 10.1007/s43152-022-00040-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 02/02/2023]
Abstract
Purpose of Review The current paradigm of idiopathic pulmonary fibrosis (IPF) pathogenesis involves recurrent injury to a sensitive alveolar epithelium followed by impaired repair responses marked by fibroblast activation and deposition of extracellular matrix. Multiple cell types are involved in this response with potential roles suggested by advances in single-cell RNA sequencing and lung developmental biology. Notably, recent work has better characterized the cell types present in the pulmonary endothelium and identified vascular changes in patients with IPF. Recent Findings Lung tissue from patients with IPF has been examined at single-cell resolution, revealing reductions in lung capillary cells and expansion of a population of vascular cells expressing markers associated with bronchial endothelium. In addition, pre-clinical models have demonstrated a fundamental role for aging and vascular permeability in the development of pulmonary fibrosis. Summary Mounting evidence suggests that the endothelium undergoes changes in the context of fibrosis, and these changes may contribute to the development and/or progression of pulmonary fibrosis. Additional studies will be needed to further define the functional role of these vascular changes.
Collapse
Affiliation(s)
| | - Tristan Kooistra
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
14
|
Ortiz-Zapater E, Signes-Costa J, Montero P, Roger I. Lung Fibrosis and Fibrosis in the Lungs: Is It All about Myofibroblasts? Biomedicines 2022; 10:biomedicines10061423. [PMID: 35740444 PMCID: PMC9220162 DOI: 10.3390/biomedicines10061423] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
In the lungs, fibrosis is a growing clinical problem that results in shortness of breath and can end up in respiratory failure. Even though the main fibrotic disease affecting the lung is idiopathic pulmonary fibrosis (IPF), which affects the interstitial space, there are many fibrotic events that have high and dangerous consequences for the lungs. Asthma, chronic obstructive pulmonary disease (COPD), excessive allergies, clearance of infection or COVID-19, all are frequent diseases that show lung fibrosis. In this review, we describe the different kinds of fibrosis and analyse the main types of cells involved-myofibroblasts and other cells, like macrophages-and review the main fibrotic mechanisms. Finally, we analyse present treatments for fibrosis in the lungs and highlight potential targets for anti-fibrotic therapies.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Faculty of Medicine-IIS INCLIVA, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | | | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Xu Q, Gao B, Liu X, Zhang X, Wu L, Xing D, Ma L, Liu J. Myocyte enhancer factor 2D promotes hepatocellular carcinoma through AMOTL2/YAP signaling that inhibited by luteolin. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:206-214. [PMID: 35698637 PMCID: PMC9187918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignancies in the world. There is a lack of effective treatment. Previous studies have shown that myocyte enhancer factor 2D (MEF2D) promotes the progression of HCC. Underlying mechanisms have not been fully elucidated. In this study, we reported experimental results obtained using double luciferase. Our results showed that AMOTL2, a negative regulator of Hippo/YAP signaling, and the MEF2 cis-acting element in the upstream region of its promoter bind to MEF2D, inhibiting its transcriptional expression. Studies confirmed that MEF2D affected the protein expression level of AMOTL2 and the YAP signaling activation. It promoted the migration and proliferation of hepatoma cells. We found that luteolin, a natural flavonoid, has anti-tumor activity in HCC cells by affecting YAP signaling transduction. In conclusion, we demonstrated that AMOTL2/YAP signaling is associated with MEF2D-related HCC progression. Luteolin is a promising anti-HCC compound for regulating this signaling.
Collapse
Affiliation(s)
- Qianqian Xu
- Department of Pharmacology, School of Pharmacy, Qingdao UniversityQingdao, Shandong, China
- Cancer Institute, The Affiliated Hospital of Qingdao University/Qingdao Cancer InstituteQingdao, Shandong, China
| | - Bing Gao
- Department of Pharmacology, School of Pharmacy, Qingdao UniversityQingdao, Shandong, China
| | - Xinlin Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University/Qingdao Cancer InstituteQingdao, Shandong, China
| | - Xin Zhang
- Department of Adult Internal Medicine, Qingdao Women and Children’s HospitalQingdao, Shandong, China
| | - Lili Wu
- Department of Pharmacology, School of Pharmacy, Qingdao UniversityQingdao, Shandong, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University/Qingdao Cancer InstituteQingdao, Shandong, China
| | - Leina Ma
- Cancer Institute, The Affiliated Hospital of Qingdao University/Qingdao Cancer InstituteQingdao, Shandong, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao UniversityQingdao, Shandong, China
| |
Collapse
|
16
|
Deng F, Wu Z, Xu M, Xia P. YAP Activates STAT3 Signalling to Promote Colonic Epithelial Cell Proliferation in DSS-Induced Colitis and Colitis Associated Cancer. J Inflamm Res 2022; 15:5471-5482. [PMID: 36164660 PMCID: PMC9508680 DOI: 10.2147/jir.s377077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIMS Yes-associated protein (YAP) is a key transcriptional coactivator of cell proliferation and differentiation. In this study, we sought to identify the roles of YAP in colonic epithelial regeneration and tumourigenesis. METHODS Murine DSS-induced colitis and YAP overexpression models were constructed via lentiviral intraperitoneal injection. Stable YAP-overexpressing cells, protein immunoprecipitation, and ChIP were used to deeply explore the molecular mechanism. RESULTS We found that the expression of YAP was dramatically diminished in the colonic crypts during the acute colitis phase, while YAP was strikingly enhanced to initiate tissue repair after DSS withdrawal. Overexpressing YAP in mice drastically accelerated epithelial regeneration, presenting with more intact structural integrity and reduced inflammatory cell infiltration in the mucosa. Further mechanistic studies showed that the expression of YAP in the nucleus was significantly increased by 2 h post-DSS removal, accompanied by upregulated protein levels of activated STAT3. Overexpression of YAP (YAPWT) elevated the expression of activated STAT3 and its transcriptional targets and strengthened the proliferation and "wound healing" ability of colonic cells. However, these effects were reversed when STAT3 was silenced in YAPWT cells. Moreover, YAP could directly interact with STAT3 in the nucleus, and c-Myc and CyclinD1 were the transcriptional targets. Finally, during colitis-associated cancer (CAC), YAPWT promoted the progression of CAC, while the phosphomimetic YAP downregulated the expression of STAT3 and inhibited the development and progression of CAC. CONCLUSION YAP activates STAT3 signalling to facilitate mucosal regeneration after DSS-induced colitis. However, excessive YAP activation in the colonic epithelium promotes CAC development.
Collapse
Affiliation(s)
- Feihong Deng
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
- Correspondence: Feihong Deng, Department of Gastroenterology, the Second Xiangya Hospital of Central South University; Research Center of Digestive Disease, Central South University, Changsha, Hunan, 410011, People’s Republic of China, Email
| | - Zengrong Wu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Mengmeng Xu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Pianpian Xia
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| |
Collapse
|