1
|
Jain KG, Liu Y, Zhao R, Muire PJ, Zhang J, Zang QS, Ji HL. Humanized L184Q Mutated Surfactant Protein C Gene Alters Alveolar Type 2 Epithelial Cell Fate. Int J Mol Sci 2024; 25:8723. [PMID: 39201410 PMCID: PMC11354303 DOI: 10.3390/ijms25168723] [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: 06/13/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Alveolar type 2 epithelial (AT2) cells synthesize surfactant protein C (SPC) and repair an injured alveolar epithelium. A mutated surfactant protein C gene (SftpcL184Q, Gene ID: 6440) in newborns has been associated with respiratory distress syndrome and pulmonary fibrosis. However, the underlying mechanisms causing Sftpc gene mutations to regulate AT2 lineage remain unclear. We utilized three-dimensional (3D) feeder-free AT2 organoids in vitro to simulate the alveolar epithelium and compared AT2 lineage characteristics between WT (C57BL/6) and SftpcL184Q mutant mice using colony formation assays, immunofluorescence, flow cytometry, qRT-PCR, and Western blot assays. The AT2 numbers were reduced significantly in SftpcL184Q mice. Organoid numbers and colony-forming efficiency were significantly attenuated in the 3D cultures of primary SftpcL184Q AT2 cells compared to those of WT mice. Podoplanin (PDPN, Alveolar type 1 cell (AT1) marker) expression and transient cell count was significantly increased in SftpcL184Q organoids compared to in the WT mice. The expression levels of CD74, heat shock protein 90 (HSP90), and ribosomal protein S3A1 (RPS3A1) were not significantly different between WT and SftpcL184Q AT2 cells. This study demonstrated that humanized SftpcL184Q mutation regulates AT2 lineage intrinsically. This regulation is independent of CD74, HSP90, and RPS3A1 pathways.
Collapse
Affiliation(s)
- Krishan G. Jain
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Yang Liu
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Runzhen Zhao
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Preeti J. Muire
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Department of Orthopedics and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL 60153, USA;
- Departments of Pathology and Radiation Oncology, Loyola University Medical Center, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Hong-Long Ji
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
2
|
Liu YB, Tan XH, Yang HH, Yang JT, Zhang CY, Jin L, Yang NSY, Guan CX, Zhou Y, Liu SK, Xiong JB. Wnt5a-mediated autophagy contributes to the epithelial-mesenchymal transition of human bronchial epithelial cells during asthma. Mol Med 2024; 30:93. [PMID: 38898476 PMCID: PMC11188189 DOI: 10.1186/s10020-024-00862-3] [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: 02/24/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) of human bronchial epithelial cells (HBECs) is essential for airway remodeling during asthma. Wnt5a has been implicated in various lung diseases, while its role in the EMT of HBECs during asthma is yet to be determined. This study sought to define whether Wnt5a initiated EMT, leading to airway remodeling through the induction of autophagy in HBECs. METHODS Microarray analysis was used to investigate the expression change of WNT5A in asthma patients. In parallel, EMT models were induced using 16HBE cells by exposing them to house dust mites (HDM) or interleukin-4 (IL-4), and then the expression of Wnt5a was observed. Using in vitro gain- and loss-of-function approaches via Wnt5a mimic peptide FOXY5 and Wnt5a inhibitor BOX5, the alterations in the expression of the epithelial marker E-cadherin and the mesenchymal marker protein were observed. Mechanistically, the Ca2+/CaMKII signaling pathway and autophagy were evaluated. An autophagy inhibitor 3-MA was used to examine Wnt5a in the regulation of autophagy during EMT. Furthermore, we used a CaMKII inhibitor KN-93 to determine whether Wnt5a induced autophagy overactivation and EMT via the Ca2+/CaMKII signaling pathway. RESULTS Asthma patients exhibited a significant increase in the gene expression of WNT5A compared to the healthy control. Upon HDM and IL-4 treatments, we observed that Wnt5a gene and protein expression levels were significantly increased in 16HBE cells. Interestingly, Wnt5a mimic peptide FOXY5 significantly inhibited E-cadherin and upregulated α-SMA, Collagen I, and autophagy marker proteins (Beclin1 and LC3-II). Rhodamine-phalloidin staining showed that FOXY5 resulted in a rearrangement of the cytoskeleton and an increase in the quantity of stress fibers in 16HBE cells. Importantly, blocking Wnt5a with BOX5 significantly inhibited autophagy and EMT induced by IL-4 in 16HBE cells. Mechanistically, autophagy inhibitor 3-MA and CaMKII inhibitor KN-93 reduced the EMT of 16HBE cells caused by FOXY5, as well as the increase in stress fibers, cell adhesion, and autophagy. CONCLUSION This study illustrates a new link in the Wnt5a-Ca2+/CaMKII-autophagy axis to triggering airway remodeling. Our findings may provide novel strategies for the treatment of EMT-related diseases.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, 410011, China.
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| | - Jian-Bing Xiong
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
3
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [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] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
4
|
Kim SY, McTeague D, Cheong SS, Hind M, Dean CH. Deciphering the impacts of modulating the Wnt-planar cell polarity (PCP) pathway on alveolar repair. Front Cell Dev Biol 2024; 12:1349312. [PMID: 38476262 PMCID: PMC10927798 DOI: 10.3389/fcell.2024.1349312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Many adult lung diseases involve dysregulated lung repair. Deciphering the molecular and cellular mechanisms that govern intrinsic lung repair is essential to develop new treatments to repair/regenerate the lungs. Aberrant Wnt signalling is associated with lung diseases including emphysema, idiopathic pulmonary fibrosis and pulmonary arterial hypertension but how Wnt signalling contributes to these diseases is still unclear. There are several alternative pathways that can be stimulated upon Wnt ligand binding, one of these is the Planar Cell Polarity (PCP) pathway which induces actin cytoskeleton remodelling. Wnt5a is known to stimulate the PCP pathway and this ligand is of particular interest in regenerative lung biology because of its association with lung diseases and its role in the alveolar stem cell niche. To decipher the cellular mechanisms through which Wnt5a and the PCP pathway affect alveolar repair we utilised a 3-D ex-vivo model of lung injury and repair, the AIR model. Our results show that Wnt5a specifically enhances the alveolar epithelial progenitor cell population following injury and surprisingly, this function is attenuated but not abolished in Looptail (Lp) mouse lungs in which the PCP pathway is dysfunctional. However, Lp tracheal epithelial cells show reduced stiffness and Lp alveolar epithelial cells are less migratory than wildtype (WT), indicating that Lp lung epithelial cells have a reduced capacity for repair. These findings provide important mechanistic insight into how Wnt5a and the PCP pathway contribute to lung repair and indicate that these components of Wnt signalling may be viable targets for the development of pro-repair treatments.
Collapse
Affiliation(s)
- Sally Yunsun Kim
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David McTeague
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Sek-Shir Cheong
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Charlotte H. Dean
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [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: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
6
|
Jain KG, Xi NM, Zhao R, Ahmad W, Ali G, Ji HL. Alveolar Type 2 Epithelial Cell Organoids: Focus on Culture Methods. Biomedicines 2023; 11:3034. [PMID: 38002035 PMCID: PMC10669847 DOI: 10.3390/biomedicines11113034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Lung diseases rank third in terms of mortality and represent a significant economic burden globally. Scientists have been conducting research to better understand respiratory diseases and find treatments for them. An ideal in vitro model must mimic the in vivo organ structure, physiology, and pathology. Organoids are self-organizing, three-dimensional (3D) structures originating from adult stem cells, embryonic lung bud progenitors, embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). These 3D organoid cultures may provide a platform for exploring tissue development, the regulatory mechanisms related to the repair of lung epithelia, pathophysiological and immunomodulatory responses to different respiratory conditions, and screening compounds for new drugs. To create 3D lung organoids in vitro, both co-culture and feeder-free methods have been used. However, there exists substantial heterogeneity in the organoid culture methods, including the sources of AT2 cells, media composition, and feeder cell origins. This article highlights the currently available methods for growing AT2 organoids and prospective improvements to improve the available culture techniques/conditions. Further, we discuss various applications, particularly those aimed at modeling human distal lung diseases and cell therapy.
Collapse
Affiliation(s)
- Krishan Gopal Jain
- Department of Surgery, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (W.A.)
- Burn and Shock Trauma Research Institute, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Nan Miles Xi
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL 60660, USA;
| | - Runzhen Zhao
- Department of Surgery, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (W.A.)
- Burn and Shock Trauma Research Institute, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Waqas Ahmad
- Department of Surgery, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (W.A.)
- Burn and Shock Trauma Research Institute, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Gibran Ali
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Hong-Long Ji
- Department of Surgery, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (W.A.)
- Burn and Shock Trauma Research Institute, Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
7
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Zhang J, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. Stem Cell Res Ther 2023; 14:185. [PMID: 37501095 PMCID: PMC10375781 DOI: 10.1186/s13287-023-03414-4] [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: 11/21/2022] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). METHODS Wild-type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony-forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. RESULTS A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of re-alveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. CONCLUSIONS Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA.
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
8
|
Ali G, Zhang M, Chang J, Zhao R, Jin Y, Ji HL. PAI-1 regulates AT2-mediated re-alveolarization and ion permeability. RESEARCH SQUARE 2023:rs.3.rs-2289657. [PMID: 36909505 PMCID: PMC10002791 DOI: 10.21203/rs.3.rs-2289657/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Background Acute lung injury is characterized by overwhelmingly elevated PAI-1 in both lung edema fluid and the circulating system. The role of increased PAI-1, encoded by Serpine1 gene, in the regeneration of injured lung epithelium has not been understood completely. This study aimed to investigate the role of Serpine1 in the regulation of alveolar type 2 epithelial cell (AT2) fate in a humanized mouse line carrying diseased mutants (Serpine1Tg). Methods Wild type (wt) and Serpine1Tg AT2 cells were either cultured as monolayers or 3D alveolospheres. Colony forming assay and total surface area of organoids were analyzed. AT1 and AT2 cells in organoids were counted by immunohistochemistry and fluorescence-activated cell sorting (FACS). To test the potential effects of elevated PAI-1 on the permeability in the epithelial monolayers, we digitized the biophysical properties of polarized AT2 monolayers grown at the air-liquid interface. Results A significant reduction in total AT2 cells harvested in Serpine1Tg mice was observed compared with wt controls. AT2 cells harvested from Serpine1Tg mice reduced significantly over the wt controls. Spheroids formed by Serpine1Tg AT2 cells were lesser than wt control. Similarly, the corresponding surface area, a readout of realveolarization of injured epithelium, was markedly reduced in Serpine1Tg organoids. FACS analysis revealed a significant suppression in the number of AT2 cells, in particular, the CD44+ subpopulation, in Serpine1Tg organoids. A lesser ratio of AT1:AT2 cells in Serpine1Tg organoids was observed compared with wt cultures. There was a significant increase in transepithelial resistance but not amiloride inhibition. Conclusions Our study suggests elevated PAI-1 in injured lungs downregulates alveolar epithelial regeneration by reducing the AT2 self-renewal, particularly in the CD44+ cells.
Collapse
Affiliation(s)
- Gibran Ali
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Mo Zhang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jianjun Chang
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, USA
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Texas Lung Injury Institute, University of Texas at Tyler Health Science Center, Tyler, TX, USA
- Corresponding Author: Dr. Hong-Long (James) Ji, M.D., Ph.D., , University of Texas at Tyler Health Science Center, 11937 US Highway 271, Tyler, TX 75708-3154, USA
| |
Collapse
|
9
|
Chen J, Gao F, Li D, Wang J. MiR26-5p inhibits pathological pulmonary microvascular angiogenesis via down-regulating WNT5A. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:812-819. [PMID: 37396938 PMCID: PMC10311967 DOI: 10.22038/ijbms.2023.68856.15006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/19/2023] [Indexed: 07/04/2023]
Abstract
Objectives Pathological micro angiogenesis is a key pathogenic factor in pulmonary diseases such as pulmonary hypertension and hepatopulmonary syndrome. More and more pieces of evidence show that excessive proliferation of pulmonary microvascular endothelial cells is the key event of pathological micro angiogenesis. The purpose of this research is to reveal the mechanism of miR26-5p regulating pulmonary microvascular hyperproliferation. Materials and Methods Hepatopulmonary syndrome rat model was made by common bile duct ligation. HE and IHC staining were used for analysis of the pathology of the rat. CCK8, transwell, and wound healing assay were used to assess miR26-5p or target gene WNT5A functioned toward PMVECs. microRNA specific mimics and inhibitors were used for up/down-regulated miR26-5p expression in PMVECs. Recombinant lentivirus was used for overexpression/knockdown WNT5A expression in PMVECs. And the regulation relationship of miR26-5p and WNT5A was analyzed by dual-luciferase reporter assay. Results qPCR showed that miR26-5p was significantly down-regulated in the course of HPS disease. Bioinformatics data showed that WNT5A was one of the potential key target genes of miR26-5p. Immunohistochemistry and qPCR analysis showed that WNT5A was largely expressed in pulmonary microvascular endothelial cells, in addition, this molecule was significantly up-regulated with the progression of the disease. Furthermore, dual luciferase reporter assay showed that miR26-5p could bind to WNT5A 3 'UTR region to inhibit WNT5A synthesis. Conclusion The results suggested MiR26-5p negatively regulated PMVECs proliferation and migration by WNT5A expression. Overexpression of miR26-5p may be a potentially beneficial strategy for HPS therapy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Anesthesiology, People’s Hospital of Chongqing Banan District, Chongqing 401320, China
- These authors contributed equally to this work
| | - Feng Gao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 400038, China
- These authors contributed equally to this work
| | - Dan Li
- Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jinquan Wang
- Department of Anesthesiology, The Ninth People’s Hospital of Chongqing, Chongqing 400700, China
| |
Collapse
|