1
|
Liu A, Wang Y, Zheng S, Bao Z, Zhu H, Yin L, Liu C, Zhao X, Zhao Z, Zhu D, Yu H. Endonuclear Circ-calm4 regulates ferroptosis via a circR-Loop of the COMP gene in pulmonary artery smooth muscle cells. Eur J Pharmacol 2024; 982:176944. [PMID: 39187041 DOI: 10.1016/j.ejphar.2024.176944] [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: 03/31/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Pulmonary hypertension (PH) is a serious pulmonary vascular disease characterized by vascular remodeling. Circular RNAs (CircRNAs) play important roles in pulmonary hypertension, but the mechanism of PH is not fully understood, particularly the roles of circRNAs located in the nucleus. Circ-calmodulin 4 (circ-calm4) is expressed in both the cytoplasm and the nucleus of pulmonary arterial smooth muscle cells (PASMCs). This study aimed to investigate the role of endonuclear circ-calm4 in PH and elucidate its underlying signaling pathway in ferroptosis. Immunoblotting, quantitative real-time polymerase chain reaction (PCR), malondialdehyde (MDA) assay, immunofluorescence, iron assay, dot blot, and chromatin immunoprecipitation (ChIP) were performed to investigate the role of endonuclear circ-calm4 in PASMC ferroptosis. Increased endonuclear circ-calm4 facilitated ferroptosis in PASMCs under hypoxic conditions. We further identified the cartilage oligomeric matrix protein (COMP) as a downstream effector of circ-calm4 that contributed to the occurrence of hypoxia-induced ferroptosis in PASMCs. Importantly, we confirmed that endonuclear circ-calm4 formed circR-loops with the promoter region of the COMP gene and negatively regulated its expression. Inhibition of COMP restored the phenotypes related to ferroptosis under hypoxia stimulation combined with antisense oligonucleotide (ASO)-circ-calm4 treatment. We conclude that the circ-calm4/COMP axis contributed to hypoxia-induced ferroptosis in PASMCs and that circ-calm4 formed circR-loops with the COMP promoter in the nucleus and negatively regulated its expression. The circ-calm4/COMP axis may be useful for the design of therapeutic strategies for protecting cellular functionality against ferroptosis and pulmonary hypertension.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Cartilage Oligomeric Matrix Protein/genetics
- Cartilage Oligomeric Matrix Protein/metabolism
- Cell Hypoxia/genetics
- Cell Nucleus/metabolism
- Cells, Cultured
- Ferroptosis/genetics
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Pulmonary Artery/cytology
- Pulmonary Artery/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Aijing Liu
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Yingqi Wang
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Shuang Zheng
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Zhitu Bao
- Department of Chest Surgery, the Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang Province, China
| | - He Zhu
- Department of Oncology, the Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Lulu Yin
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Chunmiao Liu
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Xiaoxu Zhao
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Ziru Zhao
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), China; College of Pharmacy, Harbin Medical University, China.
| | - Hang Yu
- Department of Basic Medicine, Harbin Medical University (Daqing), Heilongjiang Province, China.
| |
Collapse
|
2
|
Xiao Y, Martinez L, Zigmond Z, Woltmann D, Singer DV, Singer HA, Vazquez-Padron RI, Salman LH. Functions for platelet factor 4 (PF4/CXCL4) and its receptors in fibroblast-myofibroblast transition and fibrotic failure of arteriovenous fistulas (AVFs). J Vasc Access 2024; 25:1911-1924. [PMID: 37589266 PMCID: PMC10998683 DOI: 10.1177/11297298231192386] [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] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Over 60% of End Stage Renal Disease (ESRD) patients are relying on hemodialysis (HD) to survive, and the arteriovenous fistula (AVF) is the preferred vascular access method for HD. However approximately half of all newly created AVF fail to mature and cannot be used without a salvage procedure. We have recently demonstrated an association between AVF maturation failure and post-operative fibrosis, while our RNA-seq study also revealed that veins that ultimately failed during AVF maturation had elevated levels of platelet factor 4 (PF4/CXCL4). However, a link between these two findings was yet to be established. METHODS In this study, we investigated potential mechanisms between PF4 levels and fibrotic remodeling in veins. We compared the local expression of PF4 and fibrosis marker integrin β6 (ITGB6) in veins that successfully underwent maturation with that in veins that ultimately failed to mature. We also measured the changes of expression level of α-smooth muscle actin (αSMA/ACTA2) and collagen (Col1/COL1A1) in venous fibroblasts upon various treatments, such as PF4 pharmacological treatment, alteration of PF4 expression, and blocking of PF4 receptors. RESULTS We found that PF4 is expressed in veins and co-localizes with αSMA. In venous fibroblasts, PF4 stimulates expression of αSMA and Col1 via different pathways. The former requires integrins αvβ5 and α5β1, while chemokine receptor CXCR3 is needed for the latter. Interestingly, we also discovered that the expression of PF4 is associated with that of ITGB6, the β subunit of integrin αvβ6. This integrin is critical for the activation of the major fibrosis factor TGFβ, and overexpression of PF4 promotes activation of the TGFβ pathway. CONCLUSIONS These results indicate that upregulation of PF4 may cause venous fibrosis both directly by stimulating fibroblast differentiation and expression of extracellular matrix (ECM) molecules and indirectly by facilitating the activation of the TGFβ pathway.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Zachary Zigmond
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Daniel Woltmann
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Diane V Singer
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Harold A Singer
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Loay H Salman
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology & Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
3
|
Wang X, Zou Z, Li K, Ren C, Yu X, Zhang Y, Zhao P, Yan S, Li Q. Design and fabrication of dual-layer PCL nanofibrous scaffolds with inductive influence on vascular cell responses. Colloids Surf B Biointerfaces 2024; 240:113988. [PMID: 38810467 DOI: 10.1016/j.colsurfb.2024.113988] [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: 01/25/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Confronted with the profound threat of cardiovascular diseases to health, vascular tissue engineering presents potential beyond the limitations of autologous and allogeneic grafts, offering a promising solution. This study undertakes an initial exploration into the impact of a natural active protein, elastin, on vascular cell behavior, by incorporating with polycaprolactone to prepare fibrous tissue engineering scaffold. The results reveal that elastin serves to foster endothelial cell adhesion and proliferation, suppress smooth muscle cell proliferation, and induce macrophage polarization. Furthermore, the incorporation of elastin contributes to heightened scaffold strength, compliance, and elongation, concomitantly lowering the elastic modulus. Subsequently, a bilayer oriented polycaprolactone (PCL) scaffold infused with elastin is proposed. This design draws inspiration from the cellular arrangement of native blood vessels, leveraging oriented fibers to guide cell orientation. The resulting fiber scaffold exhibits commendable mechanical properties and cell infiltration capacity, imparting valuable insights for the rapid endothelialization of vascular scaffolds.
Collapse
Affiliation(s)
- Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zifan Zou
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Cuihong Ren
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaorong Yu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Yang Zhang
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Peng Zhao
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Shujie Yan
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
4
|
Yang Q, Lai B, Xie H, Deng M, Li J, Yang Y, Wan J, Liao B, Liu F. Identification of differentially expressed ER stress-related genes and their association with pulmonary arterial hypertension. Respir Res 2024; 25:220. [PMID: 38789967 PMCID: PMC11127292 DOI: 10.1186/s12931-024-02849-4] [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: 01/08/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a complex and progressive illness that has a multifaceted origin, significant fatality rates, and profound effects on health. The pathogenesis of PAH is poorly defined due to the insufficient understanding of the combined impact of endoplasmic reticulum (ER) stress and immune infiltration, both of which play vital roles in PAH development. This study aims to identify potential ER stress-related biomarkers in PAH and investigate their involvement in immune infiltration. METHODS The GEO database was used to download gene expression profiles. Genes associated with ER stress were obtained from the MSigDB database. Weighted gene co-expression network analysis (WGCNA), GO, KEGG, and protein-protein interaction (PPI) were utilized to conduct screening of hub genes and explore potential molecular mechanisms. Furthermore, the investigation also delved into the presence of immune cells in PAH tissues and the correlation between hub genes and the immune system. Finally, we validated the diagnostic value and expression levels of the hub genes in PAH using subject-workup characterization curves and real-time quantitative PCR. RESULTS In the PAH and control groups, a total of 31 genes related to ER stress were found to be differentially expressed. The enrichment analysis revealed that these genes were primarily enriched in reacting to stress in the endoplasmic reticulum, dealing with unfolded proteins, transporting proteins, and processing proteins within the endoplasmic reticulum. EIF2S1, NPLOC4, SEC61B, SYVN1, and DERL1 were identified as the top 5 hub genes in the PPI network. Immune infiltration analysis revealed that these hub genes were closely related to immune cells. The receiver operating characteristic (ROC) curves revealed that the hub genes exhibited excellent diagnostic efficacy for PAH. The levels of SEC61B, NPLOC4, and EIF2S1 expression were in agreement with the findings of bioinformatics analysis in the PAH group. CONCLUSIONS Potential biomarkers that could be utilized are SEC61B, NPLOC4, and EIF2S1, as identified in this study. The infiltration of immune cells was crucial to the development and advancement of PAH. This study provided new potential therapeutic targets for PAH.
Collapse
Affiliation(s)
- Qi Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Banghui Lai
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Hao Xie
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Mingbin Deng
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jun Li
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yan Yang
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| | - Feng Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
5
|
Zhang X, Qin H, Ma Q, Zhang J, Tian H, Meng Y. CircST6GAL1 knockdown alleviates pulmonary arterial hypertension by regulating miR-509-5p/multiple C2 and transmembrane domain containing 2 axis. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13771. [PMID: 38747117 PMCID: PMC11094577 DOI: 10.1111/crj.13771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Hypertension is a main contributing factor of cardiovascular diseases; deregulated circular RNAs are involved in the pathogenesis of pulmonary arterial hypertension (PAH). Herein, we evaluated the function and mechanism of circST6GAL1 in PAH process. METHODS Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic environment for functional analysis. The cell counting kit-8, 5-ethynyl-2'-deoxyuridine, wound healing, and flow cytometry assays were used to investigate cell proliferation, migration, and apoptosis. qRT-PCR and Western blotting analyses were used for level measurement of genes and proteins. The binding between miR-509-5p and circST6GAL1 or multiple C2 and transmembrane domain containing 2 (MCTP2) was analyzed by dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays. The monocrotaline (MCT)-induced PAH mouse models were established for in vivo assay. RESULTS CircST6GAL1 was highly expressed in PAH patients and hypoxia-induced HPASMCs. Functionally, circST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs. Mechanistically, circST6GAL1 directly targeted miR-509-5p, and MCTP2 was a target of miR-509-5p. Rescue assays showed that the regulatory effects of circST6GAL1 deficiency on hypoxia-induced HPASMCs were abolished. Moreover, forced expression of miR-509-5p suppressed HPASMC proliferation and migration and induced cell apoptosis under hypoxia stimulation, while these effects were abolished by MCTP2 overexpression. Moreover, circST6GAL1 silencing improved MCT-induced pulmonary vascular remodeling and PAH. CONCLUSION CircST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs, and alleviated pulmonary vascular remodeling in MCT-induced PAH mouse models through the miR-509-5p/MCTP2 axis, indicating a potential therapeutic target for PAH.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Hao Qin
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Qiang Ma
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Junbo Zhang
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Hongyan Tian
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yan Meng
- Department of Peripheral Vascular DiseasesThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
6
|
Su H, Zhu H, Wang S, Li Y, Yan C, Wang J, Ying K. CircItgb5 promotes synthetic phenotype of pulmonary artery smooth muscle cells via interacting with miR-96-5p and Uba1 in monocrotaline-induced pulmonary arterial hypertension. Respir Res 2023; 24:165. [PMID: 37344798 DOI: 10.1186/s12931-023-02480-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a rare but fatal cardiopulmonary disease mainly characterized by pulmonary vascular remodeling. Aberrant expression of circRNAs has been reported to play a crucial role in pulmonary vascular remodeling. The existing literature predominantly centers on studies that examined the sponge mechanism of circRNAs. However, the mechanism of circRNAs in regulating PAH-related protein remains largely unknown. This study aimed to investigate the effect of circItgb5 on pulmonary vascular remodeling and the underlying functional mechanism. MATERIALS AND METHODS High-throughput circRNAs sequencing was used to detect circItgb5 expression in control and PDGF-BB-treated pulmonary arterial smooth muscle cells (PASMCs). Localization of circItgb5 in PASMCs was determined via the fluorescence in situ hybridization assay. Sanger sequencing was applied to analyze the circularization of Itgb5. The identification of proteins interacting with circItgb5 was achieved through a RNA pull-down assay. To assess the impact of circItgb5 on PASMCs proliferation, an EdU assay was employed. Additionally, the cell cycle of PASMCs was examined using a flow cytometry assay. Western blotting was used to detect biomarkers associated with the phenotypic switch of PASMCs. Furthermore, a monocrotaline (MCT)-induced PAH rat model was established to explore the effect of silencing circItgb5 on pulmonary vascular remodeling. RESULTS CircItgb5 was significantly upregulated in PDGF-BB-treated PASMCs and was predominately localized in the cytoplasm of PASMCs. In vivo experiments revealed that the knockdown of circItgb5 attenuated MCT-induced pulmonary vascular remodeling and right ventricular hypertrophy. In vitro experiments revealed that circItgb5 promoted the transition of PASMCs to synthetic phenotype. Mechanistically, circItgb5 sponged miR-96-5p to increase mTOR level and interacted with Uba1 protein to activate the Ube2n/Mdm2/ACE2 pathway. CONCLUSIONS CircItgb5 promoted the transition of PASMCs to synthetic phenotype by interacting with miR-96-5p and Uba1 protein. Knockdown of circItgb5 mitigated pulmonary arterial pressure, pulmonary vascular remodeling and right ventricular hypertrophy. Overall, circItgb5 has the potential for application as a therapeutic target for PAH.
Collapse
Affiliation(s)
- Hua Su
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Huiqi Zhu
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Sihao Wang
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Yeping Li
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Chao Yan
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Jiaoyan Wang
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China
| | - Kejing Ying
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, No. 3 Qingchun Road East, Hangzhou, China.
| |
Collapse
|
7
|
Huang W, Liu H, Pan Y, Wang X, Yang H, Wang D, Lin J, Zhang H. A modified primary culture method of rat pulmonary vein smooth muscle cells. J Cardiothorac Surg 2023; 18:146. [PMID: 37069582 PMCID: PMC10111653 DOI: 10.1186/s13019-023-02233-1] [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: 10/31/2022] [Accepted: 04/02/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Although the pressure of pulmonary vein increases before pulmonary artery in pulmonary hypertension due to left heart disease (PH-LHD), only a few studies have assessed pulmonary vein smooth muscle cells (PVSMCs) because of the lack of a simple and feasible isolation method. METHODS In this study, we introduced a simple method to obtain PVSMCs. Primary pulmonary veins were removed by puncture needle cannula guidance. Then, PVSMCs were cultured by the tissue explant method and purified by the differential adhesion method. The cells were characterized by hematoxylin-eosin (HE) staining, immunohistochemistry, western blotting, and immunofluorescence to observe the morphology and verify the expression of alpha-smooth muscle actin (α-SMA). RESULTS The HE staining results showed that the pulmonary vein media was thinner than the pulmonary artery, the intima and adventitia of the pulmonary vein were removed by this method, and the obtained cells with good activity exhibited morphological characteristics of smooth muscle cells. In addition, higher α-SMA expression was observed in the cells obtained by our isolation method than in the traditional method. CONCLUSION This study established a simple and feasible method to isolate and culture PVSMCs that might facilitate the cytological experiments for PH-LHD.
Collapse
Affiliation(s)
- Wenhui Huang
- Critical Care Medicine, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
- Anesthesiology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, 350004, P.R. China
| | - Hongjin Liu
- Department of Cardiovascular Surgery, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Yichao Pan
- Department of Cardiovascular Surgery, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Xueying Wang
- Critical Care Medicine, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Hongwei Yang
- Department of Cardiovascular Surgery, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Danjie Wang
- Critical Care Medicine, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Jing Lin
- Department of Cardiovascular Surgery, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Hui Zhang
- Critical Care Medicine, Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China.
- Critical Care Medicine, Union Hospital of Fujian Medical University, NO.29 Xinquan Road, Gulou District, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
8
|
Pulmonary Vascular Remodeling in Pulmonary Hypertension. J Pers Med 2023; 13:jpm13020366. [PMID: 36836600 PMCID: PMC9967990 DOI: 10.3390/jpm13020366] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Pulmonary vascular remodeling is the critical structural alteration and pathological feature in pulmonary hypertension (PH) and involves changes in the intima, media and adventitia. Pulmonary vascular remodeling consists of the proliferation and phenotypic transformation of pulmonary artery endothelial cells (PAECs) and pulmonary artery smooth muscle cells (PASMCs) of the middle membranous pulmonary artery, as well as complex interactions involving external layer pulmonary artery fibroblasts (PAFs) and extracellular matrix (ECM). Inflammatory mechanisms, apoptosis and other factors in the vascular wall are influenced by different mechanisms that likely act in concert to drive disease progression. This article reviews these pathological changes and highlights some pathogenetic mechanisms involved in the remodeling process.
Collapse
|
9
|
Mao J, Ma L. Research progress on the mechanism of phenotypic transformation of pulmonary artery smooth muscle cells induced by hypoxia. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:750-757. [PMID: 36915980 PMCID: PMC10262008 DOI: 10.3724/zdxbyxb-2022-0282] [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: 05/30/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022]
Abstract
Phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs) is a key factor in pulmonary vascular remodeling. Inhibiting or reversing phenotypic transformation can inhibit pulmonary vascular remodeling and control the progression of hypoxic pulmonary hypertension. Recent studies have shown that hypoxia causes intracellular peroxide metabolism to induce oxidative stress, induces multi-pathway signal transduction, including those related to autophagy, endoplasmic reticulum stress and mitochondrial dysfunction, and also induces non-coding RNA regulation of cell marker protein expression, resulting in PASMCs phenotypic transformation. This article reviews recent research progress on mechanisms of hypoxia-induced phenotypic transformation of PASMCs, which may be helpful for finding targets to inhibit phenotypic transformation and to improve pulmonary vascular remodeling diseases such as hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Jiaqi Mao
- 1. Medical Institute of Qinghai University, Xining 810001, China
- 2. Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China
| | - Lan Ma
- 2. Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China
| |
Collapse
|
10
|
Mark PR, Murray SA, Yang T, Eby A, Lai A, Lu D, Zieba J, Rajasekaran S, VanSickle EA, Rossetti LZ, Guidugli L, Watkins K, Wright MS, Bupp CP, Prokop JW. Autosomal recessive LRP1-related syndrome featuring cardiopulmonary dysfunction, bone dysmorphology, and corneal clouding. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006169. [PMID: 36307211 PMCID: PMC9632358 DOI: 10.1101/mcs.a006169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 08/22/2022] [Indexed: 01/25/2023] Open
Abstract
We provide the first study of two siblings with a novel autosomal recessive LRP1-related syndrome identified by rapid genome sequencing and overlapping multiple genetic models. The patients presented with respiratory distress, congenital heart defects, hypotonia, dysmorphology, and unique findings, including corneal clouding and ascites. Both siblings had compound heterozygous damaging variants, c.11420G > C (p.Cys3807Ser) and c.12407T > G (p.Val4136Gly) in LRP1, in which segregation analysis helped dismiss additional variants of interest. LRP1 analysis using multiple human/mouse data sets reveals a correlation to patient phenotypes of Peters plus syndrome with additional severe cardiomyopathy and blood vessel development complications linked to neural crest cells.
Collapse
Affiliation(s)
- Paul R. Mark
- Division of Medical Genetics, Spectrum Health, Grand Rapids, Michigan 49503, USA;,Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA
| | | | - Tao Yang
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan 49503, USA
| | - Alexandra Eby
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA;,Department of Science, Davenport University, Grand Rapids, Michigan 49512, USA
| | - Angela Lai
- Neonatal Intensive Care Unit, Bronson Methodist Hospital, Kalamazoo, Michigan 49007, USA
| | - Di Lu
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan 49503, USA
| | - Jacob Zieba
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA;,Genetics and Genome Sciences Program, Michigan State University, East Lansing, Michigan 48824, USA
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA;,Office of Research, Spectrum Health, Grand Rapids, Michigan 49503, USA
| | | | - Linda Z. Rossetti
- Division of Medical Genetics, Spectrum Health, Grand Rapids, Michigan 49503, USA;,Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA
| | - Lucia Guidugli
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Kelly Watkins
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Meredith S. Wright
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Caleb P. Bupp
- Division of Medical Genetics, Spectrum Health, Grand Rapids, Michigan 49503, USA;,Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA
| | - Jeremy W. Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan 49503, USA;,Genetics and Genome Sciences Program, Michigan State University, East Lansing, Michigan 48824, USA;,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
11
|
Vascular peroxidase 1 promotes phenotypic transformation of pulmonary artery smooth muscle cells via ERK pathway in hypoxia-induced pulmonary hypertensive rats. Life Sci 2022; 307:120910. [PMID: 36029851 DOI: 10.1016/j.lfs.2022.120910] [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: 04/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
AIMS Vascular peroxidase 1 (VPO1) plays an important role in mediation of vascular remodeling with pulmonary arterial hypertension (PAH). This study aims to determine whether VPO1 can promote phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs) and the underlying mechanisms. MAIN METHODS Sprague-Dawley (SD) rats were exposed to 10 % O2 for 21 days to establish the model of vascular remodeling in pulmonary arterial hypertension. PASMCs were incubated with 3 % O2 for 48 h to induce phenotypic transformation. Western blot was performed to detect the expressions of target proteins. The 5-ethynyl-2'-deoxyuridine (EdU) assay was conducted to measure the proliferation of PASMCs. KEY FINDINGS In the rats exposed to hypoxia, there were increases in right ventricular systolic pressure, pulmonary vascular remodeling and phenotypic transformation of PASMCs (the down-regulated contractile proteins of α-smooth muscle actin, smooth muscle 22α while the up-regulated synthetic proteins of osteopontin, cyclinD1), accompanied by up-regulation of VPO1, increase of hypochlorous acid (HOCl) production and elevation of the phosphorylation of ERK. In the cultured PASMCs exposed to hypoxia, similar results were achieved but they were reversed by VPO1 small interfering RNA (VPO1 siRNA) or HOCl inhibitor. Replacement of hypoxia with NaOCl could induce PASMCs phenotypic transformation and activate the ERK signaling. Furthermore, ERK inhibitor (PD98059) could also attenuate hypoxia-induced PASMCs phenotypic transformation. SIGNIFICANCE VPO1 play a pivotal role in promotion of phenotypic transformation of PASMCs under hypoxic condition through activation of VPO1/HOCl/ERK pathway. It might serve as a potential target for prevention of pulmonary vascular remodeling.
Collapse
|
12
|
Liu Z, Andraska E, Akinbode D, Mars W, Alvidrez RIM. LRP1 in the Vascular Wall. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Calvier L, Herz J, Hansmann G. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension. JACC Basic Transl Sci 2022; 7:164-180. [PMID: 35257044 PMCID: PMC8897182 DOI: 10.1016/j.jacbts.2021.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022]
Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases.
Collapse
Key Words
- ApoE, apolipoprotein E
- Apoer2
- BMP
- BMPR, bone morphogenetic protein receptor
- BMPR2
- COPD, chronic obstructive pulmonary disease
- CTGF, connective tissue growth factor
- HDL, high-density lipoprotein
- KO, knockout
- LDL receptor related protein
- LDL, low-density lipoprotein
- LDLR
- LDLR, low-density lipoprotein receptor
- LRP
- LRP, low-density lipoprotein receptor–related protein
- LRP1
- LRP1B
- LRP2
- LRP4
- LRP5
- LRP6
- LRP8
- MEgf7
- Mesd, mesoderm development
- PAH
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PDGF
- PDGFR-β, platelet-derived growth factor receptor-β
- PH, pulmonary hypertension
- PPARγ
- PPARγ, peroxisome proliferator-activated receptor gamma
- PVD
- RV, right ventricle/ventricular
- RVHF
- RVSP, right ventricular systolic pressure
- TGF-β1
- TGF-β1, transforming growth factor β1
- TGFBR, transforming growth factor β1 receptor
- TNF, tumor necrosis factor receptor
- VLDLR
- VLDLR, very low density lipoprotein receptor
- VSMC, vascular smooth muscle cell
- Wnt
- apolipoprotein E receptor 2
- endothelial cell
- gp330
- low-density lipoprotein receptor
- mRNA, messenger RNA
- megalin
- monocyte
- multiple epidermal growth factor-like domains 7
- pulmonary arterial hypertension
- pulmonary vascular disease
- right ventricle heart failure
- smooth muscle cell
- very low density lipoprotein receptor
- β-catenin
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
West J, Rathinasabapathy A, Chen X, Shay S, Gladson S, Talati M. Overexpression of Msx1 in Mouse Lung Leads to Loss of Pulmonary Vessels Following Vascular Hypoxic Injury. Cells 2021; 10:cells10092306. [PMID: 34571956 PMCID: PMC8471093 DOI: 10.3390/cells10092306] [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: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive lung disease caused by thickening of the pulmonary arterial wall and luminal obliteration of the small peripheral arteries leading to increase in vascular resistance which elevates pulmonary artery pressure that eventually causes right heart failure and death. We have previously shown that transcription factor Msx1 (mainly expressed during embryogenesis) is strongly upregulated in transformed lymphocytes obtained from PAH patients, especially IPAH. Under pathological conditions, Msx1 overexpression can cause cell dedifferentiation or cell apoptosis. We hypothesized that Msx1 overexpression contributes to loss of small pulmonary vessels in PAH. In IPAH lung, MSX1 protein localization was strikingly increased in muscularized remodeled pulmonary vessels, whereas it was undetectable in control pulmonary arteries. We developed a transgenic mouse model overexpressing MSX1 (MSX1OE) by about 4-fold and exposed these mice to normoxic, sugen hypoxic (3 weeks) or hyperoxic (100% 02 for 3 weeks) conditions. Under normoxic conditions, compared to controls, MSX1OE mice demonstrated a 30-fold and 2-fold increase in lung Msx1 mRNA and protein expression, respectively. There was a significant retinal capillary dropout (p < 0.01) in MSX1OE mice, which was increased further (p < 0.03) with sugen hypoxia. At baseline, the number of pulmonary vessels in MSX1OE mice was similar to controls. In sugen-hypoxia-treated MSX1OE mice, the number of small (0-25 uM) and medium (25-50 uM) size muscularized vessels increased approximately 2-fold (p < 0.01) compared to baseline controls; however, they were strikingly lower (p < 0.001) in number than in sugen-hypoxia-treated control mice. In MSX1OE mouse lung, 104 genes were upregulated and 67 genes were downregulated compared to controls. Similarly, in PVECs, 156 genes were upregulated and 320 genes were downregulated from siRNA to MSX1OE, and in PVSMCs, 65 genes were upregulated and 321 genes were downregulated from siRNA to MSX1OE (with control in the middle). Many of the statistically significant GO groups associated with MSX1 expression in lung, PVECs, and PVSMCs were similar, and were involved in cell cycle, cytoskeletal and macromolecule organization, and programmed cell death. Overexpression of MSX1 suppresses many cell-cycle-related genes in PVSMCs but induces them in PVECs. In conclusion, overexpression of Msx1 leads to loss of pulmonary vessels, which is exacerbated by sugen hypoxia, and functional consequences of Msx1 overexpression are cell-dependent.
Collapse
Affiliation(s)
| | | | | | | | | | - Megha Talati
- Correspondence: ; Tel.: +1-615-322-8095; Fax: +1-615-343-7448
| |
Collapse
|
15
|
Nohawica M, Errachid A, Wyganowska-Swiatkowska M. Adipose-PAS interactions in the context of its localised bio-engineering potential (Review). Biomed Rep 2021; 15:70. [PMID: 34276988 PMCID: PMC8278035 DOI: 10.3892/br.2021.1446] [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: 02/03/2021] [Accepted: 05/05/2021] [Indexed: 11/24/2022] Open
Abstract
Adipocytes are a known source of stem cells. They are easy to harvest, and are a suitable candidate for autogenous grafts. Adipose derived stem cells (ADSCs) have multiple target tissues which they can differentiate into, including bone and cartilage. In adipose tissue, ADSCs are able to differentiate, as well as providing energy and a supply of cytokines/hormones to manage the hypoxic and lipid/hormone saturated adipose environment. The plasminogen activation system (PAS) controls the majority of proteolytic activities in both adipose and wound healing environments, allowing for rapid cellular migration and tissue remodelling. While the primary activation pathway for PAS occurs through the urokinase plasminogen activator (uPA), which is highly expressed by endothelial cells, its function is not limited to enabling revascularisation. Proteolytic activity is dependent on protease activation, localisation, recycling mechanisms and substrate availability. uPA and uPA activated plasminogen allows pluripotent cells to arrive to new local environments and fulfil the niche demands. However, overstimulation, the acquisition of a migratory phenotype and constant protein turnover can be unconducive to the formation of structured hard and soft tissues. To maintain a suitable healing pattern, the proteolytic activity stimulated by uPA is modulated by plasminogen activator inhibitor 1. Depending on the physiological settings, different parts of the remodelling mechanism are activated with varying results. Utilising the differences within each microenvironment to recreate a desired niche is a valid therapeutic bio-engineering approach. By controlling the rate of protein turnover combined with a receptive stem cell lineage, such as ADSC, a novel avenue on the therapeutic opportunities may be identified, which can overcome limitations, such as scarcity of stem cells, low angiogenic potential or poor host tissue adaptation.
Collapse
Affiliation(s)
- Michal Nohawica
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| | - Abdelmounaim Errachid
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
- Earth and Life Institute, University Catholique of Louvain, Louvain-la-Neuve B-1348, Belgium
| | - Marzena Wyganowska-Swiatkowska
- Chair and Department of Dental Surgery and Periodontology, Poznan University of Medicinal Sciences, Poznan, Greater Poland 60-812, Poland
| |
Collapse
|
16
|
Mao X, Yang X, Chen X, Yu S, Yu S, Zhang B, Ji Y, Chen Y, Ouyang Y, Luo W. Single-cell transcriptome analysis revealed the heterogeneity and microenvironment of gastrointestinal stromal tumors. Cancer Sci 2021; 112:1262-1274. [PMID: 33393143 PMCID: PMC7935798 DOI: 10.1111/cas.14795] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/15/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the human gastrointestinal tract. In this study, we performed single-cell RNA sequencing (RNA-seq) on intra- and peri-tumor tissues from GIST patients with the aim of discovering the heterogeneity of tumor cells in GIST and their interactions with other cells. We found four predominating cell types in GIST tumor tissue, including T cells, macrophages, tumor cells, and NK cells. Tumor cells could be clustered into two groups: one was highly proliferating and associated with high risk of metastasis, the other seemed "resting" and associated with low risk. Their clinical relevance and prognostic values were confirmed by RNA-seq of 65 GIST samples. T cells were the largest cell type in our single-cell data. Two groups of CD8+ effector memory (EM) cells were in the highest clonal expansion and performed the highest cytotoxicity but were also the most exhausted among all T cells. A group of macrophages were found polarized to possess both M1 and M2 signatures, and increased along with tumor progression. Cell-to-cell interaction analysis revealed that adipose endothelial cells had high interactions with tumor cells to facilitate their progression. Macrophages were at the center of the tumor microenvironment, recruiting immune cells to the tumor site and having most interactions with both tumor and nontumor cells. In conclusion, we obtained an overview of the GIST microenvironment and revealed the heterogeneity of each cell type and their relevance to risk classifications, which provided a novel theoretical basis for learning and curing GISTs.
Collapse
Affiliation(s)
- Xiaofan Mao
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Xuezhu Yang
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Xiangping Chen
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Sifei Yu
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Si Yu
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Beiying Zhang
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Yong Ji
- Gastroenterology, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Yihao Chen
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Ying Ouyang
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China.,Medical Engineering Technology Research and Development Center of Immune Repertoire in Foshan, The First People's Hospital of Foshan & Sun Yat-Sen University Foshan Hospital, Foshan, China
| |
Collapse
|
17
|
Liu N, Xue Y, Tang J, Zhang M, Ren X, Fu J. The dynamic change of phenotypic markers of smooth muscle cells in an animal model of cerebral small vessel disease. Microvasc Res 2021; 133:104061. [PMID: 32827495 DOI: 10.1016/j.mvr.2020.104061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/22/2020] [Accepted: 08/15/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND The pathological character of cerebral small vessel disease (CSVD) is the dysfunction of cerebral small arteries caused by risk factors. A switch from the contractile phenotype to the synthetic phenotype of vascular smooth muscle cells (SMCs) can decrease the contractility of arteries. The alteration of the vascular wall extracellular matrix (ECM) is found to regulate the process. We speculated that SMCs phenotype changes may also occur in CSVD induced by hypertension and the alteration of ECM especially fibronectin and laminin may regulate the process. METHOD Male spontaneously hypertensive rats (SHR) were used as a CSVD animal model. SMCs phenotypic markers and the ECM expression of the cerebral small arteries of SHR at different ages were evaluated by immunofluorescence. The phenotype changes of primary brain microvascular SMCs cultured on laminin-coating dish or fibronectin-coating dish were evaluated by western blot. RESULT A switch from the contractile phenotype to synthetic phenotype in SHR at 10 and 22 weeks of age was observed. Meanwhile, increased expression of fibronectin and a temporary decline of laminin was found in small arteries of SHR at 22 weeks. In vitro experiments also convinced that SMCs cultured on a fibronectin-coating dish failed to maintain contractile phenotype. While at 50 weeks, significant drops of both synthetic and contractile phenotypic markers were witnessed in SHR, with high expressions of four kinds of ECM. CONCLUSION SMCs in cerebral small arteries exhibited a switch from the contractile phenotype to synthetic phenotype during the chronic process of hypertension and aging. Moreover, the change of fibronectin and laminin may regulate the process.
Collapse
MESH Headings
- Age Factors
- Animals
- Biomarkers/metabolism
- Cells, Cultured
- Cerebral Arteries/metabolism
- Cerebral Arteries/pathology
- Cerebral Arteries/physiopathology
- Cerebral Small Vessel Diseases/etiology
- Cerebral Small Vessel Diseases/metabolism
- Cerebral Small Vessel Diseases/pathology
- Cerebral Small Vessel Diseases/physiopathology
- Disease Models, Animal
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Fibronectins/metabolism
- Hypertension/complications
- Hypertension/metabolism
- Hypertension/physiopathology
- Laminin/metabolism
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Rats, Inbred SHR
- Rats, Inbred WKY
- Vascular Remodeling
- Vasoconstriction
- Rats
Collapse
Affiliation(s)
- Na Liu
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Miaoyi Zhang
- Department of Neurology, North Huashan hospital, Fudan University, No.108 Lu Xiang Road, Shanghai 201900, China
| | - Xue Ren
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, China; Department of Neurology, North Huashan hospital, Fudan University, No.108 Lu Xiang Road, Shanghai 201900, China.
| |
Collapse
|
18
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
19
|
Auderset L, Pitman KA, Cullen CL, Pepper RE, Taylor BV, Foa L, Young KM. Low-Density Lipoprotein Receptor-Related Protein 1 (LRP1) Is a Negative Regulator of Oligodendrocyte Progenitor Cell Differentiation in the Adult Mouse Brain. Front Cell Dev Biol 2020; 8:564351. [PMID: 33282858 PMCID: PMC7691426 DOI: 10.3389/fcell.2020.564351] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a large, endocytic cell surface receptor that is highly expressed by oligodendrocyte progenitor cells (OPCs) and LRP1 expression is rapidly downregulated as OPCs differentiate into oligodendrocytes (OLs). We report that the conditional deletion of Lrp1 from adult mouse OPCs (Pdgfrα-CreER :: Lrp1fl/fl) increases the number of newborn, mature myelinating OLs added to the corpus callosum and motor cortex. As these additional OLs extend a normal number of internodes that are of a normal length, Lrp1-deletion increases adult myelination. OPC proliferation is also elevated following Lrp1 deletion in vivo, however, this may be a secondary, homeostatic response to increased OPC differentiation, as our in vitro experiments show that LRP1 is a direct negative regulator of OPC differentiation, not proliferation. Deleting Lrp1 from adult OPCs also increases the number of newborn mature OLs added to the corpus callosum in response to cuprizone-induced demyelination. These data suggest that the selective blockade of LRP1 function on adult OPCs may enhance myelin repair in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Loic Auderset
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Lisa Foa
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
20
|
Angiotensin-converting enzyme 2 augments the effects of endothelial progenitor cells-exosomes on vascular smooth muscle cell phenotype transition. Cell Tissue Res 2020; 382:509-518. [PMID: 32852610 DOI: 10.1007/s00441-020-03259-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022]
Abstract
Phenotype transition of vascular smooth muscle cells (VSMCs) is implicated in vascular diseases. Angiotensin-converting enzyme 2 (ACE2) is a perspective cardiovascular target due to its ability of converting angiotensin (Ang II) to Ang (1-7). Our group recently showed that ACE2 can regulate the function of endothelial progenitor cell-derived exosomes (EPC-EXs). Here, we investigate whether ACE2 could affect the role of EPC-EXs on phenotype transition of VSMCs. After co-incubation with EXs released from EPC overexpressed ACE2 (EPC-EXsACE2), the ACE2 level and Ang II/Ang (1-7), proliferation/migration, phenotype gene, cytokine and NF-κB level on VSMCs were assessed. To determine the EX uptake route, VSMCs were pretreated with inhibitors. We found that (1) EPC-EXs and EPC-EXsACE2 were uptaken by VSMCs dominantly through caveolin-dependent endocytosis. (2) EPC-EXsACE2 remarkably increased the ACE2 level and decreased Ang II/Ang (1-7) in VSMCs activated by Ang II, whereas EPC-EXsACE2 pretreated by proteinase A blocked this effect. (3) EPC-EXsACE2 had better effects than EPC-EXs on reducing proliferation/migration activities and cytokine (MCP-1, TNF-α) secretion of Ang II-activated VSMCs. (4) EPC-EXs attenuated Ang II-induced VSMC synthetic phenotype change as evidenced by upregulated expressions of calponin and a-SMA and downregulated expressions of CRBP-1 and MYH10, associated with a decreased NF-κB level. EPC-EXsACE2 augmented these effects, which were attenuated by ACE2 inhibitor (DX600). In conclusion, EPC-EXsACE2 reduced Ang II-induced VSMC phenotype change by conveying functional ACE2 to downregulate the activated NF-κB pathway.
Collapse
|
21
|
Zha LH, Zhou J, Tan Y, Guo S, Zhang MQ, Li S, Yan P, Yu ZX. NLRC3 inhibits PDGF-induced PASMCs proliferation via PI3K-mTOR pathway. J Cell Physiol 2020; 235:9557-9567. [PMID: 32383265 DOI: 10.1002/jcp.29763] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/23/2020] [Indexed: 12/15/2022]
Abstract
Few studies about nucleotide-oligomerization domain-like receptor subfamily C3 (NLRC3) in PASMCs have been conducted. This research aimed to investigate the role of NLRC3 on platelet-derived growth factor (PDGF)-induced proliferation of pulmonary artery smooth muscle cells (PASMCs) and its underlying mechanism. We found that the proliferation of PASMCs stimulated with PDGF decreased when phosphoinositide 3-kinase (PI3K) or mammalian target of rapamycin (mTOR) inhibitors pretreatment. Overexpression of NLRC3 inhibited the proliferation of PASMCs and the phosphorylation of PI3K and mTOR while knocking down NLRC3 reversed this effect. Targeted to PI3K or mTOR can also reverse the effect of NLRC3. Activation of PI3K increased the phosphorylation of mTOR while inhibition of PI3K reduced it. Our data suggest that PDGF can induce abnormal proliferation of PASMCs, and NLRC3 suppresses activation of the PI3K-mTOR signaling thus inhibits PASMCs proliferation. These findings unveiled the effect of NLRC3 as an inhibitor of the PI3K-mTOR pathway mediating protection against PASMCs proliferation.
Collapse
Affiliation(s)
- Li-Huang Zha
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Zhou
- Department of Medical Science Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yilong Tan
- Department of Ultrasonography, Zhuzhou Hospital, Central South University, Zhuzhou, Hunan, China
| | - Shuhong Guo
- Department of Cardiology, Zhuzhou Hospital, Central South University, Zhuzhou, Hunan, China
| | - Men-Qiu Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sheng Li
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Yan
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zai-Xin Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Schnieder J, Mamazhakypov A, Birnhuber A, Wilhelm J, Kwapiszewska G, Ruppert C, Markart P, Wujak L, Rubio K, Barreto G, Schaefer L, Wygrecka M. Loss of LRP1 promotes acquisition of contractile-myofibroblast phenotype and release of active TGF-β1 from ECM stores. Matrix Biol 2019; 88:69-88. [PMID: 31841706 DOI: 10.1016/j.matbio.2019.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022]
Abstract
In healing tissue, fibroblasts differentiate to α-smooth muscle actin (SMA)-expressing contractile-myofibroblasts, which pull the wound edges together ensuring proper tissue repair. Uncontrolled expansion of the myofibroblast population may, however, lead to excessive tissue scarring and finally to organ dysfunction. Here, we demonstrate that the loss of low-density lipoprotein receptor-related protein (LRP) 1 overactivates the JNK1/2-c-Jun-Fra-2 signaling pathway leading to the induction of α-SMA and periostin expression in human lung fibroblasts (hLF). These changes are accompanied by increased contractility of the cells and the integrin- and protease-dependent release of active transforming growth factor (TGF)-β1 from the extracellular matrix (ECM) stores. Liberation of active TGF-β1 from the ECM further enhances α-SMA and periostin expression thus accelerating the phenotypic switch of hLF. Global gene expression profiling of LRP1-depleted hLF revealed that the loss of LRP1 affects cytoskeleton reorganization, cell-ECM contacts, and ECM production. In line with these findings, fibrotic changes in the skin and lung of Fra-2 transgenic mice were associated with LRP1 depletion and c-Jun overexpression. Altogether, our results suggest that dysregulation of LRP1 expression in fibroblasts in healing tissue may lead to the unrestrained expansion of contractile myofibroblasts and thereby to fibrosis development. Further studies identifying molecules, which regulate LRP1 expression, may provide new therapeutic options for largely untreatable human fibrotic diseases.
Collapse
Affiliation(s)
- Jennifer Schnieder
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Argen Mamazhakypov
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Departments of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | | | - Clemens Ruppert
- Departments of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Philipp Markart
- Department of Pulmonary Medicine, Fulda Hospital, University Medicine Marburg, Campus Fulda, Fulda, Germany
| | - Lukasz Wujak
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Karla Rubio
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Guillermo Barreto
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany; Brain and Lung Epigenetics, Laboratoire Croissance, Réparation et Régénération Tissulaires (CRRET), Université Paris Est Créteil (UPEC), Créteil, France
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt Am Main, Germany
| | - Malgorzata Wygrecka
- Departments of Biochemistry and Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| |
Collapse
|