1
|
Hiraide T, Tsuda N, Momoi M, Shinya Y, Sano M, Fukuda K, Shibahara J, Kuramoto J, Kanai Y, Kosaki K, Hakamata Y, Kataoka M. CXCL12/CXCR4 pathway as a novel therapeutic target for RNF213-associated pulmonary arterial hypertension. Sci Rep 2024; 14:26604. [PMID: 39496725 PMCID: PMC11535198 DOI: 10.1038/s41598-024-77388-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/22/2024] [Indexed: 11/06/2024] Open
Abstract
Genetic backgrounds of patients with pulmonary arterial hypertension (PAH) were not fully investigated. A variant of c.14429G > A (p.Arg4810Lys) in the ring finger protein 213 gene (RNF213) was recently identified as a risk allele for poor treatment response and poor clinical prognosis in patients with PAH. However, the molecular mechanisms of the RNF213 p.Arg4810Lys variant in development of PAH are unknown. We investigated the underlying molecular mechanisms of RNF213-associated vasculopathy using an in vivo mouse model. RNF213+/p.Arg4828Lys mice, harboring the heterozygous RNF213 p.Arg4828Lys variant corresponding to the p.Arg4810Lys variant in humans, were created using the CRISPR-Cas9 system to recapitulate the genetic status of PAH patients. RNF213+/p.Arg4828Lys mice had a significant elevation of the right ventricular systolic pressure, hypertrophy of the right ventricle, and increased thickness of the pulmonary arterial medial wall compared with wild-type mice after 3 months of exposure to a hypoxic environment. C-X-C motif chemokine ligand 12 (CXCL12), a C-X-C chemokine receptor type 4 (CXCR4) ligand, was significantly elevated in the lungs of RNF213+/p.Arg4828Lys mice, and PAH was ameliorated by the administration of a CXCR4 antagonist. CXCL12-CXCR4 is an angiogenic chemokine axis, and immunohistochemistry demonstrated an increase in CXCR4 in vimentin-positive spindle-shaped cells in adventitia and interstitial lesions in RNF213+/p.Arg4828Lys mice and lung specimens from severe PAH patients with the RNF213 p.Arg4810Lys variant. We confirmed a cause-and-effect relationship between the RNF213 p.Arg4810Lys variant and PAH via the CXCL12-CXCR4 pathway. The findings in this study suggest that targeting this pathway might be a novel therapeutic strategy for RNF213-associated vasculopathy.
Collapse
Affiliation(s)
- Takahiro Hiraide
- Department of Cardiology, Keio University School of Medicine, Shinanomachi 35, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Noboru Tsuda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mizuki Momoi
- Department of Cardiology, Keio University School of Medicine, Shinanomachi 35, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshiki Shinya
- Department of Cardiology, Keio University School of Medicine, Shinanomachi 35, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Motoaki Sano
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinanomachi 35, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Junji Shibahara
- Department of Pathology, School of Medicine, Kyorin University, Mitaka, Japan
| | - Junko Kuramoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Yoji Hakamata
- Department of Basic Science, School of Veterinary Nursing and Technology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Masaharu Kataoka
- Department of Cardiology, Keio University School of Medicine, Shinanomachi 35, Shinjuku-ku, Tokyo, 160-8582, Japan.
- The Second Department of Internal Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| |
Collapse
|
2
|
Zhang H, Liu D, Xu QF, Wei J, Zhao Y, Xu DF, Wang Y, Liu YJ, Zhu XY, Jiang L. Endothelial RSPO3 mediates pulmonary endothelial regeneration by LGR4-dependent activation of β-catenin and ILK signaling pathways after inflammatory vascular injury. Int J Biol Macromol 2024; 269:131805. [PMID: 38677673 DOI: 10.1016/j.ijbiomac.2024.131805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Endothelial repair is essential for restoring tissue fluid homeostasis following lung injury. R-spondin3 (RSPO3), a secreted protein mainly produced by endothelial cells (ECs), has shown its protective effect on endothelium. However, the specific mechanisms remain unknown. To explore whether and how RSPO3 regulates endothelial regeneration after inflammatory vascular injury, the role of RSPO3 in sepsis-induced pulmonary endothelial injury was investigated in EC-specific RSPO3 knockdown, inducible EC-specific RSPO3 deletion mice, EC-specific RSPO3 overexpression mice, systemic RSPO3-administration mice, in isolated mouse lung vascular endothelial cells (MLVECs), and in plasma from septic patients. Here we show that plasma RSPO3 levels are decreased in septic patients and correlated with endothelial injury markers and PaO2/FiO2 index. Both pulmonary EC-specific knockdown of RSPO3 and inducible EC-specific RSPO3 deletion inhibit pulmonary ECs proliferation and exacerbate ECs injury, whereas intra-pulmonary EC-specific RSPO3 overexpression promotes endothelial recovery and attenuates ECs injury during endotoxemia. We show that RSPO3 mediates pulmonary endothelial regeneration by a LGR4-dependent manner. Except for β-catenin, integrin-linked kinase (ILK)/Akt is also identified as a novel downstream effector of RSPO3/LGR4 signaling. These results conclude that EC-derived RSPO3 mediates pulmonary endothelial regeneration by LGR4-dependent activation of β-catenin and ILK signaling pathways after inflammatory vascular injury.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China
| | - Di Liu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China
| | - Qing-Feng Xu
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai 200438, PR China
| | - Juan Wei
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai 200438, PR China
| | - Ying Zhao
- Department of Anesthesiology, Zhejiang Cancer Hospital, 310022, PR China
| | - Dun-Feng Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China
| | - Yu-Jian Liu
- School of Kinesiology, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, The Key Laboratory of Exercise and Health Sciences of Ministry of Education Shanghai University of Sport, Shanghai 200438, PR China
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, PR China.
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
3
|
Chen Y, Liu J, Zhang Q, Chai L, Chen H, Li D, Wang Y, Qiu Y, Shen N, Zhang J, Wang Q, Wang J, Xie X, Li S, Li M. Activation of CaMKII/HDAC4 by SDF1 contributes to pulmonary arterial hypertension via stabilization Runx2. Eur J Pharmacol 2024; 970:176483. [PMID: 38479721 DOI: 10.1016/j.ejphar.2024.176483] [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: 08/31/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/02/2024]
Abstract
Stromal derived factor 1 (SDF1) has been shown to be involved in the pathogenesis of pulmonary artery hypertension (PAH). However, the detailed molecular mechanisms remain unclear. To address this, we utilized primary cultured rat pulmonary artery smooth muscle cells (PASMCs) and monocrotaline (MCT)-induced PAH rat models to investigate the mechanisms of SDF1 driving PASMCs proliferation and pulmonary arterial remodeling. SDF1 increased runt-related transcription factor 2 (Runx2) acetylation by Calmodulin (CaM)-dependent protein kinase II (CaMKII)-dependent HDAC4 cytoplasmic translocation, elevation of Runx2 acetylation conferred its resistance to proteasome-mediated degradation. The accumulation of Runx2 further upregulated osteopontin (OPN) expression, finally leading to PASMCs proliferation. Blocking SDF1, suppression of CaMKII, inhibition the nuclear export of HDAC4 or silencing Runx2 attenuated pulmonary arterial remodeling and prevented PAH development in MCT-induced PAH rat models. Our study provides novel sights for SDF1 induction of PASMCs proliferation and suggests that targeting SDF1/CaMKII/HDAC4/Runx2 axis has potential value in the management of PAH.
Collapse
Affiliation(s)
- Yuqian Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
4
|
Newcomb G, Farkas L. Endothelial cell clonality, heterogeneity and dysfunction in pulmonary arterial hypertension. Front Med (Lausanne) 2023; 10:1304766. [PMID: 38126077 PMCID: PMC10731016 DOI: 10.3389/fmed.2023.1304766] [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: 09/29/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Our understanding of the pathophysiology of pulmonary arterial hypertension (PAH) has evolved over recent years, with the recognition that endothelial cell (EC) dysfunction and inflammation play an integral role in the development of this disease. ECs within the pulmonary vasculature play a unique role in maintaining vascular integrity and barrier function, regulating gas exchange, and contributing to vascular tone. Using single-cell transcriptomics, research has shown that there are multiple, unique EC subpopulations with different phenotypes. In response to injury or certain stressors such as hypoxia, there can be a dysregulated response with aberrant endothelial injury repair involving other pulmonary vascular cells and even immune cells. This aberrant signaling cascade is potentially a primary driver of pulmonary arterial remodeling in PAH. Recent studies have examined the role of EC clonal expansion, immune dysregulation, and genetic mutations in the pathogenesis of PAH. This review summarizes the existing literature on EC subpopulations and the intricate mechanisms through which ECs develop aberrant physiologic phenotypes and contribute to PAH. Our goal is to provide a framework for understanding the unique pulmonary EC biology and pathophysiology that is involved in the development of PAH.
Collapse
Affiliation(s)
- Geoffrey Newcomb
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Laszlo Farkas
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
5
|
Tan C, Norden PR, Yu W, Liu T, Ujiie N, Lee SK, Yan X, Dyakiv Y, Aoto K, Ortega S, De Plaen IG, Sampath V, Kume T. Endothelial FOXC1 and FOXC2 promote intestinal regeneration after ischemia-reperfusion injury. EMBO Rep 2023; 24:e56030. [PMID: 37154714 PMCID: PMC10328078 DOI: 10.15252/embr.202256030] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Pieter R Norden
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Wei Yu
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Naoto Ujiie
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Sun Kyong Lee
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Xiaocai Yan
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Kazushi Aoto
- Department of BiochemistryHamamatsu University School of MedicineHamamatsuJapan
| | - Sagrario Ortega
- Mouse Genome Editing Unit, Biotechnology ProgramSpanish National Cancer Research CentreMadridSpain
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Venkatesh Sampath
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
6
|
Jiang CY, Wu LW, Liu YW, Feng B, Ye LC, Huang X, He YY, Shen Y, Zhu YF, Zhou XL, Jiang DJ, Qi HK, Zhang H, Yan Y. Identification of ACKR4 as an immune checkpoint in pulmonary arterial hypertension. Front Immunol 2023; 14:1153573. [PMID: 37449198 PMCID: PMC10337759 DOI: 10.3389/fimmu.2023.1153573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Objective Inflammation is recognized as a contributor in the development of pulmonary arterial hypertension (PAH), and the recruitment and functional capacity of immune cells are well-orchestrated by chemokines and their receptors. This study is aimed at identification of critical chemokines in the progression of PAH via transcriptomic analysis. Methods Differentially expressed genes (DEGs) from lungs of PAH patients were achieved compared to controls based on Gene Expression Omnibus (GEO) database. Gene set enrichment analysis (GSEA) was applied for functional annotation and pathway enrichement. The abundance of immune cells was estimated by the xCell algorithm. Weighted correlation network analysis (WGCNA) was used to construct a gene expression network, based on which a diagnostic model was generated to determine its accuracy to distinguish PAH from control subjects. Target genes were then validated in lung of hypoxia-induce pulmonary hypertension (PH) mouse model. Results ACKR4 (atypical chemokine receptor 4) was downregulated in PAH lung tissues in multiple datasets. PAH relevant biological functions and pathways were enriched in patients with low-ACKR4 level according to GSEA enrichment analysis. Immuno-infiltration analysis revealed a negative correlation of activated dendritic cells, Th1 and macrophage infiltration with ACKR4 expression. Three gene modules were associated with PAH via WGCNA analysis, and a model for PAH diagnosis was generated using CXCL12, COL18A1 and TSHZ2, all of which correlated with ACKR4. The ACKR4 expression was also downregulated in lung tissues of our experimental PH mice compared to that of controls. Conclusions The reduction of ACKR4 in lung tissues of human PAH based on transcriptomic data is consistent with the alteration observed in our rodent PH. The correlation with immune cell infiltration and functional annotation indicated that ACKR4 might serve as a protective immune checkpoint for PAH.
Collapse
Affiliation(s)
- Chen-Yu Jiang
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Wei Wu
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi-Wei Liu
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei Feng
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin-Cai Ye
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Huang
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang-Yang He
- School of Pharmacy, Henan University, Kaifeng, Henan, China
| | - Yi Shen
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi-Fan Zhu
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing-Liang Zhou
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dai-Ji Jiang
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hai-Kun Qi
- School of Biomedical Engineering, Shanghaitech University, Shanghai, China
| | - Hao Zhang
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Yan
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center (SCMC), School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Shen Y, Dong Z, Fan F, Li K, Zhu S, Dai R, Huang J, Xie N, He L, Gong Z, Yang X, Tan J, Liu L, Yu F, Tang Y, You Z, Xi J, Wang Y, Kong W, Zhang Y, Fu Y. Targeting cytokine-like protein FAM3D lowers blood pressure in hypertension. Cell Rep Med 2023:101072. [PMID: 37301198 DOI: 10.1016/j.xcrm.2023.101072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 03/08/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Current antihypertensive options still incompletely control blood pressure, suggesting the existence of uncovered pathogenic mechanisms. Here, whether cytokine-like protein family with sequence similarity 3, member D (FAM3D) is involved in hypertension etiology is evaluated. A case-control study exhibits that FAM3D is elevated in patients with hypertension, with a positive association with odds of hypertension. FAM3D deficiency significantly ameliorates angiotensin II (AngII)-induced hypertension in mice. Mechanistically, FAM3D directly causes endothelial nitric oxide synthase (eNOS) uncoupling and impairs endothelium-dependent vasorelaxation, whereas 2,4-diamino-6-hydroxypyrimidine to induce eNOS uncoupling abolishes the protective effect of FAM3D deficiency against AngII-induced hypertension. Furthermore, antagonism of formyl peptide receptor 1 (FPR1) and FPR2 or the suppression of oxidative stress blunts FAM3D-induced eNOS uncoupling. Translationally, targeting endothelial FAM3D by adeno-associated virus or intraperitoneal injection of FAM3D-neutralizing antibodies markedly ameliorates AngII- or deoxycorticosterone acetate (DOCA)-salt-induced hypertension. Conclusively, FAM3D causes eNOS uncoupling through FPR1- and FPR2-mediated oxidative stress, thereby exacerbating the development of hypertension. FAM3D may be a potential therapeutic target for hypertension.
Collapse
Affiliation(s)
- Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Zhigang Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Fangfang Fan
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Cardiology, Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China
| | - Kaiyin Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Cardiology, Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China
| | - Shirong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Rongbo Dai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Nan Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Guangdong 518057, China
| | - Li He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Xueyuan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiaai Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Limei Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yida Tang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Zhen You
- Department of Biliary Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianzhong Xi
- Department of Biomedicine, College of Engineering, Peking University, Beijing 100871, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, and Key Laboratory of Medical Immunology of Ministry of Health, Peking University, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China.
| | - Yan Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Cardiology, Institute of Cardiovascular Disease, Peking University First Hospital, Beijing 100034, China.
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China.
| |
Collapse
|
8
|
Wang S, Wang M, Peng H, Tian Y, Guo H, Wang J, Yu H, Xue E, Chen X, Wang X, Fan M, Zhang Y, Wang X, Qin X, Wu Y, Li J, Ye Y, Chen D, Hu Y, Wu T. Synergism of cell adhesion regulatory genes and instant air pollutants on blood pressure elevation. CHEMOSPHERE 2023; 312:136992. [PMID: 36334751 DOI: 10.1016/j.chemosphere.2022.136992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Accumulating evidence suggests that an instant exposure to particulate matter (PM) may elevate blood pressure (BP), where cell-adhesion regulatory genes may be involved in the interplay. However, few studies to date critically examined their interaction, and it remained unclear whether these genes modified the association. To assess the association between instant PM exposure and BP, and to examine whether single-nucleotide polymorphisms (SNPs) mapped in four cell adhesion regulatory genes modify the relationship, a cross-sectional study was performed, based on the baseline of an ongoing family-based cohort in Beijing, China. A total of 4418 persons from 2089 families in Northern China were included in the analysis. Four tagged SNPs in cell adhesion regulatory genes were selected among ZFHX3, CXCL12, RASGRP1 and MIR146A. A generalized additive model (GAM) with a Gaussian link was adopted to estimate the change in blood pressure after instant PM2.5 or PM10 exposure. A cross-product term of PM2.5/PM10 and genotype was incorporated into the GAM model to test for interaction. The study observed that an instant exposure to either PM2.5 or PM10 was found to be associated with elevated systolic blood pressure (SBP). On average, a 10 μg/m3 increase in instant exposure to PM2.5 and PM10 concentration corresponded to 0.140% (95% CI: 0.014%-0.265%, P = 0.029) and 0.173% (95% CI: 0.080%-0.266%, P < 0.001) higher SBP. However, diastolic blood pressure (DBP) was not elevated as the PM2.5 or PM10 concentration increased (P > 0.05). A synergetic interaction on SBP was observed between SNPs in four cell adhesion regulatory genes (rs2910164 in MIR146A, rs2297630 in CXCL12, rs7403531 in RASGRP1, and rs7193343 in ZFHX3) and instant PM2.5 exposure (Pfor interaction <0.05). Briefly, as carriers of risk alleles in each of these four genes increased, an enhanced association was found between instant PM2.5 exposure and SBP.
Collapse
Affiliation(s)
- Siyue Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Mengying Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Hexiang Peng
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Yaohua Tian
- Department of Maternal and Child Health, School of Public Health, Huazhong University of Science and Technology, 430030, China
| | - Huangda Guo
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Jiating Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Huan Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Enci Xue
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Xi Chen
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Xueheng Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Meng Fan
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Yi Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Xiaochen Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Xueying Qin
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Yiqun Wu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Jin Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Ying Ye
- Department of Local Diseases Control and Prevention, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, 350001, China
| | - Dafang Chen
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Yonghua Hu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
| | - Tao Wu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, 100191, China; Institute of Reproductive and Child Health/Key Laboratory of Reproductive Health, National Health Commission of the People's China.
| |
Collapse
|
9
|
Cantu A, Gutierrez MC, Dong X, Leek C, Sajti E, Lingappan K. Remarkable sex-specific differences at single-cell resolution in neonatal hyperoxic lung injury. Am J Physiol Lung Cell Mol Physiol 2023; 324:L5-L31. [PMID: 36283964 PMCID: PMC9799156 DOI: 10.1152/ajplung.00269.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 01/12/2023] Open
Abstract
Exposure to supraphysiological concentrations of oxygen (hyperoxia) predisposes to bronchopulmonary dysplasia (BPD), which is characterized by abnormal alveolarization and pulmonary vascular development, in preterm neonates. Neonatal hyperoxia exposure is used to recapitulate the phenotype of human BPD in murine models. Male sex is considered an independent predictor for the development of BPD, but the main mechanisms underlying sexually dimorphic outcomes are unknown. Our objective was to investigate sex-specific and cell-type specific transcriptional changes that drive injury in the neonatal lung exposed to hyperoxia at single-cell resolution and delineate the changes in cell-cell communication networks in the developing lung. We used single-cell RNA sequencing (scRNAseq) to generate transcriptional profiles of >35,000 cells isolated from the lungs of neonatal male and female C57BL/6 mice exposed to 95% [Formula: see text] between PND1-5 (saccular stage of lung development) or normoxia and euthanized at PND7 (alveolar stage of lung development). ScRNAseq identified 22 cell clusters with distinct populations of endothelial, epithelial, mesenchymal, and immune cells. Our data identified that the distal lung vascular endothelium (composed of aerocytes and general capillary endothelial cells) is exquisitely sensitive to hyperoxia exposure with the emergence of an intermediate capillary endothelial population with both general capillaries (gCap) and aerocytes or alveolar capillaries (aCap) markers. We also identified a myeloid-derived suppressor cell population from the lung neutrophils. Sex-specific differences were evident in all lung cell subpopulations but were striking among the lung immune cells. Finally, we identified that the specific intercellular communication networks and the ligand-receptor pairs that are impacted by neonatal hyperoxia exposure.
Collapse
Affiliation(s)
- Abiud Cantu
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Manuel C Gutierrez
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Xiaoyu Dong
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Connor Leek
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eniko Sajti
- Department of Pediatrics, University of California, La Jolla, California
| | - Krithika Lingappan
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Luo X, Hang C, Zhang Z, Le K, Ying Y, Lv Y, Yan L, Huang Y, Ye L, Xu X, Zhong Y, Du L. PVECs-Derived Exosomal microRNAs Regulate PASMCs via FoxM1 Signaling in IUGR-induced Pulmonary Hypertension. J Am Heart Assoc 2022; 11:e027177. [PMID: 36533591 PMCID: PMC9798821 DOI: 10.1161/jaha.122.027177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Intrauterine growth restriction (IUGR) is closely related to systemic or pulmonary hypertension (PH) in adulthood. Aberrant crosstalk between pulmonary vascular endothelial cells (PVECs) and pulmonary arterial smooth muscle cells (PASMCs) that is mediated by exosomes plays an essential role in the progression of PH. FoxM1 (Forkhead box M1) is a key transcription factor that governs many important biological processes. Methods and Results IUGR-induced PH rat models were established. Transwell plates were used to coculture PVECs and PASMCs. Exosomes were isolated from PVEC-derived medium, and a microRNA (miRNA) screening was proceeded to identify effects of IUGR on small RNAs enclosed within exosomes. Dual-Luciferase assay was performed to validate the predicted binding sites of miRNAs on FoxM1 3' untranslated region. FoxM1 inhibitor thiostrepton was used in IUGR-induced PH rats. In this study, we found that FoxM1 expression was remarkably increased in IUGR-induced PH, and PASMCs were regulated by PVECs through FoxM1 signaling in a non-contact way. An miRNA screening showed that miR-214-3p, miR-326-3p, and miR-125b-2-3p were downregulated in PVEC-derived exosomes of the IUGR group, which were associated with overexpression of FoxM1 and more significant proliferation and migration of PASMCs. Dual-Luciferase assay demonstrated that the 3 miRNAs directly targeted FoxM1 3' untranslated region. FoxM1 inhibition blocked the PVECs-PASMCs crosstalk and reversed the abnormal functions of PASMCs. In vivo, treatment with thiostrepton significantly reduced the severity of PH. Conclusions Transmission of exosomal miRNAs from PVECs regulated the functions of PASMCs via FoxM1 signaling, and FoxM1 may serve as a potential therapeutic target in IUGR-induced PH.
Collapse
Affiliation(s)
- Xiaofei Luo
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Chengcheng Hang
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Ziming Zhang
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Kaixing Le
- Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Yuhan Ying
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Ying Lv
- Department of Pediatric Health Care, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Lingling Yan
- Department of Pediatrics, The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvincePeople’s Republic of China
| | - Yajie Huang
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Lixia Ye
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Xuefeng Xu
- Department of Rheumatology Immunology & Allergy, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Ying Zhong
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| | - Lizhong Du
- Department of Neonatology, The Children’s HospitalZhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouZhejiang ProvincePeople’s Republic of China
| |
Collapse
|
11
|
Liu B, Peng Y, Yi D, Machireddy N, Dong D, Ramirez K, Dai J, Vanderpool R, Zhu MM, Dai Z, Zhao YY. Endothelial PHD2 deficiency induces nitrative stress via suppression of caveolin-1 in pulmonary hypertension. Eur Respir J 2022; 60:2102643. [PMID: 35798360 PMCID: PMC9791795 DOI: 10.1183/13993003.02643-2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nitrative stress is a characteristic feature of the pathology of human pulmonary arterial hypertension. However, the role of nitrative stress in the pathogenesis of obliterative vascular remodelling and severe pulmonary arterial hypertension remains largely unclear. METHOD Our recently identified novel mouse model (Egln1Tie2Cre, Egln1 encoding prolyl hydroxylase 2 (PHD2)) has obliterative vascular remodelling and right heart failure, making it an excellent model to use in this study to examine the role of nitrative stress in obliterative vascular remodelling. RESULTS Nitrative stress was markedly elevated whereas endothelial caveolin-1 (Cav1) expression was suppressed in the lungs of Egln1Tie2Cre mice. Treatment with a superoxide dismutase mimetic, manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride or endothelial Nos3 knockdown using endothelial cell-targeted nanoparticle delivery of CRISPR-Cas9/guide RNA plasmid DNA inhibited obliterative pulmonary vascular remodelling and attenuated severe pulmonary hypertension in Egln1Tie2Cre mice. Genetic restoration of Cav1 expression in Egln1Tie2Cre mice normalised nitrative stress, reduced pulmonary hypertension and improved right heart function. CONCLUSION These data suggest that suppression of Cav1 expression secondary to PHD2 deficiency augments nitrative stress through endothelial nitric oxide synthase activation, which contributes to obliterative vascular remodelling and severe pulmonary hypertension. Thus, a reactive oxygen/nitrogen species scavenger might have therapeutic potential for the inhibition of obliterative vascular remodelling and severe pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Yi Peng
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Narsa Machireddy
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daoyin Dong
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Jingbo Dai
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rebecca Vanderpool
- College of Medicine Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Maggie M Zhu
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, Dept of Internal Medicine, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Critical Care, Dept of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Dept of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Zhiyu Dai and You-Yang Zhao contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
12
|
Chen X, He Y, Yu Z, Zuo J, Huang Y, Ruan Y, Zheng X, Ma Y. Polydatin Glycosides Improve Monocrotaline-Induced Pulmonary Hypertension Injury by Inhibiting Endothelial-To-Mesenchymal Transition. Front Pharmacol 2022; 13:862017. [PMID: 35370672 PMCID: PMC8972160 DOI: 10.3389/fphar.2022.862017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: To study the effect of polydatin on the injury of pulmonary arterial hypertension (PAH) induced by monocrotaline (MCT).Methods: SD rats were induced to develop PAH injury by a single subcutaneous injection of MCT (60 mg/kg). From the second day, rats in the administration group were orally given sildenafil (20 mg/kg) and polydatin (30 or 60 mg/kg) for 3 weeks. At the end of the experiment, right ventricular hypertrophy (RVH) index of SD rats was calculated, pathological damage was assessed by HE staining, transcription levels of target genes were detected by RT-PCR and Elisa, and expression levels of Endothelial-to-mesenchymal transition (EndMT) related proteins were detected by immunohistochemistry (IHC) and immunofluorescence (IF). Finally, molecular docking analysis was used to verify the interaction of polydatin on the main targets.Results: Polydatin could significantly restore the body function, reduce MCT-induced PAH injury, reduce serum biochemical indices; polydatin could effectively inhibit EndMT process by decreasing the expression of N-cadherin, β-catenin and vimentin; polydatin could down-regulate TAGLN expression and increase PECAM1 expression to reduce pulmonary vascular remodeling. The interaction between polydatin and EndMT target was confirmed by molecular docking operation.Conclusion: Pharmacological experiments combined with Combining molecular docking was first used to clarify that polydatin can reduce the pulmonary endothelial dysfunction and pulmonary vascular remodeling induced by MCT by inhibiting EndMT. The results of the study provide new ideas for the further treatment of PAH injury.
Collapse
Affiliation(s)
- Xing Chen
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
- *Correspondence: Xing Chen, ; Xiaoyuan Zheng, ; Yu Ma,
| | - Yao He
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
| | - Zhijie Yu
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
| | - Jianli Zuo
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
| | - Yan Huang
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
| | - Yi Ruan
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
| | - Xiaoyuan Zheng
- Pharmacy Department, Chongqing Emergency Medical Center, Chongqing, China
- Pharmacy Department, Chongqing University Central Hospital, Chongqing, China
- *Correspondence: Xing Chen, ; Xiaoyuan Zheng, ; Yu Ma,
| | - Yu Ma
- Chongqing Emergency Medical Center, Chongqing, China
- *Correspondence: Xing Chen, ; Xiaoyuan Zheng, ; Yu Ma,
| |
Collapse
|
13
|
Abstract
Pulmonary hypertension is an enigmatic, deleterious disease driven by multiple heterogeneous causes with a burgeoning proportion of older patients with complex, chronic comorbidities without adequate treatment options. The underlying endothelial pathophenotypes that direct vasoconstriction and panvascular remodeling remain both controversial and incompletely defined. This review discusses emerging concepts centered on endothelial senescence in pulmonary vascular disease. This principle proposes a more heterogeneous, dynamic pulmonary endothelium in disease; it provides a potentially unifying feature of endothelial dysfunction in pulmonary hypertension irrespective of cause; and it supports a clinically relevant link between aging and pulmonary hypertension like other chronic illnesses. Thus, taking cues from studies on aging and age-related diseases, we present possible opportunities and barriers to diagnostic and therapeutic targeting of senescence in pulmonary hypertension.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA
| |
Collapse
|
14
|
Liu B, Yi D, Pan J, Dai J, Zhu MM, Zhao Y, Oh SP, Fallon MB, Dai Z. Suppression of BMP signaling by PHD2 deficiency in Pulmonary Arterial hypertension. Pulm Circ 2022; 12:e12056. [PMID: 35506101 PMCID: PMC9052986 DOI: 10.1002/pul2.12056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/11/2022] Open
Abstract
BMP signaling deficiency is evident in the lungs of patients with pulmonary arterial hypertension. We demonstrated that PHD2 deficiency suppresses BMP signaling in the lung endothelial cells, suggesting the novel mechanisms of dysregulated BMP signaling in the development of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Jiakai Pan
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Jingbo Dai
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Maggie M. Zhu
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - You‐Yang Zhao
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PharmacologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - S. Paul Oh
- Department of Neurobiology, Barrow Aneurysm and AVM Research CenterBarrow Neurological InstitutePhoenixArizonaUSA
| | - Michael B. Fallon
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| |
Collapse
|
15
|
Thomas S, Manivannan S, Garg V, Lilly B. Single-Cell RNA Sequencing Reveals Novel Genes Regulated by Hypoxia in the Lung Vasculature. J Vasc Res 2022; 59:163-175. [PMID: 35294950 PMCID: PMC9117417 DOI: 10.1159/000522340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/25/2022] [Indexed: 11/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic progressive disease with significant morbidity and mortality. The disease is characterized by vascular remodeling that includes increased muscularization of distal blood vessels and vessel stiffening associated with changes in extracellular matrix deposition. In humans, chronic hypoxia causes PAH, and hypoxia-induced rodent models of PAH have been used for years to study the disease. With the development of single-cell RNA sequencing technology, it is now possible to examine hypoxia-dependent transcriptional changes in vivo at a cell-specific level. In this study, we used single-cell RNA sequencing to compare lungs from wild-type (Wt) mice exposed to hypoxia for 28 days to normoxia-treated control mice. We additionally examined mice deficient for Notch3, a smooth muscle-enriched gene linked to PAH. Data analysis revealed that hypoxia promoted cell number changes in immune and endothelial cell types in the lung, activated the innate immunity pathway, and resulted in specific changes in gene expression in vascular cells. Surprisingly, we found limited differences in lungs from mice deficient for Notch3 compared to Wt controls. These findings provide novel insight into the effects of chronic hypoxia exposure on gene expression and cell phenotypes in vivo and identify unique changes to cells of the vasculature.
Collapse
Affiliation(s)
- Shelby Thomas
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|