1
|
Diao RX, Lv WY, Wang YC, Shen QL, Tang KH, Luo XX, Jin YY. Aquaporin-1 Facilitates Macrophage M1 Polarization by Enhancing Glycolysis Through the Activation of HIF1α in Lipopolysaccharide-Induced Acute Kidney Injury. Inflammation 2024:10.1007/s10753-024-02154-8. [PMID: 39365391 DOI: 10.1007/s10753-024-02154-8] [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: 05/21/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
This study aimed to investigate how aquaporin 1 (AQP1) modulates hypoxia-inducible factor-1α (HIF1α) to promote glycolysis and drive the M1 polarization of macrophages. Within 12 h post-treatment with LPS to induce acute kidney injury in rats, a significant upregulation of AQP1 and HIF1α protein levels was noted in serum and kidney tissues. This elevation corresponded with a decrease in blood glucose concentrations and an enhancement of glycolytic activity relative to the control group. Furthermore, there was a pronounced reduction in the circulating levels of the anti-inflammatory cytokine IL-10, accompanied by an upregulation in the levels of the pro-inflammatory cytokines IL-6 and TNF-α. The administration of an HIF1α inhibitor reversed these effects, which did not affect the production of AQP1 protein. In cellular assays, AQP1 knockdown mitigated the increase in HIF1α expression induced by LPS. Furthermore, the suppression of HIF1α with PX-478 led to decreased expression levels of Hexokinase 2 (HK2) and Lactate Dehydrogenase A (LDHA), indicating that AQP1 regulates glycolysis through HIF1α. M1 polarization of macrophages was reduced by AQP1 knockdown and was further diminished by the addition of an HIF1α inhibitor. Inhibition of the glycolytic process not only weakened M1 polarization but also promoted M2 polarization, thereby reducing the release of inflammatory cytokines. These findings provide a novel perspective for developing therapeutic strategies that target AQP1 and HIF1α, potentially improving the treatment of sepsis-associated AKI.
Collapse
Affiliation(s)
- Ru-Xue Diao
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wu-Yang Lv
- Department of Clinical Laboratory, Shangluo Central Hospital, Shanxi, China
| | - Yu-Chen Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiu-Ling Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai-Hong Tang
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao-Xiao Luo
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying-Yu Jin
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Lv W, Liao J, Li C, Liu D, Luo X, Diao R, Wang Y, Jin Y. Aquaporin 1 is renoprotective in septic acute kidney injury by attenuating inflammation, apoptosis and fibrosis through inhibition of P53 expression. Front Immunol 2024; 15:1443108. [PMID: 39238634 PMCID: PMC11374652 DOI: 10.3389/fimmu.2024.1443108] [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: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
Sepsis associated Acute kidney injury (AKI) is a common clinical syndrome characterized by suddenly decreased in renal function and urinary volume. This study was designed to investigate the role of Aquaporin 1 (AQP1) and P53 in the development of sepsis-induced AKI and their potential regulatory mechanisms. Firstly, transcriptome sequencing analysis of mice kidney showed AQP1 expression was reduced and P53 expression was elevated in Cecal ligation and puncture (CLP)-induced AKI compared with controls. Bioinformatics confirmed that AQP1 expression was remarkably decreased and P53 expression was obviously elevated in renal tissues or peripheral blood of septic AKI patients. Moreover, we found in vivo experiments that AQP1 mRNA levels were dramatically decreased and P53 mRNA significantly increased following the increased expression of inflammation, apoptosis, fibrosis, NGAL and KIM-1 at various periods in septic AKI. Meanwhile, AQP1 and P53 protein levels increased significantly first and then decreased gradually in kidney tissue and serum of rats in different stages of septic AKI. Most importantly, in vivo and vitro experiments demonstrated that silencing of AQP1 greatly exacerbates renal or cellular injury by up-regulating P53 expression promoting inflammatory response, apoptosis and fibrosis. Overexpression of AQP1 prevented the elevation of inflammation, apoptosis and fibrosis by down-regulating P53 expression in Lipopolysaccharide (LPS)-induced AKI or HK-2 cells. Therefore, our results suggested that AQP1 plays a protective role in modulating AKI and can attenuate inflammatory response, apoptosis and fibrosis via downregulating P53 in septic AKI or LPS-induced HK-2cells. The pharmacological targeting of AQP1 mediated P53 expression might be identified as potential targets for the early treatment of septic AKI.
Collapse
Affiliation(s)
- Wuyang Lv
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Clinical Laboratory, Shangluo Central Hospital, Shangluo, Shaanxi, China
| | - Jia Liao
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Cuicui Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongyang Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoxiao Luo
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - RuXue Diao
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - YuChen Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingyu Jin
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Yun X, Niedermeyer S, Andrade MR, Jiang H, Suresh K, Kolb T, Damarla M, Shimoda LA. Aquaporin 1 confers apoptosis resistance in pulmonary arterial smooth muscle cells from the SU5416 hypoxia rat model. Physiol Rep 2024; 12:e16156. [PMID: 39175041 PMCID: PMC11341275 DOI: 10.14814/phy2.16156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 08/24/2024] Open
Abstract
Pulmonary hypertension (PH) arises from increased pulmonary vascular resistance due to contraction and remodeling of the pulmonary arteries. The structural changes include thickening of the smooth muscle layer from increased proliferation and resistance to apoptosis. The mechanisms underlying apoptosis resistance in PH are not fully understood. In cancer cells, high expression of aquaporin 1 (AQP1), a water channel, is associated with apoptosis resistance. We showed AQP1 protein was expressed in pulmonary arterial smooth muscle cells (PASMCs) and upregulated in preclinical PH models. In this study, we used PASMCs isolated from control male rats and the SU5416 plus hypoxia (SuHx) model to test the role of AQP1 in modulating susceptibility to apoptosis. We found the elevated level of AQP1 in PASMCs from SuHx rats was necessary for resistance to apoptosis and that apoptosis resistance could be conferred by increasing AQP1 in control PASMCs. In exploring the downstream pathways involved, we found AQP1 levels influence the expression of Bcl-2, with enhanced AQP1 levels corresponding to increased Bcl-2 expression, reducing the ratio of BAX to Bcl-2, consistent with apoptosis resistance. These results provide a mechanism by which AQP1 can regulate PASMC fate.
Collapse
MESH Headings
- Animals
- Aquaporin 1/metabolism
- Aquaporin 1/genetics
- Apoptosis
- Male
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/cytology
- Rats
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Pyrroles/pharmacology
- Indoles/pharmacology
- Hypoxia/metabolism
- Rats, Sprague-Dawley
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Cells, Cultured
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Disease Models, Animal
Collapse
Affiliation(s)
- Xin Yun
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Shannon Niedermeyer
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Manuella Ribas Andrade
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Karthik Suresh
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Todd Kolb
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care MedicineJohns Hopkins School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
4
|
Agarwal S, Fineman J, Cornfield DN, Alvira CM, Zamanian RT, Goss K, Yuan K, Bonnet S, Boucherat O, Pullamsetti S, Alcázar MA, Goncharova E, Kudryashova TV, Nicolls MR, de Jesús Pérez V. Seeing pulmonary hypertension through a paediatric lens: a viewpoint. Eur Respir J 2024; 63:2301518. [PMID: 38575157 PMCID: PMC11187317 DOI: 10.1183/13993003.01518-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024]
Abstract
Pulmonary hypertension (PH) is a life-threating condition associated with abnormally elevated pulmonary pressures and right heart failure. Current epidemiological data indicate that PH aetiologies are different between the adult and paediatric population. The most common forms of PH in adults are PH from left heart disease or chronic lung disease, followed by pulmonary arterial hypertension (PAH) [1]; in paediatric patients, PH is most often associated with developmental lung disorders and congenital heart disease (CHD) [2, 3]. In contrast to adults with PH, wherein patients worsen over time despite therapy, PH in children can improve with growth. For example, in infants with bronchopulmonary dysplasia (BPD) and PH morbidity and mortality are high, but with lung growth and ensuring no ongoing lung injury pulmonary vascular disease can improve as evidenced by discontinuation of vasodilator therapy in almost two-thirds of BPD-PH survivors by age 5 years [3, 4]. Paediatric pulmonary hypertension (PH) offers unique genetic and developmental insights that can help in the discovery of novel mechanisms and targets to treat adult PH https://bit.ly/3TMm6bi
Collapse
Affiliation(s)
- Stuti Agarwal
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Jeffrey Fineman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - David N Cornfield
- Division of Pediatric Pulmonary, Asthma, and Sleep Medicine, Stanford University, Palo Alto, CA, USA
| | - Cristina M Alvira
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - Roham T Zamanian
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Kara Goss
- Department of Medicine and Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Ke Yuan
- Boston Children's Hospital, Boston, MA, USA
| | - Sebastien Bonnet
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Olivier Boucherat
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Soni Pullamsetti
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | - Tatiana V Kudryashova
- University of Pittsburgh Heart, Blood, and Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
5
|
Niedermeyer S, Yun X, Trujillo M, Jiang H, Andrade MR, Kolb TM, Suresh K, Damarla M, Shimoda LA. A novel interaction between aquaporin 1 and caspase-3 in pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2024; 326:L638-L645. [PMID: 38375595 PMCID: PMC11380937 DOI: 10.1152/ajplung.00017.2024] [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: 01/15/2024] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024] Open
Abstract
Pulmonary hypertension (PH) is a condition in which remodeling of the pulmonary vasculature leads to hypertrophy of the muscular vascular wall and extension of muscle into nonmuscular arteries. These pathological changes are predominantly due to the abnormal proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), enhanced cellular functions that have been linked to increases in the cell membrane protein aquaporin 1 (AQP1). However, the mechanisms underlying the increased AQP1 abundance have not been fully elucidated. Here we present data that establishes a novel interaction between AQP1 and the proteolytic enzyme caspase-3. In silico analysis of the AQP1 protein reveals two caspase-3 cleavage sites on its C-terminal tail, proximal to known ubiquitin sites. Using biotin proximity ligase techniques, we establish that AQP1 and caspase-3 interact in both human embryonic kidney (HEK) 293A cells and rat PASMCs. Furthermore, we demonstrate that AQP1 levels increase and decrease with enhanced caspase-3 activity and inhibition, respectively. Ultimately, further work characterizing this interaction could provide the foundation for novel PH therapeutics.NEW & NOTEWORTHY Pulmonary arterial smooth muscle cells (PASMCs) are integral to pulmonary vascular remodeling, a characteristic of pulmonary arterial hypertension (PAH). PASMCs isolated from robust animal models of disease demonstrate enhanced proliferation and migration, pathological functions associated with increased abundance of the membrane protein aquaporin 1 (AQP1). We present evidence of a novel interaction between the proteolytic enzyme caspase-3 and AQP1, which may control AQP1 abundance. These data suggest a potential new target for novel PAH therapies.
Collapse
Affiliation(s)
- Shannon Niedermeyer
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Marielena Trujillo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Manuella R Andrade
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Todd M Kolb
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
6
|
Cheng K, Pan J, Liu Q, Ji Y, Liu L, Guo X, Wang Q, Li S, Sun J, Gong M, Zhang Y, Yuan Y. Exosomal lncRNA XIST promotes perineural invasion of pancreatic cancer cells via miR-211-5p/GDNF. Oncogene 2024; 43:1341-1352. [PMID: 38454138 DOI: 10.1038/s41388-024-02994-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Perineural invasion (PNI) is an essential form of tumor metastasis in multiple malignant cancers, such as pancreatic cancer, prostate cancer, and head and neck cancer. Growing evidence has revealed that pancreatic cancer recurrence and neuropathic pain positively correlate with PNI. Therefore, targeting PNI is a proper strategy for pancreatic cancer treatment. Exosomal lncRNA derived from pancreatic cancer cells is an essential component of the tumor microenvironment. However, whether exosomal lncXIST derived from pancreatic cancer cells can promote PNI and its exact mechanism remains to be elucidated. We show that lncXIST mediates nerve-tumor crosstalk via exosomal delivery. Our data reveal that exosomal lncXIST derived from pancreatic cancer cells is delivered to neural cells and promotes their release of glial-cell-line-derived neurotrophic factor (GDNF), essential in facilitating the PNI of pancreatic cancer. Mechanistically, microRNA-211-5p negatively regulates GDNF, and lncXIST serves as a miR-211-5p sponge. The function of exosomes in the dynamic interplay between nerves and cancer is confirmed in both in vivo and in vitro PNI models. Therefore, targeting pancreatic cancer cell-derived exosomal lncXIST may provide clues for a promising approach for developing a new strategy to combat PNI of pancreatic cancer.
Collapse
Affiliation(s)
- Ke Cheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Qinlong Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Yuke Ji
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Xiangqian Guo
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 47500, China
| | - Qiang Wang
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Software, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, 47500, China
| | - Shao Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jinyue Sun
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Miaomiao Gong
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| | - Ying Zhang
- Sixth Department of liver disease, Dalian Public Health Clinical Center, Dalian, 116044, China.
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
7
|
彭 威, 张 泽, 肖 云. [Research progress on bioinformatics in pulmonary arterial hypertension]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:425-431. [PMID: 38660909 PMCID: PMC11057300 DOI: 10.7499/j.issn.1008-8830.2310076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/26/2024] [Indexed: 04/26/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease characterized by abnormal pulmonary vascular remodeling and increased right ventricular pressure load, posing a significant threat to patient health. While some pathological mechanisms of PAH have been revealed, the deeper mechanisms of pathogenesis remain to be elucidated. In recent years, bioinformatics has provided a powerful tool for a deeper understanding of the complex mechanisms of PAH through the integration of techniques such as multi-omics analysis, artificial intelligence, and Mendelian randomization. This review focuses on the bioinformatics methods and technologies used in PAH research, summarizing their current applications in the study of disease mechanisms, diagnosis, and prognosis assessment. Additionally, it analyzes the existing challenges faced by bioinformatics and its potential applications in the clinical and basic research fields of PAH in the future.
Collapse
Affiliation(s)
| | - 泽盈 张
- 中南大学湘雅二医院心血管内科,湖南长沙410007
| | | |
Collapse
|
8
|
Welch CL, Aldred MA, Balachandar S, Dooijes D, Eichstaedt CA, Gräf S, Houweling AC, Machado RD, Pandya D, Prapa M, Shaukat M, Southgate L, Tenorio-Castano J, Chung WK. Defining the clinical validity of genes reported to cause pulmonary arterial hypertension. Genet Med 2023; 25:100925. [PMID: 37422716 PMCID: PMC10766870 DOI: 10.1016/j.gim.2023.100925] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Srimmitha Balachandar
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- NIHR BioResource for Translational Research - Rare Diseases, Department of Haemotology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Divya Pandya
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Matina Prapa
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Memoona Shaukat
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Jair Tenorio-Castano
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IDiPAZ, Universidad Autonoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; ITHACA, European Reference Network, Brussels, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY; Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
9
|
Hu Y, Zhao Y, Li P, Lu H, Li H, Ge J. Hypoxia and panvascular diseases: exploring the role of hypoxia-inducible factors in vascular smooth muscle cells under panvascular pathologies. Sci Bull (Beijing) 2023; 68:1954-1974. [PMID: 37541793 DOI: 10.1016/j.scib.2023.07.032] [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: 04/25/2023] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
As an emerging discipline, panvascular diseases are a set of vascular diseases with atherosclerosis as the common pathogenic hallmark, which mostly affect vital organs like the heart, brain, kidney, and limbs. As the major responser to the most common stressor in the vasculature (hypoxia)-hypoxia-inducible factors (HIFs), and the primary regulator of pressure and oxygen delivery in the vasculature-vascular smooth muscle cells (VSMCs), their own multifaceted nature and their interactions with each other are fascinating. Abnormally active VSMCs (e.g., atherosclerosis, pulmonary hypertension) or abnormally dysfunctional VSMCs (e.g., aneurysms, vascular calcification) are associated with HIFs. These widespread systemic diseases also reflect the interdisciplinary nature of panvascular medicine. Moreover, given the comparable proliferative characteristics exhibited by VSMCs and cancer cells, and the delicate equilibrium between angiogenesis and cancer progression, there is a pressing need for more accurate modulation targets or combination approaches to bolster the effectiveness of HIF targeting therapies. Based on the aforementioned content, this review primarily focused on the significance of integrating the overall and local perspectives, as well as temporal and spatial balance, in the context of the HIF signaling pathway in VSMC-related panvascular diseases. Furthermore, the review discussed the implications of HIF-targeting drugs on panvascular disorders, while considering the trade-offs involved.
Collapse
Affiliation(s)
- Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yongchao Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Peng Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Hua Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
10
|
Lv W, Liang L, Liu D, Li C, Jia L, Jin Y. Aquaporin 1 Facilitates Ferroptosis, M1 Polarization, Mitochondrial Dysfunction, and Autophagy Damage on Lipopolysaccharide-Induced Macrophage Through Down-Regulation of P53 Signaling Pathway. DNA Cell Biol 2023; 42:456-480. [PMID: 37379471 PMCID: PMC10457627 DOI: 10.1089/dna.2023.0016] [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: 01/20/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/30/2023] Open
Abstract
This study was designed to investigate the role of aquaporin 1 (AQP1) in ferroptosis, macrophage polarization, mitochondrial dysfunction, and impaired autophagy of lipopolysaccharide (LPS)-stimulated RAW264.7 cells and explored the underlying mechanisms. Si-AQP1-mediated AQP1 silencing RAW264.7 cells was constructed. Si-P53-mediated P53 silencing or pcDNA-P53 overexpression RAW264.7 cells was constructed. Assays of ATP, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Mitochondrial membrane potential (JC-1) staining were performed to evaluate mitochondrial biological function. Assays of flow cytometry, reactive oxygen species (ROS) staining, western blot (WB), RT-qPCR, malondialdehyde (MDA), glutathione (GSH), and total superoxide dismutase (SOD) were performed to detect cell ferroptosis, macrophage polarization, and impaired autophagy. The involvement of the P53 pathway was revealed by WB. The results showed that LPS (30 μg/mL) could induce ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage in RAW264.7 cells. Meanwhile, the expression of AQP1 was increased and the expression of P53 was decreased. In addition, Pifithrin-α (PIF; 15 μM), a P53 inhibitor, significantly aggravated ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage as well as up-regulation of AQP1 protein expression in LPS-induced RAW264.7 cells. Interestingly, this phenomenon was markedly alleviated by Kevetrin hydrochloride (70 μM), a P53 agonist. Mechanistically, silencing AQP1 significantly alleviated ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy damage by up-regulating the expression of P53 in LPS-stimulated RAW264.7 cells. Indeed, inhibition of P53 expression by PIF treatment dramatically reversed this effect on the basis of LPS+si-AQP1. Therefore, we concluded for the first time that AQP1 can promote ferroptosis, M1 polarization, mitochondrial dysfunction, and autophagy impairment by inhibiting the expression of P53 in LPS-stimulated RAW264.7 cells, and AQP1 or P53 may be considered as a crucial determiner that can regulate the biological behavior of RAW264.7 cells stimulated by LPS.
Collapse
Affiliation(s)
- Wuyang Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Liang
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongyang Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cuicui Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liao Jia
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingyu Jin
- Department of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Lv W, Xue L, Liang L, Liu D, Li C, Liao J, Jin Y. Endotoxin induced acute kidney injury modulates expression of AQP1, P53 and P21 in rat kidney, heart, lung and small intestine. PLoS One 2023; 18:e0288507. [PMID: 37490500 PMCID: PMC10368293 DOI: 10.1371/journal.pone.0288507] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
This study was designed to explore whether aquaporin 1(AQP1), P53 and P21 can be used as diagnostic biomarkers of lipopolysaccharide (LPS)-induced acute kidney injury (AKI) and potential indicators of sepsis-induced multiple organ injury. Bioinformatics results demonstrated that AQP1, P53, P21 was dramatically elevated 6h after Cecal ligation and puncture (CLP)-AKI in rat renal tissue. The expression of AQP1, P53, P21, NGAL and KIM-1 in kidney were increased significantly at first and then decreased gradually in LPS-induced AKI rats. Histopathological sections showed swelling of tubular epithelial cells and destruction of basic structures as well as infiltration of numerous inflammatory cells in LPS-induced AKI. Moreover, the expressions of AQP1, P53 and P21 in heart were significantly increased in LPS treatment rats, while the AQP1 expressions in lung and small intestine were significantly decreased. The level of NGAL mRNA in heart, lung and small intestine was firstly increased and then decreased during LPS treatment rats, but the expression of KIM-1 mRNA was not affected. Therefore, our results suggest that AQP1, P53 and P21 is remarkably upregulated in LPS-induced AKI, which may be considered as a potential novel diagnostic biomarker of Septic AKI. NGAL may serve as a biomarker of sepsis-induced multiple organ damage during the process of LPS-induced AKI.
Collapse
Affiliation(s)
- Wuyang Lv
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Longge Xue
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lei Liang
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dongyang Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Cuicui Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia Liao
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingyu Jin
- Department of Clinical Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
12
|
Liang K, Chang S, Chen Y, Tsai W, Wang K. A cluster of heritable pulmonary arterial hypertension cases in a family with all three siblings carrying the same novel AQP1 c.273C>G variant-a case report. Pulm Circ 2023; 13:e12211. [PMID: 37007933 PMCID: PMC10064854 DOI: 10.1002/pul2.12211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/27/2023] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Approximately 25%-30% of patients diagnosed with idiopathic pulmonary arterial hypertension (PAH) have a clustered underlying Mendelian genetic cause and should be classified as heritable PAH (HPAH). The sixth World Symposium on Pulmonary Hypertension listed AQP1 as a PAH-related gene. AQP1 and its protein product Aquaporin-1 (AQP1) are found in abundance within pulmonary artery smooth muscle cells. Here, we report a family affected by HPAH with all three siblings carrying the same novel missense variant of AQP1 c.273C>G (p.Ile91Met). The youngest brother and the older sister both had dyspnea and edema and were diagnosed with HPAH about 10 years ago. In 2021, they received genetic tests that revealed all three siblings carried the same novel variant of AQP1 (c.273C>G). The brother in between these two siblings, although originally claimed to be asymptomatic, raised awareness. He then sought medical examination and confirmed the diagnosis of HPAH as well. This report on all three siblings carrying the same novel variant of AQP1 (c.273C>G) highlighted the importance of genetic testing and counseling for family members when PAH was first detected.
Collapse
Affiliation(s)
- Kae‐Woei Liang
- Cardiovascular CenterTaichung Veterans General HospitalTaichungTaiwan
- Institute of Clinical Medicine and Faculty of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Post‐Baccalaureate MedicineNational Chung Hsing UniversityTaichungTaiwan
| | | | - Yu‐Wei Chen
- Cardiovascular CenterTaichung Veterans General HospitalTaichungTaiwan
- Institute of Clinical Medicine and Faculty of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Department of Post‐Baccalaureate MedicineNational Chung Hsing UniversityTaichungTaiwan
| | - Wan‐Jane Tsai
- Center for Pulmonary Arterial Hypertension and Pulmonary Vascular DiseaseChina Medical University HospitalTaichungTaiwan
| | - Kuo‐Yang Wang
- Center for Pulmonary Arterial Hypertension and Pulmonary Vascular DiseaseChina Medical University HospitalTaichungTaiwan
| |
Collapse
|
13
|
Volkart S, Kym U, Braissant O, Delgado-Eckert E, Al-Samir S, Angresius R, Huo Z, Holland-Cunz S, Gros SJ. AQP1 in the Gastrointestinal Tract of Mice: Expression Pattern and Impact of AQP1 Knockout on Colonic Function. Int J Mol Sci 2023; 24:ijms24043616. [PMID: 36835026 PMCID: PMC9959819 DOI: 10.3390/ijms24043616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Aquaporin 1 (AQP1) is one of thirteen known mammalian aquaporins. Its main function is the transport of water across cell membranes. Lately, a role of AQP has been attributed to other physiological and pathological functions including cell migration and peripheral pain perception. AQP1 has been found in several parts of the enteric nervous system, e.g., in the rat ileum and in the ovine duodenum. Its function in the intestine appears to be multifaceted and is still not completely understood. The aim of the study was to analyze the distribution and localization of AQP1 in the entire intestinal tract of mice. AQP1 expression was correlated with the hypoxic expression profile of the various intestinal segments, intestinal wall thickness and edema, as well as other aspects of colon function including the ability of mice to concentrate stools and their microbiome composition. AQP1 was found in a specific pattern in the serosa, the mucosa, and the enteric nervous system throughout the gastrointestinal tract. The highest amount of AQP1 in the gastrointestinal tract was found in the small intestine. AQP1 expression correlated with the expression profiles of hypoxia-dependent proteins such as HIF-1α and PGK1. Loss of AQP1 through knockout of AQP1 in these mice led to a reduced amount of bacteroidetes and firmicutes but an increased amount of the rest of the phyla, especially deferribacteres, proteobacteria, and verrucomicrobia. Although AQP-KO mice retained gastrointestinal function, distinct changes regarding the anatomy of the intestinal wall including intestinal wall thickness and edema were observed. Loss of AQP1 might interfere with the ability of the mice to concentrate their stool and it is associated with a significantly different composition of the of the bacterial stool microbiome.
Collapse
Affiliation(s)
- Stefanie Volkart
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Urs Kym
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Olivier Braissant
- Microcalorimetry Unit, Department of Biomedical Engineering, University of Basel, 4001 Basel, Switzerland
| | - Edgar Delgado-Eckert
- Computational Physiology and Biostatistics, Department of Biomedical Engineering at University of Basel and University Children’s Hospital Basel, 4056 Basel, Switzerland
| | - Samer Al-Samir
- Vegetative Physiologie 4220, Zentrum Physiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Rebecca Angresius
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4056 Basel, Switzerland
- Correspondence:
| |
Collapse
|
14
|
Slaven JE, Wilkerson M, Soltis AR, Rittase WB, Bradfield DT, Bylicky M, Cary L, Tsioplaya A, Bouten R, Dalgard C, Day RM. Transcriptomic Profiling and Pathway Analysis of Mesenchymal Stem Cells Following Low Dose-Rate Radiation Exposure. Antioxidants (Basel) 2023; 12:antiox12020241. [PMID: 36829800 PMCID: PMC9951969 DOI: 10.3390/antiox12020241] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Low dose-rate radiation exposure can occur in medical imaging, as background from environmental or industrial radiation, and is a hazard of space travel. In contrast with high dose-rate radiation exposure that can induce acute life-threatening syndromes, chronic low-dose radiation is associated with Chronic Radiation Syndrome (CRS), which can alter environmental sensitivity. Secondary effects of chronic low dose-rate radiation exposure include circulatory, digestive, cardiovascular, and neurological diseases, as well as cancer. Here, we investigated 1-2 Gy, 0.66 cGy/h, 60Co radiation effects on primary human mesenchymal stem cells (hMSC). There was no significant induction of apoptosis or DNA damage, and cells continued to proliferate. Gene ontology (GO) analysis of transcriptome changes revealed alterations in pathways related to cellular metabolism (cholesterol, fatty acid, and glucose metabolism), extracellular matrix modification and cell adhesion/migration, and regulation of vasoconstriction and inflammation. Interestingly, there was increased hypoxia signaling and increased activation of pathways regulated by iron deficiency, but Nrf2 and related genes were reduced. The data were validated in hMSC and human lung microvascular endothelial cells using targeted qPCR and Western blotting. Notably absent in the GO analysis were alteration pathways for DNA damage response, cell cycle inhibition, senescence, and pro-inflammatory response that we previously observed for high dose-rate radiation exposure. Our findings suggest that cellular gene transcription response to low dose-rate ionizing radiation is fundamentally different compared to high-dose-rate exposure. We hypothesize that cellular response to hypoxia and iron deficiency are driving processes, upstream of the other pathway regulation.
Collapse
Affiliation(s)
- John E. Slaven
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Matthew Wilkerson
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Anthony R. Soltis
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - W. Bradley Rittase
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Dmitry T. Bradfield
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Michelle Bylicky
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Lynnette Cary
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Alena Tsioplaya
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Roxane Bouten
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| | - Clifton Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3236; Fax: +1-301-295-3220
| |
Collapse
|
15
|
Fan L, Wu P, Li X, Tie L. Aquaporins in Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1398:125-135. [PMID: 36717490 DOI: 10.1007/978-981-19-7415-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Recent studies have shown that aquaporins (AQPs) are involved in the regulation of cardiovascular function and the development of related diseases, especially in cerebral ischemia, congestive heart failure, hypertension, and angiogenesis. Therefore, further studies are needed to elucidate the mechanism accounting for the association between AQPs and vascular function-related diseases, which may lead to novel approaches to the prevention and treatment of those diseases. Here we will discuss the expression and physiological roles of AQPs in vascular tissues and summarize recent progress in the research on AQPs related cardiovascular diseases.
Collapse
Affiliation(s)
- Lu Fan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, China
| | - Pin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, China
| | - Xuejun Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, China.
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, China.
| |
Collapse
|
16
|
Sulforaphane alleviated vascular remodeling in hypoxic pulmonary hypertension via inhibiting inflammation and oxidative stress. J Nutr Biochem 2023; 111:109182. [PMID: 36220525 DOI: 10.1016/j.jnutbio.2022.109182] [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: 11/28/2021] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/05/2022]
Abstract
Hypoxic pulmonary hypertension (HPH) is a cardiopulmonary disease featured by pulmonary vascular remodeling, which is due to abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) and dysfunction of endothelial cells (ECs). Sulforaphane (SFN) is a natural isothiocyanate extracted from cruciferous vegetables with promising anti-inflammatory and anti-oxidative activities. This study aimed to explore the effect and mechanism of SFN on HPH. Male mice were exposed to persistent chronic hypoxia for 4 weeks to induce HPH. The results demonstrated that SFN repressed the increased right ventricular systolic pressure (RVSP) and attenuated the right ventricular hypertrophy and pulmonary arteries remodeling in HPH mice. In particular, after SFN treatment, the CD68 positive cells in lung sections were reduced; TNF-α and IL-6 levels in lungs and serum declined; activation of NF-κB in PASMCs was inhibited in response to hypoxia. Besides, SFN enhanced the superoxide dismutase (SOD) activity in serum, SOD2 expression, total glutathione levels, and GSH/GSSG ratio in PASMCs, along with a decrease in malondialdehyde (MDA) contents in serum and ROS production in PASMCs after hypoxia exposure. Notably, SFN, as an Nrf2 activator, reversed the reduction in Nrf2 expression in hypoxic PASMCs. In vitro, SFN treatment inhibited hyperproliferation and promoted apoptosis of PASMCs under hypoxia conditions. SFN also prevented the apoptosis of pulmonary microvascular ECs caused by hypoxia. Therefore, these data suggested that SFN could significantly restrain the inflammation and oxidative stress, thereby inhibiting PASMCs proliferation, promoting PASMCs apoptosis, and reversing hypoxia injury in ECs to improve pulmonary vascular remodeling.
Collapse
|
17
|
Wang D, Ji Y, Wang R, Cheng K, Liu L, Wu N, Tang Q, Zheng X, Li J, Zhu Z, Wang Q, Zhang X, Li R, Pan J, Sui Z, Yuan Y. Lycopene Ameliorates Hypoxic Pulmonary Hypertension via Suppression of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9179427. [PMID: 39282152 PMCID: PMC11401662 DOI: 10.1155/2022/9179427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 09/18/2024]
Abstract
Hypoxic pulmonary hypertension (HPH) is a progressive cardiopulmonary system disease characterized by pulmonary vascular remodeling. Its occurrence and progression are closely related to oxidative stress. Lycopene, extracted from red vegetables and fruits, exhibits a particularly high antioxidant capacity that is beneficial for cardiovascular diseases. Nevertheless, the role and mechanism of lycopene in HPH remain unknown. Here, we found that lycopene reversed the elevated right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and pulmonary vascular remodeling induced by hypoxia in rats. In vitro, lycopene caused lower proliferation and migration of PASMCs, with higher apoptosis. Consistent with the antiproliferative result of lycopene on hypoxic PASMCs, the hippo signaling pathway associated with cell growth was activated. Furthermore, lycopene reduced malondialdehyde (MDA) levels and enhanced superoxide dismutase (SOD) activity in the lungs and serum of rats under hypoxia conditions. The expression of NOX4 in the lungs was also significantly decreased. Hypoxic PASMCs subjected to lycopene showed decreased reactive oxygen species (ROS) production and NOX4 expression. Importantly, lycopene repressed HIF-1α expression both in the lungs and PASMCs in response to hypoxia in the absence of a significant change of HIF-1α mRNA. Compared with 2ME2 (a HIF-1α inhibitor) alone treatment, lycopene treatment did not significantly change PASMC proliferation, NOX4 expression, and ROS production after 2ME2 blocked HIF-1α, suggesting the inhibitory effect of lycopene on HIF-1α-NOX4-ROS axis and the targeted effect on HIF-1α. After CHX blocked protein synthesis, lycopene promoted the protein degradation of HIF-1α. MG-132, a proteasome inhibitor, notably reversed the decrease in HIF-1α protein level induced by lycopene in response to hypoxia. Therefore, lycopene suppressed hypoxia-induced oxidative stress through HIF-1α-NOX4-ROS axis, thereby alleviating HPH. Our findings will provide a new research direction for clinical HPH therapies.
Collapse
Affiliation(s)
- Dingyou Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Yuke Ji
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Rui Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Ke Cheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Na Wu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Qing Tang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Xu Zheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Junxia Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Zhilong Zhu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Qinghua Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Xueyan Zhang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Runbo Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Zheng Sui
- Department of Vasculocardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
18
|
Chen D, Zhang HF, Yuan TY, Sun SC, Wang RR, Wang SB, Fang LH, Lyu Y, Du GH. Puerarin-V prevents the progression of hypoxia- and monocrotaline-induced pulmonary hypertension in rodent models. Acta Pharmacol Sin 2022; 43:2325-2339. [PMID: 35190697 PMCID: PMC9433387 DOI: 10.1038/s41401-022-00865-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiopulmonary disease characterized by a progressive increase in pulmonary vascular resistance. One of the initial pathogenic factors of PH is pulmonary arterial remodeling under various stimuli. Current marketed drugs against PH mainly relieve symptoms without significant improvement in overall prognosis. Discovering and developing new therapeutic drugs that interfere with vascular remodeling is in urgent need. Puerarin is an isoflavone compound extracted from the root of Kudzu vine, which is widely used in the treatment of cardiovascular diseases. In the present study, we evaluated the efficacy of puerarin in the treatment of experimental PH. PH was induced in rats by a single injection of MCT (50 mg/kg, sc), and in mice by exposure to hypoxia (10% O2) for 14 days. After MCT injection the rats were administered puerarin (10, 30, 100 mg · kg-1 · d-1, i.g.) for 28 days, whereas hypoxia-treated mice were pre-administered puerarin (60 mg · kg-1 · d-1, i.g.) for 7 days. We showed that puerarin administration exerted significant protective effects in both experimental PH rodent models, evidenced by significantly reduced right ventricular systolic pressure (RVSP) and lung injury, improved pulmonary artery blood flow as well as pulmonary vasodilation and contraction function, inhibited inflammatory responses in lung tissues, improved resistance to apoptosis and abnormal proliferation in lung tissues, attenuated right ventricular injury and remodeling, and maintained normal function of the right ventricle. We revealed that MCT and hypoxia treatment significantly downregulated BMPR2/Smad signaling in the lung tissues and PPARγ/PI3K/Akt signaling in the lung tissues and right ventricles, which were restored by puerarin administration. In addition, we showed that a novel crystal type V (Puer-V) exerted better therapeutic effects than the crude form of puerarin (Puer). Furthermore, Puer-V was more efficient than bosentan (a positive control drug) in alleviating the abnormal structural changes and dysfunction of lung tissues and right ventricles. In conclusion, this study provides experimental evidence for developing Puer-V as a novel therapeutic drug to treat PH.
Collapse
Affiliation(s)
- Di Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hui-Fang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tian-Yi Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shu-Chan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ran-Ran Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shou-Bao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lian-Hua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yang Lyu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
19
|
Rodor J, Chen SH, Scanlon JP, Monteiro JP, Caudrillier A, Sweta S, Stewart KR, Shmakova A, Dobie R, Henderson BEP, Stewart K, Hadoke PWF, Southwood M, Moore SD, Upton PD, Morrell NW, Li Z, Chan SY, Handen A, Lafyatis R, de Rooij LPMH, Henderson NC, Carmeliet P, Spiroski AM, Brittan M, Baker AH. Single-cell RNA sequencing profiling of mouse endothelial cells in response to pulmonary arterial hypertension. Cardiovasc Res 2022; 118:2519-2534. [PMID: 34528097 PMCID: PMC9400412 DOI: 10.1093/cvr/cvab296] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Endothelial cell (EC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension (PAH). We aimed to characterize EC dynamics in PAH at single-cell resolution. METHODS AND RESULTS We carried out single-cell RNA sequencing (scRNA-seq) of lung ECs isolated from an EC lineage-tracing mouse model in Control and SU5416/hypoxia-induced PAH conditions. EC populations corresponding to distinct lung vessel types, including two discrete capillary populations, were identified in both Control and PAH mice. Differential gene expression analysis revealed global PAH-induced EC changes that were confirmed by bulk RNA-seq. This included upregulation of the major histocompatibility complex class II pathway, supporting a role for ECs in the inflammatory response in PAH. We also identified a PAH response specific to the second capillary EC population including upregulation of genes involved in cell death, cell motility, and angiogenesis. Interestingly, four genes with genetic variants associated with PAH were dysregulated in mouse ECs in PAH. To compare relevance across PAH models and species, we performed a detailed analysis of EC heterogeneity and response to PAH in rats and humans through whole-lung PAH scRNA-seq datasets, revealing that 51% of up-regulated mouse genes were also up-regulated in rat or human PAH. We identified promising new candidates to target endothelial dysfunction including CD74, the knockdown of which regulates EC proliferation and barrier integrity in vitro. Finally, with an in silico cell ordering approach, we identified zonation-dependent changes across the arteriovenous axis in mouse PAH and showed upregulation of the Serine/threonine-protein kinase Sgk1 at the junction between the macro- and microvasculature. CONCLUSION This study uncovers PAH-induced EC transcriptomic changes at a high resolution, revealing novel targets for potential therapeutic candidate development.
Collapse
Affiliation(s)
- Julie Rodor
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Shiau Haln Chen
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Jessica P Scanlon
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - João P Monteiro
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Axelle Caudrillier
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sweta Sweta
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Katherine Ross Stewart
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Alena Shmakova
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Beth E P Henderson
- Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kevin Stewart
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Patrick W F Hadoke
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mark Southwood
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Stephen D Moore
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Paul D Upton
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Nick W Morrell
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ziwen Li
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Stephen Y Chan
- Divisions of Cardiology and Rheumatology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adam Handen
- Divisions of Cardiology and Rheumatology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Divisions of Cardiology and Rheumatology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Laura P M H de Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, Leuven Cancer Institute (LKI), VIB and KU Leuven, Leuven 3000, Belgium
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Center for Cancer Biology, Leuven Cancer Institute (LKI), VIB and KU Leuven, Leuven 3000, Belgium
| | - Ana Mishel Spiroski
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
20
|
Shangzu Z, Dingxiong X, ChengJun M, Yan C, Yangyang L, Zhiwei L, Ting Z, Zhiming M, Yiming Z, Liying Z, Yongqi L. Aquaporins: Important players in the cardiovascular pathophysiology. Pharmacol Res 2022; 183:106363. [PMID: 35905892 DOI: 10.1016/j.phrs.2022.106363] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/15/2022]
Abstract
Aquaporin is a membrane channel protein widely expressed in body tissues, which can control the input and output of water in cells. AQPs are differentially expressed in different cardiovascular tissues and participate in water transmembrane transport, cell migration, metabolism, inflammatory response, etc. The aberrant expression of AQPs highly correlates with the onset of ischemic heart disease, myocardial ischemia-reperfusion injury, heart failure, etc. Despite much attention to the regulatory role of AQPs in the cardiovascular system, the translation of AQPs into clinical application still faces many challenges, including clarification of the localization of AQPs in the cardiovascular system and mechanisms mediating cardiovascular pathophysiology, as well as the development of cardiovascular-specific AQPs modulators.Therefore, in this study, we comprehensively reviewed the critical roles of AQP family proteins in maintaining cardiovascular homeostasis and described the underlying mechanisms by which AQPs mediated the outcomes of cardiovascular diseases. Meanwhile, AQPs serve as important therapeutic targets, which provide a wide range of opportunities to investigate the mechanisms of cardiovascular diseases and the treatment of those diseases.
Collapse
Affiliation(s)
- Zhang Shangzu
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Xie Dingxiong
- Gansu Institute of Cardiovascular Diseases, LanZhou,China
| | - Ma ChengJun
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Chen Yan
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Li Yangyang
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Liu Zhiwei
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhou Ting
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Miao Zhiming
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhang Yiming
- Gansu University of traditional Chinese Medicine, LanZhou, China
| | - Zhang Liying
- Gansu University of traditional Chinese Medicine, LanZhou, China; Gansu Institute of Cardiovascular Diseases, LanZhou,China.
| | - Liu Yongqi
- Gansu University of traditional Chinese Medicine, LanZhou, China; Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, China.
| |
Collapse
|
21
|
Remes A, Körbelin J, Arnold C, Rohwedder C, Heckmann MB, Mairbauerl H, Frank D, Korff T, Frey N, Trepel M, Müller OJ. AAV-mediated gene transfer of inducible nitric oxide synthase (iNOS) to an animal model of pulmonary hypertension. Hum Gene Ther 2022; 33:959-967. [PMID: 35850528 DOI: 10.1089/hum.2021.230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by progressive obstruction of pulmonary arteries due to inflammatory processes, cellular proliferation, and extracellular matrix deposition and vasoconstriction. As treatment options are limited, we studied gene transfer of an inducible nitric oxide synthase (iNOS) using adeno-associated virus (AAV) vectors specifically targeted to endothelial cells of pulmonary vessels in a murine model of PH. Adult mice were intravenously injected with AAV vectors expressing iNOS. Mice were subjected to hypoxia for three weeks and sacrificed afterwards. We found elevated levels of iNOS both in lung tissue and pulmonary endothelial cells in hypoxic controls which could be further increased by AAV-mediated iNOS gene transfer. This additional increase in iNOS was associated with decreased wall thickness of pulmonary vessels, less macrophage infiltration, and reduced molecular markers of fibrosis. Taken together, using a tissue-targeted approach, we show that AAV-mediated iNOS overexpression in endothelial cells of the pulmonary vasculature significantly decreases vascular remodeling in a murine model of PH, suggesting upregulation of iNOS as promising target for treatment of PH.
Collapse
Affiliation(s)
- Anca Remes
- Department of Internal Medicine III, University of Kiel, and German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Germany, Kiel, Germany;
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Department of Oncology, Hematology and Bone Marrow Transplantation, Martinistr. 52, Division of Pneumology, Hamburg, Germany, 20246;
| | - Caroline Arnold
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany, Heidelberg, Germany;
| | - Carolin Rohwedder
- Internal Medicine III, University Hospital Heidelberg, Germany, and German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany;
| | - Markus Benjamin Heckmann
- Internal Medicine III, University Hospital Heidelberg, Germany, and German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany;
| | - Heimo Mairbauerl
- Medical Clinic VII, Heidelberg University, Germany and Translational Lung Research Center, part of the German Center for Lung Research (DZL), University of Heidelberg, Germany, Heidelberg, Germany;
| | - Derk Frank
- Department of Internal Medicine III, University of Kiel, and German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Germany, Kiel, Germany;
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany, Heidelberg, Germany;
| | - Norbert Frey
- Internal Medicine III, University Hospital Heidelberg, Germany, and German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany;
| | - Martin Trepel
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf Germany, Hamburg, Germany.,Department of Hematology and Oncology, University Medical Center Augsburg, Germany, Ausburg, Germany;
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, and German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Germany, Kiel, Germany;
| |
Collapse
|
22
|
Description of Two New Cases of AQP1 Related Pulmonary Arterial Hypertension and Review of the Literature. Genes (Basel) 2022; 13:genes13050927. [PMID: 35627312 PMCID: PMC9141352 DOI: 10.3390/genes13050927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe clinical condition characterized by an increase in mean pulmonary artery pressure, which leads to a right ventricular hypertrophy and potentially heart failure and death. In the last several years, many genes have been associated with PAH, particularly in idiopathic and heritable forms but also in associated forms. Here we described the identification of two unrelated families in which the AQP1 variant was found from a cohort of 300 patients. The variants were identified by whole exome sequencing (WES). In the first family, the variant was detected in three affected members from a hereditary PAH, and in the second family the proband had PAH associated with scleroderma. In addition, we have reviewed all cases published in the literature thus far of patients with PAH and AQP1 variants. Functional studies have led to some contradictory conclusions, and the evidence of the relationship of AQP1 and PAH is still limited. However, we describe two further families with PAH and variants in AQP1, expanding both the number of cases and the clinically associated phenotype. We provide further evidence of the association of AQP1 and the development of hereditary and associated forms of PAH.
Collapse
|
23
|
Aldred MA, Morrell NW, Guignabert C. New Mutations and Pathogenesis of Pulmonary Hypertension: Progress and Puzzles in Disease Pathogenesis. Circ Res 2022; 130:1365-1381. [PMID: 35482831 PMCID: PMC9897592 DOI: 10.1161/circresaha.122.320084] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex multifactorial disease with poor prognosis characterized by functional and structural alterations of the pulmonary circulation causing marked increase in pulmonary vascular resistance, ultimately leading to right heart failure and death. Mutations in the gene encoding BMPRII-a receptor for the TGF-β (transforming growth factor-beta) superfamily-account for over 70% of families with PAH and ≈20% of sporadic cases. In recent years, however, less common or rare mutations in other genes have been identified. This review will consider how these newly discovered PAH genes could help to provide a better understanding of the molecular and cellular bases of the maintenance of the pulmonary vascular integrity, as well as their role in the PAH pathogenesis underlying occlusion of arterioles in the lung. We will also discuss how insights into the genetic contributions of these new PAH-related genes may open up new therapeutic targets for this, currently incurable, cardiopulmonary disorder.
Collapse
Affiliation(s)
- Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicholas W Morrell
- University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, UK
| | - Christophe Guignabert
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France,Université Paris-Saclay, Faculté de Médecine, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
24
|
Yu B, Wang X, Song Y, Xie G, Jiao S, Shi L, Cao X, Han X, Qu A. The role of hypoxia-inducible factors in cardiovascular diseases. Pharmacol Ther 2022; 238:108186. [PMID: 35413308 DOI: 10.1016/j.pharmthera.2022.108186] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/29/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. During the development of cardiovascular diseases, hypoxia plays a crucial role. Hypoxia-inducible factors (HIFs) are the key transcription factors for adaptive hypoxic responses, which orchestrate the transcription of numerous genes involved in angiogenesis, erythropoiesis, glycolytic metabolism, inflammation, and so on. Recent studies have dissected the precise role of cell-specific HIFs in the pathogenesis of hypertension, atherosclerosis, aortic aneurysms, pulmonary arterial hypertension, and heart failure using tissue-specific HIF-knockout or -overexpressing animal models. More importantly, several compounds developed as HIF inhibitors or activators have been in clinical trials for the treatment of renal cancer or anemia; however, little is known on the therapeutic potential of these inhibitors for cardiovascular diseases. The purpose of this review is to summarize the recent advances on HIFs in the pathogenesis and pathophysiology of cardiovascular diseases and to provide evidence of potential clinical therapeutic targets.
Collapse
Affiliation(s)
- Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Yanting Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China; Department of Pathology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Guomin Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Shiyu Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Li Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Xuejie Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Xinyao Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, PR China; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing 100069, PR China.
| |
Collapse
|
25
|
Wang R, Pan J, Han J, Gong M, Liu L, Zhang Y, Liu Y, Wang D, Tang Q, Wu N, Wang L, Yan J, Li H, Yuan Y. Melatonin Attenuates Dasatinib-Aggravated Hypoxic Pulmonary Hypertension via Inhibiting Pulmonary Vascular Remodeling. Front Cardiovasc Med 2022; 9:790921. [PMID: 35402542 PMCID: PMC8987569 DOI: 10.3389/fcvm.2022.790921] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Dasatinib treatment is approved as first-line therapy for chronic myeloid leukemia. However, pulmonary hypertension (PH) is a highly morbid and often fatal side-effect of dasatinib, characterized by progressive pulmonary vascular remodeling. Melatonin exerts strong antioxidant capacity against the progression of cardiovascular system diseases. The present work aimed to investigate the effect of melatonin on dasatinib-aggravated hypoxic PH and explore its possible mechanisms. Dasatinib-aggravated rat experimental model of hypoxic PH was established by utilizing dasatinib under hypoxia. The results indicated that melatonin could attenuate dasatinib-aggravated pulmonary pressure and vascular remodeling in rats under hypoxia. Additionally, melatonin attenuated the activity of XO, the content of MDA, the expression of NOX4, and elevated the activity of CAT, GPx, and SOD, the expression of SOD2, which were caused by dasatinib under hypoxia. In vitro, dasatinib led to decreased LDH activity and production of NO in human pulmonary microvascular endothelial cells (HPMECs), moreover increased generation of ROS, and expression of NOX4 both in HPMECs and primary rat pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. Dasatinib up-regulated the expression of cleaved caspase-3 and the ratio of apoptotic cells in HPMECs, and also elevated the percentage of S phase and the expression of Cyclin D1 in primary PASMCs under hypoxia. Melatonin ameliorated dasatinib-aggravated oxidative damage and apoptosis in HPMECs, meanwhile reduced oxidative stress level, proliferation, and repressed the stability of HIF1-α protein in PASMCs under hypoxia. In conclusion, melatonin significantly attenuates dasatinib-aggravated hypoxic PH by inhibiting pulmonary vascular remodeling in rats. The possible mechanisms involved protecting endothelial cells and inhibiting abnormal proliferation of smooth muscle cells. Our findings may suggest that melatonin has potential clinical value as a therapeutic approach to alleviate dasatinib-aggravated hypoxic PH.
Collapse
Affiliation(s)
- Rui Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinzhen Han
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Miaomiao Gong
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yunlong Zhang
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Liu
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Dingyou Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qing Tang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Na Wu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lin Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinsong Yan
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
- Jinsong Yan,
| | - Hua Li
- College of Pharmacy, Dalian Medical University, Dalian, China
- Hua Li,
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- *Correspondence: Yuhui Yuan,
| |
Collapse
|
26
|
Yun X, Philip NM, Jiang H, Smith Z, Huetsch JC, Damarla M, Suresh K, Shimoda LA. Upregulation of Aquaporin 1 Mediates Increased Migration and Proliferation in Pulmonary Vascular Cells From the Rat SU5416/Hypoxia Model of Pulmonary Hypertension. Front Physiol 2021; 12:763444. [PMID: 34975522 PMCID: PMC8718640 DOI: 10.3389/fphys.2021.763444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by exuberant vascular remodeling leading to elevated pulmonary arterial pressure, maladaptive right ventricular remodeling, and eventual death. The factors controlling pulmonary arterial smooth muscle cell (PASMC) and endothelial cell hyperplasia and migration, hallmark features of the vascular remodeling observed in PAH, remain poorly understood. We previously demonstrated that hypoxia upregulates the expression of aquaporin 1 (AQP1), a water channel, in PASMCs, and that this upregulation was required for hypoxia-induced migration and proliferation. However, whether the same is true in a model of severe PAH and in pulmonary microvascular endothelial cells (MVECs) is unknown. In this study, we used the SU5416 plus hypoxia (SuHx) rat model of severe pulmonary hypertension, which mimics many of the features of human PAH, to determine whether AQP1 levels were altered in PASMCs and MVECs and contributed to a hyperproliferative/hypermigratory phenotype. Rats received a single injection of SU5416 (20 mg/kg) and then were placed in 10% O2 for 3 weeks, followed by a return to normoxic conditions for an additional 2 weeks. We found that AQP1 protein levels were increased in both PASMCs and MVECs from SuHx rats, even in the absence of sustained hypoxic exposure, and that in MVECs, the increase in protein expression was associated with upregulation of AQP1 mRNA levels. Silencing of AQP1 had no significant effect on PASMCs from control animals but normalized enhanced migration and proliferation observed in cells from SuHx rats. Loss of AQP1 also reduced migration and proliferation in MVECs from SuHx rats. Finally, augmenting AQP1 levels in MVECs from control rats using forced expression was sufficient to increase migration and proliferation. These results demonstrate a key role for enhanced AQP1 expression in mediating abnormal migration and proliferation in pulmonary vascular cells from a rodent model that reflects many of the features of human PAH.
Collapse
|
27
|
Cao J, Yang L, Wang L, Zhao Q, Wu D, Li M, Mu Y. Heat shock protein 70 attenuates hypoxia‑induced apoptosis of pulmonary microvascular endothelial cells isolated from neonatal rats. Mol Med Rep 2021; 24:690. [PMID: 34328190 PMCID: PMC8365595 DOI: 10.3892/mmr.2021.12327] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/15/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary microvascular endothelial cell (PMVEC) apoptosis is the initial stage of adult pulmonary hypertension (PH), which involves high pulmonary arterial pressure and pulmonary vascular remodeling. However, the mechanism regulating PMVEC apoptosis and its involvement in the early stages of neonatal hypoxic PH (HPH) pathogenesis are currently unclear. The present study aimed to investigate the effects of heat shock protein 70 (HSP70) on hypoxia‑induced apoptosis in PMVECs. PMVECs isolated from neonatal Sprague‑Dawley rats were transfected with lentivirus with or without HSP70, or treated with the synthetic HSP70 inhibitor N‑formyl‑3,4‑methylenedioxy‑benzylidene-g-butyrolactam under hypoxic conditions (5% O2) for 24, 48 or 72 h. PMVEC apoptosis was evaluated by performing flow cytometry and mitochondrial membrane potential (MMP) assays. The expression levels of HSP70, hypoxia‑inducible factor‑1α (HIF‑1α) and apoptosis‑associated proteins were determined by conducting reverse transcription‑quantitative PCR and western blotting. Following 24, 48 or 72 h of hypoxia, the apoptotic rates of PMVECs were significantly elevated compared with cells under normoxic conditions. The MMP was significantly reduced, whereas the mRNA and protein expression levels of HIF‑1α, cytochrome c (cyt C), caspase‑3 and HSP70 were enhanced by hypoxia compared with those under normoxic conditions. Additionally, the mRNA and protein expression levels of B‑cell lymphoma 2 (Bcl‑2) were significantly downregulated in the hypoxia group compared with those in the normoxia group. In hypoxic PMVECs, HSP70 overexpression decreased the apoptotic rate and the expression levels of cyt C, downregulated the expression levels of caspase‑3 and HIF‑1α, and increased the MMP and the expression levels of Bcl‑2. HSP70 inhibition resulted in the opposite outcomes compared with those of HSP70 overexpression. Therefore, the results of the present study suggested that HSP70 may inhibit mitochondrial pathway‑mediated apoptosis in isolated neonatal rat PMVECs in early‑stage hypoxia, which may be associated with HSP70‑mediated HIF‑1α downregulation. Overall, HSP70 may be protective against neonatal HPH through the HSP70/HIF‑1α pathway.
Collapse
Affiliation(s)
- Jing Cao
- Department of Echocardiography, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
- Department of Neonatology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Lingjie Yang
- Department of Echocardiography, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Le Wang
- Department of Neonatology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Qian Zhao
- School of Pediatrics, Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Dian Wu
- School of Pediatrics, Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Mingxia Li
- Department of Neonatology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| | - Yuming Mu
- Department of Echocardiography, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang Uyghur Autonomous Region 830054, P.R. China
| |
Collapse
|
28
|
Gong M, Wang X, Mu L, Wang Y, Pan J, Yuan X, Zhou H, Xing J, Wang R, Sun J, Liu Q, Zhang X, Wang L, Chen Y, Pei Y, Li S, Liu L, Zhao Y, Yuan Y. Steroid receptor coactivator-1 enhances the stemness of glioblastoma by activating long noncoding RNA XIST/miR-152/KLF4 pathway. Cancer Sci 2021; 112:604-618. [PMID: 33090636 PMCID: PMC7894023 DOI: 10.1111/cas.14685] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) recurrence is attributed to the presence of therapy-resistant glioblastoma stem cells. Steroid receptor coactivator-1 (SRC-1) acts as an oncogenic regulator in many human tumors. The relationship between SRC-1 and GBM has not yet been studied. Herein, we investigate the role of SRC-1 in GBM. In this study, we found that SRC-1 expression is positively correlated with grades of glioma and inversely correlated with glioma patient's prognosis. Steroid receptor coactivator-1 promotes the proliferation, migration, and tumor growth of GBM cells. Notably, SRC-1 knockdown suppresses the stemness of GBM cells. Mechanistically, long noncoding RNA X-inactive specific transcript (XIST) is regulated by SRC-1 at the posttranscriptional level and mediates the function of SRC-1 in promoting stemness-like properties of GBM. Steroid receptor coactivator-1 can promote the expression of Kruppel-like factor 4 (KLF4) through the XIST/microRNA (miR)-152 axis. Additionally, arenobufagin and bufalin, SRC small molecule inhibitors, can reduce the proliferation and stemness of GBM cells. This study reveals SRC-1 promotes the stemness of GBM by activating the long noncoding RNA XIST/miR-152/KLF4 pathway and provides novel markers for diagnosis and therapy of GBM.
Collapse
Affiliation(s)
- Miaomiao Gong
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xun Wang
- Department of NeurosurgeryThe Third People’s Hospital of DalianDalianChina
| | - Lin Mu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yueyue Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jinjin Pan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xiaocheng Yuan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Haoran Zhou
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jinshan Xing
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Rui Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Jian Sun
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Qiwang Liu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Xiya Zhang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Lin Wang
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yiying Chen
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yandong Pei
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shao Li
- College of Basic Medical SciencesDalian Medical UniversityDalianChina
| | - Liang Liu
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Yongshun Zhao
- The First Affiliated HospitalDalian Medical UniversityDalianChina
| | - Yuhui Yuan
- The Second Affiliated HospitalInstitute of Cancer Stem CellDalian Medical UniversityDalianChina
| |
Collapse
|
29
|
Yadav E, Yadav N, Hus A, Yadav JS. Aquaporins in lung health and disease: Emerging roles, regulation, and clinical implications. Respir Med 2020; 174:106193. [PMID: 33096317 DOI: 10.1016/j.rmed.2020.106193] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/17/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Aquaporins (AQPs) aka water channels are a family of conserved transmembrane proteins (~30 kDa monomers) expressed in various organ systems. Of the 13 AQPs (AQP0 through AQP12) in the human body, four (AQPs 1, 3, 4, and 5) are expressed in the respiratory system. These channels are conventionally known for mediating transcellular fluid movements. Certain AQPs (aquaglyceroporins) have the capability to transport glycerol and potentially other solutes. There is an emerging body of literature unveiling the non-conventional roles of AQPs such as in cell proliferation and migration, gas permeation, signal potentiation, etc. Initial gene knock-out studies established a physiological role for lung AQPs, particularly AQP5, in maintaining homeostasis, by mediating fluid secretion from submucosal glands onto the airway surface liquid (ASL) lining. Subsequent studies have highlighted the functional significance of AQPs, particularly AQP1 and AQP5 in lung pathophysiology and diseases, including but not limited to chronic and acute lung injury, chronic obstructive pulmonary disease (COPD), other inflammatory lung conditions, and lung cancer. AQP1 has been suggested as a potential prognostic marker for malignant mesothelioma. Recent efforts are directed toward exploiting AQPs as targets for diagnosis, prevention, intervention, and/or treatment of various lung conditions. Emerging information on regulatory pathways and directed mechanistic research are posited to unravel novel strategies for these clinical implications. Future considerations should focus on development of AQP inhibitors, blockers, and modulators for therapeutic needs, and better understanding the role of lung-specific AQPs in inter-individual susceptibility to chronic lung diseases such as COPD and cancer.
Collapse
Affiliation(s)
- Ekta Yadav
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Niket Yadav
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, 22908-0738, USA
| | - Ariel Hus
- Department of Biology, University of Miami, Coral Gables, Florida, 33146, USA
| | - Jagjit S Yadav
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
30
|
Li ZM, Xu SY, Feng YZ, Cheng YR, Xiong JB, Zhou Y, Guan CX. The role of NOX4 in pulmonary diseases. J Cell Physiol 2020; 236:1628-1637. [PMID: 32780450 DOI: 10.1002/jcp.30005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/26/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) is a subtype of the NOX family, which is mainly expressed in the pulmonary vasculature and pulmonary endothelial cells in the respiratory system. NOX4 has unique characteristics, and is a constitutively active enzyme that primarily produces hydrogen peroxide. The signaling pathways associated with NOX4 are complicated. Negative and positive feedback play significant roles in regulating NOX4 expression. The role of NOX4 is controversial because NOX4 plays a protective or damaging role in different respiratory diseases. This review summarizes the structure, enzymatic properties, regulation, and signaling pathways of NOX4. This review then introduces the roles of NOX4 in different diseases in the respiratory system, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, and pulmonary fibrosis.
Collapse
Affiliation(s)
- Zi-Ming Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Sheng-Ya Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yi-Zhuo Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Rui Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Hasan B, Tuyghun E, Yang Y, Tuerxun P, Li X. Comprehensive network analysis to identify the molecular pathogenesis of pulmonary hypertension. Minerva Cardioangiol 2020; 68:319-325. [PMID: 32319267 DOI: 10.23736/s0026-4725.20.05111-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pulmonary hypertension (PAH) is a chronic progressive disease that may lead to right heart failure and eventually death. At present, great progress had been achieved in the treatment of pulmonary hypertension. However, pulmonary hypertension cannot be fundamentally cured, and its pathogenesis is still unclear. METHODS A multifactor-driven dysfunction module of pulmonary hypertension has been constructed in order to explore its potential pathogenesis. We performed differential expression analysis, coexpression analysis, enrichment analysis and hypergeometric test to calculate the potential regulatory effects of multiple factors on the module. RESULTS Four modules and corresponding hub genes were identified. In addition, we also obtained a series of ncRNA (MALAT1 and miR-17-5p) and transcription factor (HIF1A). Network analysis revealed that MALAT1, NFKB1 and RELA targeting IL1B of module 4 and IL6 of module 1 to participate in the occurrence and development of pulmonary hypertension through Toll-like receptor signaling pathway. CONCLUSIONS It is necessary to identify disease-related disorders by integrating multiple regulatory factors. The regulatory network may play an important role in PAH. The results not only provided new methods and ideas for follow-up research, but also helps researchers to have a deeper understanding of potential pathogenesis for PAH.
Collapse
Affiliation(s)
- Bilal Hasan
- Laboratory of Pulmonary Hypertension, Department of Cardiology, Traditional Chinese Hospital Affiliated Xinjiang Medical University, Urumqi, China
| | - Ehbal Tuyghun
- Laboratory of Pulmonary Physiology and Pathology, Department of Cardiology, Traditional Chinese Hospital Affiliated Xinjiang Medical University, Urumqi, China
| | - Yan Yang
- Department of Cardiology, Traditional Chinese Hospital Affiliated Xinjiang Medical University, Urumqi, China
| | - Paerhati Tuerxun
- Department of Cardiology, Traditional Chinese Hospital Affiliated Xinjiang Medical University, Urumqi, China
| | - Xiufen Li
- Department of Cardiology, Traditional Chinese Hospital Affiliated Xinjiang Medical University, Urumqi, China -
| |
Collapse
|
32
|
Vassiliou AG, Keskinidou C, Kotanidou A, Frantzeskaki F, Dimopoulou I, Langleben D, Orfanos SE. Decreased bone morphogenetic protein type II receptor and BMP-related signalling molecules' expression in aquaporin 1-silenced human pulmonary microvascular endothelial cells. Hellenic J Cardiol 2020; 62:84-86. [PMID: 32304816 DOI: 10.1016/j.hjc.2020.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- Alice G Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece
| | - Chrysi Keskinidou
- First Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece; Medical School of Democritus University of Thrace, 6(th) km Alexandroupolis-Dragana, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece; First Department of Critical Care Medicine & Pulmonary Services, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece
| | - Frantzeska Frantzeskaki
- Second Department of Critical Care, National & Kapodistrian University of Athens Medical School, "Attikon" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine & Pulmonary Services, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece
| | - David Langleben
- Center for Pulmonary Vascular Disease, Division of Cardiology, Azrieli Heart Center, and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece; First Department of Critical Care Medicine & Pulmonary Services, National & Kapodistrian University of Athens Medical School, Evangelismos Hospital, Athens, Greece; Second Department of Critical Care, National & Kapodistrian University of Athens Medical School, "Attikon" Hospital, Athens, Greece.
| |
Collapse
|
33
|
Chen J, Zhang H, Yu W, Chen L, Wang Z, Zhang T. Expression of pulmonary arterial elastin in rats with hypoxic pulmonary hypertension using H2S. J Recept Signal Transduct Res 2020; 40:383-387. [PMID: 32160810 DOI: 10.1080/10799893.2020.1738482] [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] [Indexed: 01/27/2023]
Abstract
Object: This study analyses the changes of pulmonary arterial elastin expression inhibited by hydrogen sulfide (H2S) in rats with hypoxic pulmonary hypertension.Method: The research used 30 healthy rats and randomly divided them into control group, hypoxia group, and hypoxia + sodium hydrosulfide group. Each group contains 10 samples. The right catheterization was selected to measure the mean pulmonary artery pressure (mPAP). The RV/LV + S ratio was calculated through separating the right ventricle and the left ventricle plus the interventricular septum. Optical microscopy was used to observe the changes of pulmonary vascular structure. The research used immunohistochemistry to express the levels of elastin and transforming growth factor beta (TGF-β).Results: The ratios of Mpap and RV/LV + S in the hypoxic group exceed the control group. The hypoxia + sodium hydrosulfide group (hypoxia + NaHS) is lower than the hypoxic group. In the hypoxic group, the elastic expressions of medium and small pulmonary artery smooth muscle cells exceed the control group. The expression of elastin in hypoxic + NaHS medium and small pulmonary artery smooth muscle cells is lower than that of the control group.The protein expression levels of α-SM-actin in muscle arterial smooth muscle of pulmonary arterioles in hypoxic group, control group and hypoxic + NaHS group were 49.84% + 6.27%, 56.84% + 6.38%, 23.82% + 3.84%, 27.51% + 3.24%, 29.00% + 4.05%, 34.72% + 3.38%.Conclusion: Hydrogen sulfide in rats with hypoxic pulmonary hypertension can inhibit the expression of elastin in its extracellular matrix, which also has remarkable regulation function in forming HPH and remodeling hypoxic pulmonary vascular structure.
Collapse
Affiliation(s)
- Juan Chen
- Department of gynaecology and obstetrics, Jinan No.1 people's Hospital, Jinan, People's Republic of China
| | - Haizhou Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University and Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Wancheng Yu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University and Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Lei Chen
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University and Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University and Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Tao Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University and Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
34
|
Zhao Y, Wang B, Zhang J, He D, Zhang Q, Pan C, Yuan Q, Shi Y, Tang H, Xu F, Wei S, Chen Y. ALDH2 (Aldehyde Dehydrogenase 2) Protects Against Hypoxia-Induced Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:2303-2319. [PMID: 31510791 DOI: 10.1161/atvbaha.119.312946] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Hypoxia-induced pulmonary hypertension (HPH) increases lipid peroxidation with generation of toxic aldehydes that are metabolized by detoxifying enzymes, including ALDH2 (aldehyde dehydrogenase 2). However, the role of lipid peroxidation and ALDH2 in HPH pathogenesis remain undefined. Approach and Results: To determine the role of lipid peroxidation and ALDH2 in HPH, C57BL/6 mice, ALDH2 transgenic mice, and ALDH2 knockout (ALDH2-/-) mice were exposed to chronic hypoxia, and recombinant tissue-specific ALDH2 overexpression adeno-associated viruses were introduced into pulmonary arteries via tail vein injection for ALDH2 overexpression. Human pulmonary artery smooth muscle cells were used to elucidate underlying mechanisms in vitro. Chronic hypoxia promoted lipid peroxidation due to the excessive production of reactive oxygen species and increased expression of lipoxygenases in lung tissues. 4-hydroxynonenal but not malondialdehyde level was increased in hypoxic lung tissues which might reflect differences in detoxifying enzymes. ALDH2 overexpression attenuated the development of HPH, whereas ALDH2 knockout aggravated it. Specific overexpression of ALDH2 using AAV1 (adeno-associated virus)-ICAM (intercellular adhesion molecule) 2p-ALDH2 and AAV2-SM22αp (smooth muscle 22 alpha)-ALDH2 viral vectors in pulmonary artery smooth muscle cells, but not endothelial cells, prevented the development of HPH. Hypoxia or 4-hydroxynonenal increased stabilization of HIF (hypoxia-inducible factor)-1α, phosphorylation of Drp1 (dynamin-related protein 1) at serine 616, mitochondrial fission, and pulmonary artery smooth muscle cells proliferation, whereas ALDH2 activation suppressed the latter 3. CONCLUSIONS Increased 4-hydroxynonenal level plays a critical role in the development of HPH. ALDH2 attenuates the development of HPH by regulating mitochondrial fission and smooth muscle cell proliferation suggesting ALDH2 as a potential new therapeutic target for pulmonary hypertension.
Collapse
Affiliation(s)
- Yu Zhao
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Bailu Wang
- Clinical Trial Center (B.W.), Qilu Hospital of Shandong University, Jinan
| | - Jian Zhang
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Dayu He
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Qun Zhang
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Chang Pan
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Qiuhuan Yuan
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Yinan Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (Y.S., H.T.)
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (Y.S., H.T.).,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangdong, China (H.T.)
| | - Feng Xu
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Shujian Wei
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| | - Yuguo Chen
- From the Department of Emergency and Chest Pain Center, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University (Y.Z., J.Z., D.H., Q.Z., C.P., Q.Y., F.X., S.W., Y.C.), Qilu Hospital of Shandong University, Jinan
| |
Collapse
|