1
|
Khan N, Kumar V, Li P, Schlapbach LJ, Boyd AW, Coulthard MG, Woodruff TM. Inhibiting Eph/ephrin signaling reduces vascular leak and endothelial cell dysfunction in mice with sepsis. Sci Transl Med 2024; 16:eadg5768. [PMID: 38657024 DOI: 10.1126/scitranslmed.adg5768] [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: 01/12/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Sepsis is a life-threatening disease caused by a dysregulated host response to infection, resulting in 11 million deaths globally each year. Vascular endothelial cell dysfunction results in the loss of endothelial barrier integrity, which contributes to sepsis-induced multiple organ failure and mortality. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors and their ephrin ligands play a key role in vascular endothelial barrier disruption but are currently not a therapeutic target in sepsis. Using a cecal ligation and puncture (CLP) mouse model of sepsis, we showed that prophylactic or therapeutic treatment of mice with EphA4-Fc, a decoy receptor and pan-ephrin inhibitor, resulted in improved survival and a reduction in vascular leak, lung injury, and endothelial cell dysfunction. EphA2-/- mice also exhibited reduced mortality and pathology after CLP compared with wild-type mice. Proteomics of plasma samples from mice with sepsis after CLP revealed dysregulation of a number of Eph/ephrins, including EphA2/ephrin A1. Administration of EphA4-Fc to cultured human endothelial cells pretreated with TNF-α or ephrin-A1 prevented loss of endothelial junction proteins, specifically VE-cadherin, with maintenance of endothelial barrier integrity. In children admitted to hospital with fever and suspected infection, we observed that changes in EphA2/ephrin A1 in serum samples correlated with endothelial and organ dysfunction. Targeting Eph/ephrin signaling may be a potential therapeutic strategy to reduce sepsis-induced endothelial dysfunction and mortality.
Collapse
Affiliation(s)
- Nemat Khan
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Pengcheng Li
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Luregn J Schlapbach
- Children's Intensive Care Research Program, Child Health Research Centre, University of Queensland, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zürich, University of Zürich, 8032 Zürich, Switzerland
| | - Andrew W Boyd
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia
| | - Mark G Coulthard
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
2
|
Qiao Z, Liao M, Xiao M, Luo S, Wang K, Niu M, Jiang H, Sun S, Xu G, Xu N, Xu Q, Liu Y. Ephrin B3 exacerbates colitis and colitis-associated colorectal cancer. Biochem Pharmacol 2024; 220:116004. [PMID: 38142837 DOI: 10.1016/j.bcp.2023.116004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Ephrin B3, a member of Eph/ephrin family, contributes to embryogenesis and carcinogenesis, but few studies have suggested whether this ligand has regulatory effect on colitis. This study was to determine whether ephrin B3 played a role in colitis and colonic carcinogenesis. Dextran sodium sulfate (DSS)-induced colitis and azoxymethane (AOM)/DSS-induced colitis-associated carcinogenesis model was established in Efnb3-deficient (Efnb3-/-) mice. Label-free quantitative proteomics were performed to identify the Efnb3-regulated proteins. Our results showed that Efnb3 knock out reduced the symptoms of DSS-induced colitis, such as disease activity index (DAI), inflammatory factors release, and dysfunction of the intestinal barrier. Quantitative proteomics revealed that Efnb3 regulated 95 proteins which clustered in the platelet degranulation, response to elevated platelet cytosolic Ca2+, MAPK signaling for integrins such as ITGB4. Furthermore, ephrin B3 inactived ITGB4/AKT signal pathway and then promoted epithelial barrier dysfunction. Simultaneously, ephrin B3 promoted Gremlin-1/NF-κB signal pathway and thereby increased inflammatory factors release. In addition, the higher level of Efnb3 in colon cancer patients is correlated with worse survival. Efnb3-/- mice exhibited susceptibility to AOM/DSS-induced colorectal cancer. Our finding discovered that Efnb3 played an important role in the development of colitis and colitis-associated colorectal cancer. Efnb3 deficiency improved the intestinal barrier by ITGB4 and suppressed inflammation via Gremlin-1/NF-κB signal pathway, which may provide a novel therapeutic strategy for the treatment of colitis and colitis-associated colorectal cancer.
Collapse
Affiliation(s)
- Zhen Qiao
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Min Liao
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mingyue Xiao
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Saiyan Luo
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kexin Wang
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mengxin Niu
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Honglv Jiang
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Suya Sun
- Department of Neurology, Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Anatomy, Histology and Embryology, Neuroscience Division, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guoqiang Xu
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - NanJie Xu
- Department of Anatomy, Histology and Embryology, Neuroscience Division, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiongming Xu
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yanli Liu
- Department of Pharmacognosy, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
3
|
Vincenzi M, Mercurio FA, Leone M. EPHA2 Receptor as a Possible Therapeutic Target in Viral Infections. Curr Med Chem 2024; 31:5670-5701. [PMID: 37828671 DOI: 10.2174/0109298673256638231003111234] [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: 04/06/2023] [Revised: 08/02/2023] [Accepted: 08/24/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND The receptor tyrosine kinase EphA2 plays a role in many diseases, like cancer, cataracts, and osteoporosis. Interestingly, it has also been linked to viral infections. OBJECTIVE Herein, current literature has been reviewed to clarify EphA2 functions in viral infections and explore its potential role as a target in antiviral drug discovery strategies. METHODS Research and review articles along with preprints connecting EphA2 to different viruses have been searched through PubMed and the web. Structures of complexes between EphA2 domains and viral proteins have been retrieved from the PDB database. RESULTS EphA2 assumes a key role in Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein Barr virus (EBV) infections by directly binding, through its ligand binding domain, viral glycoproteins. For human cytomegalovirus (HCMV), the role of EphA2 in maintaining virus latency state, through cooperation with specific viral proteins, has also been speculated. In certain cells, with high EphA2 expression levels, following ligand stimulation, receptor activation might contribute to severe symptoms accompanying a few viral infections, including lung injuries often related to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). CONCLUSION Since EphA2 works as a host receptor for certain viruses, it might be worth more deeply investigating known compounds targeting its extracellular ligand binding domain as antiviral therapeutics. Due to EphA2's function in inflammation, its possible correlation with SARS-CoV-2 cannot be excluded, but more experimental studies are needed in this case to undoubtedly attribute the role of this receptor in viral infections.
Collapse
Affiliation(s)
- Marian Vincenzi
- Institute of Biostructures and Bioimaging, National Research Council of Italy (CNR-IBB), Naples, Italy
| | - Flavia Anna Mercurio
- Institute of Biostructures and Bioimaging, National Research Council of Italy (CNR-IBB), Naples, Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging, National Research Council of Italy (CNR-IBB), Naples, Italy
| |
Collapse
|
4
|
Zeng J, Wu Q, Xiong S, Lu C, Zhang Z, Huang H, Xiong Y, Luo T. Inhibition of EphA2 protects against atherosclerosis by synergizing with statins to mitigate macrophage inflammation. Biomed Pharmacother 2023; 169:115885. [PMID: 37984301 DOI: 10.1016/j.biopha.2023.115885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
Statins are highly prevalent in patients with coronary artery disease. Statins exert their anti-inflammatory effects on the vascular wall and circulating levels of pro-inflammatory cytokines. However, increasing attention revealed the exacerbation of macrophage inflammation induced by statins, and a clear mechanistic explanation of whether the detrimental effects of statins on macrophage inflammatory phenotypes outweigh the beneficial effects is has not yet been established. Here, RNA-sequencing and RT-qPCR analyses demonstrated that statins significantly upregulated EphA2, Nlrp3, IL-1β and TNF-α expression in macrophages. Mechanistically, we found that atorvastatin reduced KLF4 binding to the EphA2 promoter using KLF4-chromatin immunoprecipitation, suppressed HDAC11-mediated deacetylation and subsequently led to enhanced EphA2 transcription. The 4D-label-free proteomics analysis further confirmed the upregulated EphA2 and inflammatory signals. Furthermore, the proinflammatory effect of atorvastatin was neutralized by an addition of recombinant Fc-ephrinA1, a selective Eph receptor tyrosine kinase inhibitor (ALW-II-41-27) or EphA2-silencing adenovirus (siEphA2). In vivo, EphA2 was identified a proatherogenic factor and apoE-/- mice placed on a high-fat diet following gastric gavage with atorvastatin exhibited a consistent elevation in EphA2 expression. We further observed that the transfection with siEphA2 in atorvastatin-treated mice significantly attenuated atherosclerotic plaque formation and abrogated statin-orchestrated macrophages proinflammatory genes expression as compared to that in atorvastatin alone. Increased plaque stability index was also observed following the addition of siEphA2, as evidenced by increased collagen and smooth muscle content and diminished lipid accumulation and macrophage infiltration. The data suggest that blockage of EphA2 provides an additional therapeutic benefit for further improving the anti-atherogenic effects of statins.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610014, China
| | - Qiao Wu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Shiqiang Xiong
- Department of Cardiology, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Cong Lu
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610014, China
| | - Zheng Zhang
- Department of Cardiology, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Hui Huang
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610014, China
| | - Yan Xiong
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610014, China
| | - Tiantian Luo
- Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610014, China.
| |
Collapse
|
5
|
Su JX, Li SJ, Zhou XF, Zhang ZJ, Yan Y, Liu SL, Qi Q. Chemotherapy-induced metastasis: molecular mechanisms and clinical therapies. Acta Pharmacol Sin 2023; 44:1725-1736. [PMID: 37169853 PMCID: PMC10462662 DOI: 10.1038/s41401-023-01093-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Chemotherapy, the most widely accepted treatment for malignant tumors, is dependent on cell death induced by various drugs including antimetabolites, alkylating agents, mitotic spindle inhibitors, antitumor antibiotics, and hormonal anticancer drugs. In addition to causing side effects due to non-selective cytotoxicity, chemotherapeutic drugs can initiate and promote metastasis, which greatly reduces their clinical efficacy. The knowledge of how they induce metastasis is essential for developing strategies that improve the outcomes of chemotherapy. Herein, we summarize the recent findings on chemotherapy-induced metastasis and discuss the underlying mechanisms including tumor-initiating cell expansion, the epithelial-mesenchymal transition, extracellular vesicle involvement, and tumor microenvironment alterations. In addition, the use of combination treatments to overcome chemotherapy-induced metastasis is also elaborated.
Collapse
Affiliation(s)
- Jin-Xuan Su
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Si-Jia Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhi-Jing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yu Yan
- Functional Experimental Teaching Center, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Song-Lin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment; MOE Key Laboratory of Tumor Molecular Biology; Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
6
|
Yan H, Wu W, Hu Y, Li J, Xu J, Chen X, Xu Z, Yang X, Yang B, He Q, Luo P. Regorafenib inhibits EphA2 phosphorylation and leads to liver damage via the ERK/MDM2/p53 axis. Nat Commun 2023; 14:2756. [PMID: 37179400 PMCID: PMC10182995 DOI: 10.1038/s41467-023-38430-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The hepatotoxicity of regorafenib is one of the most noteworthy concerns for patients, however the mechanism is poorly understood. Hence, there is a lack of effective intervention strategies. Here, by comparing the target with sorafenib, we show that regorafenib-induced liver injury is mainly due to its nontherapeutic target Eph receptor A2 (EphA2). EphA2 deficiency attenuated liver damage and cell apoptosis under regorafenib treatment in male mice. Mechanistically, regorafenib inhibits EphA2 Ser897 phosphorylation and reduces ubiquitination of p53 by altering the intracellular localization of mouse double minute 2 (MDM2) by affecting the extracellular signal-regulated kinase (ERK)/MDM2 axis. Meanwhile, we found that schisandrin C, which can upregulate the phosphorylation of EphA2 at Ser897 also has protective effect against the toxicity in vivo. Collectively, our findings identify the inhibition of EphA2 Ser897 phosphorylation as a key cause of regorafenib-induced hepatotoxicity, and chemical activation of EphA2 Ser897 represents a potential therapeutic strategy to prevent regorafenib-induced hepatotoxicity.
Collapse
Affiliation(s)
- Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wentong Wu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhuai Hu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China
- Laboratory of Fruit Quality Biology/Zhejiang Provincial Key Laboratory of Horticultural Plant Integrative Biology/The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou, 310058, China
| | - Jinjin Li
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jiangxin Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xueqin Chen
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, 310002, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310018, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
| |
Collapse
|
7
|
Pantzke J, Koch A, Zimmermann EJ, Rastak N, Offer S, Bisig C, Bauer S, Oeder S, Orasche J, Fiala P, Stintz M, Rüger CP, Streibel T, Di Bucchianico S, Zimmermann R. Processing of carbon-reinforced construction materials releases PM 2.5 inducing inflammation and (secondary) genotoxicity in human lung epithelial cells and fibroblasts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 98:104079. [PMID: 36796551 DOI: 10.1016/j.etap.2023.104079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Building demolition following domestic fires or abrasive processing after thermal recycling can release particles harmful for the environment and human health. To mimic such situations, particles release during dry-cutting of construction materials was investigated. A reinforcement material consisting of carbon rods (CR), carbon concrete composite (C³) and thermally treated C³ (ttC³) were physicochemically and toxicologically analyzed in monocultured lung epithelial cells, and co-cultured lung epithelial cells and fibroblasts at the air-liquid interface. C³ particles reduced their diameter to WHO fibre dimensions during thermal treatment. Caused by physical properties or by polycyclic aromatic hydrocarbons and bisphenol A found in the materials, especially the released particles of CR and ttC³ induced an acute inflammatory response and (secondary) DNA damage. Transcriptome analysis indicated that CR and ttC³ particles carried out their toxicity via different mechanisms. While ttC³ affected pro-fibrotic pathways, CR was mostly involved in DNA damage response and in pro-oncogenic signaling.
Collapse
Affiliation(s)
- Jana Pantzke
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Arne Koch
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany
| | - Elias J Zimmermann
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Narges Rastak
- Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Svenja Offer
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Christoph Bisig
- Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Stefanie Bauer
- Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jürgen Orasche
- Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Petra Fiala
- Department of Mechanical Process Engineering, Technical University of Dresden, 01187 Dresden, Germany
| | - Michael Stintz
- Department of Mechanical Process Engineering, Technical University of Dresden, 01187 Dresden, Germany
| | - Christopher P Rüger
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Department Life, Light & Matter (LLM), University of Rostock, 18051 Rostock, Germany
| | - Thorsten Streibel
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center, Chair of Analytical Chemistry, University of Rostock, 18059 Rostock, Germany; Joint Mass Spectrometry Center, Comprehensive Molecular Analytics, Helmholtz Zentrum München, 85764 Neuherberg, Germany; Department Life, Light & Matter (LLM), University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
8
|
Shin JM, Han MS, Park JH, Lee SH, Kim TH, Lee SH. The EphA1 and EphA2 Signaling Modulates the Epithelial Permeability in Human Sinonasal Epithelial Cells and the Rhinovirus Infection Induces Epithelial Barrier Dysfunction via EphA2 Receptor Signaling. Int J Mol Sci 2023; 24:ijms24043629. [PMID: 36835041 PMCID: PMC9962399 DOI: 10.3390/ijms24043629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Deficiencies in epithelial barrier integrity are involved in the pathogenesis of chronic rhinosinusitis (CRS). This study aimed to investigate the role of ephrinA1/ephA2 signaling on sinonasal epithelial permeability and rhinovirus-induced epithelial permeability. This role in the process of epithelial permeability was evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to rhinovirus infection. EphrinA1 treatment increased epithelial permeability, which was associated with decreased expression of ZO-1, ZO-2, and occludin. These effects of ephrinA1 were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. Furthermore, rhinovirus infection upregulated the expression levels of ephrinA1 and ephA2, increasing epithelial permeability, which was suppressed in ephA2-deficient cells. These results suggest a novel role of ephrinA1/ephA2 signaling in epithelial barrier integrity in the sinonasal epithelium, suggesting their participation in rhinovirus-induced epithelial dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | - Sang Hag Lee
- Correspondence: ; Tel.: +82-2-920-5486; Fax: +82-2-925-5233
| |
Collapse
|
9
|
Stigmasterol protects human brain microvessel endothelial cells against ischemia-reperfusion injury through suppressing EPHA2 phosphorylation. Chin J Nat Med 2023; 21:127-135. [PMID: 36871980 DOI: 10.1016/s1875-5364(23)60390-5] [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: 09/17/2022] [Indexed: 03/07/2023]
Abstract
Stigmasterol is a plant sterol with anti-apoptotic, anti-oxidative and anti-inflammatory effect through multiple mechanisms. In this study, we further assessed whether it exerts protective effect on human brain microvessel endothelial cells (HBMECs) against ischemia-reperfusion injury and explored the underlying mechanisms. HBMECs were used to establish an in vitro oxygen and glucose deprivation/reperfusion (OGD/R) model, while a middle cerebral artery occlusion (MCAO) model of rats were constructed. The interaction between stigmasterol and EPHA2 was detected by surface plasmon resonance (SPR) and cellular thermal shift assay (CETSA). The results showed that 10 μmol·L-1 stigmasterol significantly protected cell viability, alleviated the loss of tight junction proteins and attenuated the blood-brain barrier (BBB) damage induced by OGD/R in thein vitro model. Subsequent molecular docking showed that stigmasterol might interact with EPHA2 at multiple sites, including T692, a critical gatekeep residue of this receptor. Exogenous ephrin-A1 (an EPHA2 ligand) exacerbated OGD/R-induced EPHA2 phosphorylation at S897, facilitated ZO-1/claudin-5 loss, and promoted BBB leakage in vitro, which were significantly attenuated after stigmasterol treatment. The rat MCAO model confirmed these protective effects in vivo. In summary, these findings suggest that stigmasterol protects HBMECs against ischemia-reperfusion injury by maintaining cell viability, reducing the loss of tight junction proteins, and attenuating the BBB damage. These protective effects are at least meditated by its interaction with EPHA2 and inhibitory effect on EPHA2 phosphorylation.
Collapse
|
10
|
Ieguchi K, Funakoshi M, Mishima T, Takizawa K, Omori T, Nakamura F, Watanabe M, Tsuji M, Kiuchi Y, Kobayashi S, Tsunoda T, Maru Y, Wada S. The Sympathetic Nervous System Contributes to the Establishment of Pre-Metastatic Pulmonary Microenvironments. Int J Mol Sci 2022; 23:ijms231810652. [PMID: 36142564 PMCID: PMC9501257 DOI: 10.3390/ijms231810652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Emerging evidence suggests that neural activity contributes to tumor initiation and its acquisition of metastatic properties. More specifically, it has been reported that the sympathetic nervous system regulates tumor angiogenesis, tumor growth, and metastasis. The function of the sympathetic nervous system in primary tumors has been gradually elucidated. However, its functions in pre-metastatic environments and/or the preparation of metastatic environments far from the primary sites are still unknown. To investigate the role of the sympathetic nervous system in pre-metastatic environments, we performed chemical sympathectomy using 6-OHDA in mice and observed a decrease in lung metastasis by attenuating the recruitment of myeloid-derived suppressor cells. Furthermore, we note that neuro-immune cell interactions could be observed in tumor-bearing mouse lungs in conjunction with the decreased expression of Sema3A. These data indicate that the sympathetic nervous system contributes to the preparation of pre-metastatic microenvironments in the lungs, which are mediated by neuro-immune cell interactions.
Collapse
Affiliation(s)
- Katsuaki Ieguchi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
| | - Masabumi Funakoshi
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Department of Peripheral Nervous System Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Taishi Mishima
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Kohtaro Takizawa
- Department of Biochemistry, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Tsutomu Omori
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Fumio Nakamura
- Department of Biochemistry, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Makoto Watanabe
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Mayumi Tsuji
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Shinichi Kobayashi
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
| | - Takuya Tsunoda
- Department of Medicine, Division of Medical Oncology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (Y.M.); (S.W.); Tel.: +81-3-5269-7417 (Y.M.); +81-3-3300-5257 (S.W.)
| | - Satoshi Wada
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Medicine, Division of Medical Oncology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Correspondence: (Y.M.); (S.W.); Tel.: +81-3-5269-7417 (Y.M.); +81-3-3300-5257 (S.W.)
| |
Collapse
|
11
|
Macrophage-Targeted Nanomedicines for ARDS/ALI: Promise and Potential. Inflammation 2022; 45:2124-2141. [PMID: 35641717 PMCID: PMC9154210 DOI: 10.1007/s10753-022-01692-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/11/2022] [Accepted: 05/24/2022] [Indexed: 11/05/2022]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are characterized by progressive lung impairment typically triggered by inflammatory processes. The mortality toll for ARDS/ALI yet remains high because of the poor prognosis, lack of disease-specific inflammation management therapies, and prolonged hospitalizations. The urgency for the development of new effective therapeutic strategies has become acutely evident for patients with coronavirus disease 2019 (COVID-19) who are highly susceptible to ARDS/ALI. We propose that the lack of target specificity in ARDS/ALI of current treatments is one of the reasons for poor patient outcomes. Unlike traditional therapeutics, nanomedicine offers precise drug targeting to inflamed tissues, the capacity to surmount pulmonary barriers, enhanced interactions with lung epithelium, and the potential to reduce off-target and systemic adverse effects. In this article, we focus on the key cellular drivers of inflammation in ARDS/ALI: macrophages. We propose that as macrophages are involved in the etiology of ARDS/ALI and regulate inflammatory cascades, they are a promising target for new therapeutic development. In this review, we offer a survey of multiple nanomedicines that are currently being investigated with promising macrophage targeting potential and strategies for pulmonary delivery. Specifically, we will focus on nanomedicines that have shown engagement with proinflammatory macrophage targets and have the potential to reduce inflammation and reverse tissue damage in ARDS/ALI.
Collapse
|
12
|
Siswanto FM, Tamura A, Sakuma R, Imaoka S. Yeast β-glucan increases etoposide sensitivity in lung cancer cell line A549 by suppressing Nrf2 via the non-canonical NF-κB pathway. Mol Pharmacol 2022; 101:257-273. [PMID: 35193967 DOI: 10.1124/molpharm.121.000475] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022] Open
Abstract
Etoposide is regarded as one of the main standard cytotoxic drugs for lung cancer. However, mutations in Keap1, the main regulator of nuclear factor erythroid 2-related factor 2 (Nrf2), are often detected in lung cancer and lead to chemoresistance. Since the aberrant activation of Nrf2 enhances drug resistance, the suppression of the Nrf2 pathway is a promising therapeutic strategy for lung cancer. We herein used the human lung adenocarcinoma cell line A549 because it harbors a Keap1 loss-of-function mutation. A treatment with β-glucan, a major component of the fungal cell wall, reduced Nrf2 protein levels, down-regulated the expression of CYP3A5, UGT1A1, and MDR1, and increased etoposide sensitivity in A549 cells. Furthermore, the ephrin type-A receptor 2 (EphA2) receptor was important for the recognition and biological activity of β-glucan in A549 cells. EphA2 signaling includes nuclear factor kappa B (NF-κB), STAT3, and p38 mitogen-activated protein kinase (MAPK). However, treatment of cells with stattic (STAT3 inhibitor) or SB203580 (p38 MAPK inhibitor) did not diminish the effects of β-glucan. In contrast, knockdown of RelB abolished the effects of β-glucan, suggesting the involvement of the non-canonical NF-κB pathway. The β-glucan effects were also attenuated by the knockdown of WDR23. The β-glucan treatment and RelB overexpression induced the expression of CUL4A, which increased WDR23 ligase activity and promoted the subsequent depletion of Nrf2. These results revealed a novel property of β-glucan as a resistance-modifying agent in addition to its widely reported immunomodulatory effects for lung cancer therapy via the EphA2-RelB-CUL4A-Nrf2 axis. Significance Statement Chemotherapeutic resistance remains a major obstacle in cancer therapy despite extensive efforts to elucidate the underlying molecular mechanisms and overcome multidrug resistance. The present study revealed a novel resistance-modifying property of β-glucan, thereby expanding our knowledge on the beneficial roles of β-glucan and providing an alternative strategy to prevent drug resistance by cancer. The present results provide evidence for the involvement of a novel mode of NF-κB and Nrf2 crosstalk in the drug resistance phenotype.
Collapse
Affiliation(s)
- Ferbian Milas Siswanto
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Japan
| | - Akiyoshi Tamura
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Japan
| | - Rika Sakuma
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Japan
| | - Susumu Imaoka
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Japan
| |
Collapse
|
13
|
Cholenic acid derivative UniPR1331 impairs tumor angiogenesis via blockade of VEGF/VEGFR2 in addition to Eph/ephrin. Cancer Gene Ther 2022; 29:908-917. [PMID: 34426652 PMCID: PMC9293752 DOI: 10.1038/s41417-021-00379-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/12/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting ones, is crucial for tumor growth and metastatization, and is considered a promising therapeutic target. Unfortunately, drugs directed against a specific proangiogenic growth factor or receptor turned out to be of limited benefit for oncology patients, likely due to the high biochemical redundancy of the neovascularization process. In this scenario, multitarget compounds that are able to simultaneously tackle different proangiogenic pathways are eagerly awaited. UniPR1331 is a 3β-hydroxy-Δ5-cholenic acid derivative, which is already known to inhibit Eph-ephrin interaction. Here, we employed an analysis pipeline consisting of molecular modeling and simulation, surface plasmon resonance spectrometry, biochemical assays, and endothelial cell models to demonstrate that UniPR1331 directly interacts with the vascular endothelial growth factor receptor 2 (VEGFR2) too. The binding of UniPR1331 to VEGFR2 prevents its interaction with the natural ligand vascular endothelial growth factor and subsequent autophosphorylation, signal transduction, and in vitro proangiogenic activation of endothelial cells. In vivo, UniPR1331 inhibits tumor cell-driven angiogenesis in zebrafish. Taken together, these data shed light on the pleiotropic pharmacological effect of UniPR1331, and point to Δ5-cholenic acid as a promising molecular scaffold for the development of multitarget antiangiogenic compounds.
Collapse
|
14
|
Lee SH, Kang SH, Han MS, Kwak JW, Kim HG, Lee TH, Lee DB, Kim TH. The Expression of ephrinA1/ephA2 Receptor Increases in Chronic Rhinosinusitis and ephrinA1/ephA2 Signaling Affects Rhinovirus-Induced Innate Immunity in Human Sinonasal Epithelial Cells. Front Immunol 2021; 12:793517. [PMID: 34975898 PMCID: PMC8716742 DOI: 10.3389/fimmu.2021.793517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
EphA2 receptor and its ephrin ligands are involved in virus infection, epithelial permeability, and chemokine secretion. We hypothesized that ephrinA1/ephA2 signaling participates in rhinovirus (RV)-induced antiviral immune response in sinonasal mucosa of patients with chronic rhinosinusitis (CRS). Therefore, we investigated the expression of ephrinA1/ephA2 in normal and inflamed sinonasal mucosa and evaluated whether they regulate chemokine secretion and the production of antiviral immune mediators including interferons (IFNs) in RV-infected human primary sinonasal epithelial cells. For this purpose, the expression and distribution of ephrinA1/ephA2 in sinonasal mucosa were evaluated with RT-qPCR, immunofluorescence, and western blot. Their roles in chemokine secretion and the production of antiviral immune mediators such as type I and III IFNs, and interferon stimulated genes were evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to RV and poly(I:C). We found that ephrinA1/ephA2 were expressed in normal mucosa and their levels increased in inflamed sinonasal mucosa of CRS patients. RV infection or poly(I:C) treatment induced chemokine secretion which were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. The production of antiviral immune mediators enhanced by rhinovirus or poly (I:C) is increased by blocking ephA2 compared with that of cells stimulated by either rhinovirus or poly(I:C) alone. In addition, blocking ephA2 attenuated RV replication in cultured cells. Taken together, these results describe a novel role of ephrinA1/ephA2 signaling in antiviral innate immune response in sinonasal epithelium, suggesting their participation in RV-induced development and exacerbations of CRS.
Collapse
Affiliation(s)
- Sang Hag Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Middleton JD, Sivakumar S, Hai T. Chemotherapy-Induced Changes in the Lung Microenvironment: The Role of MMP-2 in Facilitating Intravascular Arrest of Breast Cancer Cells. Int J Mol Sci 2021; 22:10280. [PMID: 34638621 PMCID: PMC8508901 DOI: 10.3390/ijms221910280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 11/18/2022] Open
Abstract
Previously, we showed that mice treated with cyclophosphamide (CTX) 4 days before intravenous injection of breast cancer cells had more cancer cells in the lung at 3 h after cancer injection than control counterparts without CTX. At 4 days after its injection, CTX is already excreted from the mice, allowing this pre-treatment design to reveal how CTX may modify the lung environment to indirectly affect cancer cells. In this study, we tested the hypothesis that the increase in cancer cell abundance at 3 h by CTX is due to an increase in the adhesiveness of vascular wall for cancer cells. Our data from protein array analysis and inhibition approach combined with in vitro and in vivo assays support the following two-prong mechanism. (1) CTX increases vascular permeability, resulting in the exposure of the basement membrane (BM). (2) CTX increases the level of matrix metalloproteinase-2 (MMP-2) in mouse serum, which remodels the BM and is functionally important for CTX to increase cancer abundance at this early stage. The combined effect of these two processes is the increased accessibility of critical protein domains in the BM, resulting in higher vascular adhesiveness for cancer cells to adhere. The critical protein domains in the vascular microenvironment are RGD and YISGR domains, whose known binding partners on cancer cells are integrin dimers and laminin receptor, respectively.
Collapse
Affiliation(s)
- Justin D. Middleton
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| | - Subhakeertana Sivakumar
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
| | - Tsonwin Hai
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (J.D.M.); (S.S.)
- Molecular, Cellular, and Developmental Biology Program, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
16
|
EphA2 signaling within integrin adhesions regulates fibrillar adhesion elongation and fibronectin deposition. Matrix Biol 2021; 103-104:1-21. [PMID: 34537369 DOI: 10.1016/j.matbio.2021.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 12/28/2022]
Abstract
The multifunctional glycoprotein fibronectin influences several crucial cellular processes and contributes to multiple pathologies. While a link exists between fibronectin-associated pathologies and the receptor tyrosine kinase EphA2, the mechanism by which EphA2 promotes fibronectin matrix remodeling remains unknown. We previously demonstrated that EphA2 deletion reduces smooth muscle fibronectin deposition and blunts fibronectin deposition in atherosclerosis without influencing fibronectin expression. We now show that EphA2 expression is required for contractility-dependent elongation of tensin- and α5β1 integrin-rich fibrillar adhesions that drive fibronectin fibrillogenesis. Mechanistically, EphA2 localizes to integrin adhesions where focal adhesion kinase mediates ligand-independent Y772 phosphorylation, and mutation of this site significantly blunts fibrillar adhesion length. EphA2 deficiency decreases smooth muscle cell contractility by enhancing p190RhoGAP activation and reducing RhoA activity, whereas stimulating RhoA signaling in EphA2 deficient cells rescues fibrillar adhesion elongation. Together, these data identify EphA2 as a novel regulator of fibrillar adhesion elongation and provide the first data identifying a role for EphA2 signaling in integrin adhesions.
Collapse
|
17
|
Ruiz P, Emond C, McLanahan ED, Joshi-Barr S, Mumtaz M. Exploring Mechanistic Toxicity of Mixtures Using PBPK Modeling and Computational Systems Biology. Toxicol Sci 2021; 174:38-50. [PMID: 31851354 DOI: 10.1093/toxsci/kfz243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Mixtures risk assessment needs an efficient integration of in vivo, in vitro, and in silico data with epidemiology and human studies data. This involves several approaches, some in current use and others under development. This work extends the Agency for Toxic Substances and Disease Registry physiologically based pharmacokinetic (PBPK) toolkit, available for risk assessors, to include a mixture PBPK model of benzene, toluene, ethylbenzene, and xylenes. The recoded model was evaluated and applied to exposure scenarios to evaluate the validity of dose additivity for mixtures. In the second part of this work, we studied toluene, ethylbenzene, and xylene (TEX)-gene-disease associations using Comparative Toxicogenomics Database, pathway analysis and published microarray data from human gene expression changes in blood samples after short- and long-term exposures. Collectively, this information was used to establish hypotheses on potential linkages between TEX exposures and human health. The results show that 236 genes expressed were common between the short- and long-term exposures. These genes could be central for the interconnecting biological pathways potentially stimulated by TEX exposure, likely related to respiratory and neuro diseases. Using publicly available data we propose a conceptual framework to study pathway perturbations leading to toxicity of chemical mixtures. This proposed methodology lends mechanistic insights of the toxicity of mixtures and when experimentally validated will allow data gaps filling for mixtures' toxicity assessment. This work proposes an approach using current knowledge, available multiple stream data and applying computational methods to advance mixtures risk assessment.
Collapse
Affiliation(s)
- Patricia Ruiz
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia
| | - Claude Emond
- BioSimulation Consulting, Inc., Newark, Delaware
| | - Evad D McLanahan
- Division of Community Health Investigations, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia
| | | | - Moiz Mumtaz
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, Atlanta, Georgia
| |
Collapse
|
18
|
Li P, Wang L, Li P, Hu F, Cao Y, Tang D, Ye G, Li H, Wang D. Silencing of long non-coding RNA XIST represses gastric cancer progression through blocking NFκB pathway via inhibiting HNF4A-mediated transcription of EPHA1. Cancer Gene Ther 2021; 28:307-320. [PMID: 33199830 DOI: 10.1038/s41417-020-00220-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/05/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is a common cancer and a leading cause of cancer-related deaths worldwide. Recent studies have supported the important role of long non-coding RNAs (lncRNAs) in GC progression. This study identified functional significance of X inactive specific transcript (XIST) in GC. The expression of XIST and EPHA1 in GC tissues and cells was measured. Then, dual luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and Chromatin Immunoprecipitation (ChIP) assay were performed to explore the interaction among XIST, EPHA1 and HNF4A. The effects of XIST on GG progression were evaluated by determining expression of proliferation- and invasion-related proteins (Ki67, PCNA, MMP-2, and MMP-9). Further, the functional role of XIST in GC with the involvement of NFκB pathway was also analyzed. Subsequently, the tumor growth in nude mice was evaluated. High expression of XIST and EPHA1 was observed in GC. XIST elevated EPHA1 expression by recruiting HNF4A. In addition, silencing of XIST inhibited GC progression in vitro and in vivo. Overexpressed XIST and EPHA1 yielded a reversed effect on cell proliferation and invasion. SN50 treatment (inhibitor of NFκB pathway) counteracted the promotive effect on GC cell proliferation and invasion mediated by XIST. The present study unveils that XIST increases the enrichment of HNF4A in the promoter region of EPHA1, thus promoting the deterioration of GC.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation/physiology
- Disease Progression
- Hepatocyte Nuclear Factor 4/antagonists & inhibitors
- Hepatocyte Nuclear Factor 4/metabolism
- Heterografts
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- RNA, Long Noncoding/antagonists & inhibitors
- RNA, Long Noncoding/biosynthesis
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Receptor, EphA1/genetics
- Receptor, EphA1/metabolism
- Signal Transduction
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Transcription, Genetic
Collapse
Affiliation(s)
- Ping Li
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of Experimental Surgery-Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University, Mannheim, 68167, Germany
| | - Liuhua Wang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| | - Pengfei Li
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
| | - Fangyong Hu
- Department of Central Laboratory, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
- Department of General Surgery, Huaian Tumor Hospital & Huaian Hospital of Huaian City, Huaian, 223200, PR China
| | - Yi Cao
- Department of Experimental Surgery-Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University, Mannheim, 68167, Germany
| | - Dong Tang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| | - Gang Ye
- Department of General Surgery, Jiangdu People's Hospital of Yangzhou, Yangzhou, 225200, PR China
| | - Hongbo Li
- Department of General Surgery, Jiangdu People's Hospital of Yangzhou, Yangzhou, 225200, PR China.
| | - Daorong Wang
- Department of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Institute of General Surgery-Yangzhou, Yangzhou University, Yangzhou, 225000, PR China
| |
Collapse
|
19
|
Tian D, Qin Q, Li M, Li X, Xu Q, Lv Q. Homocysteine Impairs Endothelial Cell Barrier Function and Angiogenic Potential via the Progranulin/EphA2 Pathway. Front Pharmacol 2021; 11:614760. [PMID: 33510642 PMCID: PMC7836014 DOI: 10.3389/fphar.2020.614760] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022] Open
Abstract
Hyperhomocysteinemia is a well-recognized independent risk factor for cardiovascular disease. To date, the mechanism of pathological plasma homocysteine (Hcy) level elevation remains to be elucidated. We aimed to investigate the levels of progranulin (PGRN), Eph-receptor tyrosine kinase-type A2 (EphA2), vascular cell adhesion molecule-1 (VCAM-1), and Hcy in patients with arteriosclerosis and investigate their functions in Hcy-injured human umbilical vein endothelial cells (HUVECs). EphA2 knockdown was induced in HUVECs by shRNA lentivirus infection with EphA2-RNAi, and bulk RNA-seq assay was performed. Then we investigated the mechanism underlying the effect of recombinant human PGRN (rhPGRN) combined with shRNA interference of EphA2 on cell proliferation, migration, and angiogenesis in Hcy-injured HUVECs. Results showed that serum EphA2, VCAM-1, and Hcy levels in acute coronary syndrome patients were significantly higher than those in chronic coronary syndrome patients (p = 0.000; p = 0.000; p = 0.033, respectively). In vitro, we demonstrated that knockdown of EphA2 significantly impaired cell adhesion and inhibited HUVECs migration and angiogenesis (p < 0.001), which was associated with reduction in VCAM1 and VE-cadherin (p < 0.05). Hcy modulated the expression of PGRN and EphA2 in a time-and dose-dependent manner. However, rhPGRN ameliorated the Hcy-induced reduction in cell viability and migration (p < 0.05). Mechanistically, we found that PGRN/EphA2 and its downstream AKT/NF-κB signaling might be the primary signal transduction pathways underlying Hcy-induced injury. The present study illustrated that PGRN plays a previously unrecognized role in Hcy-induced endothelial injury, which is achieved through its interaction with EphA2 signaling, implying a promising therapeutic target for cardiovascular disease.
Collapse
Affiliation(s)
- Dan Tian
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Disease, Shanghai, China
| | - Mingfei Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Disease, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Xu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianzhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Lee JY, Stevens RP, Kash M, Zhou C, Koloteva A, Renema P, Paudel SS, Stevens T. KD025 Shifts Pulmonary Endothelial Cell Bioenergetics and Decreases Baseline Lung Permeability. Am J Respir Cell Mol Biol 2020; 63:519-530. [PMID: 32628869 PMCID: PMC7528923 DOI: 10.1165/rcmb.2019-0435oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
KD025 is a ROCK2 inhibitor currently being tested in clinical trials for the treatment of fibrotic lung diseases. The therapeutic effects of KD025 are partly due to its inhibition of profibrotic pathways and fat metabolism. However, whether KD025 affects pulmonary microvascular endothelial cell (PMVEC) function is unknown, despite evidence that alveolar-capillary membrane disruption constitutes major causes of death in fibrotic lung diseases. We hypothesized that KD025 regulates PMVEC metabolism, pH, migration, and survival, a series of interrelated functional characteristics that determine pulmonary barrier integrity. We used PMVECs isolated from Sprague Dawley rats. KD025 dose-dependently decreased lactate production and glucose consumption. The inhibitory effect of KD025 was more potent compared with other metabolic modifiers, including 2-deoxy-glucose, extracellular acidosis, dichloroacetate, and remogliflozin. Interestingly, KD025 increased oxidative phosphorylation, whereas 2-deoxy-glucose did not. KD025 also decreased intracellular pH and induced a compensatory increase in anion exchanger 2. KD025 inhibited PMVEC migration, but fasudil (nonspecific ROCK inhibitor) did not. We tested endothelial permeability in vivo using Evans Blue dye in the bleomycin pulmonary fibrosis model. Baseline permeability was decreased in KD025-treated animals independent of bleomycin treatment. Under hypoxia, KD025 increased PMVEC necrosis as indicated by increased lactate dehydrogenase release and propidium iodide uptake and decreased ATP; it did not affect Annexin V binding. ROCK2 knockdown had no effect on PMVEC metabolism, pH, and migration, but it increased nonapoptotic caspase-3 activity. Together, we report that KD025 promotes oxidative phosphorylation; decreases glycolysis, intracellular pH, and migration; and strengthens pulmonary barrier integrity in a ROCK2-independent manner.
Collapse
Affiliation(s)
- Ji Young Lee
- Department of Physiology and Cell Biology
- Department of Internal Medicine
- Division of Pulmonary and Critical Care Medicine
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Reece P. Stevens
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Mary Kash
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Chun Zhou
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Anna Koloteva
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Phoibe Renema
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Sunita S. Paudel
- Department of Physiology and Cell Biology
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Physiology and Cell Biology
- Department of Internal Medicine
- Center for Lung Biology
- College of Medicine, and
- University of South Alabama, Mobile, Alabama
| |
Collapse
|
21
|
Vreeken D, Zhang H, van Zonneveld AJ, van Gils JM. Ephs and Ephrins in Adult Endothelial Biology. Int J Mol Sci 2020; 21:ijms21165623. [PMID: 32781521 PMCID: PMC7460586 DOI: 10.3390/ijms21165623] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022] Open
Abstract
Eph receptors and their ephrin ligands are important guidance molecules during neurological and vascular development. In recent years, it has become clear that the Eph protein family remains functional in adult physiology. A subset of Ephs and ephrins is highly expressed by endothelial cells. As endothelial cells form the first barrier between the blood and surrounding tissues, maintenance of a healthy endothelium is crucial for tissue homeostasis. This review gives an overview of the current insights of the role of ephrin ligands and receptors in endothelial function and leukocyte recruitment in the (patho)physiology of adult vascular biology.
Collapse
|
22
|
Chen Y, Zhang L, Zhang Y, Bai T, Song J, Qian W, Hou X. EphrinA1/EphA2 Promotes Epithelial Hyperpermeability Involving in Lipopolysaccharide-induced Intestinal Barrier Dysfunction. J Neurogastroenterol Motil 2020; 26:397-409. [PMID: 32606260 PMCID: PMC7329149 DOI: 10.5056/jnm19095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022] Open
Abstract
Background/Aims Lipopolysaccharide (LPS) is the key factor inducing mucosal and systemic inflammation in various intestinal and parenteral diseases, which could initially disrupt the epithelial barrier function. EphrinA1/ephA2 is speculated to increase the epithelial permeability for its "repulsive interaction" between adjacent cells. This study aim to investigate the role of ephrinA1/ephA2 in LPS-induced epithelial hyperpermeability. Methods In vivo model challenged with oral LPS in C57BL/6 mice and in vitro model exposed to LPS in Caco2 monolayer were established. The barrier function was assessed including expression of tight junction proteins (occludin and claudin-1), transepithelial electrical resistance, and permeability to macromolecules (fluorescein isothiocyanate-labeled fluorescent dextran 4 kDa [FD4]). Moreover, the expression and phosphorylation of ephrinA1/ephA2 were quantified, and its roles in the process of epithelial barrier disruption were confirmed via stimulating ephA2 with ephrinA1-Fc chimera (ephrinA1-Fc) and inactivating ephA2 with ephA2-Fc chimera (ephA2-Fc), or ephA2 monoclonal antibody (ephA2-mab), as well as inhibiting extracellular signal-regulated kinase 1/2 (ERK1/2) with PD98059. Results LPS induced significant barrier dysfunction with dismissed occludin and claudin-1 expression, reduced transepithelial electrical resistance and increased FD4 permeability, accompanied by upregulated ephrinA1/ephA2 pathway and phosphorylation of ephA2 receptor. Furthermore, ephA2-Fc, and ephA2-mab ameliorated LPS-induced epithelial hyperpermeability, which was also inhibited by PD98059. Additionally, ephrinA1-Fc led to apparent epithelial leakage in Caco2 monolayer by promoting the phosphorylation of ERK1/2, which could be obviously blocked by ephA2-mab and PD98059. Conclusion EphrinA1/ephA2 promotes epithelial hyperpermeability with an ERK1/2-dependent pathway, which involves in LPS-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Yuhua Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongbo Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Gastrointestinal Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Zhang A, Xing J, Xia T, Zhang H, Fang M, Li S, Du Y, Li XC, Zhang Z, Zeng MS. EphA2 phosphorylates NLRP3 and inhibits inflammasomes in airway epithelial cells. EMBO Rep 2020; 21:e49666. [PMID: 32352641 DOI: 10.15252/embr.201949666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 12/16/2022] Open
Abstract
Inflammasomes are intracellular complexes that form in the cytosol of inflammatory cells. NLRP3 is one of the sensor proteins in the complex that can recognize a wide variety of stimuli ranging from microbial components to environmental particulates. Here, we report that in mouse airway epithelial cells (AECs), inflammasome activation is inhibited by EphA2, a member of the transmembrane tyrosine kinase receptor family, via tyrosine phosphorylation of NLRP3 in a model of reovirus infection. We find that EphA2 depletion markedly enhances interleukin-1β (IL-1β) and interleukin-18 (IL-18) production in response to the virus. EphA2-/- mice show stronger inflammatory infiltration and enhanced inflammasome activation upon viral infection, and aggravated asthma symptoms upon ovalbumin (ova) induction. Mechanistically, EphA2 binds to NLRP3 and induces its phosphorylation at Tyr132, thereby interfering with ASC speck formation and blocking the activation of the NLRP3-inflammasome. These data demonstrate that reovirus employs EphA2 to suppress inflammasome activation in AECs and that EphA2 deficiency causes a pathological exacerbation of asthma in an ova-induced asthma model.
Collapse
Affiliation(s)
- Ao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Junji Xing
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Tianliang Xia
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hua Zhang
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mingli Fang
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.,Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Shibing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yong Du
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Xian C Li
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.,Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science Center and Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.,Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
24
|
Park BH, Shin MH, Douglas IS, Chung KS, Song JH, Kim SY, Kim EY, Jung JY, Kang YA, Chang J, Kim YS, Park MS. Erythropoietin-Producing Hepatoma Receptor Tyrosine Kinase A2 Modulation Associates with Protective Effect of Prone Position in Ventilator-induced Lung Injury. Am J Respir Cell Mol Biol 2019; 58:519-529. [PMID: 29216437 DOI: 10.1165/rcmb.2017-0143oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The erythropoietin-producing hepatoma (Eph) receptor tyrosine kinase A2 (EphA2) and its ligand, ephrinA1, play a pivotal role in inflammation and tissue injury by modulating the epithelial and endothelial barrier integrity. Therefore, EphA2 receptor may be a potential therapeutic target for modulating ventilator-induced lung injury (VILI). To support this hypothesis, here, we analyzed EphA2/ephrinA1 signaling in the process of VILI and determined the role of EphA2/ephrinA1 signaling in the protective mechanism of prone positioning in a VILI model. Wild-type mice were ventilated with high (24 ml/kg; positive end-expiratory pressure, 0 cm; 5 h) tidal volume in a supine or prone position. Anti-EphA2 receptor antibody or IgG was administered to the supine position group. Injury was assessed by analyzing the BAL fluid, lung injury scoring, and transmission electron microscopy. Lung lysates were evaluated using cytokine/chemokine ELISA and Western blotting of EphA2, ephrinA1, PI3Kγ, Akt, NF-κB, and P70S6 kinase. EphA2/ephrinA1 expression was higher in the supine high tidal volume group than in the control group, but it did not increase upon prone positioning or anti-EphA2 receptor antibody treatment. EphA2 antagonism reduced the extent of VILI and downregulated the expression of PI3Kγ, Akt, NF-κB, and P70S6 kinase. These findings demonstrate that EphA2/ephrinA1 signaling is involved in the molecular mechanism of VILI and that modulation of EphA2/ehprinA1 signaling by prone position or EphA2 antagonism may be associated with the lung-protective effect. Our data provide evidence for EphA2/ehprinA1 as a promising therapeutic target for modulating VILI.
Collapse
Affiliation(s)
- Byung Hoon Park
- 1 Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Gyeonggi Provincial Medical Center Paju Hospital, Paju City, Gyeonggi-Do, Republic of Korea
| | - Mi Hwa Shin
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Ivor S Douglas
- 3 Division of Pulmonary Sciences and Critical Care Medicine, Denver Health Medical Center, University of Colorado School of Medicine, Denver, Colorado
| | - Kyung Soo Chung
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Joo Han Song
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Song Yee Kim
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Eun Young Kim
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Ji Ye Jung
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Young Ae Kang
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Joon Chang
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Young Sam Kim
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| | - Moo Suk Park
- 2 Division of Pulmonology, The Institute of Chest Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; and
| |
Collapse
|
25
|
Wu B, Rockel JS, Lagares D, Kapoor M. Ephrins and Eph Receptor Signaling in Tissue Repair and Fibrosis. Curr Rheumatol Rep 2019; 21:23. [PMID: 30980212 DOI: 10.1007/s11926-019-0825-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Fibrosis is a pathological feature of many human diseases that affect multiple organs. The development of anti-fibrotic therapies has been a difficult endeavor due to the complexity of signaling pathways associated with fibrogenic processes, complicating the identification and modulation of specific targets. Evidence suggests that ephrin ligands and Eph receptors are crucial signaling molecules that contribute to physiological wound repair and the development of tissue fibrosis. Here, we discuss recent advances in the understanding of ephrin and Eph signaling in tissue repair and fibrosis. RECENT FINDINGS Ephrin-B2 is implicated in fibrosis of multiple organs. Intercepting its signaling may help counteract fibrosis. Ephrins and Eph receptors are candidate mediators of fibrosis. Ephrin-B2, in particular, promotes fibrogenic processes in multiple organs. Thus, therapeutic strategies targeting Ephrin-B2 signaling could yield new ways to treat organ fibrosis.
Collapse
Affiliation(s)
- Brian Wu
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jason S Rockel
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mohit Kapoor
- The Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. .,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
Feng G, Sun B, Liu HX, Liu QH, Zhao L, Wang TL. EphA2 antagonism alleviates LPS-induced acute lung injury via Nrf2/HO-1, TLR4/MyD88 and RhoA/ROCK pathways. Int Immunopharmacol 2019; 72:176-185. [PMID: 30986645 DOI: 10.1016/j.intimp.2019.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 01/09/2023]
Abstract
Eph receptor tyrosine kinases have a wide range of biological functions and have gradually been recognized increasingly as key regulators of inflammation and injury diseases. Although previous studies suggested that EphA2 receptor may be involved in the regulation of inflammation and vascular permeability in injured lung, the detailed effects of EphA2 on LPS-induced acute lung injury (ALI) are still inadequate and the underlying mechanism remains poorly understood. In this study, we detected the effects of EphA2 antagonism on inflammation, pulmonary vascular permeability and oxidative stress in LPS-induced ALI and investigate the potential mechanism. Our results showed that EphA2 antagonism markedly inhibited the cytokines release and inflammatory cells infiltration in BALF, prevented the LPS-induced elevations of MPO activity and MDA level in lung tissues. Our study also found that EphA2 antagonism significantly decreased the wet/dry ratios, reduced the Evans blue albumin extravasation in lung tissues and obviously alleviated the LPS-induced increment of pulmonary vascular permeability. Mechanistically, EphA2 antagonism significantly increased the activation of Nrf2 along with its target antioxidant enzyme HO-1 and inhibited the expressions of TLR4/MyD88 in lung tissues and A549 alveolar epithelial cells. Furthermore, EphA2 antagonism dramatically inhibited the LPS-evoked activations of RhoA/ROCK in lung tissues. In conclusion, our data indicate that EphA2 receptor plays an essential role in LPS-induced ALI and EphA2 antagonism has protective effects against LPS-induced ALI via Nrf2/HO-1, TLR4/MyD88 and RhoA/ROCK pathways. These results suggest that antagonism of EphA2 may be an effective therapeutic strategy for the treatment of ALI.
Collapse
Affiliation(s)
- Guang Feng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Bo Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, PR China
| | - Hai-Xia Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Qing-Hai Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Lei Zhao
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Tian-Long Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China.
| |
Collapse
|
27
|
Ieguchi K, Maru Y. Roles of EphA1/A2 and ephrin-A1 in cancer. Cancer Sci 2019; 110:841-848. [PMID: 30657619 PMCID: PMC6398892 DOI: 10.1111/cas.13942] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/22/2022] Open
Abstract
The biological functions of the Eph/ephrin system have been intensively investigated and well documented so far since its discovery in 1987. Although the Eph/ephrin system has been implicated in pathological settings such as Alzheimer's disease and cancer, the molecular mechanism of the Eph/ephrin system in those diseases is not well understood. Especially in cancer, recent studies have demonstrated that most of Eph and ephrin are up‐ or down‐regulated in various types of cancer, and have been implicated in tumor progression, tumor malignancy, and prognosis. However, they lack consistency and are in controversy. The localization patterns of EphA1 and EphA2 in mouse lungs are very similar, and both knockout mice showed similar phenotypes in the lungs. Ephrin‐A1 that is a membrane‐anchored ligand for EphAs was co‐localized with EphA1 and EphA2 in lung vascular endothelial cells. We recently uncovered the molecular mechanism of ephrin‐A1‐induced lung metastasis by understanding the physiological function of ephrin‐A1 in lungs. This review focuses on the function of EphA1, EphA2, and ephrin‐A1 in tumors and an establishment of pre‐metastatic microenvironment in the lungs.
Collapse
Affiliation(s)
- Katsuaki Ieguchi
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| |
Collapse
|
28
|
Patil MA, Upadhyay AK, Hernandez-Lagunas L, Good R, Carpenter TC, Sucharov CC, Nozik-Grayck E, Kompella UB. Targeted delivery of YSA-functionalized and non-functionalized polymeric nanoparticles to injured pulmonary vasculature. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1059-S1066. [PMID: 30450979 DOI: 10.1080/21691401.2018.1528984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ephrin type-A receptor 2 (EphA2) is a transmembrane receptor which is upregulated in injured lungs, including those treated with bleomycin. YSA peptide (YSAYPDSVPMMS), a mimic of ephrin ligands, binds to EphA2 receptors on cell surface with high affinity. In this study, we assessed the ability of YSA-functionalized and non-functionalized poly (dl-lactide-co-glycolide) (PLGA) nanoparticles to enhance delivery to bleomycin treated cultured vascular endothelial cells and, in a bleomycin induced lung injury mouse model. Nanoparticles were loaded with a lipophilic fluorescent dye. Human umbilical vein endothelial cells (HUVEC) with or without 2-day bleomycin pretreatment (25 µg/ml) and adult mice with or without intratracheal instillation of bleomycin (0.1 U) were dosed with nanoparticles. Mice received nanoparticles via tail vein injection 4 days after bleomycin treatment. Three days after nanoparticle injection, tissues (lung, heart, kidney, spleen, liver, brain, eyes and whole blood) were harvested and quantified for fluorescence using IVIS imaging. Mean particle uptake increased with time and concentration for both types of particles in HUVEC, with the uptake being higher for YSA-functionalized nanoparticles. Bleomycin treatment increased the 3-h uptake of both types of nanoparticles in HUVEC by about two-fold, with the YSA-functionalized nanoparticle uptake being 1.66-fold compared to non-functionalized nanoparticles (p < .05). In mice, bleomycin injury resulted in 2.3- and 4.7-fold increase in the lung levels of non-functionalized and YSA-functionalized nanoparticles (p < .05), respectively, although the differences between the two particle types were not significant. In conclusion, PLGA nanoparticle delivery to cultured vascular endothelial cells and mouse lungs in vivo is higher following bleomycin treatment, with the delivery tending to be higher for YSA functionalized nanoparticles.
Collapse
Affiliation(s)
- Madhoosudan A Patil
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Arun K Upadhyay
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Laura Hernandez-Lagunas
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA
| | - Ryan Good
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA
| | - Todd C Carpenter
- c Division of Pediatric Critical Care Medicine, Department of Pediatrics , University of Colorado School of Medicine , Aurora , CO , USA
| | - Carmen C Sucharov
- d Division of Cardiology, Department of Medicine , University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| | - Eva Nozik-Grayck
- b Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine , University of Colorado Denver , Denver , CO , USA.,c Division of Pediatric Critical Care Medicine, Department of Pediatrics , University of Colorado School of Medicine , Aurora , CO , USA
| | - Uday B Kompella
- a Department of Pharmaceutical Sciences , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,e Department of Ophthalmology , University of Colorado Anschutz Medical Campus , Aurora , Colorado.,f Department of Bioengineering , University of Colorado Anschutz Medical Campus , Aurora , CO , USA.,g Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus , Aurora , CO , USA
| |
Collapse
|
29
|
Good RJ, Hernandez-Lagunas L, Allawzi A, Maltzahn JK, Vohwinkel CU, Upadhyay AK, Kompella UB, Birukov KG, Carpenter TC, Sucharov CC, Nozik-Grayck E. MicroRNA dysregulation in lung injury: the role of the miR-26a/EphA2 axis in regulation of endothelial permeability. Am J Physiol Lung Cell Mol Physiol 2018; 315:L584-L594. [PMID: 30024304 PMCID: PMC6230876 DOI: 10.1152/ajplung.00073.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that regulate gene expression in many diseases, although the contribution of miRNAs to the pathophysiology of lung injury remains obscure. We hypothesized that dysregulation of miRNA expression drives the changes in key genes implicated in the development of lung injury. To test our hypothesis, we utilized a model of lung injury induced early after administration of intratracheal bleomycin (0.1 U). Wild-type mice were treated with bleomycin or PBS, and lungs were collected at 4 or 7 days. A profile of lung miRNA was determined by miRNA array and confirmed by quantitative PCR and flow cytometry. Lung miR-26a was significantly decreased 7 days after bleomycin injury, and, on the basis of enrichment of predicted gene targets, it was identified as a putative regulator of cell adhesion, including the gene targets EphA2, KDR, and ROCK1, important in altered barrier function. Lung EphA2 mRNA, and protein increased in the bleomycin-injured lung. We further explored the miR-26a/EphA2 axis in vitro using human lung microvascular endothelial cells (HMVEC-L). Cells were transfected with miR-26a mimic and inhibitor, and expression of gene targets and permeability was measured. miR-26a regulated expression of EphA2 but not KDR or ROCK1. Additionally, miR-26a inhibition increased HMVEC-L permeability, and the disrupted barrier integrity due to miR-26a was blocked by EphA2 knockdown, shown by VE-cadherin staining. Our data suggest that miR-26a is an important epigenetic regulator of EphA2 expression in the pulmonary endothelium. As such, miR-26a may represent a novel therapeutic target in lung injury by mitigating EphA2-mediated changes in permeability.
Collapse
Affiliation(s)
- Ryan J. Good
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Laura Hernandez-Lagunas
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Ayed Allawzi
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Joanne K. Maltzahn
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Christine U. Vohwinkel
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Arun K. Upadhyay
- 4Department of Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Uday B. Kompella
- 4Department of Pharmaceutical Sciences, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Konstantin G. Birukov
- 5Department of Anesthesiology and Medicine, University of Maryland, Baltimore, Maryland
| | - Todd C. Carpenter
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Carmen C. Sucharov
- 3Cardiology, Department of Pediatrics and Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| | - Eva Nozik-Grayck
- 1Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado,2Pediatric Critical Care Medicine, University of Colorado Denver Anschutz Medical Center, Aurora, Colorado
| |
Collapse
|
30
|
Funk SD, Finney AC, Yurdagul A, Pattillo CB, Orr AW. EphA2 stimulates VCAM-1 expression through calcium-dependent NFAT1 activity. Cell Signal 2018; 49:30-38. [PMID: 29793020 DOI: 10.1016/j.cellsig.2018.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023]
Abstract
Endothelial cell activation by proinflammatory stimuli drives leukocyte recruitment through enhanced expression of counter-receptors such as vascular cell adhesion molecule-1 (VCAM-1). We previously demonstrated that activation of the receptor tyrosine kinase EphA2 with its ligand ephrin-A1 induces VCAM-1 expression. Here, we sought to characterize the proinflammatory signaling pathways involved. Analysis of over-represented transcription factors in ephrin-A1-induced genes identified multiple potential transcriptional regulators, including the Rel family members nuclear factor-κB (NF-κB/p65) and nuclear factor of activated T-cells (NFAT). While ephrin-A1 failed to induce endothelial NF-κB activation, NF-κB inhibitors prevented ephrin-A1-induced VCAM-1 expression, suggesting basal NF-κB activity is required. In contrast, ephrin-A1 induced a robust EphA2-dependent increase in NFAT activation, and mutation of the NF-κB/NFAT-binding sites in the VCAM-1 promoter blunted ephrin-A1-induced promoter activity. NFAT activation classically occurs through calcium-dependent calcineurin activation, and inhibiting NFAT signaling with calcineurin inhibitors (cyclosporine A, FK506) or direct NFAT inhibitors (A-285222) was sufficient to block ephrin-A1-induced VCAM-1 expression. Consistent with robust NFAT activation, ephrin-A1-induced an EphA2-dependent calcium influx in endothelial cells that was required for ephrin-A1-induced NFAT activation and VCAM-1 expression. This work provides the first data showing EphA2-dependent calcium influx and NFAT activation and identifies NFAT as a novel EphA2-dependent proinflammatory pathway in endothelial activation.
Collapse
Affiliation(s)
- Steven Daniel Funk
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States; Department of Internal Medicine, Renal Division, Washington University, St. Louis, MO 63110, United States
| | - Alexandra C Finney
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States
| | - Arif Yurdagul
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States; Department of Medicine, Columbia University, New York, NY 10027, United States
| | - Christopher B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States
| | - A Wayne Orr
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States; Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, United States.
| |
Collapse
|
31
|
Zeng L, Li K, Wei H, Hu J, Jiao L, Yu S, Xiong Y. A Novel EphA2 Inhibitor Exerts Beneficial Effects in PI-IBS in Vivo and in Vitro Models via Nrf2 and NF-κB Signaling Pathways. Front Pharmacol 2018; 9:272. [PMID: 29662452 PMCID: PMC5890185 DOI: 10.3389/fphar.2018.00272] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Though the detailed pathological mechanism of post-infectious irritable bowel syndrome (PI-IBS) remains unclear, accumulating evidence indicates that oxidative stress and inflammation are implicated in the process of PI-IBS. Oxidative stress and inflammation are regulated by Nrf2 and NF-κB signaling pathways, respectively. EphA2, a member of Eph receptor family, promotes oxidative stress and inflammatory responses via regulation of Nrf2 and NF-κB signaling pathways in various types of human diseases. Understanding the mechanisms by which EphA2 regulate oxidative stress and inflammation in PI-IBS is important for the development of new strategies to treat PI-IBS. However, the effects of ALW-II-41-27, a novel EphA2 inhibitor on PI-IBS and the underlying molecular mechanisms have never been studied. In the present study, we showed that ALW-II-41-27 decreased gastrointestinal motility and abdominal withdrawal reflex (AWR) scores, markedly reduced the levels of oxidative stress markers [4-hydroxy-2-nonenal (4-HNE), protein carbonyl, and 8-hydroxy-2-de-axyguanine (8-OHdG)] and proinflammatory cytokines (TNF-α, IL-6, IL-17, and ICAM-1), and remarkably increased the level of anti-inflammatory cytokine (IL-10) in serum and colon of Trichinella spiralis-infected mice. Moreover, ALW-II-41-27 was effective in suppressing oxidative stress and inflammation in LPS-treated NCM460 colonic cells. Treatment of ALW-II-41-27 reversed the activation of NF-κB and inactivation of Nrf2 in LPS-treated NCM460 cells. Importantly, these protective effects of ALW-II-41-27 were partially inhibited by EphA2 KO and abolished by EphA2 overexpression. In conclusion, EphA2 may represent a promising therapeutic target for patients with PI-IBS and ALW-II-41-27 might function as a novel therapeutic agent for PI-IBS.
Collapse
Affiliation(s)
- Li Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Kaixue Li
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Hong Wei
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Jingjing Hu
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Lu Jiao
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| | - Shaoyong Yu
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ying Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
32
|
Ma T, Liu X, Cen Z, Xin C, Guo M, Zou C, Song W, Xie R, Wang K, Zhou H, Zhang J, Wang Z, Bian C, Cui K, Li J, Wei YQ, Li J, Zhou X. MicroRNA-302b negatively regulates IL-1β production in response to MSU crystals by targeting IRAK4 and EphA2. Arthritis Res Ther 2018; 20:34. [PMID: 29482609 PMCID: PMC5828083 DOI: 10.1186/s13075-018-1528-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/26/2018] [Indexed: 02/05/2023] Open
Abstract
Background Interleukin-1β (IL-1β) is a pivotal proinflammatory cytokine that is strongly associated with the inflammation of gout. However, the underlying mechanism through which the production of IL-1β is regulated has not been fully elucidated. Our previous work identified that miR-302b had an important immune regulatory role in bacterial lung infections. This study was conducted to evaluate the function of miR-302b on monosodium urate (MSU) crystal-induced inflammation and its mechanism. Methods The expression pattern and the immune-regulatory role of miR-302b were evaluated both in vitro and in vivo. The functional targets of miR-302b were predicted by bioinformatics, and then validated by genetic approaches. In addition, the clinical feature of miR-302b was analyzed using serum samples of patients with gouty arthritis. Results The extremely high expression of miR-302b was observed in both macrophages and mouse air membranes treated with MSU. Intriguingly, overexpression of miR-302b regulated NF-κB and caspase-1 signaling, leading to significantly attenuate MSU-induced IL-1β. By genetic analysis, miR-302b exhibited inhibitory function on IRAK4 and EphA2 by binding to their 3′-UTR regions. Corporately silencing IRAK4 and EphA2 largely impaired MSU-induced IL-1β protein production. Moreover, it was also found that miR-302b and EphA2 suppressed the migration of macrophages. Finally, it was observed that high expression of miR-302b was a general feature in patients with gouty arthritis. Conclusions These results suggest that miR-302b can regulate IL-1β production in MSU-induced inflammation by targeting NF-κB and caspase-1 signaling, and may be a potential therapeutic target for gouty arthritis. Electronic supplementary material The online version of this article (10.1186/s13075-018-1528-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teng Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xiao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhifu Cen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mingfeng Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Chaoyu Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Wenpeng Song
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Rou Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Kailun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Jun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Ce Bian
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaijun Cui
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yu-Quan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
33
|
Lee SH, Shin JH, Song JH, Leem AY, Park MS, Kim YS, Chang J, Chung KS. Clinical implications of the plasma EphA2 receptor level in critically ill patients with septic shock. Sci Rep 2017; 7:17612. [PMID: 29242524 PMCID: PMC5730544 DOI: 10.1038/s41598-017-17909-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 12/04/2017] [Indexed: 12/25/2022] Open
Abstract
The Eph/ephrin receptor ligand system is known to play a role in inflammation induced by infection, injury, and inflammatory diseases. The present study aimed to evaluate plasma EphA2 receptor levels in critically ill patients with sepsis. This study was a prospective cohort study evaluating samples and clinical data from the medical intensive care unit (MICU) of a 2000-bed university tertiary referral hospital in South Korea. Positive correlations of the plasma EphA2 receptor level with the acute physiology and chronic health evaluation (APACHE) II score and the sequential organ failure assessment (SOFA) score were observed. The area under the curve (AUC) for the plasma EphA2 receptor level on a receiver operating characteristic curve was 0.690 (95% confidence interval [CI], 0.608-0.764); the AUCs for the APACHE II score and SOFA scores were 0.659 (95% CI, 0.576-0.736) and 0.745 (95% CI, 0.666-0.814), respectively. A Cox proportional hazard model identified an association between an increased plasma EphA2 receptor level (>51.5 pg mL-1) and increased risk of 28-day mortality in the MICU (hazard ratio = 3.22, 95% CI, 1.709-6.049). An increased plasma EphA2 receptor level was associated with sepsis severity and 28-day mortality among sepsis patients.
Collapse
Affiliation(s)
- Su Hwan Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hye Shin
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joo Han Song
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ah Young Leem
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moo Suk Park
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Sam Kim
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Chang
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Soo Chung
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Institute of Chest Diseases, Yonsei University College of Medicine, Seoul, Korea.
- Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
34
|
All-trans retinoic acid attenuates bleomycin-induced pulmonary fibrosis via downregulating EphA2-EphrinA1 signaling. Biochem Biophys Res Commun 2017; 491:721-726. [PMID: 28743499 DOI: 10.1016/j.bbrc.2017.07.122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 01/19/2023]
Abstract
The role of all-trans retinoic acid (ATRA) in pulmonary fibrosis is relatively unknown, although this metabolite modulates cell differentiation, proliferation, and development. We aimed to evaluate the role of ATRA in bleomycin-induced pulmonary fibrosis, and whether the mechanism involves EphA2-EphrinA1 and PI3K-Akt signaling. We evaluated three groups of mice: a control group (intraperitoneal DMSO injection 3 times weekly after PBS instillation), bleomycin group (intraperitoneal DMSO injection 3 times weekly after bleomycin instillation), and bleomycin + ATRA group (intraperitoneal ATRA injection 3 times weekly after bleomycin instillation). The cell counts and protein concentration in the bronchoalveolar lavage fluid (BALF), changes in histopathology, Ashcroft score, hydroxyproline assay, expression of several signal pathway proteins including EphA2-EphrinA1, and PI3K-Akt, and cytokine levels were compared among the groups. We found that bleomycin significantly increased the protein concentration in the BALF, Ashcroft score in lung tissue, and hydroxyproline contents in lung lysates. Furthermore, bleomycin upregulated EphA2, EphrinA1, PI3K 110γ, Akt, IL-6 and TNF-α. However, administration of ATRA attenuated the upregulation of EphA2-EphrinA1 and PI3K-Akt after bleomycin instillation, and decreased pulmonary fibrosis. In addition, ATRA suppressed IL-6 and TNF-α production induced by bleomycin-induced injury. Collectively, these data suggest that ATRA attenuates bleomycin-induced pulmonary fibrosis by regulating EphA2-EphrinA1 and PI3K-Akt signaling.
Collapse
|
35
|
Finney AC, Funk SD, Green JM, Yurdagul A, Rana MA, Pistorius R, Henry M, Yurochko A, Pattillo CB, Traylor JG, Chen J, Woolard MD, Kevil CG, Orr AW. EphA2 Expression Regulates Inflammation and Fibroproliferative Remodeling in Atherosclerosis. Circulation 2017; 136:566-582. [PMID: 28487392 DOI: 10.1161/circulationaha.116.026644] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/03/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Atherosclerotic plaque formation results from chronic inflammation and fibroproliferative remodeling in the vascular wall. We previously demonstrated that both human and mouse atherosclerotic plaques show elevated expression of EphA2, a guidance molecule involved in cell-cell interactions and tumorigenesis. METHODS Here, we assessed the role of EphA2 in atherosclerosis by deleting EphA2 in a mouse model of atherosclerosis (Apoe-/-) and by assessing EphA2 function in multiple vascular cell culture models. After 8 to 16 weeks on a Western diet, male and female mice were assessed for atherosclerotic burden in the large vessels, and plasma lipid levels were analyzed. RESULTS Despite enhanced weight gain and plasma lipid levels compared with Apoe-/- controls, EphA2-/-Apoe-/- knockout mice show diminished atherosclerotic plaque formation, characterized by reduced proinflammatory gene expression and plaque macrophage content. Although plaque macrophages express EphA2, EphA2 deletion does not affect macrophage phenotype, inflammatory responses, and lipid uptake, and bone marrow chimeras suggest that hematopoietic EphA2 deletion does not affect plaque formation. In contrast, endothelial EphA2 knockdown significantly reduces monocyte firm adhesion under flow. In addition, EphA2-/-Apoe-/- mice show reduced progression to advanced atherosclerotic plaques with diminished smooth muscle and collagen content. Consistent with this phenotype, EphA2 shows enhanced expression after smooth muscle transition to a synthetic phenotype, and EphA2 depletion reduces smooth muscle proliferation, mitogenic signaling, and extracellular matrix deposition both in atherosclerotic plaques and in vascular smooth muscle cells in culture. CONCLUSIONS Together, these data identify a novel role for EphA2 in atherosclerosis, regulating both plaque inflammation and progression to advanced atherosclerotic lesions. Cell culture studies suggest that endothelial EphA2 contributes to atherosclerotic inflammation by promoting monocyte firm adhesion, whereas smooth muscle EphA2 expression may regulate the progression to advanced atherosclerosis by regulating smooth muscle proliferation and extracellular matrix deposition.
Collapse
Affiliation(s)
- Alexandra C Finney
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Steven D Funk
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Jonette M Green
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Arif Yurdagul
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Mohammad Atif Rana
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Rebecca Pistorius
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Miriam Henry
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Andrew Yurochko
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Christopher B Pattillo
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - James G Traylor
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Jin Chen
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Matthew D Woolard
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Christopher G Kevil
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - A Wayne Orr
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.).
| |
Collapse
|
36
|
Ghori A, Freimann FB, Nieminen-Kelhä M, Kremenetskaia I, Gertz K, Endres M, Vajkoczy P. EphrinB2 Activation Enhances Vascular Repair Mechanisms and Reduces Brain Swelling After Mild Cerebral Ischemia. Arterioscler Thromb Vasc Biol 2017; 37:867-878. [PMID: 28254815 DOI: 10.1161/atvbaha.116.308620] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/15/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Cerebral edema caused by the disruption of the blood-brain barrier is a major complication after stroke. Therefore, strategies to accelerate and enhance neurovascular recovery after stroke are of prime interest. Our main aim was to study the role of ephrinB2/EphB4 signaling in mediating the vascular repair and in blood-brain barrier restoration after mild cerebral ischemia occlusion/reperfusion. APPROACH AND RESULTS Here, we show that the guidance molecule ephrinB2 plays a key role in neurovascular protection and blood-brain barrier restoration after stroke. In a focal stroke model, we characterize the stroke-induced damage to cerebral blood vessels and their subsequent endogenous repair on a cellular, molecular, and functional level. EphrinB2 and its tyrosine kinase receptor EphB4 are upregulated early after stroke by endothelial cells and perivascular support cells, in parallel to their reassembly during neurovascular recovery. Using both retroviral and pharmacological approaches, we show that the inhibition of ephrinB2/EphB4 signaling suppresses post-middle cerebral artery occlusion neurovascular repair mechanisms resulting in an aggravation of brain swelling. In contrast, the activation of ephrinB2 after brain ischemia leads to an increased pericyte recruitment and increased endothelial-pericyte interaction, resulting in an accelerated neurovascular repair after ischemia. CONCLUSIONS We show that reducing swelling could result in improved outcome because of reduction in damaged brain tissue. We also identify a novel role for ephrinB2/EphB4 signaling in the maintenance of the neurovascular homeostasis and provide a novel therapeutic approach in reducing brain swelling after stroke.
Collapse
Affiliation(s)
- Adnan Ghori
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Florian B Freimann
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Melina Nieminen-Kelhä
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Irina Kremenetskaia
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Karen Gertz
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Matthias Endres
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.)
| | - Peter Vajkoczy
- From the Department of Neurosurgery (A.G., F.B.F., M.N.-K., I.K., P.V.), Center for Stroke Research (K.G., M.E., P.V.), Department of Neurology (M.E.), and German Center for Neurodegenerative Diseases (M.E.), Charité, Universitätsmedizin Berlin, Germany; German Center for Cardiovascular Research (DZHK), Max-Delbrück-Centrum Für Molekulare Medizin Berlin-Buch, Germany (M.E.); and Freie Universität Berlin, Germany (A.G.).
| |
Collapse
|
37
|
EPHA4-FC TREATMENT REDUCES ISCHEMIA/REPERFUSION-INDUCED INTESTINAL INJURY BY INHIBITING VASCULAR PERMEABILITY. Shock 2016; 45:184-91. [PMID: 26771935 DOI: 10.1097/shk.0000000000000494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The inflammatory response is characterized by increased endothelial permeability, which permits the passage of fluid and inflammatory cells into interstitial spaces. The Eph/ephrin receptor ligand system plays a role in inflammation through a signaling cascade, which modifies Rho-GTPase activity. We hypothesized that blocking Eph/ephrin signaling using an EphA4-Fc would result in decreased inflammation and tissue injury in a model of ischemia/reperfusion (I/R) injury. Mice undergoing intestinal I/R pretreated with the EphA4-Fc had significantly reduced intestinal injury compared to mice injected with the control Fc. This reduction in I/R injury was accompanied by significantly reduced neutrophil infiltration, but did not affect intestinal inflammatory cytokine generation. Using microdialysis, we identified that intestinal I/R induced a marked increase in systemic vascular leakage, which was completely abrogated in EphA4-Fc-treated mice. Finally, we confirmed the direct role of Eph/ephrin signaling in endothelial leakage by demonstrating that EphA4-Fc inhibited tumor necrosis factor-α-induced vascular permeability in human umbilical vein endothelial cells. This study identifies that Eph/ephrin interaction induces proinflammatory signaling in vivo by inducing vascular leak and neutrophil infiltration, which results in tissue injury in intestinal I/R. Therefore, therapeutic targeting of Eph/ephrin interaction using inhibitors, such as EphA4-Fc, may be a novel method to prevent tissue injury in acute inflammation by influencing endothelial integrity and by controlling vascular leak.
Collapse
|
38
|
Shiuan E, Chen J. Eph Receptor Tyrosine Kinases in Tumor Immunity. Cancer Res 2016; 76:6452-6457. [PMID: 27811149 DOI: 10.1158/0008-5472.can-16-1521] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022]
Abstract
The family of Eph receptor tyrosine kinases and their ephrin ligands regulate a diverse array of physiologic processes, such as axonal guidance, bone remodeling, and immune cell development and trafficking. Eph/ephrin interactions have also been implicated in various pathologic processes, including inflammation, cancer, and tumor angiogenesis. Because Eph receptors play prominent roles in both the immune system and cancer, they likely impact the tumor immune microenvironment, an area in which Eph receptors remain understudied. Here, we provide the first comprehensive review of Eph receptors in the context of tumor immunity. With the recent rise of cancer immunotherapies as promising therapeutic interventions, further elucidation of the roles of Eph receptors in the tumor immune microenvironment will be critical for understanding and developing novel targets against tumor immune evasion. Cancer Res; 76(22); 6452-7. ©2016 AACR.
Collapse
Affiliation(s)
- Eileen Shiuan
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee
| | - Jin Chen
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee. .,Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Cell & Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
39
|
Inhibition of EphA2/EphrinA1 signal attenuates lipopolysaccharide-induced lung injury. Clin Sci (Lond) 2016; 130:1993-2003. [PMID: 27549114 DOI: 10.1042/cs20160360] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/22/2016] [Indexed: 01/30/2023]
Abstract
Eph-Ephrin signalling mediates various cellular processes, including vasculogenesis, angiogenesis, cell migration, axon guidance, fluid homoeostasis and repair after injury. Although previous studies have demonstrated that stimulation of the EphA receptor induces increased vascular permeability and inflammatory response in lung injury, the detailed mechanisms of EphA2 signalling are unknown. In the present study, we evaluated the role of EphA2 signalling in mice with lipopolysaccharide (LPS)-induced lung injury. Acute LPS exposure significantly up-regulated EphA2 and EphrinA1 expression. Compared with LPS+IgG mice (IgG instillation after LPS exposure), LPS+EphA2 mAb mice [EphA2 monoclonal antibody (mAb) instillation posttreatment after LPS exposure] had attenuated lung injury and reduced cell counts and protein concentration of bronchoalveolar lavage fluid (BALF). EphA2 mAb posttreatment down-regulated the expression of phosphoinositide 3-kinases (PI3K) 110γ, phospho-Akt, phospho-NF-κB p65, phospho-Src and phospho-S6K in lung lysates. In addition, inhibiting the EphA2 receptor augmented the expression of E-cadherin, which is involved in cell-cell adhesion. Our study identified EphA2 receptor as an unrecognized modulator of several signalling pathways-including PI3K-Akt-NF-kB, Src-NF-κB, E-cadherin and mTOR-in LPS-induced lung injury. These results suggest that EphA2 receptor inhibitors may function as novel therapeutic agents for LPS-induced lung injury.
Collapse
|
40
|
Hong JY, Shin MH, Chung KS, Kim EY, Jung JY, Kang YA, Kim YS, Kim SK, Chang J, Park MS. EphA2 Receptor Signaling Mediates Inflammatory Responses in Lipopolysaccharide-Induced Lung Injury. Tuberc Respir Dis (Seoul) 2015; 78:218-26. [PMID: 26175775 PMCID: PMC4499589 DOI: 10.4046/trd.2015.78.3.218] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/30/2015] [Accepted: 04/27/2015] [Indexed: 01/14/2023] Open
Abstract
Background Eph receptors and ephrin ligands have several functions including angiogenesis, cell migration, axon guidance, fluid homeostasis, oncogenesis, inflammation and injury repair. The EphA2 receptor potentially mediates the regulation of vascular permeability and inflammation in response to lung injury. Methods Mice were divided into 3 experimental groups to study the role of EphA2 signaling in the lipopolysaccharide (LPS)-induced lung injury model i.e., IgG+phosphate-buffered saline (PBS) group (IgG instillation before PBS exposure), IgG+LPS group (IgG instillation before LPS exposure) and EphA2 monoclonal antibody (mAb)+LPS group (EphA2 mAb pretreatment before LPS exposure). Results EphA2 and ephrinA1 were upregulated in LPS-induced lung injury. The lung injury score of the EphA2 mAb+LPS group was lower than that of the IgG+LPS group (4.30±2.93 vs. 11.45±1.20, respectively; p=0.004). Cell counts (EphA2 mAb+LPS: 11.33×104±8.84×104 vs. IgG+LPS: 208.0×104±122.6×104; p=0.018) and total protein concentrations (EphA2 mAb+LPS: 0.52±0.41 mg/mL vs. IgG+LPS: 1.38±1.08 mg/mL; p=0.192) were decreased in EphA2 mAb+LPS group, as compared to the IgG+LPS group. In addition, EphA2 antagonism reduced the expression of phospho-p85, phosphoinositide 3-kinase 110γ, phospho-Akt, nuclear factor κB, and proinflammatory cytokines. Conclusion This results of the study indicated a role for EphA2-ephrinA1 signaling in the pathogenesis of LPS-induced lung injury. Furthermore, EphA2 antagonism inhibits the phosphoinositide 3-kinase-Akt pathway and attenuates inflammation.
Collapse
Affiliation(s)
- Ji Young Hong
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Hwa Shin
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Soo Chung
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Young Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Ye Jung
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ae Kang
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sam Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Se Kyu Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Joon Chang
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Moo Suk Park
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
41
|
Aschner Y, Zemans RL, Yamashita CM, Downey GP. Matrix metalloproteinases and protein tyrosine kinases: potential novel targets in acute lung injury and ARDS. Chest 2014; 146:1081-1091. [PMID: 25287998 DOI: 10.1378/chest.14-0397] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) and ARDS fall within a spectrum of pulmonary disease that is characterized by hypoxemia, noncardiogenic pulmonary edema, and dysregulated and excessive inflammation. While mortality rates have improved with the advent of specialized ICUs and lung protective mechanical ventilation strategies, few other therapies have proven effective in the management of ARDS, which remains a significant clinical problem. Further development of biomarkers of disease severity, response to therapy, and prognosis is urgently needed. Several novel pathways have been identified and studied with respect to the pathogenesis of ALI and ARDS that show promise in bridging some of these gaps. This review will focus on the roles of matrix metalloproteinases and protein tyrosine kinases in the pathobiology of ALI in humans, and in animal models and in vitro studies. These molecules can act independently, as well as coordinately, in a feed-forward manner via activation of tyrosine kinase-regulated pathways that are pivotal in the development of ARDS. Specific signaling events involving proteolytic processing by matrix metalloproteinases that contribute to ALI, including cytokine and chemokine activation and release, neutrophil recruitment, transmigration and activation, and disruption of the intact alveolar-capillary barrier, will be explored in the context of these novel molecular pathways.
Collapse
Affiliation(s)
- Yael Aschner
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine and Pediatrics, National Jewish Health, Denver, CO; Division of Pulmonary Sciences and Critical Care Medicine, Departments of Medicine, University of Colorado Denver, Aurora, CO
| | - Rachel L Zemans
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine and Pediatrics, National Jewish Health, Denver, CO; Division of Pulmonary Sciences and Critical Care Medicine, Departments of Medicine, University of Colorado Denver, Aurora, CO
| | - Cory M Yamashita
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Gregory P Downey
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine and Pediatrics, National Jewish Health, Denver, CO; Division of Pulmonary Sciences and Critical Care Medicine, Departments of Medicine, University of Colorado Denver, Aurora, CO; Immunology, University of Colorado Denver, Aurora, CO.
| |
Collapse
|
42
|
Ende G, Poitz DM, Wiedemann E, Augstein A, Friedrichs J, Giebe S, Weinert S, Werner C, Strasser RH, Jellinghaus S. TNF-α-mediated adhesion of monocytes to endothelial cells-The role of ephrinA1. J Mol Cell Cardiol 2014; 77:125-35. [PMID: 25451169 DOI: 10.1016/j.yjmcc.2014.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 10/13/2014] [Accepted: 10/20/2014] [Indexed: 12/19/2022]
Abstract
The ligand ephrin A1 is more often discussed to play a role in the development of the atherosclerotic plaque and in this context especially in the monocyte adhesion to endothelial cells. As tumor necrosis factor-α (TNF-α) is known to induce monocyte adhesion to endothelium and ephrin A1 expression, the present study focuses on the involvement of ephrin A1 in TNF-α-mediated monocyte adhesion. The analysis of different members of the Eph/ephrin system in TNF-α-treated human umbilical vein endothelial cells (HUVEC) revealed that especially ephrinA1 was found to be highly regulated by TNF-α compared to other members of the Eph family. This effect is also present in arterial endothelial cells from the umbilical artery and from the coronary artery. This regulation is dependent on NFκB-activation as shown by the expression of a constitutive-active IκB-mutant. By using siRNA-mediated silencing and adenoviral overexpression of ephrinA1 in HUVEC, the involvement of ephrinA1 in the TNF-α triggered monocyte adhesion to endothelial cells could be demonstrated. In addition, these results could be verified by quantitative adhesion measurement using atomic force microscopy-based single-cell force spectroscopy and under flow conditions. Furthermore, this effect is mediated via the EphA4 receptor. EphrinA1 does not influence the mRNA or protein expression of the adhesion receptors VCAM-1 and ICAM-1 in endothelial cells. However, the surface presentation of these adhesion receptors is modulated in an ephrinA1-dependent manner. In conclusion, these data demonstrate that ephrinA1 plays an important role in the TNF-α-mediated adhesion of monocytes to endothelial cells, which might be of great importance in the context of atherosclerosis.
Collapse
Affiliation(s)
- Georg Ende
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany.
| | - David M Poitz
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - Elisa Wiedemann
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - Antje Augstein
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - Jens Friedrichs
- Institute for Biofunctional Polymer Materials Dresden, Leibniz Institute of Polymer Research, Germany
| | - Sindy Giebe
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Sönke Weinert
- Internal Medicine, Department of Cardiology, Angiology and Pneumology, Magdeburg University, Magdeburg, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials Dresden, Leibniz Institute of Polymer Research, Germany
| | - Ruth H Strasser
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | | |
Collapse
|
43
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
44
|
DuSablon A, Kent S, Coburn A, Virag J. EphA2-receptor deficiency exacerbates myocardial infarction and reduces survival in hyperglycemic mice. Cardiovasc Diabetol 2014; 13:114. [PMID: 25166508 PMCID: PMC4147179 DOI: 10.1186/s12933-014-0114-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/12/2014] [Indexed: 02/04/2023] Open
Abstract
Background We have previously shown that EphrinA1/EphA expression profile changes in response to myocardial infarction (MI), exogenous EphrinA1-Fc administration following MI positively influences wound healing, and that deletion of the EphA2 Receptor (EphA2-R) exacerbates injury and remodeling. To determine whether or not ephrinA1-Fc would be of therapeutic value in the hyperglycemic infarcted heart, it is critical to evaluate how ephrinA1/EphA signaling changes in the hyperglycemic myocardium in response to MI. Methods Streptozotocin (STZ)-induced hyperglycemia in wild type (WT) and EphA2-receptor mutant (EphA2-R-M) mice was initiated by an intraperitoneal injection of STZ (150 mg/kg) 10 days before surgery. MI was induced by permanent ligation of the left anterior descending coronary artery and analyses were performed at 4 days post-MI. ANOVAs with Student-Newman Keuls multiple comparison post-hoc analysis illustrated which groups were significantly different, with significance of at least p < 0.05. Results Both WT and EphA2-R-M mice responded adversely to STZ, but only hyperglycemic EphA2-R-M mice had lower ejection fraction (EF) and fractional shortening (FS). At 4 days post-MI, we observed greater post-MI mortality in EphA2-R-M mice compared with WT and this was greater still in the EphA2-R-M hyperglycemic mice. Although infarct size was greater in hyperglycemic WT mice vs normoglycemic mice, there was no difference between hyperglycemic EphA2-R-M mice and normoglycemic EphA2-R-M mice. The hypertrophic response that normally occurs in viable myocardium remote to the infarct was noticeably absent in epicardial cardiomyocytes and cardiac dysfunction worsened in hyperglycemic EphA2-R-M hearts post-MI. The characteristic interstitial fibrotic response in the compensating myocardium remote to the infarct also did not occur in hyperglycemic EphA2-R-M mouse hearts to the same extent as that observed in the hyperglycemic WT mouse hearts. Differences in neutrophil and pan-leukocyte infiltration and serum cytokines implicate EphA2-R in modulation of injury and the differences in ephrinA1 and EphA6-R expression in governing this are discussed. Conclusions We conclude that EphA2-mutant mice are more prone to hyperglycemia-induced increased injury, decreased survival, and worsened LV remodeling due to impaired wound healing.
Collapse
|
45
|
Lennon FE, Mirzapoiazova T, Mambetsariev N, Mambetsariev B, Salgia R, Singleton PA. Transactivation of the receptor-tyrosine kinase ephrin receptor A2 is required for the low molecular weight hyaluronan-mediated angiogenesis that is implicated in tumor progression. J Biol Chem 2014; 289:24043-58. [PMID: 25023279 DOI: 10.1074/jbc.m114.554766] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis or the formation of new blood vessels is important in the growth and metastatic potential of various cancers. Therefore, understanding the mechanism(s) by which angiogenesis occurs can have important therapeutic implications in numerous malignancies. We and others have demonstrated that low molecular weight hyaluronan (LMW-HA, ∼2500 Da) promotes endothelial cell (EC) barrier disruption and angiogenesis. However, the mechanism(s) by which this occurs is poorly defined. Our data indicate that treatment of human EC with LMW-HA induced CD44v10 association with the receptor-tyrosine kinase, EphA2, transactivation (tyrosine phosphorylation) of EphA2, and recruitment of the PDZ domain scaffolding protein, PATJ, to the cell periphery. Silencing (siRNA) CD44, EphA2, PATJ, or Dbs (RhoGEF) expression blocked LMW-HA-mediated angiogenesis (EC proliferation, migration, and tubule formation). In addition, silencing EphA2, PATJ, Src, or Dbs expression blocked LMW-HA-mediated RhoA activation. To translate our in vitro findings, we utilized a novel anginex/liposomal targeting of murine angiogenic endothelium with either CD44 or EphA2 siRNA and observed inhibition of LMW-HA-induced angiogenesis in implanted Matrigel plugs. Taken together, these results indicate LMW-HA-mediated transactivation of EphA2 is required for PATJ and Dbs membrane recruitment and subsequent RhoA activation required for angiogenesis. These results suggest that targeting downstream effectors of LMW-HA could be a useful therapeutic intervention for angiogenesis-associated diseases including tumor progression.
Collapse
Affiliation(s)
- Frances E Lennon
- From the Department of Medicine, Section of Pulmonary and Critical Care and
| | | | | | - Bolot Mambetsariev
- From the Department of Medicine, Section of Pulmonary and Critical Care and
| | - Ravi Salgia
- Section of Hematology and Oncology University of Chicago, Chicago Illinois 60637
| | | |
Collapse
|
46
|
Nasreen N, Khodayari N, Sriram PS, Patel J, Mohammed KA. Tobacco smoke induces epithelial barrier dysfunction via receptor EphA2 signaling. Am J Physiol Cell Physiol 2014; 306:C1154-66. [PMID: 24717580 DOI: 10.1152/ajpcell.00415.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Erythropoietin-producing human hepatocellular carcinoma (Eph) receptors are the largest family of receptor tyrosine kinases (RTKs) that mediate various cellular and developmental processes. The degrees of expression of these key molecules control the cell-cell interactions. Although the role of Eph receptors and their ligand Ephrins is well studied in developmental processes, their function in tobacco smoke (TS)-induced epithelial barrier dysfunction is unknown. We hypothesized that TS may induce permeability in bronchial airway epithelial cell (BAEpC) monolayer by modulating receptor EphA2 expression, actin cytoskeleton, adherens junction, and focal adhesion proteins. Here we report that in BAEpCs, acute TS exposure significantly upregulated EphA2 and EphrinA1 expression, disrupted the actin filaments, decreased E-cadherin expression, and increased protein permeability, whereas the focal adhesion protein paxillin was unaffected. Silencing the receptor EphA2 expression with silencing interference RNA (siRNA) significantly attenuated TS-induced hyperpermeability in BAEpCs. In addition, when BAEpC monolayer was transfected with EphA2-expressing plasmid and treated with recombinant EphrinA1, the transepithelial electrical resistance decreased significantly. Furthermore, TS downregulated E-cadherin expression and induced hyperpermeability across BAEpC monolayer in a Erk1/Erk2, p38, and JNK MAPK-dependent manner. TS induced hyperpermeability in BAEpC monolayer by targeting cell-cell adhesions, and interestingly cell-matrix adhesions were unaffected. The present data suggest that TS causes significant damage to the BAEpCs via induction of EphA2 and downregulation of E-cadherin. Induction of EphA2 in the BAEpCs exposed to TS may be an important signaling event in the pathogenesis of TS-induced epithelial injury.
Collapse
Affiliation(s)
- Najmunnisa Nasreen
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida; and North Florida/South Georgia Veterans Health Care System, Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Nazli Khodayari
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida; and North Florida/South Georgia Veterans Health Care System, Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Peruvemba S Sriram
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida; and North Florida/South Georgia Veterans Health Care System, Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Jawaharlal Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida; and North Florida/South Georgia Veterans Health Care System, Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| | - Kamal A Mohammed
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida; and North Florida/South Georgia Veterans Health Care System, Malcom Randall Veterans Affairs Medical Center, University of Florida, Gainesville, Florida
| |
Collapse
|
47
|
Jiang H, Li X, Zhang X, Liu Y, Huang S, Wang X. EphA2 knockdown attenuates atherosclerotic lesion development in ApoE(-/-) mice. Cardiovasc Pathol 2014; 23:169-74. [PMID: 24561077 DOI: 10.1016/j.carpath.2014.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The inflammatory response of vascular endothelial cells plays important roles in the initiation and progression of atherosclerotic lesions. EphA2 receptor activation promotes the endothelial cell inflammatory response, and its expression is increased in the endothelial cell layer of atherosclerotic plaques. However, the association between EphA2 and atherosclerosis has not been determined. METHODS Eight-week-old male ApoE(-/-) mice were systemically infected with adenoassociated virus serotype 9 carrying a small hairpin RNA specifically targeting the EphA2 gene to knock down EphA2 expression in aortic endothelial cells. These mice were then fed a high-cholesterol diet for 12 weeks. Blood was collected for the measurement of plasma lipids. The aortas were harvested to evaluate the atherosclerotic lesion size, macrophage components, and expression of proinflammatory genes using Oil Red O staining, immunofluorescence staining, and molecular biology analysis. RESULTS The lesions formed in the entire aorta and aortic sinus of the ApoE(-/-) mice with EphA2 knockdown were significantly smaller than those in the control mice (10.7%±3.1% versus 25.1%±4.2%; 0.51±0.02mm(2) versus 0.85±0.03mm(2); n=10; P<.05). Furthermore, the lesions in the ApoE(-/-) mice with EphA2 knockdown displayed reduced inflammation compared with the control mice, as reflected by the decreased macrophage infiltration (8.2%±2.9% versus 22.7%±4%; n=10; P<.05); decreased nuclear factor-κβ activation; and diminished expression of vascular cell adhesion molecule-1, E-selectin, and monocyte chemotactic protein-1 (all P<.05). CONCLUSIONS Our data demonstrate that the EphA2 receptor silencing attenuates the extent and inflammation of atherosclerotic lesions in ApoE(-/-) mice. Thus, EphA2 knockdown in endothelial cells represents a novel therapeutic strategy for patients with atherosclerosis.
Collapse
Affiliation(s)
- Hong Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China.
| | - Xinyun Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoli Zhang
- Department of Histology and Embryology, School of Medicine, Shandong University, Jinan, China
| | - Yan Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Shanying Huang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Xiaowei Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
48
|
ADAM12-cleaved ephrin-A1 contributes to lung metastasis. Oncogene 2013; 33:2179-90. [DOI: 10.1038/onc.2013.180] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/22/2022]
|
49
|
Thundyil J, Manzanero S, Pavlovski D, Cully TR, Lok KZ, Widiapradja A, Chunduri P, Jo DG, Naruse C, Asano M, Launikonis BS, Sobey CG, Coulthard MG, Arumugam TV. Evidence that the EphA2 receptor exacerbates ischemic brain injury. PLoS One 2013; 8:e53528. [PMID: 23308246 PMCID: PMC3538581 DOI: 10.1371/journal.pone.0053528] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/28/2012] [Indexed: 12/17/2022] Open
Abstract
Ephrin (Eph) signaling within the central nervous system is known to modulate axon guidance, synaptic plasticity, and to promote long-term potentiation. We investigated the potential involvement of EphA2 receptors in ischemic stroke-induced brain inflammation in a mouse model of focal stroke. Cerebral ischemia was induced in male C57Bl6/J wild-type (WT) and EphA2-deficient (EphA2−/−) mice by middle cerebral artery occlusion (MCAO; 60 min), followed by reperfusion (24 or 72 h). Brain infarction was measured using triphenyltetrazolium chloride staining. Neurological deficit scores and brain infarct volumes were significantly less in EphA2−/− mice compared with WT controls. This protection by EphA2 deletion was associated with a comparative decrease in brain edema, blood-brain barrier damage, MMP-9 expression and leukocyte infiltration, and higher expression levels of the tight junction protein, zona occludens-1. Moreover, EphA2−/− brains had significantly lower levels of the pro-apoptotic proteins, cleaved caspase-3 and BAX, and higher levels of the anti-apoptotic protein, Bcl-2 as compared to WT group. We confirmed that isolated WT cortical neurons express the EphA2 receptor and its ligands (ephrin-A1–A3). Furthermore, expression of all four proteins was increased in WT primary cortical neurons following 24 h of glucose deprivation, and in the brains of WT mice following stroke. Glucose deprivation induced less cell death in primary neurons from EphA2−/− compared with WT mice. In conclusion, our data provide the first evidence that the EphA2 receptor directly contributes to blood-brain barrier damage and neuronal death following ischemic stroke.
Collapse
Affiliation(s)
- John Thundyil
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Silvia Manzanero
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Dale Pavlovski
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Tanya R. Cully
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Ker-Zhing Lok
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Alexander Widiapradja
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Prasad Chunduri
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Chie Naruse
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Masahide Asano
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Bradley S. Launikonis
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Christopher G. Sobey
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Mark G. Coulthard
- Academic Discipline of Paediatrics and Child Health, University of Queensland, Royal Children’s Hospital, Herston, Queensland, Australia
- Paediatric Intensive Care Unit, Royal Children's Hospital, Herston, Queensland, Australia
- Queensland Children’s Medical Research Institute, Royal Children's Hospital, Herston, Queensland, Australia
| | - Thiruma V. Arumugam
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
- * E-mail:
| |
Collapse
|
50
|
Funk SD, Orr AW. Ephs and ephrins resurface in inflammation, immunity, and atherosclerosis. Pharmacol Res 2013; 67:42-52. [DOI: 10.1016/j.phrs.2012.10.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 10/04/2012] [Accepted: 10/10/2012] [Indexed: 01/13/2023]
|