1
|
Taskinen JH, Holopainen M, Ruhanen H, van der Stoel M, Käkelä R, Ikonen E, Keskitalo S, Varjosalo M, Olkkonen VM. Functional omics of ORP7 in primary endothelial cells. BMC Biol 2024; 22:292. [PMID: 39695567 DOI: 10.1186/s12915-024-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Many members of the oxysterol-binding protein-related protein (ORP) family have been characterized in detail over the past decades, but the lipid transport and other functions of ORP7 still remain elusive. What is known about ORP7 points toward an endoplasmic reticulum and plasma membrane-localized protein, which also interacts with GABA type A receptor-associated protein like 2 (GABARAPL2) and unlipidated Microtubule-associated proteins 1A/1B light chain 3B (LC3B), suggesting a further autophagosomal/lysosomal association. Functional roles of ORP7 have been suggested in cholesterol efflux, hypercholesterolemia, and macroautophagy. We performed a hypothesis-free multi-omics analysis of chemical ORP7 inhibition utilizing transcriptomics and lipidomics as well as proximity biotinylation interactomics to characterize ORP7 functions in a primary cell type, human umbilical vein endothelial cells (HUVECs). Moreover, assays on angiogenesis, cholesterol efflux, and lipid droplet quantification were conducted. RESULTS Pharmacological inhibition of ORP7 leads to an increase in gene expression related to lipid metabolism and inflammation, while genes associated with cell cycle and cell division were downregulated. Lipidomic analysis revealed increases in ceramides and lysophosphatidylcholines as well as saturated and monounsaturated triacylglycerols. Significant decreases were seen in all cholesteryl ester and in some unsaturated triacylglycerol species, compatible with the detected decrease of mean lipid droplet area. Along with the reduced lipid stores, ATP-binding cassette subfamily G member 1 (ABCG1)-mediated cholesterol efflux and angiogenesis decreased. Interactomics revealed an interaction of ORP7 with AKT1, a central metabolic regulator. CONCLUSIONS The transcriptomics results suggest an increase in prostanoid as well as oxysterol synthesis, which could be related to the observed upregulation of proinflammatory genes. We envision that the defective angiogenesis in HUVECs subjected to ORP7 inhibition could be the result of an unfavorable plasma membrane lipid composition and/or reduced potential for cell division. To conclude, the present study suggests multifaceted functions of ORP7 in lipid homeostasis, angiogenic tube formation, and gene expression of lipid metabolism, inflammation, and cell cycle in primary endothelial cells.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Minna Holopainen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Miesje van der Stoel
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014, Helsinki, Finland
| | - Elina Ikonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Salla Keskitalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
| | - Markku Varjosalo
- Proteomics Unit Viikki, Institute of Biotechnology, HiLIFE and Biocenter Finland, University of Helsinki, Viikinkaari 1, 00790, Helsinki, Finland
- Systems Biology/Pathology Research Group, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
2
|
Santos AB, Carona A, Ettcheto M, Camins A, Falcão A, Fortuna A, Bicker J. Krüppel-like factors: potential roles in blood-brain barrier dysfunction and epileptogenesis. Acta Pharmacol Sin 2024; 45:1765-1776. [PMID: 38684799 PMCID: PMC11335766 DOI: 10.1038/s41401-024-01285-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/07/2024] [Indexed: 05/02/2024] Open
Abstract
Epilepsy is a chronic and debilitating neurological disorder, known for the occurrence of spontaneous and recurrent seizures. Despite the availability of antiseizure drugs, 30% of people with epilepsy experience uncontrolled seizures and drug resistance, evidencing that new therapeutic options are required. The process of epileptogenesis involves the development and expansion of tissue capable of generating spontaneous recurrent seizures, during which numerous events take place, namely blood-brain barrier (BBB) dysfunction, and neuroinflammation. The consequent cerebrovascular dysfunction results in a lower seizure threshold, seizure recurrence, and chronic epilepsy. This suggests that improving cerebrovascular health may interrupt the pathological cycle responsible for disease development and progression. Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors, encountered in brain endothelial cells, glial cells, and neurons. KLFs are known to regulate vascular function and changes in their expression are associated with neuroinflammation and human diseases, including epilepsy. Hence, KLFs have demonstrated various roles in cerebrovascular dysfunction and epileptogenesis. This review critically discusses the purpose of KLFs in epileptogenic mechanisms and BBB dysfunction, as well as the potential of their pharmacological modulation as therapeutic approach for epilepsy treatment.
Collapse
Affiliation(s)
| | - Andreia Carona
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Miren Ettcheto
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Antoni Camins
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
3
|
Li Q, Kang J, Liu N, Huang J, Zhang X, Pang K, Zhang S, Wang M, Zhao Y, Dong S, Li H, Zhao D, Lu F, Zhang W. Hydrogen sulfide improves endothelial barrier function by modulating the ubiquitination degradation of KLF4 through TRAF7 S-sulfhydration in diabetic aorta. Free Radic Biol Med 2024; 216:118-138. [PMID: 38479633 DOI: 10.1016/j.freeradbiomed.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
Anomalous vascular endothelium significantly contributes to various cardiovascular diseases. VE-cadherin plays a vital role in governing the endothelial barrier. Krüppel-like factor 4(KLF4), as a transcription factor, which binds the VE-cadherin promoter and enhances its transcription. Tumor necrosis factor receptor-associated factor 7 (TRAF7) is an E3 ubiquitin ligase that has been shown to modulate the degradation of KLF4. H2S can covalently modify cysteine residues on proteins through S-sulfhydration, thereby influencing the structure and functionality of the target protein. However, the role of S-sulfhydration on endothelial barrier integrity remains to be comprehensively elucidated. This study aims to investigate whether protein S-sulfhydration in the endothelium regulates endothelial integrity and its underlying mechanism. In this study, we observed that protein S-sulfhydration was reduced in the endothelium during diabetes and TRAF7 was the main target. Overexpression of TRAF7-Cys327 mutant could mitigate the endothelial barrier damage by weakening TRAF7 interaction with KLF4 and reducing ubiquitination degradation of KLF4. In conclusion, our research demonstrates that H2S plays a pivotal role in regulating S-sulfhydration of TRAF7 at Cys327. This regulation effectively inhibits the ubiquitin-mediated degradation of KLF4, resulting in an upregulation of VE-cadherin levels. This molecular mechanism contributes to the prevention of endothelial barrier damage.
Collapse
Affiliation(s)
- Qianzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Jiaxin Kang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Jiayi Huang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Xueya Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Kemiao Pang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Hongxia Li
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China.
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
4
|
Mylonas KS, Peroulis M, Kapetanakis EI, Kapelouzou A. Myocardial Expression of Pluripotency, Longevity, and Proinflammatory Genes in the Context of Hypercholesterolemia and Statin Treatment. J Clin Med 2024; 13:1994. [PMID: 38610757 PMCID: PMC11012955 DOI: 10.3390/jcm13071994] [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: 01/31/2024] [Revised: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Background: This study sought to assess the effect of statin therapy on myocardial inflammation in a White New Zealand rabbit model of atherogenesis. Methods: The mRNA expression levels of pro-inflammatory, pluripotency, and aging-related markers were quantified following a controlled feeding protocol and statin treatments. Results: Following high-cholesterol diet induction, we observed significant upregulation in the myocardial mRNA levels of MYD88, NF-κB, chemokines (CCL4, CCL20, and CCR2), IFN-γ, interleukins (IL-1β, IL-2, IL-4, IL-8, IL-10, and IL-18), and novel markers (klotho, KFL4, NANOG, and HIF1α). In contrast, HOXA5 expression was diminished following a hyperlipidemic diet. Both statin treatments significantly influenced the markers studied. Nevertheless, rosuvastatin administration resulted in a greater reduction in MYD88, NF-kB, chemokines (CCL4, CCL20, and CCR2), and interleukins IL-1β, IL-8, KLF4, NANOG, and HIF1α than fluvastatin. Fluvastatin, on the other hand, led to a stronger decrease in IL-4. Downregulation of IL-2 and IL-18 and upregulation of IFNβ and HOXA5 were comparable between the two statins. Notably, rosuvastatin had a stronger effect on the upregulation of klotho and IL-10. Conclusion: Overall, statin therapy significantly attenuated inflammatory, pluripotency, and klotho expression in myocardial tissue under atherogenic conditions. Our findings also highlight the differential efficacy of rosuvastatin over fluvastatin in curtailing proatherogenic inflammation, which could have profound implications for the clinical management of cardiovascular disease.
Collapse
Affiliation(s)
- Konstantinos S Mylonas
- Department of Cardiac Surgery, Onassis Cardiac Surgery Center, 356 Leof. Andreas Syngros, 17674 Athens, Greece
| | - Michail Peroulis
- Vascular Surgery Unit, Department of Surgery, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Emmanouil I Kapetanakis
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Alkistis Kapelouzou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
5
|
Rudraraju M, Shan S, Liu F, Tyler J, Caldwell RB, Somanath PR, Narayanan SP. Pharmacological Modulation of β-Catenin Preserves Endothelial Barrier Integrity and Mitigates Retinal Vascular Permeability and Inflammation. J Clin Med 2023; 12:7145. [PMID: 38002758 PMCID: PMC10672253 DOI: 10.3390/jcm12227145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Compromised blood-retinal barrier (BRB) integrity is a significant factor in ocular diseases like uveitis and retinopathies, leading to pathological vascular permeability and retinal edema. Adherens and tight junction (AJ and TJ) dysregulation due to retinal inflammation plays a pivotal role in BRB disruption. We investigated the potential of ICG001, which inhibits β-catenin-mediated transcription, in stabilizing cell junctions and preventing BRB leakage. In vitro studies using human retinal endothelial cells (HRECs) showed that ICG001 treatment improved β-Catenin distribution within AJs post lipopolysaccharide (LPS) treatment and enhanced monolayer barrier resistance. The in vivo experiments involved a mouse model of LPS-induced ocular inflammation. LPS treatment resulted in increased albumin leakage from retinal vessels, elevated vascular endothelial growth factor (VEGF) and Plasmalemmal Vesicle-Associated Protein (PLVAP) expression, as well as microglia and macroglia activation. ICG001 treatment (i.p.) effectively mitigated albumin leakage, reduced VEGF and PLVAP expression, and reduced the number of activated microglia/macrophages. Furthermore, ICG001 treatment suppressed the surge in inflammatory cytokine synthesis induced by LPS. These findings highlight the potential of interventions targeting β-Catenin to enhance cell junction stability and improve compromised barrier integrity in various ocular inflammatory diseases, offering hope for better management and treatment options.
Collapse
Affiliation(s)
- Madhuri Rudraraju
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Shengshuai Shan
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Fang Liu
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Jennifer Tyler
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | - Ruth B. Caldwell
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics, Clinical and Administrative Pharmacy Department, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
6
|
Chakraborty S, Singh A, Wang L, Wang X, Sanborn MA, Ye Z, Maienschein-Cline M, Mukhopadhyay A, Ganesh BB, Malik AB, Rehman J. Trained immunity of alveolar macrophages enhances injury resolution via KLF4-MERTK-mediated efferocytosis. J Exp Med 2023; 220:e20221388. [PMID: 37615937 PMCID: PMC10450795 DOI: 10.1084/jem.20221388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 05/19/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023] Open
Abstract
Recent studies suggest that training of innate immune cells such as tissue-resident macrophages by repeated noxious stimuli can heighten host defense responses. However, it remains unclear whether trained immunity of tissue-resident macrophages also enhances injury resolution to counterbalance the heightened inflammatory responses. Here, we studied lung-resident alveolar macrophages (AMs) prechallenged with either the bacterial endotoxin or with Pseudomonas aeruginosa and observed that these trained AMs showed greater resilience to pathogen-induced cell death. Transcriptomic analysis and functional assays showed greater capacity of trained AMs for efferocytosis of cellular debris and injury resolution. Single-cell high-dimensional mass cytometry analysis and lineage tracing demonstrated that training induces an expansion of a MERTKhiMarcohiCD163+F4/80low lung-resident AM subset with a proresolving phenotype. Reprogrammed AMs upregulated expression of the efferocytosis receptor MERTK mediated by the transcription factor KLF4. Adoptive transfer of these trained AMs restricted inflammatory lung injury in recipient mice exposed to lethal P. aeruginosa. Thus, our study has identified a subset of tissue-resident trained macrophages that prevent hyperinflammation and restore tissue homeostasis following repeated pathogen challenges.
Collapse
Affiliation(s)
- Sreeparna Chakraborty
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Abhalaxmi Singh
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Li Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Xinge Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois College of Medicine, Chicago, IL, USA
| | - Mark A. Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zijing Ye
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Balaji B. Ganesh
- Research Resources Center, University of Illinois Chicago, Chicago, Illinois, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois College of Medicine, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
7
|
Mylonas KS, Peroulis M, Kapelouzou A. Transfection of Vein Grafts with Early Growth Response Factor-1 Oligodeoxynucleotide Decoy: Effects on Stem-Cell Genes and Toll-like Receptor-Mediated Inflammation. Int J Mol Sci 2023; 24:15866. [PMID: 37958848 PMCID: PMC10647335 DOI: 10.3390/ijms242115866] [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: 10/05/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The long-term patency of vein grafts is challenged by intimal hyperplasia. We sought to explore the intricate relationships between the transcription factor Egr-1, toll-like receptors (TLRs), and stem cell genes and also assessed oligodeoxynucleotide decoys (ODNs) as a strategy to prevent vein graft failures. A total of 42 New Zealand white rabbits were fed hyperlipidemic chow and classified into three groups. A double-stranded Egr-1 ODN was synthesized and infused in vein grafts prior to anastomosis with the common carotid artery. All vein grafts were retrieved at the conclusion of the predefined experimental period. Real-time quantitative polymerase chain reaction was performed to estimate expression patterns for several genes of interest. MYD88, TLR2-4, TLR8, NF-kB, TNF-α, IFNβ, and IFNγ; chemokines CCL4, CCL20, CCR2; numerous interleukins; and stem cell genes KFL4, NANOG, HOXA5, and HIF1α were universally downregulated in the ODN arm compared with the controls. By understanding these multifaceted interactions, our study offers actionable insights that may pave the way for innovative interventions in vascular reconstructions.
Collapse
Affiliation(s)
| | - Michail Peroulis
- Department of Surgery, Vascular Surgery Unit, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Alkistis Kapelouzou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
8
|
Liu J, Chen H, Li X, Song C, Wang L, Wang D. Micro-Executor of Natural Products in Metabolic Diseases. Molecules 2023; 28:6202. [PMID: 37687031 PMCID: PMC10488769 DOI: 10.3390/molecules28176202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Obesity, diabetes, and cardiovascular diseases are the major chronic metabolic diseases that threaten human health. In order to combat these epidemics, there remains a desperate need for effective, safe, and easily available therapeutic strategies. Recently, the development of natural product research has provided new methods and options for these diseases. Numerous studies have demonstrated that microRNAs (miRNAs) are key regulators of metabolic diseases, and natural products can improve lipid and glucose metabolism disorders and cardiovascular diseases by regulating the expression of miRNAs. In this review, we present the recent advances involving the associations between miRNAs and natural products and the current evidence showing the positive effects of miRNAs for natural product treatment in metabolic diseases. We also encourage further research to address the relationship between miRNAs and natural products under physiological and pathological conditions, thus leading to stronger support for drug development from natural products in the future.
Collapse
Affiliation(s)
- Jinxin Liu
- Food and Pharmacy College, Xuchang University, Xuchang 461000, China; (J.L.); (C.S.)
| | - Huanwen Chen
- Center for Agricultural and Rural Development, Zhangdian District, Zibo 255000, China;
| | - Xiaoli Li
- Zibo Digital Agriculture and Rural Development Center, Zibo 255000, China;
| | - Chunmei Song
- Food and Pharmacy College, Xuchang University, Xuchang 461000, China; (J.L.); (C.S.)
| | - Li Wang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Deguo Wang
- Food and Pharmacy College, Xuchang University, Xuchang 461000, China; (J.L.); (C.S.)
- Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
| |
Collapse
|
9
|
Liu K, Dou R, Yang C, Di Z, Shi D, Zhang C, Song J, Fang Y, Huang S, Xiang Z, Zhang W, Wang S, Xiong B. Exosome-transmitted miR-29a induces colorectal cancer metastasis by destroying the vascular endothelial barrier. Carcinogenesis 2023; 44:356-367. [PMID: 36939367 DOI: 10.1093/carcin/bgad013] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/05/2023] [Accepted: 03/17/2023] [Indexed: 03/21/2023] Open
Abstract
Metastasis is the leading cause of colorectal cancer treatment failure and mortality. Communication between endothelium and tumor cells in the tumor microenvironment is required for cancer metastasis. Tumor-derived exosomes have been shown to increase vascular permeability by delivering microRNA (miRNA) to vascular endothelial cells, facilitating cancer metastasis. The mechanism by which Epithelial-mesenchymal transition (EMT) tumor cell-derived exosomes influence vascular permeability remains unknown. MicroRNA-29a (miR-29a) expression is up-regulated in colorectal cancer (CRC) tissues, which is clinically significant in metastasis. Exosomal miR-29a secreted by EMT-CRC cells has been found to decrease the expression of Zonula occlusion 1 (ZO-1), Claudin-5, and Occludin via targeting Kruppel-like factor 4 (KLF4). In vitro co-culture investigations further revealed that EMT-cancer cells release exosomal miR-29a, which alters vascular endothelial permeability. Furthermore, exosomal miR-29a promoted liver metastases in CRC mice. Our findings demonstrate that EMT-CRC cells may transport exosomal miR-29a to endothelial cells in the tumor microenvironment (TME). As a result, increased vascular permeability promotes the development and metastasis of CRC. Exosomal miR-29a has the potential to be a predictive marker for tumor metastasis as well as a viable therapeutic target for CRC.
Collapse
Affiliation(s)
- Keshu Liu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Rongzhang Dou
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Ziyang Di
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Dongdong Shi
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Chunxiao Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Jialin Song
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Yan Fang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Sihao Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Zhenxian Xiang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Weisong Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| |
Collapse
|
10
|
Kotlyarov S. Effects of Atherogenic Factors on Endothelial Cells: Bioinformatics Analysis of Differentially Expressed Genes and Signaling Pathways. Biomedicines 2023; 11:1216. [PMID: 37189834 PMCID: PMC10135807 DOI: 10.3390/biomedicines11041216] [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: 03/11/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Background: Atherosclerosis is a serious medical condition associated with high morbidity and mortality rates. It develops over many years as a complex chain of events in the vascular wall involving various cells and is influenced by many factors of clinical interest. (2) Methods: In this study, we performed a bioinformatic analysis of Gene Expression Omnibus (GEO) datasets to investigate the gene ontology of differentially expressed genes (DEGs) in endothelial cells exposed to atherogenic factors such as tobacco smoking, oscillatory shear, and oxidized low-density lipoproteins (oxLDL). DEGs were identified using the limma R package, and gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) network analysis were performed. (3) Results: We studied biological processes and signaling pathways involving DEGs in endothelial cells under the influence of atherogenic factors. GO enrichment analysis demonstrated that the DEGs were mainly involved in cytokine-mediated signaling pathway, innate immune response, lipid biosynthetic process, 5-lipoxygenase activity, and nitric-oxide synthase activity. KEGG pathway enrichment analysis showed that common pathways included tumor necrosis factor signaling pathway, NF-κB signaling pathway, NOD-like receptor signaling pathway, lipid and atherosclerosis, lipoprotein particle binding, and apoptosis. (4) Conclusions: Atherogenic factors such as smoking, impaired flow, and oxLDL contribute to impaired innate immune response, metabolism, and apoptosis in endothelial cells, potentially leading to the development of atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
11
|
Hashimoto Y, Greene C, Munnich A, Campbell M. The CLDN5 gene at the blood-brain barrier in health and disease. Fluids Barriers CNS 2023; 20:22. [PMID: 36978081 PMCID: PMC10044825 DOI: 10.1186/s12987-023-00424-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The CLDN5 gene encodes claudin-5 (CLDN-5) that is expressed in endothelial cells and forms tight junctions which limit the passive diffusions of ions and solutes. The blood-brain barrier (BBB), composed of brain microvascular endothelial cells and associated pericytes and end-feet of astrocytes, is a physical and biological barrier to maintain the brain microenvironment. The expression of CLDN-5 is tightly regulated in the BBB by other junctional proteins in endothelial cells and by supports from pericytes and astrocytes. The most recent literature clearly shows a compromised BBB with a decline in CLDN-5 expression increasing the risks of developing neuropsychiatric disorders, epilepsy, brain calcification and dementia. The purpose of this review is to summarize the known diseases associated with CLDN-5 expression and function. In the first part of this review, we highlight the recent understanding of how other junctional proteins as well as pericytes and astrocytes maintain CLDN-5 expression in brain endothelial cells. We detail some drugs that can enhance these supports and are being developed or currently in use to treat diseases associated with CLDN-5 decline. We then summarise mutagenesis-based studies which have facilitated a better understanding of the physiological role of the CLDN-5 protein at the BBB and have demonstrated the functional consequences of a recently identified pathogenic CLDN-5 missense mutation from patients with alternating hemiplegia of childhood. This mutation is the first gain-of-function mutation identified in the CLDN gene family with all others representing loss-of-function mutations resulting in mis-localization of CLDN protein and/or attenuated barrier function. Finally, we summarize recent reports about the dosage-dependent effect of CLDN-5 expression on the development of neurological diseases in mice and discuss what cellular supports for CLDN-5 regulation are compromised in the BBB in human diseases.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| | - Chris Greene
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland
| | - Arnold Munnich
- Institut Imagine, INSERM UMR1163, Université Paris Cité, Paris, F-75015, France
- Departments of Pediatric Neurology and Medical Genetics, Hospital Necker Enfants Malades, Université Paris Cité, Paris, F-75015, France
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| |
Collapse
|
12
|
Kotlyarov S, Kotlyarova A. Participation of Krüppel-like Factors in Atherogenesis. Metabolites 2023; 13:metabo13030448. [PMID: 36984888 PMCID: PMC10052737 DOI: 10.3390/metabo13030448] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Atherosclerosis is an important problem in modern medicine, the keys to understanding many aspects of which are still not available to clinicians. Atherosclerosis develops as a result of a complex chain of events in which many cells of the vascular wall and peripheral blood flow are involved. Endothelial cells, which line the vascular wall in a monolayer, play an important role in vascular biology. A growing body of evidence strengthens the understanding of the multifaceted functions of endothelial cells, which not only organize the barrier between blood flow and tissues but also act as regulators of hemodynamics and play an important role in regulating the function of other cells in the vascular wall. Krüppel-like factors (KLFs) perform several biological functions in various cells of the vascular wall. The large family of KLFs in humans includes 18 members, among which KLF2 and KLF4 are at the crossroads between endothelial cell mechanobiology and immunometabolism, which play important roles in both the normal vascular wall and atherosclerosis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
13
|
Li Z, Solomonidis EG, Berkeley B, Tang MNH, Stewart KR, Perez-Vicencio D, McCracken IR, Spiroski AM, Gray GA, Barton AK, Sellers SL, Riley PR, Baker AH, Brittan M. Multi-species meta-analysis identifies transcriptional signatures associated with cardiac endothelial responses in the ischaemic heart. Cardiovasc Res 2023; 119:136-154. [PMID: 36082978 PMCID: PMC10022865 DOI: 10.1093/cvr/cvac151] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/04/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
AIM Myocardial infarction remains the leading cause of heart failure. The adult human heart lacks the capacity to undergo endogenous regeneration. New blood vessel growth is integral to regenerative medicine necessitating a comprehensive understanding of the pathways that regulate vascular regeneration. We sought to define the transcriptomic dynamics of coronary endothelial cells following ischaemic injuries in the developing and adult mouse and human heart and to identify new mechanistic insights and targets for cardiovascular regeneration. METHODS AND RESULTS We carried out a comprehensive meta-analysis of integrated single-cell RNA-sequencing data of coronary vascular endothelial cells from the developing and adult mouse and human heart spanning healthy and acute and chronic ischaemic cardiac disease. We identified species-conserved gene regulatory pathways aligned to endogenous neovascularization. We annotated injury-associated temporal shifts of the endothelial transcriptome and validated four genes: VEGF-C, KLF4, EGR1, and ZFP36. Moreover, we showed that ZFP36 regulates human coronary endothelial cell proliferation and defined that VEGF-C administration in vivo enhances clonal expansion of the cardiac vasculature post-myocardial infarction. Finally, we constructed a coronary endothelial cell meta-atlas, CrescENDO, to empower future in-depth research to target pathways associated with coronary neovascularization. CONCLUSION We present a high-resolution single-cell meta-atlas of healthy and injured coronary endothelial cells in the mouse and human heart, revealing a suite of novel targets with great potential to promote vascular regeneration, and providing a rich resource for therapeutic development.
Collapse
Affiliation(s)
- Ziwen Li
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Emmanouil G Solomonidis
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Katherine Ross Stewart
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Daniel Perez-Vicencio
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ian R McCracken
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ana-Mishel Spiroski
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Anna K Barton
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Stephanie L Sellers
- Division of Cardiology, Centre for Heart Lung Innovation, Providence Research, University of British Columbia, Vancouver, Canada
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
14
|
Li B, Huang X, Wei J, Huang H, Liu Z, Hu J, Zhang Q, Chen Y, Cui Y, Chen Z, Guo X, Huang Q. Role of moesin and its phosphorylation in VE-cadherin expression and distribution in endothelial adherens junctions. Cell Signal 2022; 100:110466. [PMID: 36100057 DOI: 10.1016/j.cellsig.2022.110466] [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: 06/09/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIM Vascular endothelial cadherin (VE-cadherin) is an important element of adherens junctions (AJs) between endothelial cells. Its expression and proper distribution are critical for AJ formation and vascular integrity. Our previous studies have demonstrated that moesin phosphorylation mediated the hyper-permeability in endothelial monolayer and microvessels. However, the role of moesin and its phosphorylation in VE-cadherin expression and distribution is not clear. METHODS AND RESULTS In vivo, expression of VE-cadherin was significantly reduced in retina and other various tissues in moesin knock out mice (Msn-/Y). In vitro, by regulating moesin expression with siRNA and adenovirus transfection, we verified that moesin has an effect on VE-cadherin expression in HUVECs, while transcription factor KLF4 may participate in this process. In addition, treatment of advanced glycation end products (AGEs) induced abnormal distribution of VE-cadherin in retinal microvessels from C57BL/6 wild type mice, and in vitro studies indicated that moesin Thr558 phosphorylation had a critical role in AGE-induced VE-cadherin internalization from cytomembrane to cytoplasm. Further investigation demonstrated that the inhibition of F-actin polymerization with cytochalasin D could abolish AGE- and Thr558 phosphor-moesin-mediated VE-cadherin internalization. CONCLUSION This study suggests that moesin regulates VE-cadherin expression through KLF4 and the state of moesin phosphorylation at Thr558 affects the integrity of VE-cadherin-based AJs. Thr558 phosphor-moesin mediates AGE-induced VE-cadherin internalization through cytoskeleton reassembling.
Collapse
Affiliation(s)
- Bingyu Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiayi Wei
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhuanhua Liu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiaqing Hu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qin Zhang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yanjia Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun Cui
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Shunde, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Liu W, Jiang X, Li X, Sun K, Yang Y, Yang M, Li S, Zhu X. LMBR1L regulates proliferation and migration of endothelial cells through Norrin/β-catenin signaling. J Cell Sci 2022; 135:274701. [PMID: 35146515 DOI: 10.1242/jcs.259468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/07/2022] [Indexed: 11/20/2022] Open
Abstract
Precise Norrin/β-catenin signaling is critical for proper angiogenesis. Dysregulation of the signaling leads various diseases, of which retinal exudative vitreoretinopathy is the most prevalent. Here, we used global knockout mouse model to show that endothelial cells-derived limb region 1 like (LMBR1L), a transmembrane protein of unknown function in angiogenesis, is essential for retinal vascular development. In vitro experiments revealed that LMBR1L depletion resulted in aberrant activation of Norrin/β-catenin signaling pathway via decreased ubiquitination of FZD4, increased Norrin co-receptor LRP5 and p-GSK3β-Ser9 expression level, which caused accumulation of β-catenin. Moreover, inhibition of LMBR1L in human retinal microvascular endothelial cells (HRECs) caused increased proliferation ability and defective cell migration, which might due to upregulated expression levels of the AJ components. Treatment of p-GSK3β-Ser9 inhibitor AR-A014418 restored the phenotypes in LMBR1L-null HRECs, which further demonstrated the important regulatory role of LMBR1L in Norrin/β-catenin signaling pathway. Taken together, our data unravels an essential role of LMBR1L in angiogenesis.
Collapse
Affiliation(s)
- Wenjing Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Xiaoyan Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Xiao Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Kuanxiang Sun
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Mu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Shujin Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China.,Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China.,Departement of Ophthalmology, First People's Hospital of Shangqiu, Shangqiu, Henan, 476000, China
| |
Collapse
|
16
|
Mastej V, Axen C, Wary A, Minshall RD, Wary KK. A requirement for Krüppel Like Factor-4 in the maintenance of endothelial cell quiescence. Front Cell Dev Biol 2022; 10:1003028. [PMID: 36425528 PMCID: PMC9679496 DOI: 10.3389/fcell.2022.1003028] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
Rationale and Goal: Endothelial cells (ECs) are quiescent and critical for maintaining homeostatic functions of the mature vascular system, while disruption of quiescence is at the heart of endothelial to mesenchymal transition (EndMT) and tumor angiogenesis. Here, we addressed the hypothesis that KLF4 maintains the EC quiescence. Methods and Results: In ECs, KLF4 bound to KLF2, and the KLF4-transctivation domain (TAD) interacted directly with KLF2. KLF4-depletion increased KLF2 expression, accompanied by phosphorylation of SMAD3, increased expression of alpha-smooth muscle actin (αSMA), VCAM-1, TGF-β1, and ACE2, but decreased VE-cadherin expression. In the absence of Klf4, Klf2 bound to the Klf2-promoter/enhancer region and autoregulated its own expression. Loss of EC-Klf4 in Rosa mT/mG ::Klf4 fl/fl ::Cdh5 CreERT2 engineered mice, increased Klf2 levels and these cells underwent EndMT. Importantly, these mice harboring EndMT was also accompanied by lung inflammation, disruption of lung alveolar architecture, and pulmonary fibrosis. Conclusion: In quiescent ECs, KLF2 and KLF4 partnered to regulate a combinatorial mechanism. The loss of KLF4 disrupted this combinatorial mechanism, thereby upregulating KLF2 as an adaptive response. However, increased KLF2 expression overdrives for the loss of KLF4, giving rise to an EndMT phenotype.
Collapse
Affiliation(s)
- Victoria Mastej
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, United States
| | - Cassondra Axen
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, United States
| | - Anita Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, United States
| | - Richard D Minshall
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, United States
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, United States
| |
Collapse
|
17
|
Wu YH, Chen WC, Tseng CK, Chen YH, Lin CK, Lee JC. Heme oxygenase-1 inhibits DENV-induced endothelial hyperpermeability and serves as a potential target against dengue hemorrhagic fever. FASEB J 2021; 36:e22110. [PMID: 34918393 DOI: 10.1096/fj.202100688rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 02/02/2023]
Abstract
Dengue virus (DENV) is a cause of vascular endothelial dysfunction and vascular leakage, which are characterized as hallmarks of dengue hemorrhagic fever or dengue shock syndrome, which become a severe global health emergency with substantial morbidity and mortality. Currently, there are still no promising therapeutics to alleviate the dengue-associated vascular hemorrhage in a clinical setting. In the present study, we first observed that heme oxygenase-1 (HO-1) expression level was highly suppressed in severe DENV-infected patients. In contrast, the overexpression of HO-1 could attenuate DENV-induced pathogenesis, including plasma leakage and thrombocytopenia, in an AG129 mouse model. Our data indicate that overexpression of HO-1 or its metabolite biliverdin can maintain endothelial integrity upon DENV infection in vitro and in vivo. We further characterized the positive regulatory effect of HO-1 on the endothelial adhesion factor vascular endothelial-cadherin to decrease DENV-induced endothelial hyperpermeability. Subsequently, we confirmed that two medicinal plant-derived compounds, andrographolide, and celastrol, widely used as a nutritional or medicinal supplement are useful to attenuate DENV-induced plasma leakage through induction of the HO-1 expression in DENV-infected AG129 mice. In conclusion, our findings reveal that induction of the HO-1 signal pathway is a promising option for the treatment of DENV-induced vascular pathologies.
Collapse
Affiliation(s)
- Yu-Hsuan Wu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Chun Chen
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Kai Tseng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Hsu Chen
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chun-Kuang Lin
- Department of Marine Biotechnology and Resources, Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, Doctoral Degree Program in Marine Biotechnology, College of Marine Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.,Graduate Institute of Medicine in College of Medicine and Graduate Institute of Natural Products in College of Pharmacy, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
18
|
Subbalakshmi AR, Sahoo S, McMullen I, Saxena AN, Venugopal SK, Somarelli JA, Jolly MK. KLF4 Induces Mesenchymal-Epithelial Transition (MET) by Suppressing Multiple EMT-Inducing Transcription Factors. Cancers (Basel) 2021; 13:5135. [PMID: 34680284 PMCID: PMC8533753 DOI: 10.3390/cancers13205135] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Epithelial-Mesenchymal Plasticity (EMP) refers to reversible dynamic processes where cells can transition from epithelial to mesenchymal (EMT) or from mesenchymal to epithelial (MET) phenotypes. Both these processes are modulated by multiple transcription factors acting in concert. While EMT-inducing transcription factors (TFs)-TWIST1/2, ZEB1/2, SNAIL1/2/3, GSC, and FOXC2-are well-characterized, the MET-inducing TFs are relatively poorly understood (OVOL1/2 and GRHL1/2). Here, using mechanism-based mathematical modeling, we show that transcription factor KLF4 can delay the onset of EMT by suppressing multiple EMT-TFs. Our simulations suggest that KLF4 overexpression can promote a phenotypic shift toward a more epithelial state, an observation suggested by the negative correlation of KLF4 with EMT-TFs and with transcriptomic-based EMT scoring metrics in cancer cell lines. We also show that the influence of KLF4 in modulating the EMT dynamics can be strengthened by its ability to inhibit cell-state transitions at the epigenetic level. Thus, KLF4 can inhibit EMT through multiple parallel paths and can act as a putative MET-TF. KLF4 associates with the patient survival metrics across multiple cancers in a context-specific manner, highlighting the complex association of EMP with patient survival.
Collapse
Affiliation(s)
- Ayalur Raghu Subbalakshmi
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | - Sarthak Sahoo
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | | | | | - Sudhanva Kalasapura Venugopal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| | - Jason A. Somarelli
- Department of Medicine, Duke University, Durham, NC 27708, USA;
- Duke Cancer Institute, Duke University, Durham, NC 27708, USA
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India; (A.R.S.); (S.S.); (S.K.V.)
| |
Collapse
|
19
|
Guo XW, Zhang H, Huang JQ, Wang SN, Lu Y, Cheng B, Dong SH, Wang YY, Li FS, Li YW. PIEZO1 Ion Channel Mediates Ionizing Radiation-Induced Pulmonary Endothelial Cell Ferroptosis via Ca 2+/Calpain/VE-Cadherin Signaling. Front Mol Biosci 2021; 8:725274. [PMID: 34568428 PMCID: PMC8458942 DOI: 10.3389/fmolb.2021.725274] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary endothelial cell dysfunction plays an important role in ionizing radiation (IR)-induced lung injury. Whether pulmonary endothelial cell ferroptosis occurs after IR and what are the underlying mechanisms remain elusive. Here, we demonstrate that 15-Gy IR induced ferroptosis characterized by lethal accumulation of reactive oxygen species (ROS), lipid peroxidation, mitochondria shrinkage, and decreased glutathione peroxidase 4 (GPX4) and SLC7A11 expression in pulmonary endothelial cells. The phenomena could be mimicked by Yoda1, a specific activator of mechanosensitive calcium channel PIEZO1. PIEZO1 protein expression was upregulated by IR in vivo and in vitro. The increased PIEZO1 expression after IR was accompanied with increased calcium influx and increased calpain activity. The effects of radiation on lung endothelial cell ferroptosis was partly reversed by inhibition of PIEZO1 activity using the selective inhibitor GsMTx4 or inhibition of downstreaming Ca2+/calpain signaling using PD151746. Both IR and activation of PIEZO1 led to increased degradation of VE-cadherin, while PD151746 blocked these effects. VE-cadherin knockdown by specific siRNA causes ferroptosis-like phenomena with increased ROS and lipid peroxidation in the lung endothelial cells. Overexpression of VE-cadherin partly recused the ferroptosis caused by IR or PIEZO1 activation as supported by decreased ROS production, lipid peroxidation and mitochondria shrinkage compared to IR or PIEZO1 activation alone. In summary, our study reveals a previously unrecognized role of PIEZO1 in modulating ferroptosis, providing a new target for future mitigation of radiation-induced lung injury.
Collapse
Affiliation(s)
- Xue-Wei Guo
- The Postgraduate Training Base of Jinzhou Medical University (The PLA Rocket Force Characteristic Medical Center), Beijing, China.,Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hao Zhang
- Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jia-Qi Huang
- The Postgraduate Training Base of Jinzhou Medical University (The PLA Rocket Force Characteristic Medical Center), Beijing, China.,Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Si-Nian Wang
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yan Lu
- Department of Neurology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Bo Cheng
- Department of Pathology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Su-He Dong
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ying-Ying Wang
- Department of Anesthesiology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Feng-Sheng Li
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yong-Wang Li
- Department of Anesthesiology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
20
|
KLF4 Upregulation in Atherosclerotic Thoracic Aortas: Exploring the Protective Effect of Colchicine-based Regimens in a Hyperlipidemic Rabbit Model. Ann Vasc Surg 2021; 78:328-335. [PMID: 34182114 DOI: 10.1016/j.avsg.2021.04.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inflammatory dysregulation of KLF4 is related to atheromatosis. In the present study, we explored the impact of colchicine-based regimens on the development of thoracic aortic atheromatosis and KLF4 expression. METHODS Twenty-eight New Zealand White rabbits were divided to 4 groups. The control group (n = 6) was fed standard chow, group A (n = 6) was fed chow enriched with 1% w/w cholesterol, group B (n = 8) was fed the same cholesterol-enriched diet plus 2 mg/kg body weight/day colchicine and 250 mg/kg body weight/day fenofibrate, while group C (n = 8) was also fed the same diet plus 2 mg/kg body weight/day colchicine and 15 mg/kg body weight/day N-acetylcysteine. After 7 weeks, all animals were euthanized, and their thoracic aortas were isolated. Atherosclerotic plaque area was estimated with morphometric analysis. KLF4 expression was quantified with quantitative RT-PCR. RESULTS Group A developed significantly more atherosclerosis compared to group B (MD: 13.67, 95% CI: 7.49-19.84) and C (MD: 20.29, 95% CI: 14.12-26.47). Colchicine with N-acetylcysteine resulted in more pronounced reduction in the extent of atherosclerotic plaques compared to colchicine/fibrate (MD: 6.62, 95% CI: 0.90-12.34). Group A exhibited significantly greater KLF4 expression compared to group B (MD: 4.94, 95% CI: 1.11-8.77) and C (MD: 9.94, 95% CI: 6.11-13.77). Combining colchicine with N-acetylcysteine instead of fenofibrate (MD: 5.00, 95% CI: 1.45-8.54) led to a more robust reduction in KLF4 expression. CONCLUSIONS In the present hyperlipidemic animal model, colchicine-based regimens curtailed de novo atherogenesis and KLF4 overexpression in thoracic aortas.
Collapse
|
21
|
Shimotsu R, Hotta K, Ikegami R, Asamura T, Tabuchi A, Masamoto K, Yagishita K, Poole DC, Kano Y. Vascular permeability of skeletal muscle microvessels in rat arterial ligation model: in vivo analysis using two-photon laser scanning microscopy. Am J Physiol Regul Integr Comp Physiol 2021; 320:R972-R983. [PMID: 33949210 DOI: 10.1152/ajpregu.00135.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 04/23/2021] [Indexed: 11/22/2022]
Abstract
Peripheral artery disease (PAD) in the lower limb compromises oxygen supply due to arterial occlusion. Ischemic skeletal muscle is accompanied by capillary structural deformation. Therefore, using novel microscopy techniques, we tested the hypothesis that endothelial cell swelling temporally and quantitatively corresponds to enhanced microvascular permeability. Hindlimb ischemia was created in male Wistar rat's by iliac artery ligation (AL). The tibialis anterior (TA) muscle microcirculation was imaged using intravenously infused rhodamine B isothiocyanate dextran fluorescent dye via two-photon laser scanning microscopy (TPLSM) and dye extravasation at 3 and 7 days post-AL quantified to assess microvascular permeability. The TA microvascular endothelial ultrastructure was analyzed by transmission electron microscopy (TEM). Compared with control (0.40 ± 0.15 μm3 × 106), using TPLSM, the volumetrically determined interstitial leakage of fluorescent dye measured at 3 (3.0 ± 0.40 μm3 × 106) and 7 (2.5 ± 0.8 μm3 × 106) days was increased (both P < 0.05). Capillary wall thickness was also elevated at 3 (0.21 ± 0.06 μm) and 7 (0.21 ± 0.08 μm) days versus control (0.11 ± 0.03 μm, both P < 0.05). Capillary endothelial cell swelling was temporally and quantitatively associated with elevated vascular permeability in the AL model of PAD but these changes occurred in the absence of elevations in protein levels of vascular endothelial growth factor (VEGF) its receptor (VEGFR2 which decreased by AL-7 day) or matrix metalloproteinase. The temporal coherence of endothelial cell swelling and increased vascular permeability supports a common upstream mediator. TPLSM, in combination with TEM, provides a sensitive and spatially discrete technique to assess the mechanistic bases for, and efficacy of, therapeutic countermeasures to the pernicious sequelae of compromised peripheral arterial function.
Collapse
Affiliation(s)
- Rie Shimotsu
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
| | - Kzuki Hotta
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, Japan
| | - Ryo Ikegami
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, Japan
- Department of Health Science, Health Science University, Yamanashi, Japan
| | - Tomoyo Asamura
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
| | - Ayaka Tabuchi
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
| | - Kazuto Masamoto
- Faculty of Informatics and Engineering, University of Electro-Communications, Chofu, Japan
- Center for Neuroscience and Biomedical Engineering (CNBE), University of Electro-Communications, Chofu, Japan
| | - Kazuyoshi Yagishita
- Clinical Center for Sports Medicine and Sports Dentistry, Hyperbaric Medical Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - David C Poole
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Yutaka Kano
- Department of Engineering Science, University of Electro-Communications, Chofu, Japan
- Center for Neuroscience and Biomedical Engineering (CNBE), University of Electro-Communications, Chofu, Japan
| |
Collapse
|
22
|
Yang C, Xiao X, Huang L, Zhou F, Chen LH, Zhao YY, Qu SL, Zhang C. Role of Kruppel-like factor 4 in atherosclerosis. Clin Chim Acta 2020; 512:135-141. [PMID: 33181148 DOI: 10.1016/j.cca.2020.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is one of the chronic progressive diseases, which is caused by vascular injury and promoted by the interaction of various inflammatory factors and inflammatory cells. In recent years, kruppel-like factor 4 (KLF4), a significant transcription factor that participated in cell growth, differentiation and proliferation, has been proved to cause substantial impacts on regulating cardiovascular disease. This paper will give a comprehensive summary to highlight KLF4 as a crucial regulator of foam cell formation, vascular smooth muscle cells (VSMCs) phenotypic transformation, macrophage polarization, endothelial cells inflammation, lymphocyte differentiation and cell proliferation in the process of atherosclerosis. Recent studies show that KLF4 may be an important "molecular switch" in the process of improving vascular injury and inflammation under harmful stimulation, suggesting that KLF4 is a latent disease biomarker for the therapeutic target of atherosclerosis and vascular disease.
Collapse
Affiliation(s)
- Chen Yang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Fan Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Hui Chen
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yu-Yan Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
23
|
Increased Serum KLF4 in Severe Atheromatosis and Extensive Aneurysmal Disease. Ann Vasc Surg 2020; 68:338-343. [DOI: 10.1016/j.avsg.2020.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 11/18/2022]
|
24
|
Anwar M, Mehta D. Post-translational modifications of S1PR1 and endothelial barrier regulation. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158760. [PMID: 32585303 PMCID: PMC7409382 DOI: 10.1016/j.bbalip.2020.158760] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
Abstract
Sphingosine-1-phosphate receptor-1 (S1PR1), a G-protein coupled receptor that is expressed in endothelium and activated upon ligation by the bioactive lipid sphingosine-1-phosphate (S1P), is an important vascular-barrier protective mechanism at the level of adherens junctions (AJ). Loss of endothelial barrier function is a central factor in the pathogenesis of various inflammatory conditions characterized by protein-rich lung edema formation, such as acute respiratory distress syndrome (ARDS). While several S1PR1 agonists are available, the challenge of arresting the progression of protein-rich edema formation remains to be met. In this review, we discuss the role of S1PRs, especially S1PR1, in regulating endothelial barrier function. We review recent findings showing that replenishment of the pool of cell-surface S1PR1 may be crucial to the effectiveness of S1P in repairing the endothelial barrier. In this context, we discuss the S1P generating machinery and mechanisms that regulate S1PR1 at the cell surface and their impact on endothelial barrier function.
Collapse
Affiliation(s)
- Mumtaz Anwar
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago Chicago, IL 60612, United States of America
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago Chicago, IL 60612, United States of America.
| |
Collapse
|
25
|
Khan MJ, Singh P, Dohare R, Jha R, Rahmani AH, Almatroodi SA, Ali S, Syed MA. Inhibition of miRNA-34a Promotes M2 Macrophage Polarization and Improves LPS-Induced Lung Injury by Targeting Klf4. Genes (Basel) 2020; 11:genes11090966. [PMID: 32825525 PMCID: PMC7563942 DOI: 10.3390/genes11090966] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/05/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an outcome of an accelerated immune response that starts initially as a defensive measure, however, due to non-canonical signaling, it later proves to be fatal not only to the affected tissue but to the whole organ system. microRNAs are known for playing a decisive role in regulating the expression of genes involved in diverse functions such as lung development, repair, and inflammation. In-silico analyses of clinical data and microRNA databases predicted a probable interaction between miRNA-34a (miR-34a), mitogen-activated protein kinase 1 (ERK), and kruppel like factor 4 (Klf4). Parallel to in silico results, here, we show that intra-tracheal instillation of lipopolysaccharides (LPS) to mice enhanced miR-34a expression in lung macrophages. Inhibition of miR-34a significantly improved lung histology, whereas over-expression of miR-34a worsened the lung injury phenotype. miR-34a over-expression in macrophages were also demonstrated to favour pro-inflammatory M1 phenotype and inhibition of M2 polarization. In a quest to confirm this likely interaction, expression profiles of Klf4 as the putative target were analyzed in different macrophage polarizing conditions. Klf4 expression was found to be prominent in the miR-34a inhibitor-treated group but down-regulated in the miR-34a mimic treated group. Immuno-histopathological analyses of lung tissue from the mice treated with miR-34a inhibitor also showed reduced inflammatory M1 markers as well as enhanced cell proliferation. The present study indicates that miR-34a intensified LPS-induced lung injury and inflammation by regulating Klf4 and macrophage polarization, which may serve as a potential therapeutic target for acute lung injury/ARDS.
Collapse
Affiliation(s)
- Mohd Junaid Khan
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (P.S.); (R.D.); (R.J.)
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (P.S.); (R.D.); (R.J.)
| | - Rishabh Jha
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (P.S.); (R.D.); (R.J.)
| | - Arshad H. Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (A.H.R.); (S.A.A.)
| | - Saleh A. Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (A.H.R.); (S.A.A.)
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences Jamia Hamdard, New Delhi 110025, India;
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India;
- Correspondence: ; Tel.: +91-995-378-6440
| |
Collapse
|
26
|
Baruah J, Chaudhuri S, Mastej V, Axen C, Hitzman R, Ribeiro IMB, Wary KK. Low-Level Nanog Expression in the Regulation of Quiescent Endothelium. Arterioscler Thromb Vasc Biol 2020; 40:2244-2264. [PMID: 32640900 PMCID: PMC7447188 DOI: 10.1161/atvbaha.120.314875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Supplemental Digital Content is available in the text. Nanog is expressed in adult endothelial cells (ECs) at a low-level, however, its functional significance is not known. The goal of our study was to elucidate the role of Nanog in adult ECs using a genetically engineered mouse model system.
Collapse
Affiliation(s)
- Jugajyoti Baruah
- From the Department of Psychiatry, Harvard Medical School, Boston, MA (J.B.).,Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA (J.B.)
| | - Suhnrita Chaudhuri
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Victoria Mastej
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Cassondra Axen
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Ryan Hitzman
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Isabella M B Ribeiro
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| | - Kishore K Wary
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago (V.M., S.C., C.A., R.H., I.M.B.R., K.K.W.)
| |
Collapse
|
27
|
Li J, Zhao Y, Choi J, Ting KK, Coleman P, Chen J, Cogger VC, Wan L, Shi Z, Moller T, Zheng X, Vadas MA, Gamble JR. Targeting miR-27a/VE-cadherin interactions rescues cerebral cavernous malformations in mice. PLoS Biol 2020; 18:e3000734. [PMID: 32502201 PMCID: PMC7299406 DOI: 10.1371/journal.pbio.3000734] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/17/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions predominantly developing in the central nervous system (CNS), with no effective treatments other than surgery. Loss-of-function mutation in CCM1/krev interaction trapped 1 (KRIT1), CCM2, or CCM3/programmed cell death 10 (PDCD10) causes lesions that are characterized by abnormal vascular integrity. Vascular endothelial cadherin (VE-cadherin), a major regulator of endothelial cell (EC) junctional integrity is strongly disorganized in ECs lining the CCM lesions. We report here that microRNA-27a (miR-27a), a negative regulator of VE-cadherin, is elevated in ECs isolated from mouse brains developing early CCM lesions and in cultured ECs with CCM1 or CCM2 depletion. Furthermore, we show miR-27a acts downstream of kruppel-like factor (KLF)2 and KLF4, two known key transcription factors involved in CCM lesion development. Using CD5-2 (a target site blocker [TSB]) to prevent the miR-27a/VE-cadherin mRNA interaction, we present a potential therapy to increase VE-cadherin expression and thus rescue the abnormal vascular integrity. In CCM1- or CCM2-depleted ECs, CD5-2 reduces monolayer permeability, and in Ccm1 heterozygous mice, it restores dermal vessel barrier function. In a neonatal mouse model of CCM disease, CD5-2 normalizes vasculature and reduces vascular leakage in the lesions, inhibits the development of large lesions, and significantly reduces the size of established lesions in the hindbrain. Furthermore, CD5-2 limits the accumulation of inflammatory cells in the lesion area. Our work has established that VE-cadherin is a potential therapeutic target for normalization of the vasculature and highlights that targeting miR-27a/VE-cadherin interaction by CD5-2 is a potential novel therapy for the devastating disease, CCM. Cerebral cavernous malformation (CCM) is a disease for which, hitherto, surgery has been the only option. This study shows that a potential therapeutic, CD5-2, inhibits lesion development and vascular leak in the brains of CCM neonatal mice by targeting the endothelial cell–specific adhesion molecule VE-cadherin and restoring the vascular integrity of CCM lesions.
Collapse
Affiliation(s)
- Jia Li
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Yang Zhao
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jaesung Choi
- Laboratory of Cardiovascular Signaling, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Ka Ka Ting
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Paul Coleman
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Victoria C. Cogger
- Aging and Alzheimers Institute and ANZAC Research Institute and Concord Hospital, Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Li Wan
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Zhongsong Shi
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Xiangjian Zheng
- Laboratory of Cardiovascular Signaling, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Mathew A. Vadas
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
| | - Jennifer R. Gamble
- Centre for the Endothelium, Vascular Biology Program, Centenary Institute, The University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
28
|
Ruiz S, Vardon-Bounes F, Buléon M, Guilbeau-Frugier C, Séguelas MH, Conil JM, Girolami JP, Tack I, Minville V. Kinin B1 receptor: a potential therapeutic target in sepsis-induced vascular hyperpermeability. J Transl Med 2020; 18:174. [PMID: 32306971 PMCID: PMC7168845 DOI: 10.1186/s12967-020-02342-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background In sepsis, the endothelial barrier becomes incompetent, with the leaking of plasma into interstitial tissues. VE-cadherin, an adherens junction protein, is the gatekeeper of endothelial cohesion. Kinins, released during sepsis, induce vascular leakage and vasodilation. They act via two G-protein coupled receptors: B1 (B1R) and B2 (B2R). B1R is inducible in the presence of pro-inflammatory cytokines, endotoxins or after tissue injury. It acts at a later stage of sepsis and elicits a sustained inflammatory response. The aim of our study was to investigate the relationships between B1R and VE-cadherin destabilization in vivo in a later phase of sepsis. Methods Experimental, prospective study in a university research laboratory. We used a polymicrobial model of septic shock by cecal ligation and puncture in C57BL6 male mice or C57BL6 male mice that received a specific B1R antagonist (R-954). We studied the influence of B1R on sepsis-induced vascular permeability 30 h after surgery for several organs, and VE-cadherin expression in the lung and kidneys by injecting R-954 just before surgery. The 96-h survival was determined in mice without treatment or in animals receiving R-954 as a “prophylactic” regimen (a subcutaneous injection of 200 µg/kg, prior to CLP and 24 h after CLP), or as a “curative” regimen (injection of 100 µg/kg at H6, H24 and H48 post-surgery). Results B1R inactivation helps to maintain MAP above 65 mmHg but induces different permeability profiles depending on whether or not organ perfusion is autoregulated. In our model, VE-cadherin was destabilized in vivo during septic shock. At a late stage of sepsis, the B1R blockade reduced the VE-cadherin disruption by limiting eNOS activation. The survival rate for mice that received R-954 after sepsis induction was higher than in animals that received an antagonist as a prophylactic treatment. Conclusions B1R antagonizing reduced mortality in our model of murine septic shock by limiting the vascular permeability induced by VE-cadherin destabilization through maintenance of the macrohemodynamics, consequently limiting organ dysfunctions.
Collapse
Affiliation(s)
- Stéphanie Ruiz
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France. .,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.
| | - Fanny Vardon-Bounes
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Marie Buléon
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Céline Guilbeau-Frugier
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.,Department of Forensic Medicine, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Biological Electron Microscopy Center, Rangueil Faculty of Medicine, Toulouse University, Toulouse, France
| | - Marie-Hélène Séguelas
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Jean-Marie Conil
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France
| | - Jean-Pierre Girolami
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| | - Ivan Tack
- Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France.,Department of Physiology, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France
| | - Vincent Minville
- Department of Anesthesiology and Intensive Care, Rangueil Hospital-University Hospital of Toulouse, 1 Avenue du Professeur Jean Poulhès TSA 50032, 31059, Toulouse Cedex 9, France.,Institute of Metabolic and Cardiovascular Diseases, INSERM/UPS UMR, 1048-I2MC, Equipe 3, Paul Sabatier University, Toulouse, France
| |
Collapse
|
29
|
Zhang X, Tang X, Ma F, Fan Y, Sun P, Zhu T, Zhang J, Hamblin MH, Chen YE, Yin KJ. Endothelium-targeted overexpression of Krüppel-like factor 11 protects the blood-brain barrier function after ischemic brain injury. Brain Pathol 2020; 30:746-765. [PMID: 32196819 DOI: 10.1111/bpa.12831] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/07/2020] [Accepted: 03/15/2020] [Indexed: 12/22/2022] Open
Abstract
Microvascular endothelial cell (EC) injury and the subsequent blood-brain barrier (BBB) breakdown are frequently seen in many neurological disorders, including stroke. We have previously documented that peroxisome proliferator-activated receptor gamma (PPARγ)-mediated cerebral protection during ischemic insults needs Krüppel-like factor 11 (KLF11) as a critical coactivator. However, the role of endothelial KLF11 in cerebrovascular function and stroke outcome is unclear. This study is aimed at investigating the regulatory role of endothelial KLF11 in BBB preservation and neurovascular protection after ischemic stroke. EC-targeted overexpression of KLF11 significantly mitigated BBB leakage in ischemic brains, evidenced by significantly reduced extravasation of BBB tracers and infiltration of peripheral immune cells, and less brain water content. Endothelial cell-selective KLF11 transgenic (EC-KLF11 Tg) mice also exhibited smaller brain infarct and improved neurological function in response to ischemic insults. Furthermore, EC-targeted transgenic overexpression of KLF11 preserved cerebral tight junction (TJ) levels and attenuated the expression of pro-inflammatory factors in mice after ischemic stroke. Mechanistically, we demonstrated that KLF11 directly binds to the promoter of major endothelial TJ proteins including occludin and ZO-1 to promote their activities. Our data indicate that KLF11 functions at the EC level to preserve BBB structural and functional integrity, and therefore, confers brain protection in ischemic stroke. KLF11 may be a novel therapeutic target for the treatment of ischemic stroke and other neurological conditions involving BBB breakdown and neuroinflammation.
Collapse
Affiliation(s)
- Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Xuelian Tang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Ping Sun
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| | - Tianqing Zhu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA, 70112
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15261
| |
Collapse
|
30
|
Downregulation of S1P Lyase Improves Barrier Function in Human Cerebral Microvascular Endothelial Cells Following an Inflammatory Challenge. Int J Mol Sci 2020; 21:ijms21041240. [PMID: 32069843 PMCID: PMC7072972 DOI: 10.3390/ijms21041240] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/30/2020] [Accepted: 02/10/2020] [Indexed: 01/08/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a key bioactive lipid that regulates a myriad of physiological and pathophysiological processes, including endothelial barrier function, vascular tone, vascular inflammation, and angiogenesis. Various S1P receptor subtypes have been suggested to be involved in the regulation of these processes, whereas the contribution of intracellular S1P (iS1P) through intracellular targets is little explored. In this study, we used the human cerebral microvascular endothelial cell line HCMEC/D3 to stably downregulate the S1P lyase (SPL-kd) and evaluate the consequences on endothelial barrier function and on the molecular factors that regulate barrier tightness under normal and inflammatory conditions. The results show that in SPL-kd cells, transendothelial electrical resistance, as a measure of barrier integrity, was regulated in a dual manner. SPL-kd cells had a delayed barrier build up, a shorter interval of a stable barrier, and, thereafter, a continuous breakdown. Contrariwise, a protection was seen from the rapid proinflammatory cytokine-mediated barrier breakdown. On the molecular level, SPL-kd caused an increased basal protein expression of the adherens junction molecules PECAM-1, VE-cadherin, and β-catenin, increased activity of the signaling kinases protein kinase C, AMP-dependent kinase, and p38-MAPK, but reduced protein expression of the transcription factor c-Jun. However, the only factors that were significantly reduced in TNFα/SPL-kd compared to TNFα/control cells, which could explain the observed protection, were VCAM-1, IL-6, MCP-1, and c-Jun. Furthermore, lipid profiling revealed that dihydro-S1P and S1P were strongly enhanced in TNFα-treated SPL-kd cells. In summary, our data suggest that SPL inhibition is a valid approach to dampenan inflammatory response and augmente barrier integrity during an inflammatory challenge.
Collapse
|
31
|
Two faces of bivalent domain regulate VEGFA responsiveness and angiogenesis. Cell Death Dis 2020; 11:75. [PMID: 32001672 PMCID: PMC6992747 DOI: 10.1038/s41419-020-2228-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 01/22/2023]
Abstract
The bivalent domain (BD) at promoter region is an unique epigenetic feature poised for activation or repression during cell differentiation in embryonic stem cell. However, the function of BDs in already differentiated cells remains exclusive. By profiling the epigenetic landscape of endothelial cells during VEGFA (vascular endothelial growth factor A) stimulation, we discovered that BDs are widespread in endothelial cells and preferentially marked genes responsive to VEGFA. The BDs responsive to VEGFA have more permissive chromatin environment comparing to other BDs. The initial activation of bivalent genes depends on RNAPII pausing release induced by EZH1 rather than removal of H3K27me3. The later suppression of bivalent gene expression depended on KDM5A recruitment by its interaction with PRC2. Importantly, EZH1 promoted both in vitro and in vivo angiogenesis by upregulating EGR3, whereas KDM5A dampened angiogenesis. Collectively, this study demonstrates a novel dual function of BDs in endothelial cells to control VEGF responsiveness and angiogenesis.
Collapse
|
32
|
Patel MM, Behar AR, Silasi R, Regmi G, Sansam CL, Keshari RS, Lupu F, Lupu C. Role of ADTRP (Androgen-Dependent Tissue Factor Pathway Inhibitor Regulating Protein) in Vascular Development and Function. J Am Heart Assoc 2019; 7:e010690. [PMID: 30571485 PMCID: PMC6404433 DOI: 10.1161/jaha.118.010690] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The physiological function of ADTRP (androgen‐dependent tissue factor pathway inhibitor regulating protein) is unknown. We previously identified ADTRP as coregulating with and supporting the anticoagulant activity of tissue factor pathway inhibitor in endothelial cells in vitro. Here, we studied the role of ADTRP in vivo, specifically related to vascular development, stability, and function. Methods and Results Genetic inhibition of Adtrp produced vascular malformations in the low‐pressure vasculature of zebrafish embryos and newborn mice: dilation/tortuosity, perivascular inflammation, extravascular proteolysis, increased permeability, and microhemorrhages, which produced partially penetrant lethality. Vascular leakiness correlated with decreased endothelial cell junction components VE‐cadherin and claudin‐5. Changes in hemostasis in young adults comprised modest decrease of tissue factor pathway inhibitor antigen and activity and increased tail bleeding time and volume. Cell‐based reporter assays revealed that ADTRP negatively regulates canonical Wnt signaling, affecting membrane events downstream of low‐density lipoprotein receptor‐related protein 6 (LRP6) and upstream of glycogen synthase kinase 3 beta. ADTRP deficiency increased aberrant/ectopic Wnt/β‐catenin signaling in vivo in newborn mice and zebrafish embryos, and upregulated matrix metallopeptidase (MMP)‐9 in endothelial cells and mast cells (MCs). Vascular lesions in newborn Adtrp−/− pups displayed accumulation of MCs, decreased extracellular matrix content, and deficient perivascular cell coverage. Wnt‐pathway inhibition reversed the increased mmp9 in zebrafish embryos, demonstrating that mmp9 expression induced by Adtrp deficiency was downstream of canonical Wnt signaling. Conclusions Our studies demonstrate that ADTRP plays a major role in vascular development and function, most likely through expression in endothelial cells and/or perivascular cells of Wnt‐regulated genes that control vascular stability and integrity.
Collapse
Affiliation(s)
- Maulin M Patel
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK.,3 Department of Cell Biology University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Amanda R Behar
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| | - Robert Silasi
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| | - Girija Regmi
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| | - Christopher L Sansam
- 2 Cell Cycle & Cancer Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| | - Ravi S Keshari
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| | - Florea Lupu
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK.,3 Department of Cell Biology University of Oklahoma Health Sciences Center Oklahoma City OK.,4 Department of Pathology University of Oklahoma Health Sciences Center Oklahoma City OK
| | - Cristina Lupu
- 1 Cardiovascular Biology Research Program Oklahoma Medical Research Foundation Oklahoma City OK
| |
Collapse
|
33
|
Vascular Wall as Source of Stem Cells Able to Differentiate into Endothelial Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31797283 DOI: 10.1007/5584_2019_421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
The traditional view of the vascular biology is changed by the discovery of vascular progenitor cells in bone marrow or peripheral blood Further complexity is due to the findings that the vessel walls harbor progenitor and stem cells, called vascular wall-resident vascular stem cells (VW-VSCs), able to differentiate to mature vascular wall cells. These immature stem/progenitor cell populations and multipotent mesenchymal lineage participate in postnatal neovascularization and vascular wall remodeling. Further studies are necessary to deepen the knowledge on characterization and biology of VW-VSCs, in particular of endothelial progenitor cells (EPCs) in order to improve their use in clinical settings for regenerative approaches.
Collapse
|
34
|
Boen JRA, Gevaert AB, De Keulenaer GW, Van Craenenbroeck EM, Segers VFM. The role of endothelial miRNAs in myocardial biology and disease. J Mol Cell Cardiol 2019; 138:75-87. [PMID: 31756323 DOI: 10.1016/j.yjmcc.2019.11.151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
The myocardium is a highly structured pluricellular tissue which is governed by an intricate network of intercellular communication. Endothelial cells are the most abundant cell type in the myocardium and exert crucial roles in both healthy myocardium and during myocardial disease. In the last decade, microRNAs have emerged as new actors in the regulation of cellular function in almost every cell type. Here, we review recent evidence on the regulatory function of different microRNAs expressed in endothelial cells, also called endothelial microRNAs, in healthy and diseased myocardium. Endothelial microRNA emerged as modulators of angiogenesis in the myocardium, they are implicated in the paracrine role of endothelial cells in regulating cardiac contractility and homeostasis, and interfere in the crosstalk between endothelial cells and cardiomyocytes.
Collapse
Affiliation(s)
- Jente R A Boen
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Andreas B Gevaert
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, ZNA Middelheim Hospital, Lindendreef 1, 2020 Antwerp, Belgium.
| | - Emeline M Van Craenenbroeck
- Research group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium.
| | - Vincent F M Segers
- Department of Cardiology, Antwerp University Hospital (UZA), Wilrijkstraat 10, Edegem, Belgium; Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
35
|
Fan Y, Lu H, Liang W, Hu W, Zhang J, Chen YE. Krüppel-like factors and vascular wall homeostasis. J Mol Cell Biol 2018; 9:352-363. [PMID: 28992202 DOI: 10.1093/jmcb/mjx037] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are major causes of death worldwide. Identification of promising targets for prevention and treatment of CVDs is paramount in the cardiovascular field. Numerous transcription factors regulate cellular function through modulation of specific genes and thereby are involved in the physiological and pathophysiological processes of CVDs. Although Krüppel-like factors (KLFs) have a similar protein structure with a conserved zinc finger domain, they possess distinct tissue and cell distribution patterns as well as biological functions. In the vascular system, KLF activities are regulated at both transcriptional and posttranscriptional levels. Growing in vitro, in vivo, and genetic epidemiology studies suggest that specific KLFs play important roles in vascular wall biology, which further affect vascular diseases. KLFs regulate various functional aspects such as cell growth, differentiation, activation, and development through controlling a whole cluster of functionally related genes and modulating various signaling pathways in response to pathological conditions. Therapeutic targeting of selective KLF family members may be desirable to achieve distinct treatment effects in the context of various vascular diseases. Further elucidation of the association of KLFs with human CVDs, their underlying molecular mechanisms, and precise protein structure studies will be essential to define KLFs as promising targets for therapeutic interventions in CVDs.
Collapse
Affiliation(s)
- Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenying Liang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Wenting Hu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
36
|
|
37
|
Differential Regulation of Zfp30 Expression in Murine Airway Epithelia Through Altered Binding of ZFP148 to rs51434084. G3-GENES GENOMES GENETICS 2018; 8:687-693. [PMID: 29242385 PMCID: PMC5919737 DOI: 10.1534/g3.117.300507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neutrophil chemotaxis to the airways is a key aspect of host response to microbes and a feature of multiple pulmonary diseases including asthma. Tight regulation of this recruitment is critical to prevent unwanted host tissue damage and inflammation. Using a mouse (Mus musculus) model of asthma applied to the Collaborative Cross population, we previously identified a lung gene expression quantitative trait locus (eQTL) for Zinc finger protein 30 (Zfp30) that was also a QTL for neutrophil recruitment and the hallmark neutrophil chemokine CXCL1. The Zfp30 eQTL is defined by three functionally distinct haplotypes. In this study, we searched for causal genetic variants that underlie the Zfp30 eQTL to gain a better understanding of this candidate repressor's regulation. First, we identified a putative regulatory region spanning 500 bp upstream of Zfp30, which contains 10 SNPs that form five haplotypes. In reporter gene assays in vitro, these haplotypes recapitulated the three previously identified in vivo expression patterns. Second, using site-directed mutagenesis followed by reporter gene assays, we identified a single variant, rs51434084, which explained the majority of variation in expression between two out of three haplotype groups. Finally, using a combination of in silico predictions and electrophoretic mobility shift assays, we identified ZFP148 as a transcription factor that differentially binds to the Zfp30 promoter region harboring rs51434084. In conclusion, we provide evidence in support of rs51434084 being a causal variant for the Zfp30 eQTL, and have identified a mechanism by which this variant alters Zfp30 expression, namely differential binding of ZFP148.
Collapse
|
38
|
KLF4 protects brain microvascular endothelial cells from ischemic stroke induced apoptosis by transcriptionally activating MALAT1. Biochem Biophys Res Commun 2018; 495:2376-2382. [DOI: 10.1016/j.bbrc.2017.11.205] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 11/30/2017] [Indexed: 02/05/2023]
|
39
|
Lin L, Han Q, Xiong Y, Li T, Liu Z, Xu H, Wu Y, Wang N, Liu X. Krüpple-like-factor 4 Attenuates Lung Fibrosis via Inhibiting Epithelial-mesenchymal Transition. Sci Rep 2017; 7:15847. [PMID: 29158503 PMCID: PMC5696468 DOI: 10.1038/s41598-017-14602-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 09/26/2017] [Indexed: 12/03/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Krüpple-like-factor 4 (KLF4), has been suggested to play an important role in the phenotype transition. However, its function in pulmonary fibrosis and EMT of human alveolar epithelial cells (AECs) remains unclear. This study aimed to examine the role of KLF4 in pulmonary fibrosis and EMT. Decreased expression of KLF4 was first observed in human IPF lung tissues and models of bleomycin-induced pulmonary fibrosis. Transgenic mice with overexpression of KLF4 were subjected to bleomycin-induced pulmonary fibrosis model and showed attenuated lung fibrosis and EMT compared to wild type group. Furthermore, the effects overexpression and knockdown of KLF4 on TGF-β1-induced EMT were examined in AECs. Adenovirus-mediated overexpression of KLF4 attenuated TGF-β1-induced EMT and activation of Smad2/3 and Dvl in AECs. Conversely, knockdown of KLF4 promoted the activation of pathways above mentioned and TGF-β1-induced EMT. Our results demonstrates that KLF4 plays an important role in bleomycin-induced lung fibrosis through suppressing TGFβ1-induced EMT. Thus, it may serve as a potential target for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Lianjun Lin
- The Geriatrics Department, Peking University First Hospital, Beijing, China
| | - Qian Han
- The Geriatrics Department, Peking University First Hospital, Beijing, China
| | - Yan Xiong
- The Pathology Department, Peking University First Hospital, Beijing, China
| | - Ting Li
- The Pathology Department, Peking University First Hospital, Beijing, China
| | - Zhonghui Liu
- The Geriatrics Department, Peking University First Hospital, Beijing, China
| | - Huiying Xu
- The Geriatrics Department, Peking University First Hospital, Beijing, China
| | - Yanping Wu
- The Geriatrics Department, Peking University First Hospital, Beijing, China
| | - Nanping Wang
- Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China.
| | - Xinmin Liu
- The Geriatrics Department, Peking University First Hospital, Beijing, China.
| |
Collapse
|
40
|
Baruah J, Hitzman R, Zhang J, Chaudhuri S, Mastej V, Wary KK. The allosteric glycogen synthase kinase-3 inhibitor NP12 limits myocardial remodeling and promotes angiogenesis in an acute myocardial infarction model. J Biol Chem 2017; 292:20785-20798. [PMID: 29070680 DOI: 10.1074/jbc.m117.814376] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/19/2017] [Indexed: 12/22/2022] Open
Abstract
A key feature of acute myocardial infarction (AMI) is an alteration in cardiac architecture. Signaling events that result in the inhibition of glycogen synthase kinase-3 (GSK-3)β represent an adaptive response that might limit the extent of adverse remodeling in the aftermath of AMI. Here, we report that an allosteric inhibitor of GSK-3β, 4-benzyl-2-(naphthalene-1-yl)-1,2,4-thiadiazolidine-3,5-dione (NP12), lessens the magnitude of adverse myocardial remodeling and promotes angiogenesis. Male and female mice 8-10 weeks old were grouped (six animals in each group) into sham surgery (sham group), left anterior descending (LAD) ligation of the coronary artery followed by intramyocardial PBS injections (control group), and LAD ligation followed by NP12 administration (NP12 group). After 7 and 14 days, the extents of fibrosis and integrity of blood vessels were determined. Intramyocardial administration of NP12 increased phosphorylation of GSK-3β, reduced fibrosis, and restored diastolic function in the mice that had experienced an AMI. Morphometric analyses revealed increased CD31+ and Ki67+ vascular structures and decreased apoptosis in these mice. NP12 administration mediated proliferation of reparative cells in the AMI hearts. In a time-course analysis, Wnt3a and NP12 stabilized β-catenin and increased expression of both Nanog and VEGFR2. Moreover, NP12 increased the expression of β-catenin and Nanog in myocardium from AMI mice. Finally, loss- and gain-of-function experiments indicated that the NP12-mediated benefit is, in part, Nanog-specific. These findings indicate that NP12 reduces fibrosis, reestablishes coronary blood flow, and improves ventricular function following an AMI. We conclude that NP12 might be useful for limiting ventricular remodeling after an AMI.
Collapse
Affiliation(s)
- Jugajyoti Baruah
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| | - Ryan Hitzman
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| | - Jane Zhang
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| | - Suhnrita Chaudhuri
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| | - Victoria Mastej
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| | - Kishore K Wary
- From the Department of Pharmacology, University of Illinois, Chicago, Illinois 60612
| |
Collapse
|
41
|
Wary A, Wary N, Baruah J, Mastej V, Wary KK. Chromatin-modifying agents convert fibroblasts to OCT4+ and VEGFR-2+ capillary tube-forming cells. PLoS One 2017; 12:e0176496. [PMID: 28467484 PMCID: PMC5415225 DOI: 10.1371/journal.pone.0176496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 04/10/2017] [Indexed: 12/13/2022] Open
Abstract
RATIONALE The human epigenome is plastic. The goal of this study was to address if fibroblast cells can be epigenetically modified to promote neovessel formation. METHODS AND RESULTS Here, we used highly abundant human adult dermal fibroblast cells (hADFCs) that were treated with the chromatin-modifying agents 5-aza-2'-deoxycytidine and trichostatin A, and subsequently subjected to differentiation by activating Wnt signaling. Our results show that these epigenetically modified hADFCs increasingly expressed β-catenin, pluripotency factor octamer-binding transcription factor-4 (OCT4, also known as POU5F1), and endothelial cell (EC) marker called vascular endothelial growth factor receptor-2 (VEGFR-2, also known as Fetal Liver Kinase-1). In microscopic analysis, β-catenin localized to cell-cell contact points, while OCT4 was found to be localized primarily to the nucleus of these cells. Furthermore, in a chromatin immunoprecipitation experiment, OCT4 bound to the VEGFR-2/FLK1 promoter. Finally, these modified hADFCs also transduced Wnt signaling. Importantly, on a two-dimensional (2D) gel substrate, a subset of the converted cells formed vascular network-like structures in the presence of VEGF. CONCLUSION Chromatin-modifying agents converted hADFCs to OCT4+ and VEGFR-2+ capillary tube-forming cells in a 2D matrix in VEGF-dependent manner.
Collapse
Affiliation(s)
- Anita Wary
- York Community High School, Elmhurst, Illinois, United States of America
| | - Neil Wary
- Illinois Mathematics and Science Academy, Aurora, Illinois, United States of America
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Victoria Mastej
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Kishore K. Wary
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
42
|
Genetic Deletion of Krüppel-Like Factor 11 Aggravates Ischemic Brain Injury. Mol Neurobiol 2017; 55:2911-2921. [PMID: 28456933 DOI: 10.1007/s12035-017-0556-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/12/2017] [Indexed: 02/02/2023]
Abstract
Krüppel-like factors (KLFs) belong to the zinc finger family of transcription factors, and their function in the CNS is largely unexplored. KLF11 is a member of the KLF family, and we have previously demonstrated that peroxisome proliferator-activated receptor gamma-mediated cerebral protection during ischemic insults needs recruitment of KLF11 as its critical coactivator. Here, we sought to determine the role of KLF11 itself in cerebrovascular function and the pathogenesis of ischemic stroke. Transient middle cerebral artery occlusion (MCAO) was performed in KLF11 knockout and wild-type control mice, and brain infarction was analyzed by TTC staining. BBB integrity was assessed by using Evans Blue and TMR-Dextran extravasation assays. KLF11 KO mice exhibited significantly larger brain infarction and poorer neurological outcomes in response to ischemic insults. Genetic deficiency of KLF11 in mice also significantly aggravated ischemia-induced BBB disruption by increasing cerebrovascular permeability and edema. Mechanistically, KLF11 was found to directly regulate IL-6 in the brains of ischemic mice. These findings suggest that KLF11 acts as a novel protective factor in ischemic stroke. Elucidating the functional importance of KLF11 in ischemia may lead us to discover novel pharmacological targets for the development of effective therapies against ischemic stroke.
Collapse
|
43
|
Döring Y, Noels H, van der Vorst EPC, Neideck C, Egea V, Drechsler M, Mandl M, Pawig L, Jansen Y, Schröder K, Bidzhekov K, Megens RTA, Theelen W, Klinkhammer BM, Boor P, Schurgers L, van Gorp R, Ries C, Kusters PJH, van der Wal A, Hackeng TM, Gäbel G, Brandes RP, Soehnlein O, Lutgens E, Vestweber D, Teupser D, Holdt LM, Rader DJ, Saleheen D, Weber C. Vascular CXCR4 Limits Atherosclerosis by Maintaining Arterial Integrity: Evidence From Mouse and Human Studies. Circulation 2017; 136:388-403. [PMID: 28450349 DOI: 10.1161/circulationaha.117.027646] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/17/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND The CXCL12/CXCR4 chemokine ligand/receptor axis controls (progenitor) cell homeostasis and trafficking. So far, an atheroprotective role of CXCL12/CXCR4 has only been implied through pharmacological intervention, in particular, because the somatic deletion of the CXCR4 gene in mice is embryonically lethal. Moreover, cell-specific effects of CXCR4 in the arterial wall and underlying mechanisms remain elusive, prompting us to investigate the relevance of CXCR4 in vascular cell types for atheroprotection. METHODS We examined the role of vascular CXCR4 in atherosclerosis and plaque composition by inducing an endothelial cell (BmxCreERT2-driven)-specific or smooth muscle cell (SMC, SmmhcCreERT2- or TaglnCre-driven)-specific deficiency of CXCR4 in an apolipoprotein E-deficient mouse model. To identify underlying mechanisms for effects of CXCR4, we studied endothelial permeability, intravital leukocyte adhesion, involvement of the Akt/WNT/β-catenin signaling pathway and relevant phosphatases in VE-cadherin expression and function, vascular tone in aortic rings, cholesterol efflux from macrophages, and expression of SMC phenotypic markers. Finally, we analyzed associations of common genetic variants at the CXCR4 locus with the risk for coronary heart disease, along with CXCR4 transcript expression in human atherosclerotic plaques. RESULTS The cell-specific deletion of CXCR4 in arterial endothelial cells (n=12-15) or SMCs (n=13-24) markedly increased atherosclerotic lesion formation in hyperlipidemic mice. Endothelial barrier function was promoted by CXCL12/CXCR4, which triggered Akt/WNT/β-catenin signaling to drive VE-cadherin expression and stabilized junctional VE-cadherin complexes through associated phosphatases. Conversely, endothelial CXCR4 deficiency caused arterial leakage and inflammatory leukocyte recruitment during atherogenesis. In arterial SMCs, CXCR4 sustained normal vascular reactivity and contractile responses, whereas CXCR4 deficiency favored a synthetic phenotype, the occurrence of macrophage-like SMCs in the lesions, and impaired cholesterol efflux. Regression analyses in humans (n=259 796) identified the C-allele at rs2322864 within the CXCR4 locus to be associated with increased risk for coronary heart disease. In line, C/C risk genotype carriers showed reduced CXCR4 expression in carotid artery plaques (n=188), which was furthermore associated with symptomatic disease. CONCLUSIONS Our data clearly establish that vascular CXCR4 limits atherosclerosis by maintaining arterial integrity, preserving endothelial barrier function, and a normal contractile SMC phenotype. Enhancing these beneficial functions of arterial CXCR4 by selective modulators might open novel therapeutic options in atherosclerosis.
Collapse
Affiliation(s)
| | - Heidi Noels
- From Institute for Cardiovascular Prevention (IPEK), LMU Munich, Germany (Y.D., E.P.C.v.d.V., C.N., V.E., M.D., M.M., Y.J., K.B., R.T.A.M., C.R., O.S., E.T., C.W.); Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Germany (H.N., L.P., W.T.); Institute for Cardiovascular Physiology, Vascular Research Centre, Goethe University, Frankfurt am Main, Germany (K.S., R.P.B.); Division of Nephrology and Immunology, RWTH Aachen University Hospital, Germany (B.M.K., P.B.); Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, the Netherlands (R.T.A.M., R.v.G., T.M.H., C.W.); Academic Medical Center, Department of Pathology and Department of Medical Biochemistry, Amsterdam University, the Netherlands (P.J.H.K., A.v.D.W., E.T.); Department of Vascular and Endovascular Surgery, LMU Munich, Germany (G.G.); DZHK (German Centre for Cardiovascular Research), partner site Frankfurt am Main, Germany (R.P.B.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (O.S., C.W.); Department of Physiology and Pharmacology, Karolinksa Institutet, Stockholm, Sweden (O.S.); Max-Plank-Institute for Molecular Biomedicine, Münster, Germany (D.V.); Institute for Laboratory Medicine, LMU Munich, Germany (D.T., L.M.H.); and Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (D.J.R., D.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Peter A, Fatykhova D, Kershaw O, Gruber AD, Rueckert J, Neudecker J, Toennies M, Bauer TT, Schneider P, Schimek M, Eggeling S, Suttorp N, Hocke AC, Hippenstiel S. Localization and pneumococcal alteration of junction proteins in the human alveolar-capillary compartment. Histochem Cell Biol 2017; 147:707-719. [PMID: 28247028 DOI: 10.1007/s00418-017-1551-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2017] [Indexed: 02/03/2023]
Abstract
Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Andrea Peter
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Department for Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355, Berlin, Germany
| | - Diana Fatykhova
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Jens Rueckert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Mario Toennies
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Torsten T Bauer
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Paul Schneider
- Department for General and Thoracic Surgery, DRK Clinics, Drontheimer Strasse 39-40, 13359, Berlin, Germany
| | - Maria Schimek
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas C Hocke
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
45
|
Sangwung P, Zhou G, Nayak L, Chan ER, Kumar S, Kang DW, Zhang R, Liao X, Lu Y, Sugi K, Fujioka H, Shi H, Lapping SD, Ghosh CC, Higgins SJ, Parikh SM, Jo H, Jain MK. KLF2 and KLF4 control endothelial identity and vascular integrity. JCI Insight 2017; 2:e91700. [PMID: 28239661 DOI: 10.1172/jci.insight.91700] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Maintenance of vascular integrity in the adult animal is needed for survival, and it is critically dependent on the endothelial lining, which controls barrier function, blood fluidity, and flow dynamics. However, nodal regulators that coordinate endothelial identity and function in the adult animal remain poorly characterized. Here, we show that endothelial KLF2 and KLF4 control a large segment of the endothelial transcriptome, thereby affecting virtually all key endothelial functions. Inducible endothelial-specific deletion of Klf2 and/or Klf4 reveals that a single allele of either gene is sufficient for survival, but absence of both (EC-DKO) results in acute death from myocardial infarction, heart failure, and stroke. EC-DKO animals exhibit profound compromise in vascular integrity and profound dysregulation of the coagulation system. Collectively, these studies establish an absolute requirement for KLF2/4 for maintenance of endothelial and vascular integrity in the adult animal.
Collapse
Affiliation(s)
- Panjamaporn Sangwung
- Cardiovascular Research Institute, Department of Medicine, and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Guangjin Zhou
- Cardiovascular Research Institute, Department of Medicine, and
| | - Lalitha Nayak
- Cardiovascular Research Institute, Department of Medicine, and.,Division of Hematology and Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - E Ricky Chan
- Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Dong-Won Kang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Rongli Zhang
- Cardiovascular Research Institute, Department of Medicine, and
| | - Xudong Liao
- Cardiovascular Research Institute, Department of Medicine, and
| | - Yuan Lu
- Cardiovascular Research Institute, Department of Medicine, and
| | - Keiki Sugi
- Cardiovascular Research Institute, Department of Medicine, and
| | - Hisashi Fujioka
- Electron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Hong Shi
- Cardiovascular Research Institute, Department of Medicine, and
| | | | - Chandra C Ghosh
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah J Higgins
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Samir M Parikh
- Center for Vascular Biology Research and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA.,Division of Cardiology, Emory University, Atlanta, Georgia, USA
| | - Mukesh K Jain
- Cardiovascular Research Institute, Department of Medicine, and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Ghaleb AM, Yang VW. Krüppel-like factor 4 (KLF4): What we currently know. Gene 2017; 611:27-37. [PMID: 28237823 DOI: 10.1016/j.gene.2017.02.025] [Citation(s) in RCA: 364] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 02/06/2023]
Abstract
Krüppel-like factor 4 (KLF4) is an evolutionarily conserved zinc finger-containing transcription factor that regulates diverse cellular processes such as cell growth, proliferation, and differentiation. Since its discovery in 1996, KLF4 has been gaining a lot of attention, particularly after it was shown in 2006 as one of four factors involved in the induction of pluripotent stem cells (iPSCs). Here we review the current knowledge about the different functions and roles of KLF4 in various tissue and organ systems.
Collapse
Affiliation(s)
- Amr M Ghaleb
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Vincent W Yang
- Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
47
|
Zhang Y, Li Q, Youn JY, Cai H. Protein Phosphotyrosine Phosphatase 1B (PTP1B) in Calpain-dependent Feedback Regulation of Vascular Endothelial Growth Factor Receptor (VEGFR2) in Endothelial Cells: IMPLICATIONS IN VEGF-DEPENDENT ANGIOGENESIS AND DIABETIC WOUND HEALING. J Biol Chem 2016; 292:407-416. [PMID: 27872190 DOI: 10.1074/jbc.m116.766832] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Indexed: 01/13/2023] Open
Abstract
The VEGF/VEGFR2/Akt/eNOS/NO pathway is essential to VEGF-induced angiogenesis. We have previously discovered a novel role of calpain in mediating VEGF-induced PI3K/AMPK/Akt/eNOS activation through Ezrin. Here, we sought to identify possible feedback regulation of VEGFR2 by calpain via its substrate protein phosphotyrosine phosphatase 1B (PTP1B), and the relevance of this pathway to VEGF-induced angiogenesis, especially in diabetic wound healing. Overexpression of PTP1B inhibited VEGF-induced VEGFR2 and Akt phosphorylation in bovine aortic endothelial cells, while PTP1B siRNA increased both, implicating negative regulation of VEGFR2 by PTP1B. Calpain inhibitor ALLN induced VEGFR2 activation, which can be completely blocked by PTP1B overexpression. Calpain activation induced by overexpression or Ca/A23187 resulted in PTP1B cleavage, which can be blocked by ALLN. Moreover, calpain activation inhibited VEGF-induced VEGFR2 phosphorylation, which can be restored by PTP1B siRNA. These data implicate calpain/PTP1B negative feedback regulation of VEGFR2, in addition to the primary signaling pathway of VEGF/VEGFR2/calpain/PI3K/AMPK/Akt/eNOS. We next examined a potential role of PTP1B in VEGF-induced angiogenesis. Endothelial cells transfected with PTP1B siRNA showed faster wound closure in response to VEGF. Aortic discs isolated from PTP1B siRNA-transfected mice also had augmented endothelial outgrowth. Importantly, PTP1B inhibition and/or calpain overexpression significantly accelerated wound healing in STZ-induced diabetic mice. In conclusion, our data for the first time demonstrate a calpain/PTP1B/VEGFR2 negative feedback loop in the regulation of VEGF-induced angiogenesis. Modulation of local PTP1B and/or calpain activities may prove beneficial in the treatment of impaired wound healing in diabetes.
Collapse
Affiliation(s)
- Yixuan Zhang
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Qiang Li
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Ji Youn Youn
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Hua Cai
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| |
Collapse
|
48
|
Zhao J, Chen C, Guo M, Tao Y, Cui P, Zhou Y, Qin N, Zheng J, Zhang J, Xu L. MicroRNA-7 Deficiency Ameliorates the Pathologies of Acute Lung Injury through Elevating KLF4. Front Immunol 2016; 7:389. [PMID: 27774091 PMCID: PMC5054040 DOI: 10.3389/fimmu.2016.00389] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/15/2016] [Indexed: 01/13/2023] Open
Abstract
Recent evidence showed that microRNA-7 (miR-7) played an important role in the pathologies of lung-related diseases. However, the potential role of miR-7 in acute lung injury (ALI) still remains poorly understood. Here, we assessed the effect of miR-7 deficiency on the pathology of ALI. We, first, found that the expression of miR-7 was upregulated in lung tissue in murine LPS-induced ALI model. Notably, we generated miR-7 knock down mice by using miRNA-Sponge technique and found that miR-7 deficiency could ameliorate the pathologies of lung as evidenced by accelerated body weight recovery, reduced level of bronchoalveolar lavage (BAL) proinflammatory cytokines and decreased number of BAL cells in ALI mice. Moreover, the proportion and number of various immune cells in BAL, including innate immune cell F4/80+ macrophages, γδT cells, NK1.1+ T cells, and CD11c+DCs, as well as adaptive immune cell CD4+ T cells and CD8+ T cells, also significantly changed, respectively. Mechanistic evidence showed that KLF4, a target molecule of miR-7, was upregulated in lung tissues in ALI model, accompanied by altered transduction of NF-κB, AKT, and ERK pathway. These data provided a previously unknown role of miR-7 in pathology of ALI, which could ultimately aid the understanding of development of ALI and the development of new therapeutic strategies against clinical inflammatory lung diseases.
Collapse
Affiliation(s)
- Juanjuan Zhao
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Chao Chen
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Yijin Tao
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - PanPan Cui
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical College , Guizhou , China
| | - Nalin Qin
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Jing Zheng
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical College , Guizhou , China
| | - Lin Xu
- Department of Immunology, Zunyi Medical College , Guizhou , China
| |
Collapse
|
49
|
Cuttano R, Rudini N, Bravi L, Corada M, Giampietro C, Papa E, Morini MF, Maddaluno L, Baeyens N, Adams RH, Jain MK, Owens GK, Schwartz M, Lampugnani MG, Dejana E. KLF4 is a key determinant in the development and progression of cerebral cavernous malformations. EMBO Mol Med 2016; 8:6-24. [PMID: 26612856 PMCID: PMC4718159 DOI: 10.15252/emmm.201505433] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cerebral cavernous malformations (CCMs) are vascular malformations located within the central nervous system often resulting in cerebral hemorrhage. Pharmacological treatment is needed, since current therapy is limited to neurosurgery. Familial CCM is caused by loss‐of‐function mutations in any of Ccm1, Ccm2, and Ccm3 genes. CCM cavernomas are lined by endothelial cells (ECs) undergoing endothelial‐to‐mesenchymal transition (EndMT). This switch in phenotype is due to the activation of the transforming growth factor beta/bone morphogenetic protein (TGFβ/BMP) signaling. However, the mechanism linking Ccm gene inactivation and TGFβ/BMP‐dependent EndMT remains undefined. Here, we report that Ccm1 ablation leads to the activation of a MEKK3‐MEK5‐ERK5‐MEF2 signaling axis that induces a strong increase in Kruppel‐like factor 4 (KLF4) in ECs in vivo. KLF4 transcriptional activity is responsible for the EndMT occurring in CCM1‐null ECs. KLF4 promotes TGFβ/BMP signaling through the production of BMP6. Importantly, in endothelial‐specific Ccm1 and Klf4 double knockout mice, we observe a strong reduction in the development of CCM and mouse mortality. Our data unveil KLF4 as a therapeutic target for CCM.
Collapse
Affiliation(s)
| | - Noemi Rudini
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Luca Bravi
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Monica Corada
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Costanza Giampietro
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy Department of Biosciences, University of Milan, Milan, Italy
| | - Eleanna Papa
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy on leave of absence at Department of Neurology, Laboratory for Molecular Neuro-Oncology University Hospital Zurich, Zurich, Switzerland
| | - Marco Francesco Morini
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy on leave of absence at Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Luigi Maddaluno
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy on leave of absence at Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Ralf H Adams
- Department of Tissue Morphogenesis, Faculty of Medicine, Max Planck Institute for Molecular Biomedicine University of Münster, Münster, Germany
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Cleveland, OH, USA Harrington Heart & Vascular Institute, Cleveland, OH, USA Department of Medicine University Hospitals Case Medical Center, Cleveland, OH, USA Case Western Reserve University School of Medicine University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Maria Grazia Lampugnani
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy Mario Negri Institute of Pharmacological Research, Milan, Italy
| | - Elisabetta Dejana
- IFOM the FIRC Institute of Molecular Oncology, Milan, Italy Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden Department of Oncology and Oncohematology, University of Milan, Milan, Italy
| |
Collapse
|
50
|
Liu H, Li G, Zhao W, Hu Y. Inhibition of MiR-92a May Protect Endothelial Cells After Acute Myocardial Infarction in Rats: Role of KLF2/4. Med Sci Monit 2016; 22:2451-62. [PMID: 27411964 PMCID: PMC4957625 DOI: 10.12659/msm.897266] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background This study was designed to investigate the effects of microRNA-92 (miR-92), Kruppel-like factor 2 (KLF2), and Kruppel-like factor 4 (KLF4) on endothelial injury after acute myocardial infarction (AMI). Material/Methods Blood samples were collected from 50 AMI patients for detection of cardiac troponin I (cTnI), heart-type fatty acid-binding protein (H-FABP), and von Willebrand factor (vWF). The Sprague-Dawley rat models of AMI (n=30) were established by ligating their left anterior descending coronary artery. The cardiac markers of AMI patients and rat models were analyzed with enzyme-linked immunosorbent assay and immunohistochemistry. Human umbilical vein endothelial cells were processed into 5 groups: control, negative control, miR-92a inhibitors, miR-92a inhibitors + KLF2 small interfering RNA (siRNA), and miR-92a inhibitors + KLF4 siRNA. Cell proliferation and apoptosis were detected using MTT assay and flow cytometry. RT-PCR and Western blot were conducted to analyze KLF2 and KLF4 expressions. Results AMI patients exhibited significantly higher expression of both endothelial injury markers (e.g., cTnI, H-FABP, vWF) and miR-92a in blood samples, when compared with controls (P<0.05). Model rats also had similar expressional tendencies, along with lower KLF2 and KLF4 expressions (P<0.05). Further, it could be observed in cellular experiments that treatment of miR-92a mimics can further upregulate endothelial injury markers, and miR-92a and both KLF2 and KLF4 were downregulated by miR-92a mimics (all, P<0.05). Also, the luciferase activity assay confirmed the direct binding of miR-92a to 3′ UTR of KLF2/4. Conclusions MiR-92a was involved in the endothelial injury process after AMI and was able to suppress KLF2 and KLF4 expression.
Collapse
Affiliation(s)
- Hongxia Liu
- Department of Clinical Laboratory Medicine, The Central Hospital of Nanyang, Nanyang, Henan, China (mainland)
| | - Guofen Li
- Cell Morphology Inspection of Clinical Laboratory Medicine, The Central Hospital of Nanyang, Nanyang, Henan, China (mainland)
| | - Wenxue Zhao
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yibo Hu
- Department of Cardiopulmonary Exercise Testing, The Central Hospital of Nanyang, Nanyang, Henan, China (mainland)
| |
Collapse
|