1
|
Liu DX, Hao SL, Yang WX. Crosstalk Between β-CATENIN-Mediated Cell Adhesion and the WNT Signaling Pathway. DNA Cell Biol 2023; 42:1-13. [PMID: 36399409 DOI: 10.1089/dna.2022.0424] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cell adhesion and stable signaling regulation are fundamental ways of maintaining homeostasis. Among them, the Wnt/β-CATENIN signaling plays a key role in embryonic development and maintenance of body dynamic homeostasis. At the same time, the key signaling molecule β-CATENIN in the Wnt signaling can also function as a cytoskeletal linker protein to regulate tissue barriers, cell migration, and morphogenesis. Dysregulation of the balance between Wnt signaling and adherens junctions can lead to disease. How β-CATENIN maintains the independence of these two functions, or mediates the interaction and balance of these two functions, has been explored and debated for a long time. In this study, we will focus on five aspects of β-CATENIN chaperone molecules, phosphorylation of β-CATENIN and related proteins, epithelial mesenchymal transition, β-CATENIN homolog protein γ-CATENIN and disease, thus deepening the understanding of the Wnt/β-CATENIN signaling and the homeostasis between cell adhesion and further addressing related disease problems.
Collapse
Affiliation(s)
- Ding-Xi Liu
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Shuang-Li Hao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Luo Y, Tang H, Zhang Z, Zhao R, Wang C, Hou W, Huang Q, Liu J. Pharmacological inhibition of epidermal growth factor receptor attenuates intracranial aneurysm formation by modulating the phenotype of vascular smooth muscle cells. CNS Neurosci Ther 2022; 28:64-76. [PMID: 34729926 PMCID: PMC8673708 DOI: 10.1111/cns.13735] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/15/2022] Open
Abstract
AIM To study the effect of pharmacological inhibition of epidermal growth factor receptor (EGFR) on intracranial aneurysm (IA) initiation. METHODS Human IA samples were analyzed for the expression of p-EGFR and alpha smooth muscle actin (α-SMA) by immunofluorescence (IF). Rat models of IA were established to evaluate the ability of the EGFR inhibitor, erlotinib, to attenuate the incidence of IA. We analyzed anterior cerebral artery tissues by pathological and proteomic detection for the expression of p-EGFR and relevant proteins, and vessel casting was used to evaluate the incidence of aneurysms in each group. Rat vascular smooth muscle cells (VSMCs) and endothelial cells were extracted and used to establish an in vitro co-culture model in a flow chamber with or without erlotinib treatment. We determined p-EGFR and relevant protein expression in VSMCs by immunoblotting analysis. RESULTS Epidermal growth factor receptor activation was found in human IA vessel walls and rat anterior cerebral artery walls. Treatment with erlotinib markedly attenuated the incidence of IA by inhibiting vascular remodeling and pro-inflammatory transformation of VSMC in rat IA vessel walls. Activation of EGFR in rat VSMCs and phenotypic modulation of rat VSMCs were correlated with the strength of shear stress in vitro, and treatment with erlotinib reduced phenotypic modulation of rat VSMCs. In vitro experiments also revealed that EGFR activation could be induced by TNF-α in human brain VSMCs. CONCLUSIONS These results suggest that EGFR plays a critical role in the initiation of IA and that the EGFR inhibitor erlotinib protects rats from IA initiation by regulating phenotypic modulation of VSMCs.
Collapse
Affiliation(s)
- Yin Luo
- Department of Biomedical EngineeringSchool of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| | - Haishuang Tang
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| | - Zhaolong Zhang
- Department of NeurologyStrategic Support Force Medical Center of PLABeijingChina
| | - Rui Zhao
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| | - Chuanchuan Wang
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| | - Wenguang Hou
- Department of Biomedical EngineeringSchool of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Qinghai Huang
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| | - Jianmin Liu
- Department of NeurosurgeryChanghai HospitalSecond Military Medical UniversityShanghaiChina
| |
Collapse
|
3
|
Wang C, Xing C, Li Z, Liu Y, Li Q, Wang Y, Hu J, Yuan L, Yang G. Bioinspired therapeutic platform based on extracellular vesicles for prevention of arterial wall remodeling in hypertension. Bioact Mater 2021; 8:494-504. [PMID: 34541415 PMCID: PMC8427223 DOI: 10.1016/j.bioactmat.2021.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/09/2021] [Accepted: 06/02/2021] [Indexed: 12/22/2022] Open
Abstract
Arterial stiffness due to the vessel remodeling is closely linked to raised blood pressure, and its physiopathologic mechanism is still not fully understood. We here aimed to explore whether extracellular vesicle (EV) mediated intercellular communication between endothelium and smooth muscle cell contribute to the blood vessel remodeling under hypertension. We here revealed that the arterial endothelial cells robustly secreted EV, which in turn could be circulated and/or directly taken up by the subendothelial smooth muscle cells (SMC). Under hypertension, the EV secretion increased and the miRNA profile changed significantly mainly due to the raised mechanical force and subsequent enhanced reactive oxygen species generation. Among the miRNA cargos in the EV, miR-320d/423-5p were found increased most significantly. In vivo delivery of miR-320d/423-5p mimics via engineered EV increased their expression in arterial vessels, recapitulating the phenotype in hypertension. In contrast, therapeutic delivery of miR-320d/423-5p inhibitors via engineered EV alleviated the phenotype in spontaneous hypertension rat model. Together, we have found that the injured endothelium due to the raised mechanical force in hypertension contributes to the arterial wall remodeling via the secreted EV. Our study has not only provided novel insights on the mechanism of hypertension associated blood vessel wall remodeling, but also shed light on therapeutic intervention of hypertension associated vascular diseases.
Collapse
Affiliation(s)
- Chen Wang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Changyang Xing
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Zhelong Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Yunnan Liu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Qiaoying Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Yixiao Wang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Jiao Hu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Guodong Yang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| |
Collapse
|
4
|
Weng J, Chen Z, Li J, He Q, Chen D, Yang L, Su H, Huang J, Yu S, Huang Q, Xu Q, Guo X. Advanced glycation end products induce endothelial hyperpermeability via β-catenin phosphorylation and subsequent up-regulation of ADAM10. J Cell Mol Med 2021; 25:7746-7759. [PMID: 34227224 PMCID: PMC8358892 DOI: 10.1111/jcmm.16659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/31/2022] Open
Abstract
Endothelial hyperpermeability is the initial event in the development of diabetic microvascular complications, and advanced glycation end products (AGEs) are suggested to cause much of the endothelial hyperpermeability associated with diabetes mellitus, but the molecular mechanism remains to be characterized. β-catenin reportedly plays dual functions in maintaining normal endothelial permeability by serving both as an adhesive component and a signal transduction component. Here, we found that AGEs induced the phosphorylation of β-catenin at residues Y654 and Y142 and the endothelial hyperpermeability was reversed when the two residues were blocked. In mechanism, phosphorylation of Y654 was blocked by Src inactivation, whereas phosphorylation of Y142 was reduced by a focal adhesion kinase inhibitor. β-catenin Y654 phosphorylation induced by AGEs facilitated the dissociation of vascular endothelial (VE)-cadherin/β-catenin and the impairment of adherens junctions (AJs), whereas β-catenin Y142 phosphorylation favoured the dissociation of β-catenin and α-catenin. Further investigation revealed that β-catenin Y142 phosphorylation was required for AGEs-mediated β-catenin nuclear translocation, and this nuclear-located β-catenin subsequently activated the TCF/LEF pathway. This pathway promotes the transcription of the Wnt target, ADAM10 (a disintegrin and metalloprotease 10), which mediates VE-cadherin shedding and leads to further impairment of AJs. In summary, our study showed the role of β-catenin Y654 and Y142 phosphorylation in AGEs-mediated endothelial hyperpermeability through VE-cadherin/β-catenin/α-catenin dissociation and up-regulation of ADAM10, thereby advancing our understanding of the underlying mechanisms of AGEs-induced microvascular hyperpermeability.
Collapse
Affiliation(s)
- Jie Weng
- Department of Pulmonary and Critical Care MedicineZhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Zhenfeng Chen
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jieyu Li
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Qi He
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Deshu Chen
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Lin Yang
- Guangzhou Special Service Sanatorium Center of the Rocket ForceGuangzhouChina
| | - Haiying Su
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Junlin Huang
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Shengxiang Yu
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Qiaobing Huang
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Qiulin Xu
- Department of Emergency and Critical MedicineGuangdong Provincial People’s HospitalGuangdong Academy of Medical ScienceGuangzhouChina
| | - Xiaohua Guo
- Department of PathophysiologyGuangdong Provincial Key Laboratory of Shock and MicrocirculationSchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
5
|
Ben-Saadon S, Gavriel M, Zaretsky U, Jaffa AJ, Grisaru D, Elad D. Tissue-engineered arterial intima model exposed to steady wall shear stresses. J Biomech 2021; 117:110236. [PMID: 33508722 DOI: 10.1016/j.jbiomech.2021.110236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
The arterial intima is continuously under pulsatile wall shear stresses (WSS) imposed by the circulating blood. The knowledge of the contribution of smooth muscle cells (SMC) to the response of endothelial cell (EC) to WSS is still incomplete. We developed a co-culture model of EC on top of SMC that mimics the inner in vivo structure of the arterial intima of large arteries. The co-cultured model, as well as a monolayer model of EC, were developed in custom-designed wells that allowed for mechanobiology experiments. Both the monolayer and co-culture models were exposed to steady flow induced WSS of up to 24 dyne/cm2 and for lengths of 60 min. Quantification of WSS induced alterations in the cytoskeletal actin filaments (F-actin) and vascular endothelial cadherin (VE-cadherin) junctions were utilized from confocal images and flow cytometry. High confluency of both models was observed even after exposure to the high WSS. The quantitive analysis revealed larger post WSS amounts of EC F-actin polymerization in the monolayer, which may be explained by the relative help of the SMC to resist the external load of WSS. The VE-cadherin demonstrated morphological alterations in the monolayer model, but without significant changes in their content. The SMC in the co-culture maintained their contractile phenotype post high WSS which is more physiological, but not post low WSS. Generally, the results of this work demonstrate the active role of SMC in the intima performance to resist flow induced WSS.
Collapse
Affiliation(s)
- Sara Ben-Saadon
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Mark Gavriel
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Uri Zaretsky
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Ariel J Jaffa
- Department of Obstetrics and Gynecology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv 64239, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Grisaru
- Department of Gynecological Oncology, Lis Maternity Hospital, Tel-Aviv Medical Center, Tel Aviv 64239, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Elad
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
6
|
Wang B, Zhang M, Urabe G, Shirasu T, Guo LW, Kent KC. PERK Inhibition Promotes Post-angioplasty Re-endothelialization via Modulating SMC Phenotype Changes. J Surg Res 2021; 257:294-305. [PMID: 32871430 PMCID: PMC11034999 DOI: 10.1016/j.jss.2020.05.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/19/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Drug-eluting stents impair post-angioplasty re-endothelialization thus compromising restenosis prevention while heightening thrombotic risks. We recently found that inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) effectively mitigated both restenosis and thrombosis in rodent models. This motivated us to determine how PERK inhibition impacts re-endothelialization. METHODS Re-endothelialization was evaluated in endothelial-denuded rat carotid arteries after balloon angioplasty and periadventitial administration of PERK inhibitor in a hydrogel. To study whether PERK in smooth muscle cells (SMCs) regulates re-endothelialization by paracrinally influencing endothelial cells (ECs), denuded arteries exposing SMCs were lentiviral-infected to silence PERK; in vitro, the extracellular vesicles isolated from the medium of PDGF-activated, PERK-upregulating human primary SMCs were transferred to human primary ECs. RESULTS Treatment with PERK inhibitor versus vehicle control accelerated re-endothelialization in denuded arteries. PERK-specific silencing in the denuded arterial wall (mainly SMCs) also enhanced re-endothelialization compared to scrambled shRNA control. In vitro, while medium transfer from PDGF-activated SMCs impaired EC viability and increased the mRNA levels of dysfunctional EC markers, either PERK inhibition or silencing in donor SMCs mitigated these EC changes. Furthermore, CXCL10, a paracrine cytokine detrimental to ECs, was increased by PDGF activation and decreased after PERK inhibition or silencing in SMCs. CONCLUSIONS Attenuating PERK activity pharmacologically or genetically provides an approach to accelerating post-angioplasty re-endothelialization in rats. The mechanism may involve paracrine factors regulated by PERK in SMCs that impact neighboring ECs. This study rationalizes future development of PERK-targeted endothelium-friendly vascular interventions.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Angioplasty, Balloon/instrumentation
- Animals
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Arteries/surgery
- Coronary Restenosis/etiology
- Coronary Restenosis/prevention & control
- Disease Models, Animal
- Drug-Eluting Stents/adverse effects
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Humans
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Paracrine Communication/drug effects
- Paracrine Communication/genetics
- Protein Kinase Inhibitors/administration & dosage
- RNA, Small Interfering/metabolism
- Rats
- Re-Epithelialization/drug effects
- Re-Epithelialization/genetics
- eIF-2 Kinase/antagonists & inhibitors
- eIF-2 Kinase/genetics
Collapse
Affiliation(s)
- Bowen Wang
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Mengxue Zhang
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Cellular and Molecular Pathology Graduate Program, Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Go Urabe
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Takuro Shirasu
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia; Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Lian-Wang Guo
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia; Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio.
| | - K Craig Kent
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
7
|
Nishihara H, Gastfriend BD, Soldati S, Perriot S, Mathias A, Sano Y, Shimizu F, Gosselet F, Kanda T, Palecek SP, Du Pasquier R, Shusta EV, Engelhardt B. Advancing human induced pluripotent stem cell-derived blood-brain barrier models for studying immune cell interactions. FASEB J 2020; 34:16693-16715. [PMID: 33124083 PMCID: PMC7686106 DOI: 10.1096/fj.202001507rr] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 12/16/2022]
Abstract
Human induced pluripotent stem cell (hiPSC)‐derived blood‐brain barrier (BBB) models established to date lack expression of key adhesion molecules involved in immune cell migration across the BBB in vivo. Here, we introduce the extended endothelial cell culture method (EECM), which differentiates hiPSC‐derived endothelial progenitor cells to brain microvascular endothelial cell (BMEC)‐like cells with good barrier properties and mature tight junctions. Importantly, EECM‐BMEC‐like cells exhibited constitutive cell surface expression of ICAM‐1, ICAM‐2, and E‐selectin. Pro‐inflammatory cytokine stimulation increased the cell surface expression of ICAM‐1 and induced cell surface expression of P‐selectin and VCAM‐1. Co‐culture of EECM‐BMEC‐like cells with hiPSC‐derived smooth muscle‐like cells or their conditioned medium further increased the induction of VCAM‐1. Functional expression of endothelial ICAM‐1 and VCAM‐1 was confirmed by T‐cell interaction with EECM‐BMEC‐like cells. Taken together, we introduce the first hiPSC‐derived BBB model that displays an adhesion molecule phenotype that is suitable for the study of immune cell interactions.
Collapse
Affiliation(s)
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Sylvain Perriot
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory, University of Artois, Lens, France
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA.,Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
8
|
Deng Y, Zhou Z, Lin S, Yu B. METTL1 limits differentiation and functioning of EPCs derived from human-induced pluripotent stem cells through a MAPK/ERK pathway. Biochem Biophys Res Commun 2020; 527:791-798. [PMID: 32430183 DOI: 10.1016/j.bbrc.2020.04.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/22/2020] [Indexed: 01/11/2023]
Abstract
Transplantation of endothelial progenitor cells (EPCs) has high therapeutic potential for ischemia-related ailments like heart attacks and claudication. Due to limited EPC sources, direct reprogramming is a fast-developing way to convert human-induced pluripotent stem cells (hiPSCs) into EPCs fit for transplantation. However, the procedural efficacy was affected by multiple factors, including epigenetic modifications. Recent studies have shown that m7G methylation mediated by Methyltransferase like 1 (METTL1) is required for mouse embryonic stem cells (mESCs) to differentiate normally. Yet, its contributions to EPC differentiation still require elucidation. Here, using immunofluorescence microscopy and Fluorescence-activated Cell Sorting (FACS), we found that the typical EPC markers were significantly increased in METTL1 knockdown (METTL1-KD) hiPSCs-derived EPCs compared to those of control types. In addition, we found that METTL1 knockdown activates the MAPK/ERK signaling pathway during EPCs differentiation from hiPSCs. Furthermore, functional properties of METTL1-KD EPCs were significantly raised above those of control hiPSCs-derived EPCs. Moreover, we proved that METTL1-KD hiPSCs-derived EPCs significantly accelerate vascular smooth muscle cell proliferation and 'phenotype switching' through a co-culture system. To sum up, our results demonstrate that METTL1-KD significantly promotes the differentiation of EPCs along with their in vitro functions, and this effect may be achieved through activation of the MAPK/ERK signaling pathway. This enhances current knowledge of EPC generation from hiPSCs and presents a new therapeutic target of vascular diseases.
Collapse
Affiliation(s)
- Yujie Deng
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhongyang Zhou
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Beixin Yu
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
9
|
Activation of CD137 signaling promotes neointimal formation by attenuating TET2 and transferrring from endothelial cell-derived exosomes to vascular smooth muscle cells. Biomed Pharmacother 2020; 121:109593. [DOI: 10.1016/j.biopha.2019.109593] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/22/2019] [Accepted: 10/26/2019] [Indexed: 12/12/2022] Open
|
10
|
Miyagawa K, Shi M, Chen PI, Hennigs JK, Zhao Z, Wang M, Li CG, Saito T, Taylor S, Sa S, Cao A, Wang L, Snyder MP, Rabinovitch M. Smooth Muscle Contact Drives Endothelial Regeneration by BMPR2-Notch1-Mediated Metabolic and Epigenetic Changes. Circ Res 2019; 124:211-224. [PMID: 30582451 DOI: 10.1161/circresaha.118.313374] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE Maintaining endothelial cells (EC) as a monolayer in the vessel wall depends on their metabolic state and gene expression profile, features influenced by contact with neighboring cells such as pericytes and smooth muscle cells (SMC). Failure to regenerate a normal EC monolayer in response to injury can result in occlusive neointima formation in diseases such as atherosclerosis and pulmonary arterial hypertension. OBJECTIVE We investigated the nature and functional importance of contact-dependent communication between SMC and EC to maintain EC integrity. METHODS AND RESULTS We found that in SMC and EC contact cocultures, BMPR2 (bone morphogenetic protein receptor 2) is required by both cell types to produce collagen IV to activate ILK (integrin-linked kinase). This enzyme directs p-JNK (phospho-c-Jun N-terminal kinase) to the EC membrane, where it stabilizes presenilin1 and releases N1ICD (Notch1 intracellular domain) to promote EC proliferation. This response is necessary for EC regeneration after carotid artery injury. It is deficient in EC-SMC Bmpr2 double heterozygous mice in association with reduced collagen IV production, decreased N1ICD, and attenuated EC proliferation, but can be rescued by targeting N1ICD to EC. Deletion of EC- Notch1 in transgenic mice worsens hypoxia-induced pulmonary hypertension, in association with impaired EC regenerative function associated with loss of precapillary arteries. We further determined that N1ICD maintains EC proliferative capacity by increasing mitochondrial mass and by inducing the phosphofructokinase PFKFB3 (fructose-2,6-bisphosphatase 3). Chromatin immunoprecipitation sequencing analyses showed that PFKFB3 is required for citrate-dependent H3K27 acetylation at enhancer sites of genes regulated by the acetyl transferase p300 and by N1ICD or the N1ICD target MYC and necessary for EC proliferation and homeostasis. CONCLUSIONS Thus, SMC-EC contact is required for activation of Notch1 by BMPR2, to coordinate metabolism with chromatin remodeling of genes that enable EC regeneration, and to maintain monolayer integrity and vascular homeostasis in response to injury.
Collapse
Affiliation(s)
- Kazuya Miyagawa
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Minyi Shi
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Pin-I Chen
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Jan K Hennigs
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Zhixin Zhao
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Mouer Wang
- Department of Medicine (M.W.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Caiyun G Li
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Toshie Saito
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Shalina Taylor
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Silin Sa
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Aiqin Cao
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Lingli Wang
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Michael P Snyder
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Marlene Rabinovitch
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| |
Collapse
|
11
|
Ovarian cancer-derived exosomes promote tumour metastasis in vivo: an effect modulated by the invasiveness capacity of their originating cells. Clin Sci (Lond) 2019; 133:1401-1419. [PMID: 31227603 DOI: 10.1042/cs20190082] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/05/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022]
Abstract
Exosomes are small nanovesicles that carry bioactive molecules which can be delivered to neighbouring cells to modify their biological functions. Studies have showed that exosomes from ovarian cancer (OVCA) cells can alter the cell migration and proliferation of cells within the tumour microenvironment, an effect modulated by the invasiveness capacity of their originating cells. Using an OVCA cell line xenograph mouse model, we showed that exosomes derived from a high invasiveness capacity cell line (exo-SKOV-3) promoted metastasis in vivo compared with exosomes from a low invasiveness capacity cell line (exo-OVCAR-3). Analysis from anin vivo imaging system (IVIS) revealed that exo-SKOV-3 formed metastatic niches, whereas exo-OVCAR-3 formed colonies of clustered cells close to the site of injection. Interestingly, kinetic parameters showed that the half-maximal stimulatory time (ST50) of tumour growth with exo-OVCAR-3 (4.0 ± 0.31 weeks) was significantly lower compared with the ST50 in mice injected with exo-SKOV-3 (4.5 ± 0.32 weeks). However, the number of metastic nodes in mice injected with exo-SKOV-3 was higher compared with exo-OVCAR-3. Using a quantitative mass spectrometry approach (SWATH MS/MS) followed by bioinformatics analysis using the Ingenuity Pathway Analysis (IPA), we identified a total of 771 proteins. Furthermore, 40 of these proteins were differentially expressed in tumour tissues from mice injected with exo-SKOV-3 compared with exo-OVCAR-3, and associated with Wnt canonical pathway (β-catenin). Finally, we identified a set of proteins which had elevated expression in the circulating exosomes in association with tumour metastasis. These observations suggest that exosomal signalling plays an important role in OVCA metastasis.
Collapse
|
12
|
The Functional Implications of Endothelial Gap Junctions and Cellular Mechanics in Vascular Angiogenesis. Cancers (Basel) 2019; 11:cancers11020237. [PMID: 30781714 PMCID: PMC6406946 DOI: 10.3390/cancers11020237] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis—the sprouting and growth of new blood vessels from the existing vasculature—is an important contributor to tumor development, since it facilitates the supply of oxygen and nutrients to cancer cells. Endothelial cells are critically affected during the angiogenic process as their proliferation, motility, and morphology are modulated by pro-angiogenic and environmental factors associated with tumor tissues and cancer cells. Recent in vivo and in vitro studies have revealed that the gap junctions of endothelial cells also participate in the promotion of angiogenesis. Pro-angiogenic factors modulate gap junction function and connexin expression in endothelial cells, whereas endothelial connexins are involved in angiogenic tube formation and in the cell migration of endothelial cells. Several mechanisms, including gap junction function-dependent or -independent pathways, have been proposed. In particular, connexins might have the potential to regulate cell mechanics such as cell morphology, cell migration, and cellular stiffness that are dynamically changed during the angiogenic processes. Here, we review the implication for endothelial gap junctions and cellular mechanics in vascular angiogenesis.
Collapse
|
13
|
Parthasarathi K. The Pulmonary Vascular Barrier: Insights into Structure, Function, and Regulatory Mechanisms. MOLECULAR AND FUNCTIONAL INSIGHTS INTO THE PULMONARY VASCULATURE 2018; 228:41-61. [DOI: 10.1007/978-3-319-68483-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
14
|
Tang R, Zhang G, Chen SY. Smooth Muscle Cell Proangiogenic Phenotype Induced by Cyclopentenyl Cytosine Promotes Endothelial Cell Proliferation and Migration. J Biol Chem 2016; 291:26913-26921. [PMID: 27821588 DOI: 10.1074/jbc.m116.741967] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 10/27/2016] [Indexed: 12/19/2022] Open
Abstract
Vascular smooth muscle cells (SMCs) and endothelial cells (ECs) are in close contact with blood vessels. SMC phenotypes can be altered during pathological vascular remodeling. However, how SMC phenotypes affect EC properties remains largely unknown. In this study, we found that PDGF-BB-induced synthetic SMCs suppressed EC proliferation and migration while exhibiting increased expression of anti-angiogenic factors, such as endostatin, and decreased pro-angiogenic factors, including CXC motif ligand 1 (CXCL1). Cyclopentenyl cytosine (CPEC), a CTP synthase inhibitor that has been reported previously to inhibit SMC proliferation and injury-induced neointima formation, induced SMC redifferentiation. Interestingly, CPEC-conditioned SMC culture medium promoted EC proliferation and migration because of an increase in CXCL1 along with decreased endostatin production in SMCs. Addition of recombinant endostatin protein or blockade of CXCL1 with a neutralizing antibody suppressed the EC proliferation and migration induced by CPEC-conditioned SMC medium. Mechanistically, CPEC functions as a cytosine derivate to stimulate adenosine receptors A1 and A2a, which further activate downstream cAMP and Akt signaling, leading to the phosphorylation of cAMP response element binding protein and, consequently, SMC redifferentiation. These data provided proof of a novel concept that synthetic SMC exhibits an anti-angiogenic SMC phenotype, whereas contractile SMC shows a pro-angiogenic phenotype. CPEC appears to be a potent stimulator for switching the anti-angiogenic SMC phenotype to the pro-angiogenic phenotype, which may be essential for CPEC to accelerate re-endothelialization for vascular repair during injury-induced vascular wall remodeling.
Collapse
Affiliation(s)
- Rui Tang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602
| | - Gui Zhang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
15
|
Náger M, Santacana M, Bhardwaj D, Valls J, Ferrer I, Nogués P, Cantí C, Herreros J. Nuclear phosphorylated Y142 β-catenin accumulates in astrocytomas and glioblastomas and regulates cell invasion. Cell Cycle 2016; 14:3644-55. [PMID: 26654598 DOI: 10.1080/15384101.2015.1104443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a fast growing brain tumor characterized by extensive infiltration into the surrounding tissue and one of the most aggressive cancers. GBM is the most common glioma (originating from glial-derived cells) that either evolves from a low grade astrocytoma or appears de novo. Wnt/β-catenin and Hepatocyte Growth Factor (HGF)/c-Met signaling are hyperactive in human gliomas, where they regulate cell proliferation, migration and stem cell behavior. We previously demonstrated that β-catenin is phosphorylated at Y142 by recombinant c-Met kinase and downstream of HGF signaling in neurons. Here we studied phosphoY142 (PY142) β-catenin and dephospho S/T β-catenin (a classical Wnt transducer) in glioma biopsies, GBM cell lines and biopsy-derived glioma cell cultures. We found that PY142 β-catenin mainly localizes in the nucleus and signals through transcriptional activation in GBM cells. Tissue microarray analysis confirmed strong nuclear PY142 β-catenin immunostaining in astrocytoma and GBM biopsies. By contrast, active β-catenin showed nuclear localization only in GBM samples. Western blot analysis of tumor biopsies further indicated that PY142 and active β-catenin accumulate independently, correlating with the expression of Snail/Slug (an epithelial-mesenchymal transition marker) and Cyclin-D1 (a regulator of cell cycle progression), respectively, in high grade astrocytomas and GBMs. Moreover, GBM cells stimulated with HGF showed increasing levels of PY142 β-catenin and Snail/Slug. Importantly, the expression of mutant Y142F β-catenin decreased cell detachment and invasion induced by HGF in GBM cell lines and biopsy-derived cell cultures. Our results identify PY142 β-catenin as a nuclear β-catenin signaling form that downregulates adhesion and promotes GBM cell invasion.
Collapse
Affiliation(s)
- Mireia Náger
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Maria Santacana
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Deepshikha Bhardwaj
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Joan Valls
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Isidre Ferrer
- c Institute of Neuropathology; Hospital de Bellvitge-IDIBELL ; Barcelona , Spain
| | - Pere Nogués
- d Neurosurgery Unit; Hospital Arnau de Vilanova ; Lleida , Spain
| | - Carles Cantí
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Judit Herreros
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| |
Collapse
|
16
|
Wylezinski LS, Hawiger J. Interleukin 2 Activates Brain Microvascular Endothelial Cells Resulting in Destabilization of Adherens Junctions. J Biol Chem 2016; 291:22913-22923. [PMID: 27601468 DOI: 10.1074/jbc.m116.729038] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Indexed: 11/06/2022] Open
Abstract
The pleiotropic cytokine interleukin 2 (IL2) disrupts the blood-brain barrier and alters brain microcirculation, underlying vascular leak syndrome that complicates cancer immunotherapy with IL2. The microvascular effects of IL2 also play a role in the development of multiple sclerosis and other chronic neurological disorders. The mechanism of IL2-induced disruption of brain microcirculation has not been determined previously. We found that both human and murine brain microvascular endothelial cells express constituents of the IL2 receptor complex. Then we established that signaling through this receptor complex leads to activation of the transcription factor, nuclear factor κB, resulting in expression of proinflammatory interleukin 6 and monocyte chemoattractant protein 1. We also discovered that IL2 induces disruption of adherens junctions, concomitant with cytoskeletal reorganization, ultimately leading to increased endothelial cell permeability. IL2-induced phosphorylation of vascular endothelial cadherin (VE-cadherin), a constituent of adherens junctions, leads to dissociation of its stabilizing adaptor partners, p120-catenin and β-catenin. Increased phosphorylation of VE-cadherin was also accompanied by a reduction of Src homology 2 domain-containing protein-tyrosine phosphatase 2, known to maintain vascular barrier function. These results unravel the mechanism of deleterious effects induced by IL2 on brain microvascular endothelial cells and may inform the development of new measures to improve IL2 cancer immunotherapy, as well as treatments for autoimmune diseases affecting the central nervous system.
Collapse
Affiliation(s)
| | - Jacek Hawiger
- From the Departments of Molecular Physiology and Biophysics and .,Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center and.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37232-2363
| |
Collapse
|
17
|
Asymmetric Wnt Pathway Signaling Facilitates Stem Cell-Like Divisions via the Nonreceptor Tyrosine Kinase FRK-1 in Caenorhabditis elegans. Genetics 2015; 201:1047-60. [PMID: 26358719 DOI: 10.1534/genetics.115.181412] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/05/2015] [Indexed: 11/18/2022] Open
Abstract
Asymmetric cell division is critical during development, as it influences processes such as cell fate specification and cell migration. We have characterized FRK-1, a homolog of the mammalian Fer nonreceptor tyrosine kinase, and found it to be required for differentiation and maintenance of epithelial cell types, including the stem cell-like seam cells of the hypodermis. A genomic knockout of frk-1, allele ok760, results in severely uncoordinated larvae that arrest at the L1 stage and have an excess number of lateral hypodermal cells that appear to have lost asymmetry in the stem cell-like divisions of the seam cell lineage. frk-1(ok760) mutants show that there are excess lateral hypodermal cells that are abnormally shaped and smaller in size compared to wild type, a defect that could be rescued only in a manner dependent on the kinase activity of FRK-1. Additionally, we observed a significant change in the expression of heterochronic regulators in frk-1(ok760) mutants. However, frk-1(ok760) mutants do not express late, nonseam hypodermal GFP markers, suggesting the seam cells do not precociously differentiate as adult-hyp7 cells. Finally, our data also demonstrate a clear role for FRK-1 in seam cell proliferation, as eliminating FRK-1 during the L3-L4 transition results in supernumerary seam cell nuclei that are dependent on asymmetric Wnt signaling. Specifically, we observe aberrant POP-1 and WRM-1 localization that is dependent on the presence of FRK-1 and APR-1. Overall, our data suggest a requirement for FRK-1 in maintaining the identity and proliferation of seam cells primarily through an interaction with the asymmetric Wnt pathway.
Collapse
|
18
|
Nephrocyte-neurocyte interaction and cellular metabolic analysis on membrane-integrated microfluidic device. Sci China Chem 2015. [DOI: 10.1007/s11426-015-5453-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
19
|
Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G, Elia L. TGFβ Triggers miR-143/145 Transfer From Smooth Muscle Cells to Endothelial Cells, Thereby Modulating Vessel Stabilization. Circ Res 2015; 116:1753-64. [PMID: 25801897 DOI: 10.1161/circresaha.116.305178] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/23/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE The miR-143/145 cluster is highly expressed in smooth muscle cells (SMCs), where it regulates phenotypic switch and vascular homeostasis. Whether it plays a role in neighboring endothelial cells (ECs) is still unknown. OBJECTIVE To determine whether SMCs control EC functions through passage of miR-143 and miR-145. METHODS AND RESULTS We used cocultures of SMCs and ECs under different conditions, as well as intact vessels to assess the transfer of miR-143 and miR-145 from one cell type to another. Imaging of cocultured cells transduced with fluorescent miRNAs suggested that miRNA transfer involves membrane protrusions known as tunneling nanotubes. Furthermore, we show that miRNA passage is modulated by the transforming growth factor (TGF) β pathway because both a specific transforming growth factor-β (TGFβ) inhibitor (SB431542) and an shRNA against TGFβRII suppressed the passage of miR-143/145 from SMCs to ECs. Moreover, miR-143 and miR-145 modulated angiogenesis by reducing the proliferation index of ECs and their capacity to form vessel-like structures when cultured on matrigel. We also identified hexokinase II (HKII) and integrin β 8 (ITGβ8)-2 genes essential for the angiogenic potential of ECs-as targets of miR-143 and miR-145, respectively. The inhibition of these genes modulated EC phenotype, similarly to miR-143 and miR-145 overexpression in ECs. These findings were confirmed by ex vivo and in vivo approaches, in which it was shown that TGFβ and vessel stress, respectively, triggered miR-143/145 transfer from SMCs to ECs. CONCLUSIONS Our results demonstrate that miR-143 and miR-145 act as communication molecules between SMCs and ECs to modulate the angiogenic and vessel stabilization properties of ECs.
Collapse
Affiliation(s)
- Montserrat Climent
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Manuela Quintavalle
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Michele Miragoli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Ju Chen
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Gianluigi Condorelli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| | - Leonardo Elia
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| |
Collapse
|
20
|
Affiliation(s)
- Valerie Z Wall
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle
| | - Karin E Bornfeldt
- From the Departments of Pathology (V.Z.W., K.E.B.) and Medicine, Division of Metabolism, Endocrinology and Nutrition (K.E.B.), Diabetes and Obesity Center of Excellence, University of Washington School of Medicine, Seattle.
| |
Collapse
|