1
|
Zhang L, Zhou J, Kong W. Extracellular matrix in vascular homeostasis and disease. Nat Rev Cardiol 2025; 22:333-353. [PMID: 39743560 DOI: 10.1038/s41569-024-01103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
The extracellular matrix is an essential component and constitutes a dynamic microenvironment of the vessel wall with an indispensable role in vascular homeostasis and disease. From early development through to ageing, the vascular extracellular matrix undergoes various biochemical and biomechanical alterations in response to diverse environmental cues and exerts precise regulatory control over vessel remodelling. Advances in novel technologies that enable the comprehensive evaluation of extracellular matrix components and cell-matrix interactions have led to the emergence of therapeutic strategies that specifically target this fine-tuned network. In this Review, we explore various aspects of extracellular matrix biology in vascular development, disorders and ageing, emphasizing the effect of the extracellular matrix on disease initiation and progression. Additionally, we provide an overview of the potential therapeutic implications of targeting the extracellular matrix microenvironment in vascular diseases.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
2
|
Lin Y, Bian L, Zhu G, Zhang B. Vitronectin promotes proliferation and metastasis of cervical cancer cells via the epithelial-mesenchymal transition. Front Oncol 2024; 14:1466264. [PMID: 39717749 PMCID: PMC11663901 DOI: 10.3389/fonc.2024.1466264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/15/2024] [Indexed: 12/25/2024] Open
Abstract
Background Vitronectin (VTN) is a multifunctional glycoprotein in blood and the extracellular, which could be an effective biomarker for many cancers. However, its role in cervical cancer is under investigated. In this study, we aimed to determine the molecular function of VTN and its potential mechanism in cervical cancer (CC). Materials and methods Up- and down-regulated VTN expression was determined in Hela and C33A cells. Reverse transcription, qRT-PCR, and Western blotting test were performed to identify VTN mRNA and protein levels, separately. CCK-8 assay and colony formation assay were carried out to evaluate proliferation abilities of CC cells. A scratch test and a transwell chamber assay were performed to determine cell migration and invasion ability. Expression levels of epithelial-mesenchymal transition (EMT)-related proteins were measured by Western blotting. Results Cell models with up- and down-regulated VTN expression in Hela and C33A cells were successfully established, as confirmed by Western blotting and qPCR. CCK-8 and colony formation assays demonstrated that VTN overexpression significantly enhanced the proliferation of both Hela and C33A cells. Wound healing and Transwell migration assays further indicated that VTN overexpression markedly promoted the migratory and invasive capabilities of these cells. Moreover, Western blotting analysis revealed that VTN overexpression led to a decrease in ZO-1 and E-cadherin protein levels and an increase in β-catenin and N-cadherin levels, whereas VTN knockdown yielded the opposite effect. These findings suggest that VTN promotes cervical cancer cell malignancy through epithelial-mesenchymal transition (EMT). Conclusion VTN plays a tumor-promoting role in CC by promoting the EMT of cervical cancer cells.
Collapse
Affiliation(s)
- Yao Lin
- Department of Gynecology and Obstetrics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Gynecology and Obstetrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lihong Bian
- Department of Gynecology and Obstetrics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Gynecology and Obstetrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Guangwei Zhu
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Bin Zhang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Gynecology and Obstetrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
3
|
Stinson MW, Laurenson AJ, Rotty JD. Macrophage migration is differentially regulated by distinct ECM components. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.27.538597. [PMID: 37162935 PMCID: PMC10168351 DOI: 10.1101/2023.04.27.538597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Macrophages are indispensable for proper immune surveillance and inflammatory regulation. They also exhibit dramatic phenotypic plasticity and are highly responsive to their local microenvironment, which includes the extracellular matrix (ECM). The present work demonstrates that two fibrous ECM glycoproteins, fibronectin (FN) and laminin (LAM), elicit distinct morphological and migratory responses to macrophages in 2D environments. Laminin 111 inhibits macrophage cell spreading, but drives them to migrate rapidly and less persistently compared to cells on fibronectin. Differential integrin engagement and ROCK/myosin II organization helps explain why macrophages alter their morphology and migration character on these two ECM components. The present study also demonstrates that laminin 111 exerts a suppressive effect toward fibronectin, as macrophages plated on a LAM/FN mixture adopt a morphology and migratory character almost identical to LAM alone. This suggests that distinct responses can be initiated downstream of receptor-ECM engagement, and that one component of the microenvironment may affect the cell's ability to sense another. Overall, macrophages appear intrinsically poised to rapidly switch between distinct migratory modes based on their ECM environments. The role of ECM composition in dictating motile and inflammatory responses in 3D and in vivo contexts warrants further study.
Collapse
|
4
|
Biasella F, Plössl K, Baird PN, Weber BHF. The extracellular microenvironment in immune dysregulation and inflammation in retinal disorders. Front Immunol 2023; 14:1147037. [PMID: 36936905 PMCID: PMC10014728 DOI: 10.3389/fimmu.2023.1147037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) as well as genetically complex retinal phenotypes represent a heterogenous group of ocular diseases, both on account of their phenotypic and genotypic characteristics. Therefore, overlaps in clinical features often complicate or even impede their correct clinical diagnosis. Deciphering the molecular basis of retinal diseases has not only aided in their disease classification but also helped in our understanding of how different molecular pathologies may share common pathomechanisms. In particular, these relate to dysregulation of two key processes that contribute to cellular integrity, namely extracellular matrix (ECM) homeostasis and inflammation. Pathological changes in the ECM of Bruch's membrane have been described in both monogenic IRDs, such as Sorsby fundus dystrophy (SFD) and Doyne honeycomb retinal dystrophy (DHRD), as well as in the genetically complex age-related macular degeneration (AMD) or diabetic retinopathy (DR). Additionally, complement system dysfunction and distorted immune regulation may also represent a common connection between some IRDs and complex retinal degenerations. Through highlighting such overlaps in molecular pathology, this review aims to illuminate how inflammatory processes and ECM homeostasis are linked in the healthy retina and how their interplay may be disturbed in aging as well as in disease.
Collapse
Affiliation(s)
- Fabiola Biasella
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | - Paul N. Baird
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Department of Surgery, Ophthalmology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| | - Bernhard H. F. Weber
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- *Correspondence: Paul N. Baird, ; Bernhard H. F. Weber,
| |
Collapse
|
5
|
Luo P, Li L, Huang J, Mao D, Lou S, Ruan J, Chen J, Tang R, Shi Y, Zhou S, Yang H. The role of SUMOylation in the neurovascular dysfunction after acquired brain injury. Front Pharmacol 2023; 14:1125662. [PMID: 37033632 PMCID: PMC10073463 DOI: 10.3389/fphar.2023.1125662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Acquired brain injury (ABI) is the most common disease of the nervous system, involving complex pathological processes, which often leads to a series of nervous system disorders. The structural destruction and dysfunction of the Neurovascular Unit (NVU) are prominent features of ABI. Therefore, understanding the molecular mechanism underlying NVU destruction and its reconstruction is the key to the treatment of ABI. SUMOylation is a protein post-translational modification (PTM), which can degrade and stabilize the substrate dynamically, thus playing an important role in regulating protein expression and biological signal transduction. Understanding the regulatory mechanism of SUMOylation can clarify the molecular mechanism of the occurrence and development of neurovascular dysfunction after ABI and is expected to provide a theoretical basis for the development of potential treatment strategies. This article reviews the role of SUMOylation in vascular events related to ABI, including NVU dysfunction and vascular remodeling, and puts forward therapeutic prospects.
Collapse
Affiliation(s)
- Pengren Luo
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Li
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiashang Huang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Deqiang Mao
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Silong Lou
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jian Ruan
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Jie Chen
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Ronghua Tang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - You Shi
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Shuai Zhou
- Department of Neurosurgery, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| | - Haifeng Yang
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, China
- *Correspondence: Shuai Zhou, ; Haifeng Yang,
| |
Collapse
|
6
|
Keasey MP, Lovins C, Jia C, Hagg T. Liver vitronectin release into the bloodstream increases due to reduced vagal muscarinic signaling after cerebral stroke in female mice. Physiol Rep 2022; 10:e15301. [PMID: 35531929 PMCID: PMC9082388 DOI: 10.14814/phy2.15301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022] Open
Abstract
Vitronectin (VTN) is a glycoprotein enriched in the blood and activates integrin receptors. VTN blood levels increase only in female mice 24 h after an ischemic stroke and exacerbate brain injury through IL-6-driven inflammation, but the VTN induction mechanism is unknown. Here, a 30 min middle cerebral artery occlusion (MCAO) in female mice induced VTN protein in the liver (normally the main source) in concert with plasma VTN. Male mice were excluded as VTN is not induced after stroke. MCAO also increased plasma VTN levels after de novo expression of VTN in the liver of VTN-/- female mice, using a hepatocyte-specific (SERPINA1) promoter. MCAO did not affect SERPINA1 or VTN mRNA in the liver, brain, or several peripheral organs, or platelet VTN, compared to sham mice. Thus, hepatocytes are the source of stroke-induced increases in plasma VTN, which is independent of transcription. The cholinergic innervation by the parasympathetic vagus nerve is a potential source of brain-liver signaling after stroke. Right-sided vagotomy at the cervical level led to increased plasma VTN levels, suggesting that VTN release is inhibited by vagal tone. Co-culture of hepatocytes with cholinergic neurons or treatment with acetylcholine, but not noradrenaline (sympathetic transmitter), suppressed VTN expression. Hepatocytes have muscarinic receptors and the M1/M3 agonist bethanechol decreased VTN mRNA and protein release in vitro via M1 receptors. Finally, systemic bethanechol treatment blocked stroke-induced plasma VTN. Thus, VTN translation and release are inhibited by muscarinic signaling from the vagus nerve and presents a novel target for lessening detrimental VTN expression.
Collapse
Affiliation(s)
- Matthew P. Keasey
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Chiharu Lovins
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Cuihong Jia
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| | - Theo Hagg
- Department of Biomedical SciencesQuillen College of MedicineEast Tennessee State UniversityJohnson CityTennesseeUnited States
| |
Collapse
|
7
|
Alfano D, Franco P, Stoppelli MP. Modulation of Cellular Function by the Urokinase Receptor Signalling: A Mechanistic View. Front Cell Dev Biol 2022; 10:818616. [PMID: 35493073 PMCID: PMC9045800 DOI: 10.3389/fcell.2022.818616] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
Abstract
Urokinase-type plasminogen activator receptor (uPAR or CD87) is a glycosyl-phosphatidyl-inositol anchored (GPI) membrane protein. The uPAR primary ligand is the serine protease urokinase (uPA), converting plasminogen into plasmin, a broad spectrum protease, active on most extracellular matrix components. Besides uPA, the uPAR binds specifically also to the matrix protein vitronectin and, therefore, is regarded also as an adhesion receptor. Complex formation of the uPAR with diverse transmembrane proteins, including integrins, formyl peptide receptors, G protein-coupled receptors and epidermal growth factor receptor results in intracellular signalling. Thus, the uPAR is a multifunctional receptor coordinating surface-associated pericellular proteolysis and signal transduction, thereby affecting physiological and pathological mechanisms. The uPAR-initiated signalling leads to remarkable cellular effects, that include increased cell migration, adhesion, survival, proliferation and invasion. Although this is beyond the scope of this review, the uPA/uPAR system is of great interest to cancer research, as it is associated to aggressive cancers and poor patient survival. Increasing evidence links the uPA/uPAR axis to epithelial to mesenchymal transition, a highly dynamic process, by which epithelial cells can convert into a mesenchymal phenotype. Furthermore, many reports indicate that the uPAR is involved in the maintenance of the stem-like phenotype and in the differentiation process of different cell types. Moreover, the levels of anchor-less, soluble form of uPAR, respond to a variety of inflammatory stimuli, including tumorigenesis and viral infections. Finally, the role of uPAR in virus infection has received increasing attention, in view of the Covid-19 pandemics and new information is becoming available. In this review, we provide a mechanistic perspective, via the detailed examination of consolidated and recent studies on the cellular responses to the multiple uPAR activities.
Collapse
|
8
|
Keasey MP, Razskazovskiy V, Jia C, Peterknecht ED, Bradshaw PC, Hagg T. PDIA3 inhibits mitochondrial respiratory function in brain endothelial cells and C. elegans through STAT3 signaling and decreases survival after OGD. Cell Commun Signal 2021; 19:119. [PMID: 34922569 PMCID: PMC8684072 DOI: 10.1186/s12964-021-00794-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Protein disulfide isomerase A3 (PDIA3, also named GRP58, ER-60, ERp57) is conserved across species and mediates protein folding in the endoplasmic reticulum. PDIA3 is, reportedly, a chaperone for STAT3. However, the role of PDIA3 in regulating mitochondrial bioenergetics and STAT3 phosphorylation at serine 727 (S727) has not been described. METHODS Mitochondrial respiration was compared in immortalized human cerebral microvascular cells (CMEC) wild type or null for PDIA3 and in whole organism C. Elegans WT or null for pdi-3 (worm homologue). Mitochondrial morphology and cell signaling pathways in PDIA3-/- and WT cells were assessed. PDIA3-/- cells were subjected to oxygen-glucose deprivation (OGD) to determine the effects of PDIA3 on cell survival after injury. RESULTS We show that PDIA3 gene deletion using CRISPR-Cas9 in cultured CMECs leads to an increase in mitochondrial bioenergetic function. In C. elegans, gene deletion or RNAi knockdown of pdi-3 also increased respiratory rates, confirming a conserved role for this gene in regulating mitochondrial bioenergetics. The PDIA3-/- bioenergetic phenotype was reversed by overexpression of WT PDIA3 in cultured PDIA3-/- CMECs. PDIA3-/- and siRNA knockdown caused an increase in phosphorylation of the S727 residue of STAT3, which is known to promote mitochondrial bioenergetic function. Increased respiration in PDIA3-/- CMECs was reversed by a STAT3 inhibitor. In PDIA3-/- CMECs, mitochondrial membrane potential and reactive oxygen species production, but not mitochondrial mass, was increased, suggesting an increased mitochondrial bioenergetic capacity. Finally, PDIA3-/- CMECs were more resistant to oxygen-glucose deprivation, while STAT3 inhibition reduced the protective effect. CONCLUSIONS We have discovered a novel role for PDIA3 in suppressing mitochondrial bioenergetic function by inhibiting STAT3 S727 phosphorylation.
Collapse
Affiliation(s)
- Matt. P. Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson City, TN 37614 USA
| | - V. Razskazovskiy
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson City, TN 37614 USA
| | - C. Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson City, TN 37614 USA
- Sandwell and West, Birmingham Hospitals NHS Trust, Birmingham, UK
| | | | - P. C. Bradshaw
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson City, TN 37614 USA
| | - T. Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, PO Box 70582, Johnson City, TN 37614 USA
| |
Collapse
|
9
|
Zhu XY, Chen YH, Zhang T, Liu SJ, Bai XY, Huang XY, Jiang M, Sun XD. Improvement of human embryonic stem cell-derived retinal pigment epithelium cell adhesion, maturation, and function through coating with truncated recombinant human vitronectin. Int J Ophthalmol 2021; 14:1160-1167. [PMID: 34414078 DOI: 10.18240/ijo.2021.08.04] [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: 01/06/2021] [Accepted: 04/21/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To explore an xeno-free and defined coating substrate suitable for the culture of H9 human embryonic stem cell-derived retinal pigment epithelial (hES-RPE) cells in vitro, and compare the behaviors and functions of hES-RPE cells on two culture substrates, laminin521 (LN-521) and truncated recombinant human vitronectin (VTN-N). METHODS hES-RPE cells were used in the experiment. The abilities of LN-521 and VTN-N at different concentrations to adhere to hES-RPE cells were compared with a high-content imaging system. Quantitative real-time polymerase chain reaction was used to evaluate RPE-specific gene expression levels midway (day 10) and at the end (day 20) of the time course. Cell polarity was observed by immunofluorescent staining for apical and basal markers of the RPE. The phagocytic ability of hES-RPE cells was identified by flow cytometry and immunofluorescence. RESULTS The cell adhesion assay showed that the ability of LN-521 to adhere to hES-RPE cells was dose-dependent. With increasing coating concentration, an increasing number of cells attached to the surface of LN-521-coated wells. In contrast, VTN-N presented a strong adhesive ability even at a low concentration. The optimal concentration of LN-521 and VTN-N required to coat and adhesion to hES-RPE cells were 2 and 0.25 µg/cm2, respectively. Furthermore, both LN-521 and VTN-N could facilitate adoption of the desired cobblestone cellular morphology with tight junction and showed polarity by the hES-RPE cells. However, hES-RPE cells cultivated in VTN-N had a greater phagocytic ability, and it took less time for these hES-RPE cells to mature. CONCLUSION VTN-N is a more suitable coating substrate for cultivating hES-RPE cells.
Collapse
Affiliation(s)
- Xin-Yue Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Yu-Hong Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Ting Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Su-Jun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xin-Yue Bai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xian-Yu Huang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Mei Jiang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| | - Xiao-Dong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.,National Clinical Research Center for Eye Diseases, Shanghai 200080, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
| |
Collapse
|
10
|
Focal Adhesion Kinase Inhibitor Inhibits the Oxidative Damage Induced by Central Venous Catheter via Abolishing Focal Adhesion Kinase-Protein Kinase B Pathway Activation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6685493. [PMID: 33748278 PMCID: PMC7943296 DOI: 10.1155/2021/6685493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 11/24/2022]
Abstract
The vascular injury induced by central venous catheter (CVC) indwelling is the basis for the occurrence and development of CVC-related complications, such as phlebitis, venous thrombosis, and catheter-related infections. Focal adhesion kinase (FAK) and FAK-protein kinase B (AKT) signaling pathway are of great significance in tissue repair after trauma. Here, we investigated the role and mechanism of the FAK inhibitor (1,2,4,5-phenyltetramine tetrahydrochloride (Y15)) in oxidative damage caused by CVC. EA.hy926 cells were divided into the control group (normal control), CVCs+scratches group (the intercepted CVC segments coculturing with scratched EA.hy926 cells), and CVCs+scratches+Y15 group (Y15 was added to the cell culture supernatant with CVCs + scratches at a final concentration of 50 μmol·L−1). New Zealand rabbits were randomly divided into the control group (normal control), CVC group (CVC was inserted through the rabbit's right jugular vein to the junction of the right atrium and superior vena cava), and CVC+Y15 group (CVC was immersed in a 50 μmol·L−1 Y15 solutions before insertion). The levels of markers and proteins related to oxidative damage in cells, cell culture supernatant, serum, and external jugular vein were measured by commercial kits and western blot, respectively. We found that Y15 treatment significantly decreased ROS and MDA levels and increased cell viability, NO, and SOD levels in a time-dependent manner in rabbit serum and cell culture supernatant. In addition, Y15 effectively reduced the CVC-induced pathological changes of damaged vascular tissues. Y15 also downregulated the levels of p-FAK Tyr 397 and p-Akt Ser 473 in damaged external jugular vein and EA.hy926 cells. These findings suggest that Y15 alleviated CVC-induced oxidative damage to blood vessels by suppressing focal FAK-Akt pathway activation.
Collapse
|
11
|
Huang T, Jones CG, Chung JH, Chen C. Microfibrous Extracellular Matrix Changes the Liver Hepatocyte Energy Metabolism via Integrins. ACS Biomater Sci Eng 2020; 6:5849-5856. [PMID: 33320566 DOI: 10.1021/acsbiomaterials.0c01311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell line-based liver models are critical tools for liver-related studies. However, the conventional monolayer culture of hepatocytes, the most widely used in vitro model, does not have the extracellular matrix (ECM), which contributes to the three-dimensional (3D) arrangement of the hepatocytes in the liver. As a result, the metabolic properties of the hepatocytes in the monolayer tissue culture may not accurately reflect those of the hepatocytes in the liver. Here, we developed a modular platform for 3D hepatocyte cultures on fibrous ECMs produced by electrospinning, a technique that can turn a polymer solution to the micro/nanofibers and has been widely used to produce scaffolds for 3D cell cultures. Metabolomics quantitation by liquid chromatography-mass spectrometry (LC-MS) indicated that Huh7 hepatocytes grown in microfibers electrospun from silk fibroin exhibited reduced glycolysis and tricarboxylic acid (TCA) cycle, as compared to the cells cultured as a monolayer. Further mechanistic studies suggested that integrins were correlated to the ECM's effects. This is the first time to report how an ECM scaffold could affect the fundamental metabolism of the hepatocytes via integrins.
Collapse
Affiliation(s)
- Tianjiao Huang
- Laboratory of Obesity and Aging Research, Cardiovascular Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Curtis G Jones
- The Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Jay H Chung
- Laboratory of Obesity and Aging Research, Cardiovascular Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Chengpeng Chen
- The Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
12
|
Lee J, Park A, Mun S, Kim HJ, Son H, Choi H, Kim D, Lee SJ, Kim JG, Kang HG. Proteomics-Based Identification of Diagnostic Biomarkers Related to Risk Factors and Pathogenesis of Ischemic Stroke. Diagnostics (Basel) 2020; 10:diagnostics10050340. [PMID: 32466277 PMCID: PMC7278009 DOI: 10.3390/diagnostics10050340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is caused by blood clot formation and consequent vessel blockage. Proteomic approaches provide a cost-effective alternative to current diagnostic methods, including computerized tomography (CT) scans and magnetic resonance imaging (MRI). To identify diagnostic biomarkers associated with ischemic stroke risk factors, we performed individual proteomic analysis of serum taken from 20 healthy controls and 20 ischemic stroke patients. We then performed SWATH analysis, a data-independent method, to assess quantitative changes in protein expression between the two experimental conditions. Our analysis identified several candidate protein biomarkers, 11 of which were validated by multiple reaction monitoring (MRM) analysis as novel diagnostic biomarkers associated with ischemic stroke risk factors. Our study identifies new biomarkers associated with the risk factors and pathogenesis of ischemic stroke which, to the best of our knowledge, were previously unknown. These markers may be effective in not only the diagnosis but also the prevention and management of ischemic stroke.
Collapse
Affiliation(s)
- Jiyeong Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Korea;
| | - Arum Park
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Sora Mun
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyo-Jin Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyunsong Son
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyebin Choi
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Doojin Kim
- Department of Laboratory Medicine, Seongnam Central Hospital, Seongnam 13161, Korea;
| | - Soo Joo Lee
- Department of Neurology, Eulji University Hospital, School of Medicine, Eulji University, Daejeon 35233, Korea; (S.J.L.); (J.G.K.)
| | - Jae Guk Kim
- Department of Neurology, Eulji University Hospital, School of Medicine, Eulji University, Daejeon 35233, Korea; (S.J.L.); (J.G.K.)
| | - Hee-Gyoo Kang
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Seongnam 13135, Korea
- Seongnam Senior Industry Innovation Center, Eulji University, Seongnam 13503, Korea
- Correspondence: ; Tel.: +82-31-740-7315; Fax: +82-31-740-7448
| |
Collapse
|
13
|
Bachmann M, Schäfer M, Mykuliak VV, Ripamonti M, Heiser L, Weißenbruch K, Krübel S, Franz CM, Hytönen VP, Wehrle-Haller B, Bastmeyer M. Induction of ligand promiscuity of αVβ3 integrin by mechanical force. J Cell Sci 2020; 133:jcs242404. [PMID: 32193334 DOI: 10.1242/jcs.242404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/12/2020] [Indexed: 12/20/2022] Open
Abstract
αVβ3 integrin can bind to multiple extracellular matrix proteins, including vitronectin (Vn) and fibronectin (Fn), which are often presented to cells in culture as homogenous substrates. However, in tissues, cells experience highly complex and changing environments. To better understand integrin ligand selection in such complex environments, we employed binary-choice substrates of Fn and Vn to dissect αVβ3 integrin-mediated binding to different ligands on the subcellular scale. Super-resolution imaging revealed that αVβ3 integrin preferred binding to Vn under various conditions. In contrast, binding to Fn required higher mechanical load on αVβ3 integrin. Integrin mutations, structural analysis and chemical inhibition experiments indicated that the degree of hybrid domain swing-out is relevant for the selection between Fn and Vn; only a force-mediated, full hybrid domain swing-out facilitated αVβ3-Fn binding. Thus, force-dependent conformational changes in αVβ3 integrin increased the diversity of available ligands for binding and therefore enhanced the ligand promiscuity of this integrin.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Michael Bachmann
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Markus Schäfer
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen 76344, Germany
| | - Vasyl V Mykuliak
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, and Fimlab Laboratories, Tampere 33014, Finland
| | - Marta Ripamonti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Lia Heiser
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
| | - Kai Weißenbruch
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
| | - Sarah Krübel
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
| | - Clemens M Franz
- DFG-Center for Functional Nanostructures, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1192, Japan
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, and Fimlab Laboratories, Tampere 33014, Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Martin Bastmeyer
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen 76344, Germany
| |
Collapse
|
14
|
Jia C, Malone HM, Keasey MP, Lovins C, Elam J, Hagg T. Blood Vitronectin Induces Detrimental Brain Interleukin-6 and Correlates With Outcomes After Stroke Only in Female Mice. Stroke 2020; 51:1587-1595. [PMID: 32312218 DOI: 10.1161/strokeaha.120.029036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background and Purpose- Women have worse stroke outcomes than men, especially after menopause. Few studies have focused on female-specific mechanisms, other than hormones. We investigated the role of the blood protein VTN (vitronectin) after ischemic stroke in mice. Methods- Adult male and female VTN knockout and wild-type littermates and C57BL/6 mice received a middle cerebral artery occlusion and the injured brain tissue analyzed 24 hours to 3 weeks later for cell loss and inflammation, as well as neurological function. Blood VTN levels were measured before and after stroke. Results- Intravenously injected VTN leaked extensively from bloodstream into brain infarct and penumbra by 24 hours after stroke. Strikingly, VTN was detrimental in female, but not male, mice, as shown by reduced brain injury (26.2±2.6% versus 13.4±3.8%; P=0.018; n=6 and 5) and forelimb dysfunction in female VTN knockout mice. Stroke increased plasma VTN 2- to 8-fold at 24 hours in females (36±4 versus 145±24 μg/mL; P<0.0001; n=10 and 7), but not males (62±8 versus 68±6; P>0.99; n=10 and 7), and returned to control levels by 7 days. Individually variable VTN levels at 24 hours correlated with stroke-induced brain injury at 7 days only in females. VTN promoted stroke-induced microglia/macrophage activation and leukocyte infiltration in females. Proinflammatory IL (interleukin)-6 greatly increased in the striatum at 24 hours in wild-type mice but was increased ≈60% less in female (739±159 versus 268±111; P=0.02; n=7 and 6), but not male (889±178 versus 1179±295; P=0.73; n=10 and 11), knockout mice. In individual wild-type females, plasma VTN levels correlated with striatal IL-6 expression at 24 hours. The female-specific effect of VTN-induced IL-6 expression following stroke was not due to gonadal hormones, as shown by ovariectomy and castration. Lastly, intrastriatal injection of IL-6 in female mice immediately before stroke reversed the VTN knockout phenotypes of reduced brain injury and microglia/macrophage activation. Conclusions- VTN plays a novel sexually dimorphic detrimental pathophysiological role in females and might ultimately be a therapeutic target to improve stroke outcomes in women.
Collapse
Affiliation(s)
- Cuihong Jia
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Hannah M Malone
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Matthew P Keasey
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Chiharu Lovins
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Jacob Elam
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Theo Hagg
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| |
Collapse
|
15
|
Ai LQY, Yuan RD, Chen X, Liu YJ, Liu WY, Zhu JY, Zhang Z, Yan J, Chen CL, Lin S, Ye J. Retinal blood vessel-origin yes-associated protein (YAP) governs astrocytic maturation via leukaemia inhibitory factor (LIF). Cell Prolif 2020; 53:e12757. [PMID: 31916327 PMCID: PMC7046482 DOI: 10.1111/cpr.12757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/26/2019] [Accepted: 12/16/2019] [Indexed: 12/31/2022] Open
Abstract
Objectives To testify that endothelial cells (ECs) induce astrocyte maturation by leukaemia inhibitory factor (LIF) secretion. Materials and Methods In vivo experiments, mice bearing floxed alleles of YAP were crossed with mice expressing a Cre recombinase driven by the endothelial Tek promoter (Tek‐Cre) to finally obtain the following three genotypes: YAPf/f, Tek‐Cre; YAPf/w, Tek‐Cre; and YAPf/f. Retinal vascularization and astrocyte network were evaluated by whole‐mount fluorescence and Western blotting. In vitro, experiments were performed in an astrocyte and human microvascular endothelial cell (HMEC‐1) coculture model to analyse the mechanisms underlying the effect of endothelial YAP on astrocytes. Results In vivo, YAPf/f;Tek‐Cre mice showed delayed angiogenesis, sparse vessels and decreased glial fibrillary acidic protein (GFAP)+ astrocytes but aberrant growth of endothelial networks and immature astrocytes (platelet‐derived growth factor A, PDGFRA+ astrocytes) overgrowth. In vitro, Yap deletion attenuated the LIF release that delayed the maturation of retinal astrocyte which was consistent with the results of HMEC‐1—astrocyte coculture. The effect of YAP overexpression on LIF‐LIFR axis in HMEC‐1 interferes the GFAP expression of astrocyte. In contrast, LIF protein rescues the astrocytic GFAP expression when EC YAP was inhibited by siRNAs. Conclusions We show that EC yes‐associated protein (YAP) is not only a critical coactivator of Hippo signalling in retinal vessel development but also plays an essential role in retinal astrocyte maturation by regulating LIF production.
Collapse
Affiliation(s)
- Li-Qian-Yu Ai
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Rong-Di Yuan
- Department of Ophthalmology, XinQiao Hospital, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yun-Jia Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Wen-Yi Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jing-Yi Zhu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Zhou Zhang
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jun Yan
- Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chun-Lin Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
16
|
Vitronectin mitigates stroke-increased neurogenesis only in female mice and through FAK-regulated IL-6. Exp Neurol 2019; 323:113088. [PMID: 31678139 DOI: 10.1016/j.expneurol.2019.113088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/03/2019] [Accepted: 10/20/2019] [Indexed: 12/18/2022]
Abstract
Vitronectin (VTN) is a blood protein produced mainly by the liver. We show that VTN leaks from the bloodstream into the injury site and neighboring subventricular zone (SVZ) following ischemic stroke (middle cerebral artery occlusion, MCAO) in adult mice. MCAO is known to increase neurogenesis after stroke. VTN inhibits this response in females, but not in males, as shown by ~70% more stroke-induced SVZ neurogenesis in female VTN-/- mice at 14 d. In female VTN-/- mice, stroke-induced expression of interleukin-6 (IL-6) at 24 h was reduced in the SVZ. The closely related leukemia inhibitory factor (LIF) or pro-neurogenic ciliary neurotrophic factor (CNTF) were not affected. The female-specific effect of VTN on IL-6 expression was not due to sex hormones, as shown by ovariectomy and castration. IL-6 injection next to the SVZ reversed the MCAO-induced increase in neurogenesis seen in VTN-/- mice. Our in vitro and vivo data suggest that plasma VTN activates focal adhesion kinase (FAK) in the SVZ following MCAO, which reduces IL-6 expression in astrocytes but increases it in other cells such as microglia/macrophages. Inducible conditional astrocytic FAK deletion increased MCAO-induced IL-6 expression in females at 24 h and blocked MCAO-induced neurogenesis at 14 d, confirming a key detrimental role of IL-6. Collectively, these data suggest that leakage of VTN into the SVZ reduces the neurogenic response to stroke in female mice by promoting IL-6 expression. Reducing VTN or VTN signaling may be an approach to promote neurogenesis for neuroprotection and cell replacement after stroke in females.
Collapse
|
17
|
The uPAR System as a Potential Therapeutic Target in the Diseased Eye. Cells 2019; 8:cells8080925. [PMID: 31426601 PMCID: PMC6721659 DOI: 10.3390/cells8080925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of vascular networks is characteristic of eye diseases associated with retinal cell degeneration and visual loss. Visual impairment is also the consequence of photoreceptor degeneration in inherited eye diseases with a major inflammatory component, but without angiogenic profile. Among the pathways with high impact on vascular/degenerative diseases of the eye, a central role is played by a system formed by the ligand urokinase-type plasminogen activator (uPA) and its receptor uPAR. The uPAR system, although extensively investigated in tumors, still remains a key issue in vascular diseases of the eye and even less studied in inherited retinal pathologies such as retinitis pigmantosa (RP). Its spectrum of action has been extended far beyond a classical pro-angiogenic function and has emerged as a central actor in inflammation. Preclinical studies in more prevalent eye diseases characterized by neovascular formation, as in retinopathy of prematurity, wet macular degeneration and rubeosis iridis or vasopermeability excess as in diabetic retinopathy, suggest a critical role of increased uPAR signaling indicating the potentiality of its modulation to counteract neovessel formation and microvascular dysfunction. The additional observation that the uPAR system plays a major role in RP by limiting the inflammatory cascade triggered by rod degeneration rises further questions about its role in the diseased eye.
Collapse
|
18
|
Cammalleri M, Dal Monte M, Locri F, Pecci V, De Rosa M, Pavone V, Bagnoli P. The urokinase-type plasminogen activator system as drug target in retinitis pigmentosa: New pre-clinical evidence in the rd10 mouse model. J Cell Mol Med 2019; 23:5176-5192. [PMID: 31251468 PMCID: PMC6653070 DOI: 10.1111/jcmm.14391] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/05/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Retinitis pigmentosa (RP) is characterized by progressive loss of vision due to photoreceptor degeneration leading to secondary inflammation. The urokinase-type plasminogen activator (uPA) system contributes to retinal inflammation, but its role in RP is unknown. In the rd10 mouse model of RP, we addressed this question with the use of the peptide UPARANT designed to interact with the uPA system. UPARANT was systemically administered from post-natal day (PD) 10 to PD30 when its efficacy in RP rescue was investigated using electroretinographic recordings, Western blot and immunocytochemistry. Temporal profile of protein expression in the uPA system was also investigated. UPARANT reduced both Müller cell gliosis and up-regulated levels of inflammatory markers and exerted major anti-apoptotic effects without influencing the autophagy cascade. Rescue from retinal cell degeneration was accompanied by improved retinal function. No scotopic phototransduction was rescued in the UPARANT-treated animals as determined by the kinetic analysis of rod-mediated a-waves and confirmed by rod photoreceptor markers. In contrast, the cone photopic b-wave was recovered and its rescue was confirmed in the whole mounts using cone arrestin antibody. Investigation of the uPA system regulation over RP progression revealed extremely low levels of uPA and its receptor uPAR both of which were recovered by HIF-1α stabilization indicating that HIF-1 regulates the expression of the uPA/uPAR gene in the retina. Ameliorative effects of UPARANT were likely to occur through an inhibitory action on up-regulated activity of the αvβ3 integrin/Rac1 pathway that was suggested as a novel target for the development of therapeutic approaches against RP.
Collapse
Affiliation(s)
| | | | - Filippo Locri
- Department of Biology, University of Pisa, Pisa, Italy
| | - Valeria Pecci
- Department of Biology, University of Pisa, Pisa, Italy
| | - Mario De Rosa
- Department of Experimental Medicine, Second University of Napoli, Napoli, Italy
| | - Vincenzo Pavone
- Department of Chemical Sciences, University of Napoli Federico II, Napoli, Italy
| | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Jia C, Keasey MP, Lovins C, Hagg T. Inhibition of astrocyte FAK-JNK signaling promotes subventricular zone neurogenesis through CNTF. Glia 2019; 66:2456-2469. [PMID: 30500112 DOI: 10.1002/glia.23498] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
Abstract
Astrocyte-derived ciliary neurotrophic factor (CNTF) promotes adult subventricular zone (SVZ) neurogenesis. We found that focal adhesion kinase (FAK) and JNK, but not ERK or P38, repress CNTF in vitro. Here, we defined the FAK-JNK pathway and its regulation of CNTF in mice, and the related leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), which promote stem cell renewal at the expense of neurogenesis. Intrastriatal injection of FAK inhibitor, FAK14, in adult male C57BL/6 mice reduced pJNK and increased CNTF expression in the SVZ-containing periventricular region. Injection of a JNK inhibitor increased CNTF without affecting LIF and IL-6, and increased SVZ proliferation and neuroblast formation. The JNK inhibitor had no effect in CNTF-/- mice, suggesting that JNK inhibits SVZ neurogenesis by repressing CNTF. Inducible deletion of FAK in astrocytes increased SVZ CNTF and neurogenesis, but not LIF and IL-6. Intrastriatal injection of inhibitors suggested that P38 reduces LIF and IL-6 expression, whereas ERK induces CNTF and LIF. Intrastriatal FAK inhibition increased LIF, possibly through ERK, and IL-6 through another pathway that does not involve P38. Systemic injection of FAK14 also inhibited JNK while increasing CNTF, but did not affect P38 and ERK activation, or LIF and IL-6 expression. Importantly, systemic FAK14 increased SVZ neurogenesis in wild-type C57BL/6 and CNTF+/+ mice, but not in CNTF-/- littermates, indicating that it acts by upregulating CNTF. These data show a surprising differential regulation of related cytokines and identify the FAK-JNK-CNTF pathway as a specific target in astrocytes to promote neurogenesis and possibly neuroprotection in neurological disorders.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
20
|
Kugaevskaya EV, Gureeva TA, Timoshenko OS, Solovyeva NI. Urokinase-Type Plasminogen Activator System in Norm and in Life-Threatening Processes (Review). ACTA ACUST UNITED AC 2018. [DOI: 10.15360/1813-9779-2018-6-61-79] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The multifunctional urokinase-type plasminogen activator system (uPA-system) includes serine proteinase — uPA or urokinase, its receptor (uPAR) and two inhibitors (PAI-1 and PAI-2). The review discusses the structural features and involvement of the system components in the development of life-threatening processes including carcinogenesis, inflammation, neurogenesis and fibrinolysis, in regulation of which the destruction of extracellular matrix (ECM), cell mobility and signaling inside and outside the cell play a decisive role. uPA triggers the processes by activating the plasminogen and its convertion into plasmin involved in the activation of matrix metalloproteinases (MMPs) in addition to the regulation of fibrinolysis. MMPs can hydrolyze all the major ECM components and therefore play a key role in invasion, metastasis, and cell mobility. MMPs activates a cassette of biologically active regulatory molecules and release them from ECM. uPAR, PAI-1 and PAI-2 are responsible for regulation of the uPA activity. In addition, being a signaling receptor, uPAR along with MMPs lead to the stimulation of a number of signaling pathways that are associated with the regulation of proliferation, apoptosis, adhesion, growth and migration of cells contributing to tumor progression, inflammation, chemotaxis, and angiogenesis. Effective participation of the uPA system components in ECM destruction and regulation of intracellular and extracellular signaling pathways demonstrates that the system significantly contributes to the regulation of various physiological and pathological processes.
Collapse
|
21
|
Plasma exosomes stimulate breast cancer metastasis through surface interactions and activation of FAK signaling. Breast Cancer Res Treat 2018; 174:129-141. [PMID: 30484103 DOI: 10.1007/s10549-018-5043-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE The interaction between malignant cells and surrounding healthy tissues is a critical factor in the metastatic progression of breast cancer (BC). Extracellular vesicles, especially exosomes, are known to be involved in inter-cellular communication during cancer progression. In the study presented herein, we aimed to evaluate the role of circulating plasma exosomes in the metastatic dissemination of BC and to investigate the underlying molecular mechanisms of this phenomenon. METHODS Exosomes isolated from plasma of healthy female donors were applied in various concentrations into the medium of MDA-MB-231 and MCF-7 cell lines. Motility and invasive properties of BC cells were examined by random migration and Transwell invasion assays, and the effect of plasma exosomes on the metastatic dissemination of BC cells was demonstrated in an in vivo zebrafish model. To reveal the molecular mechanism of interaction between plasma exosomes and BC cells, a comparison between un-treated and enzymatically modified exosomes was performed, followed by mass spectrometry, gene ontology, and pathway analysis. RESULTS Plasma exosomes stimulated the adhesive properties, two-dimensional random migration, and transwell invasion of BC cells in vitro as well as their in vivo metastatic dissemination in a dose-dependent manner. This stimulatory effect was mediated by interactions of surface exosome proteins with BC cells and consequent activation of focal adhesion kinase (FAK) signaling in the tumor cells. CONCLUSIONS Plasma exosomes have a potency to stimulate the metastasis-promoting properties of BC cells. This pro-metastatic property of normal plasma exosomes may have impact on the course of the disease and on its prognosis.
Collapse
|
22
|
Jia C, Keasey MP, Malone HM, Lovins C, Sante RR, Razskazovskiy V, Hagg T. Vitronectin from brain pericytes promotes adult forebrain neurogenesis by stimulating CNTF. Exp Neurol 2018; 312:20-32. [PMID: 30408465 DOI: 10.1016/j.expneurol.2018.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/17/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022]
Abstract
Vitronectin (VTN) is a glycoprotein in the blood and affects hemostasis. VTN is also present in the extracellular matrix of various organs but little is known about its function in healthy adult tissues. We show, in adult mice, that VTN is uniquely expressed by approximately half of the pericytes of subventricular zone (SVZ) where neurogenesis continues throughout life. Intracerebral VTN antibody injection or VTN knockout reduced neurogenesis as well as expression of pro-neurogenic CNTF, and anti-neurogenic LIF and IL-6. Conversely, injections of VTN, or plasma from VTN+/+, but not VTN-/- mice, increased these cytokines. VTN promoted SVZ neurogenesis when LIF and IL-6 were suppressed by co-administration of a gp130 inhibitor. Unexpectedly, VTN inhibited FAK signaling and VTN-/- mice had increased FAK signaling in the SVZ. Further, an FAK inhibitor or VTN increased CNTF expression, but not in conditional astrocytic FAK knockout mice, suggesting that VTN increases CNTF through FAK inhibition in astrocytes. These results identify a novel role of pericyte-derived VTN in the brain, where it regulates SVZ neurogenesis through co-expression of CNTF, LIF and IL-6. VTN-integrin-FAK and gp130 signaling may provide novel targets to induce neurogenesis for cell replacement therapies.
Collapse
Affiliation(s)
- Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Hannah M Malone
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Richard R Sante
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Vlad Razskazovskiy
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.
| |
Collapse
|