1
|
Farhangniya M, Samadikuchaksaraei A, Mohamadi Farsani F. Exploring Co-expression Modules-Traits Correlation through Weighted Gene Co-expression Network Analysis: A Promising Approach in Wound Healing Research. Med J Islam Repub Iran 2024; 38:82. [PMID: 39678778 PMCID: PMC11644100 DOI: 10.47176/mjiri.38.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Indexed: 12/17/2024] Open
Abstract
Background The skin is the biggest organ in the body and has several important functions in protection and regulation. However, wound development can disrupt the natural healing process, leading to challenges such as chronic wounds, persistent infections, and impaired angiogenesis. These issues not only affect individuals' well-being but also pose significant economic burdens on healthcare systems. Despite advancements in wound care research, managing chronic wounds remains a pressing concern, with obstacles such as persistent infection and impaired angiogenesis hindering the healing process. Understanding the complex genetic pathways involved in wound healing is crucial for developing effective therapeutic strategies and reducing the socio-economic impact of chronic wounds. Weighted Gene Co-Expression Network Analysis (WGCNA) offers a promising approach to uncovering key genes and modules associated with different stages of wound healing, providing valuable insights for targeted interventions to enhance tissue repair and promote efficient wound healing. Methods Data collection involved retrieving microarray gene expression datasets from the Gene Expression Omnibus website, with 65 series selected according to inclusion and exclusion criteria. Preprocessing of raw data was performed using the Robust MultiArray Averaging approach for background correction, normalization, and gene expression calculation. Weighted Gene Co-Expression Network Analysis was employed to identify co-expression patterns among genes associated with wound healing processes. This involved steps such as network construction, topological analysis, module identification, and association with clinical traits. Functional analysis included enrichment analysis and identification of hub genes through gene-gene functional interaction network analysis using the GeneMANIA database. Results The analysis using WGCNA indicated significant correlations between wound healing and the black, brown, and light green modules. These modules were further examined for their relevance to wound healing traits and subjected to functional enrichment analysis. A total of 16 genes were singled out as potential hub genes critical for wound healing. These hub genes were then scrutinized, revealing a gene-gene functional interaction network within the module network based on the KEGG enrichment database. Noteworthy pathways such as MAPK, EGFR, and ErbB signaling pathways, as well as essential cellular processes including autophagy and mitophagy, emerged as the most notable significant pathways. Conclusion We identified consensus modules relating to wound healing across nine microarray datasets. Among these, 16 hub genes were uncovered within the brown and black modules. KEGG enrichment analysis identified co-expression genes within these modules and highlighted pathways most closely associated with the development of wound healing traits, including autophagy and mitophagy. The hub genes identified in this study represent potential candidates for future research endeavors. These findings serve as a stepping stone toward further exploration of the implications of these co-expressed modules on wound healing traits.
Collapse
Affiliation(s)
- Mansoureh Farhangniya
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Health Metrics Research Center, Iranian Institute for Health Sciences Research, ACECR, Tehran, Iran
| | - Ali Samadikuchaksaraei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
Cui W, Chen S, Hu T, Zhou T, Qiu C, Jiang L, Cheng X, Ji J, Yao K, Han H. Nanoceria-Mediated Cyclosporin A Delivery for Dry Eye Disease Management through Modulating Immune-Epithelial Crosstalk. ACS NANO 2024; 18:11084-11102. [PMID: 38632691 DOI: 10.1021/acsnano.3c11514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Dry eye disease (DED) affects a substantial worldwide population with increasing frequency. Current single-targeting DED management is severely hindered by the existence of an oxidative stress-inflammation vicious cycle and complicated intercellular crosstalk within the ocular microenvironment. Here, a nanozyme-based eye drop, namely nanoceria loading cyclosporin A (Cs@P/CeO2), is developed, which possesses long-term antioxidative and anti-inflammatory capacities due to its regenerative antioxidative activity and sustained release of cyclosporin A (CsA). In vitro studies showed that the dual-functional Cs@P/CeO2 not only inhibits cellular reactive oxygen species production, sequentially maintaining mitochondrial integrity, but also downregulates inflammatory processes and repolarizes macrophages. Moreover, using flow cytometric and single-cell sequencing data, the in vivo therapeutic effect of Cs@P/CeO2 was systemically demonstrated, which rebalances the immune-epithelial communication in the corneal microenvironment with less inflammatory macrophage polarization, restrained oxidative stress, and enhanced epithelium regeneration. Collectively, our data proved that the antioxidative and anti-inflammatory Cs@P/CeO2 may provide therapeutic insights into DED management.
Collapse
Affiliation(s)
- Wenyu Cui
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Sheng Chen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, P. R. China
| | - Tianyi Hu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P. R. China
| | - Tinglian Zhou
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Chen Qiu
- MOE Laboratory of Biosystems Homeostasis & Protection and iCell Biotechnology Regenerative Biomedicine Laboratory of College of Life Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Luyang Jiang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Xiaoyu Cheng
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Haijie Han
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| |
Collapse
|
3
|
Vittum Z, Cocchiaro S, Mensah SA. Basal endothelial glycocalyx's response to shear stress: a review of structure, function, and clinical implications. Front Cell Dev Biol 2024; 12:1371769. [PMID: 38562144 PMCID: PMC10982814 DOI: 10.3389/fcell.2024.1371769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The endothelial glycocalyx encompasses the entire endothelial cell, transducing extracellular signals and regulating vascular permeability and barrier functions. The apical glycocalyx, which forms the lumen of the vessel, and the basal glycocalyx, at the smooth muscle cell interface, are often investigated separately as they are exposed to vastly different stimuli. The apical glycocalyx directly senses fluid shear forces transmitting them intracellularly through connection to the cytoskeleton of the endothelial cell. The basal glycocalyx has demonstrated sensitivity to shear due to blood flow transmitted through the cytoskeleton, promoting alternate signaling processes. In this review, we discuss current literature on the basal glycocalyx's response to shear stress in the context of mechanotransduction and remodeling. The possible implications of basal glycocalyx degradation in pathologies are also explored. Finally, this review seeks to highlight how addressing the gaps discussed would improve our wholistic understanding of the endothelial glycocalyx and its role in maintaining vascular homeostasis.
Collapse
Affiliation(s)
- Zoe Vittum
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Samantha Cocchiaro
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Solomon A. Mensah
- Biomedical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
- Mechanical Engineering Department, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
4
|
Maldonado H, Savage BD, Barker HR, May U, Vähätupa M, Badiani RK, Wolanska KI, Turner CMJ, Pemmari T, Ketomäki T, Prince S, Humphries MJ, Ruoslahti E, Morgan MR, Järvinen TAH. Systemically administered wound-homing peptide accelerates wound healing by modulating syndecan-4 function. Nat Commun 2023; 14:8069. [PMID: 38057316 PMCID: PMC10700342 DOI: 10.1038/s41467-023-43848-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
CAR (CARSKNKDC) is a wound-homing peptide that recognises angiogenic neovessels. Here we discover that systemically administered CAR peptide has inherent ability to promote wound healing: wounds close and re-epithelialise faster in CAR-treated male mice. CAR promotes keratinocyte migration in vitro. The heparan sulfate proteoglycan syndecan-4 regulates cell migration and is crucial for wound healing. We report that syndecan-4 expression is restricted to epidermis and blood vessels in mice skin wounds. Syndecan-4 regulates binding and internalisation of CAR peptide and CAR-mediated cytoskeletal remodelling. CAR induces syndecan-4-dependent activation of the small GTPase ARF6, via the guanine nucleotide exchange factor cytohesin-2, and promotes syndecan-4-, ARF6- and Cytohesin-2-mediated keratinocyte migration. Finally, we show that genetic ablation of syndecan-4 in male mice eliminates CAR-induced wound re-epithelialisation following systemic administration. We propose that CAR peptide activates syndecan-4 functions to selectively promote re-epithelialisation. Thus, CAR peptide provides a therapeutic approach to enhance wound healing in mice; systemic, yet target organ- and cell-specific.
Collapse
Affiliation(s)
- Horacio Maldonado
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Bryan D Savage
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Harlan R Barker
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Ulrike May
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Rahul K Badiani
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Katarzyna I Wolanska
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Craig M J Turner
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK
| | - Toini Pemmari
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Tuomo Ketomäki
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Stuart Prince
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Erkki Ruoslahti
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA and Center for Nanomedicine, University of California (UCSB), Santa Barbara, CA, USA
| | - Mark R Morgan
- Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK.
| | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University & Tampere University Hospital, Tampere, Finland.
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA and Center for Nanomedicine, University of California (UCSB), Santa Barbara, CA, USA.
| |
Collapse
|
5
|
Valdivia A, Avalos AM, Leyton L. Thy-1 (CD90)-regulated cell adhesion and migration of mesenchymal cells: insights into adhesomes, mechanical forces, and signaling pathways. Front Cell Dev Biol 2023; 11:1221306. [PMID: 38099295 PMCID: PMC10720913 DOI: 10.3389/fcell.2023.1221306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/25/2023] [Indexed: 12/17/2023] Open
Abstract
Cell adhesion and migration depend on the assembly and disassembly of adhesive structures known as focal adhesions. Cells adhere to the extracellular matrix (ECM) and form these structures via receptors, such as integrins and syndecans, which initiate signal transduction pathways that bridge the ECM to the cytoskeleton, thus governing adhesion and migration processes. Integrins bind to the ECM and soluble or cell surface ligands to form integrin adhesion complexes (IAC), whose composition depends on the cellular context and cell type. Proteomic analyses of these IACs led to the curation of the term adhesome, which is a complex molecular network containing hundreds of proteins involved in signaling, adhesion, and cell movement. One of the hallmarks of these IACs is to sense mechanical cues that arise due to ECM rigidity, as well as the tension exerted by cell-cell interactions, and transduce this force by modifying the actin cytoskeleton to regulate cell migration. Among the integrin/syndecan cell surface ligands, we have described Thy-1 (CD90), a GPI-anchored protein that possesses binding domains for each of these receptors and, upon engaging them, stimulates cell adhesion and migration. In this review, we examine what is currently known about adhesomes, revise how mechanical forces have changed our view on the regulation of cell migration, and, in this context, discuss how we have contributed to the understanding of signaling mechanisms that control cell adhesion and migration.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Deng Q, Liu L, Tang R, Xian D, Zhong J. A newly improved method of primary cell culture: Tissue block with continuous adhesion subculture in skin fibroblast. Acta Histochem 2023; 125:152090. [PMID: 37639809 DOI: 10.1016/j.acthis.2023.152090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Fibroblasts (FBs) have been widely used as a typical in vitro cell model for investigating the biological processes and cell pathophysiological mechanisms. However, FBs are prone to senescence in cell culture process after several passages. Thus, a new approach to cell culture is quite required to enhance the viability of cells. OBJECTIVE To explore a novel method of cell culture based on skin FBs. METHODS Dermal tissue blocks were obtained from BALB/c neonatal mice and randomly divided into experimental group and control group. The experimental group received the newly improved culture method, namely, continuous adherence subculture of tissue block (CASTB) method; while the traditional subculture method was applied in the control group. Cells at 1st, 5th and 10th passages were collected and identified by using histological/immunohistochemical and western blot analysis. Cellular viability, proliferation, senescence and apoptosis were analyzed through application of cell growth curve, CCK-8 assay, Ki67 assay, PCNA protein analysis, β-galactosidase staining, flow cytometry and western blot analysis. RESULTS Cells under two culture patterns exhibited spindle/irregular shape and vimentin positive expression. With the increase of passage times, the cellular growth rate in the control group gradually decreased, but no alterations emerged from the experimental group. CASTB method remarkably promoted cell growth and proliferation. Moreover, a greatly lower apoptosis and senescence tendency appeared in the experimental group than the control group with passages increasing. CONCLUSION The method of CASTB is superior to traditional subculture, offering a large number of primary FBs with higher efficiency and success rate and being worth of further popularization and application.
Collapse
Affiliation(s)
- Qiyan Deng
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lumei Liu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ran Tang
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Dehai Xian
- Department of Anatomy, Southwest Medical University, Luzhou 646000, China.
| | - Jianqiao Zhong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
7
|
Carvelli L, Hermo L, O’Flaherty C, Oko R, Pshezhetsky AV, Morales CR. Effects of Heparan sulfate acetyl-CoA: Alpha-glucosaminide N-acetyltransferase (HGSNAT) inactivation on the structure and function of epithelial and immune cells of the testis and epididymis and sperm parameters in adult mice. PLoS One 2023; 18:e0292157. [PMID: 37756356 PMCID: PMC10529547 DOI: 10.1371/journal.pone.0292157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Heparan sulfate (HS), an abundant component of the apical cell surface and basement membrane, belongs to the glycosaminoglycan family of carbohydrates covalently linked to proteins called heparan sulfate proteoglycans. After endocytosis, HS is degraded in the lysosome by several enzymes, including heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT), and in its absence causes Mucopolysaccharidosis III type C (Sanfilippo type C). Since endocytosis occurs in epithelial cells of the testis and epididymis, we examined the morphological effects of Hgsnat inactivation in these organs. In the testis, Hgsnat knockout (Hgsnat-Geo) mice revealed statistically significant decrease in tubule and epithelial profile area of seminiferous tubules. Electron microscopy (EM) analysis revealed cross-sectional tubule profiles with normal and moderately to severely altered appearances. Abnormalities in Sertoli cells and blood-testis barrier and the absence of germ cells in some tubules were noted along with altered morphology of sperm, sperm motility parameters and a reduction in fertilization rates in vitro. Along with quantitatively increased epithelial and tubular profile areas in the epididymis, EM demonstrated significant accumulations of electrolucent lysosomes in the caput-cauda regions that were reactive for cathepsin D and prosaposin antibodies. Lysosomes with similar storage materials were also found in basal, clear and myoid cells. In the mid/basal region of the epithelium of caput-cauda regions of KO mice, large vacuolated cells, unreactive for cytokeratin 5, a basal cell marker, were identified morphologically as epididymal mononuclear phagocytes (eMPs). The cytoplasm of the eMPs was occupied by a gigantic lysosome suggesting an active role of these cells in removing debris from the epithelium. Some eMPs were found in proximity to T-lymphocytes, a feature of dendritic cells. Taken together, our results reveal that upon Hgsnat inactivation, morphological alterations occur to the testis affecting sperm morphology and motility parameters and abnormal lysosomes in epididymal epithelial cells, indicative of a lysosomal storage disease.
Collapse
Affiliation(s)
- Lorena Carvelli
- IHEM-CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Cristian O’Flaherty
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Department of Surgery (Urology Division), McGill University, Montréal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Alexey V. Pshezhetsky
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Ahn S, Sharma U, Kasuba KC, Strohmeyer N, Müller DJ. Engineered Biomimetic Fibrillar Fibronectin Matrices Regulate Cell Adhesion Initiation, Migration, and Proliferation via α5β1 Integrin and Syndecan-4 Crosstalk. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300812. [PMID: 37357136 PMCID: PMC10460904 DOI: 10.1002/advs.202300812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Indexed: 06/27/2023]
Abstract
Cells regulate adhesion to the fibrillar extracellular matrix (ECM) of which fibronectin is an essential component. However, most studies characterize cell adhesion to globular fibronectin substrates at time scales long after cells polarize and migrate. To overcome this limitation, a simple and scalable method to engineer biomimetic 3D fibrillar fibronectin matrices is introduced and how they are sensed by fibroblasts from the onset of attachment is characterized. Compared to globular fibronectin substrates, fibroblasts accelerate adhesion initiation and strengthening within seconds to fibrillar fibronectin matrices via α5β1 integrin and syndecan-4. This regulation, which additionally accelerates on stiffened fibrillar matrices, involves actin polymerization, actomyosin contraction, and the cytoplasmic proteins paxillin, focal adhesion kinase, and phosphoinositide 3-kinase. Furthermore, this immediate sensing and adhesion of fibroblast to fibrillar fibronectin guides migration speed, persistency, and proliferation range from hours to weeks. The findings highlight that fibrillar fibronectin matrices, compared to widely-used globular fibronectin, trigger short- and long-term cell decisions very differently and urge the use of such matrices to better understand in vivo interactions of cells and ECMs. The engineered fibronectin matrices, which can be printed onto non-biological surfaces without loss of function, open avenues for various cell biological, tissue engineering and medical applications.
Collapse
Affiliation(s)
- Seungkuk Ahn
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Upnishad Sharma
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Krishna Chaitanya Kasuba
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Nico Strohmeyer
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Daniel J. Müller
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| |
Collapse
|
9
|
Integrin receptor trafficking in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:271-302. [PMID: 36813362 DOI: 10.1016/bs.pmbts.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Integrins are a family of 24 different heterodimers that are indispensable for multicellular life. Cell polarity, adhesion and migration are controlled by integrins delivered to the cell surface which in turn is regulated by the exo- and endocytic trafficking of integrins. The deep integration between trafficking and cell signaling determines the spatial and temporal output from any biochemical cue. Integrin trafficking plays a key role in development and many pathological conditions, especially cancer. Several novel regulators of integrin traffic have been discovered in recent times, including a novel class of integrin carrying vesicles, the intracellular nanovesicles (INVs). The tight regulation of trafficking pathways by cell signaling, where kinases phosphorylate key small GTPases in the trafficking pathway enable coordination of cell response to the extracellular milieu. Integrin heterodimer expression and trafficking differ in different tissues and contexts. In this Chapter, we discuss recent studies on integrin trafficking and its contribution to normal physiological and pathophysiological states.
Collapse
|
10
|
Cegarra C, Cameron B, Chaves C, Dabdoubi T, Do TM, Genêt B, Roudières V, Shi Y, Tchepikoff P, Lesuisse D. An innovative strategy to identify new targets for delivering antibodies to the brain has led to the exploration of the integrin family. PLoS One 2022; 17:e0274667. [PMID: 36108060 PMCID: PMC9477330 DOI: 10.1371/journal.pone.0274667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Increasing brain exposure of biotherapeutics is key to success in central nervous system disease drug discovery. Accessing the brain parenchyma is especially difficult for large polar molecules such as biotherapeutics and antibodies because of the blood-brain barrier. We investigated a new immunization strategy to identify novel receptors mediating transcytosis across the blood-brain barrier.
Method
We immunized mice with primary non-human primate brain microvascular endothelial cells to obtain antibodies. These antibodies were screened for their capacity to bind and to be internalized by primary non-human primate brain microvascular endothelial cells and Human Cerebral Microvascular Endothelial Cell clone D3. They were further evaluated for their transcytosis capabilities in three in vitro blood-brain barrier models. In parallel, their targets were identified by two different methods and their pattern of binding to human tissue was investigated using immunohistochemistry.
Results
12 antibodies with unique sequence and internalization capacities were selected amongst more than six hundred. Aside from one antibody targeting Activated Leukocyte Cell Adhesion Molecule and one targeting Striatin3, most of the other antibodies recognized β1 integrin and its heterodimers. The antibody with the best transcytosis capabilities in all blood-brain barrier in vitro models and with the best binding capacity was an anti-αnβ1 integrin. In comparison, commercial anti-integrin antibodies performed poorly in transcytosis assays, emphasizing the originality of the antibodies derived here. Immunohistochemistry studies showed specific vascular staining on human and non-human primate tissues.
Conclusions
This transcytotic behavior has not previously been reported for anti-integrin antibodies. Further studies should be undertaken to validate this new mechanism in vivo and to evaluate its potential in brain delivery.
Collapse
Affiliation(s)
- Céline Cegarra
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
- * E-mail:
| | | | - Catarina Chaves
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | | | - Tuan-Minh Do
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Bruno Genêt
- Integrated Drug Discovery, Sanofi, Vitry-Sur-Seine, France
| | - Valérie Roudières
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Yi Shi
- Histology, Translational Sciences, Sanofi, Vitry-Sur-Seine, France
| | | | - Dominique Lesuisse
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| |
Collapse
|
11
|
Meecham A, Cutmore LC, Protopapa P, Rigby LG, Marshall JF. Ligand-bound integrin αvβ6 internalisation and trafficking. Front Cell Dev Biol 2022; 10:920303. [PMID: 36092709 PMCID: PMC9448872 DOI: 10.3389/fcell.2022.920303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The integrin αvβ6 is expressed at low levels in most normal healthy tissue but is very often upregulated in a disease context including cancer and fibrosis. Integrins use endocytosis and trafficking as a means of regulating their surface expression and thus their functions, however little is known of how this process is regulated in the context of αvβ6. As αvβ6 is a major target for the development of therapeutics in cancer and fibrosis, understanding these dynamics is critical in the development of αvβ6-targeted therapies. Following development of a flow cytometry-based assay to measure ligand (A20FMDV2 or LAP)-bound αvβ6 endocytosis, an siRNA screen was performed to identify which genes were responsible for internalising αvβ6. These data identified 15 genes (DNM2, CBLB, DNM3, CBL, EEA1, CLTC, ARFGAP3, CAV1, CYTH2, CAV3, CAV2, IQSEC1, AP2M1, TSG101) which significantly decreased endocytosis, predominantly within dynamin-dependent pathways. Inhibition of these dynamin-dependent pathways significantly reduced αvβ6-dependent migration (αvβ6-specific migration was 547 ± 128 under control conditions, reduced to 225 ± 73 with clathrin inhibition, and 280 ± 51 with caveolin inhibition). Colocalization studies of αvβ6 with endosome markers revealed that up to 6 h post-internalisation of ligand, αvβ6 remains in Rab11-positive endosomes in a perinuclear location, with no evidence of αvβ6 degradation up to 48 h post exposure to A20FMDV2. Additionally, 60% of ligand-bound αvβ6 was recycled back to the surface by 6 h. With studies ongoing using conjugated A20FMDV2 to therapeutically target αvβ6 in cancer and fibrosis, these data have important implications. Binding of A20FMDV2 seemingly removes much of the αvβ6 from the cell membrane, and upon its recycling, a large fraction appears to still be in the ligand-bound state. While these results are observed with A20FMDV2, these data will be of value in the design of αvβ6-specific therapeutics and potentially the types of therapeutic load.
Collapse
Affiliation(s)
- Amelia Meecham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- University of California, San Diego, San Diego, CA, United States
| | - Lauren C. Cutmore
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pantelitsa Protopapa
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lauren G. Rigby
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Huang S, Wang Y, Xie S, Lai Y, Mo C, Zeng T, Kuang S, Zhou C, Zeng Z, Chen Y, Huang S, Gao L, Lv Z. Isoliquiritigenin alleviates liver fibrosis through caveolin-1-mediated hepatic stellate cells ferroptosis in zebrafish and mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154117. [PMID: 35489326 DOI: 10.1016/j.phymed.2022.154117] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Liver fibrosis is a major disease that threatens people's health around the world. However, there is a lack of effective treatment to completely reverse liver fibrosis. Liver transplantation is currently the only curative option for patients with advanced cirrhosis. Ferroptosis is a newly discovered type of cell death and plays an important role in the process of liver fibrosis, but the specific mechanism needs to be clarified. HYPOTHESIS/PURPOSE To explore the regulatory mechanism of isoliquiritigenin (ISL) in the process of liver fibrosis and the relationship between Cav-1 and ferroptosis. METHODS In this research, zebrafish, HSC-T6 cells, and mice were used as the research object. Different ROS probes to visually detect the content and distribution of ROS in live zebrafish and cells. Lentivirus and siRNA-mediated transfection techniques were used for the construction of Cav-1 overexpression and knockdown cell lines to verify the important role of Cav-1 in vitro. RESULTS Generally, we first elucidated that ISL relieved liver fibrosis by inducing hepatic stellate cells (HSCs) ferroptosis through repressing GPX4 expression and increasing the expression of TFR and DMT1, thus producing a large number of ROS, we also found that Cav-1 exerted its anti-hepatic fibrosis effect by promoting HSCs ferroptosis. CONCLUSION Our results have shown that Cav-1-mediated HSCs ferroptosis is necessary for ISL to play an anti-fibrotic effect in vitro and in vivo.
Collapse
Affiliation(s)
- Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Yuhua Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Chan Mo
- Medical Laboratory of the Third affiliated Hospital of Shenzhen University, Shenzhen, 518001, China
| | - Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Shanshan Kuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Zhiyun Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China
| | - Shaohui Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China; The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China; Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong,510515, China.
| |
Collapse
|
13
|
Hu P, Leyton L, Hagood JS, Barker TH. Thy-1-Integrin Interactions in cis and Trans Mediate Distinctive Signaling. Front Cell Dev Biol 2022; 10:928510. [PMID: 35733855 PMCID: PMC9208718 DOI: 10.3389/fcell.2022.928510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Thy-1 is a cell surface glycosylphosphatidylinositol (GPI)-anchored glycoprotein that bears a broad mosaic of biological roles across various cell types. Thy-1 displays strong physiological and pathological implications in development, cancer, immunity, and tissue fibrosis. Quite uniquely, Thy-1 is capable of mediating integrin-related signaling through direct trans- and cis-interaction with integrins. Both interaction types have shown distinctive roles, even when interacting with the same type of integrin, where binding in trans or in cis often yields divergent signaling events. In this review, we will revisit recent progress and discoveries of Thy-1–integrin interactions in trans and in cis, highlight their pathophysiological consequences and explore other potential binding partners of Thy-1 within the integrin regulation/signaling paradigm.
Collapse
Affiliation(s)
- Ping Hu
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile and Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - James S. Hagood
- Department of Pediatrics, Division of Pulmonology, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Program for Rare and Interstitial Lung Disease, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Thomas H. Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
- *Correspondence: Thomas H. Barker,
| |
Collapse
|
14
|
Shaik F, Balderstone MJM, Arokiasamy S, Whiteford JR. Roles of Syndecan-4 in cardiac injury and repair. Int J Biochem Cell Biol 2022; 146:106196. [PMID: 35331918 DOI: 10.1016/j.biocel.2022.106196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
The heparan sulphate proteoglycan Syndecan-4 belongs to a 4-member family of transmembrane receptors. Genetic deletion of Syndecan-4 in mice causes negligible developmental abnormalities however when challenged these animals show distinct phenotypes. Synedcan-4 is expressed in many cell types in the heart and its expression is elevated in response to cardiac injury and recent studies have suggested roles for Syndecan-4 in repair mechanisms within the damaged heart. The purpose of this review is to explore these biological insights into the role of Syndecan-4 in both the injured heart and later during cardiac repair and remodeling.
Collapse
Affiliation(s)
- Faheem Shaik
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK
| | - Michaela J M Balderstone
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK
| | - Samantha Arokiasamy
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK.
| | - James R Whiteford
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK.
| |
Collapse
|
15
|
Nardo WD, Miotto PM, Bayliss J, Nie S, Keenan SN, Montgomery MK, Watt MJ. Proteomic analysis reveals exercise training induced remodelling of hepatokine secretion and uncovers syndecan-4 as a regulator of hepatic lipid metabolism. Mol Metab 2022; 60:101491. [PMID: 35381388 PMCID: PMC9034320 DOI: 10.1016/j.molmet.2022.101491] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/04/2022] Open
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) is linked to impaired lipid metabolism and systemic insulin resistance, which is partly mediated by altered secretion of liver proteins known as hepatokines. Regular physical activity can resolve NAFLD and improve its metabolic comorbidities, however, the effects of exercise training on hepatokine secretion and the metabolic impact of exercise-regulated hepatokines in NAFLD remain unresolved. Herein, we examined the effect of endurance exercise training on hepatocyte secreted proteins with the aim of identifying proteins that regulate metabolism and reduce NAFLD severity. Methods C57BL/6 mice were fed a high-fat diet for six weeks to induce NAFLD. Mice were exercise trained for a further six weeks, while the control group remained sedentary. Hepatocytes were isolated two days after the last exercise bout, and intracellular and secreted proteins were detected using label-free mass spectrometry. Hepatocyte secreted factors were applied to skeletal muscle and liver ex vivo and insulin action and fatty acid metabolism were assessed. Syndecan-4 (SDC4), identified as an exercise-responsive hepatokine, was overexpressed in the livers of mice using adeno-associated virus. Whole-body energy homeostasis was assessed by indirect calorimetry and skeletal muscle and liver metabolism was assessed using radiometric techniques. Results Proteomics analysis detected 2657 intracellular and 1593 secreted proteins from mouse hepatocytes. Exercise training remodelled the hepatocyte proteome, with differences in 137 intracellular and 35 secreted proteins. Bioinformatic analysis of hepatocyte secreted proteins revealed enrichment of tumour suppressive proteins and proteins involved in lipid metabolism and mitochondrial function, and suppression of oncogenes and regulators of oxidative stress. Hepatocyte secreted factors from exercise trained mice improved insulin action in skeletal muscle and increased hepatic fatty acid oxidation. Hepatocyte-specific overexpression of SDC4 reduced hepatic steatosis, which was associated with reduced hepatic fatty acid uptake, and blunted pro-inflammatory and pro-fibrotic gene expression. Treating hepatocytes with recombinant ectodomain of SDC4 (secreted form) recapitulated these effects with reduced fatty acid uptake, lipid storage and lipid droplet accumulation. Conclusions Remodelling of hepatokine secretion is an adaptation to regular exercise training that induces changes in metabolism in the liver and skeletal muscle. SDC4 is a novel exercise-responsive hepatokine that decreases fatty acid uptake and reduces steatosis in the liver. By understanding the proteomic changes in hepatocytes with exercise, these findings have potential for the discovery of new therapeutic targets for NAFLD. Exercise training remodels hepatokine secretion. Exercise regulated secreted factors improve insulin action in skeletal muscle. Syndecan-4 (SDC4) is a novel exercise-induced hepatokine. SDC4 reduces hepatic fatty acid uptake and hepatic steatosis.
Collapse
|
16
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
17
|
Zhang Z, Li Y, Zhang T, Yang X, Fan K, Wang D, Li S, Hu Y, Fu W. Titanium implants modified by laser microtexturing enhance the bioactivity of gastric epithelial cells and fibroblast cells. J Appl Biomater Funct Mater 2021; 19:22808000211064951. [PMID: 34905988 DOI: 10.1177/22808000211064951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The clinical application of anastomotic instruments improves the efficiency of the digestive tract surgery. However, the stapler with titanium nails implanted is still controversial in terms of anastomotic complications, and further improvement and optimization are needed. The purpose of this study was to explore the optimal microtextured parameters that could enhance the bioactivity of titanium implants in vitro. Laser microtexturing technology was used to construct the groove-type microstructural surfaces with different parameters, and human gastric mucosal epithelial cells (GES-1 cells) and mouse fibroblasts (3T3 cells) were cultured on the surface of the titanium plates in vitro. The data of cell adhesion, cell proliferation and cell activity were obtained and statistically analyzed. The textured titanium plates meet the expected design. GES-1 and 3T3 cell adhesion were better in the surface of titanium plates in microstructural group than that in the polished group. GES-1 and 3T3 cells also showed higher proliferative activity in the microstructural group compared with the polished group. The laser textured titanium plates have good groove-type microstructure, which increase the surface roughness, change the surface wettability, promote the adhesion, proliferating and orderly growth of GES-1 and 3T3 cells, and show good biological properties.
Collapse
Affiliation(s)
- Zhaoxiong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yuanchun Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Ting Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Xiaoyang Yang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| | - Kaihu Fan
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Daohan Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Shuliang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, Shandong, People's Republic of China.,Department of Gastrointestinal Surgery, The Second Hospital of Liaocheng Affiliated to Shandong First Medical University, Linqing, Shandong, People's Republic of China
| | - Yahui Hu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, National Demonstration Center for Experimental Mechanical and Electrical Engineering Education (Tianjin University of Technology), Tianjin, People's Republic of China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
18
|
Black AR, Black JD. The complexities of PKCα signaling in cancer. Adv Biol Regul 2021; 80:100769. [PMID: 33307285 PMCID: PMC8141086 DOI: 10.1016/j.jbior.2020.100769] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 01/06/2023]
Abstract
Protein kinase C α (PKCα) is a ubiquitously expressed member of the PKC family of serine/threonine kinases with diverse functions in normal and neoplastic cells. Early studies identified anti-proliferative and differentiation-inducing functions for PKCα in some normal tissues (e.g., regenerating epithelia) and pro-proliferative effects in others (e.g., cells of the hematopoietic system, smooth muscle cells). Additional well documented roles of PKCα signaling in normal cells include regulation of the cytoskeleton, cell adhesion, and cell migration, and PKCα can function as a survival factor in many contexts. While a majority of tumors lose expression of PKCα, others display aberrant overexpression of the enzyme. Cancer-related mutations in PKCα are uncommon, but rare examples of driver mutations have been detected in certain cancer types (e. g., choroid gliomas). Here we review the role of PKCα in various cancers, describe mechanisms by which PKCα affects cancer-related cell functions, and discuss how the diverse functions of PKCα contribute to tumor suppressive and tumor promoting activities of the enzyme. We end the discussion by addressing mutations and expression of PKCα in tumors and the clinical relevance of these findings.
Collapse
Affiliation(s)
- Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
19
|
Syndecan-4 as a Pathogenesis Factor and Therapeutic Target in Cancer. Biomolecules 2021; 11:biom11040503. [PMID: 33810567 PMCID: PMC8065655 DOI: 10.3390/biom11040503] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is an important cause of morbidity and mortality worldwide. Advances in research on the biology of cancer revealed alterations in several key pathways underlying tumorigenesis and provided molecular targets for developing new and improved existing therapies. Syndecan-4, a transmembrane heparan sulfate proteoglycan, is a central mediator of cell adhesion, migration and proliferation. Although several studies have demonstrated important roles of syndecan-4 in cell behavior and its interactions with growth factors, extracellular matrix (ECM) molecules and cytoskeletal signaling proteins, less is known about its role and expression in multiple cancer. The data summarized in this review demonstrate that high expression of syndecan-4 is an unfavorable biomarker for estrogen receptor-negative breast cancer, glioma, liver cancer, melanoma, osteosarcoma, papillary thyroid carcinoma and testicular, kidney and bladder cancer. In contrast, in neuroblastoma and colorectal cancer, syndecan-4 is downregulated. Interestingly, syndecan-4 expression is modulated by anticancer drugs. It is upregulated upon treatment with zoledronate and this effect reduces invasion of breast cancer cells. In our recent work, we demonstrated that the syndecan-4 level was reduced after trastuzumab treatment. Similarly, syndecan-4 levels are also reduced after panitumumab treatment. Together, the data found suggest that syndecan-4 level is crucial for understanding the changes involving in malignant transformation, and also demonstrate that syndecan-4 emerges as an important target for cancer therapy and diagnosis.
Collapse
|
20
|
Gopal S, Arokiasamy S, Pataki C, Whiteford JR, Couchman JR. Syndecan receptors: pericellular regulators in development and inflammatory disease. Open Biol 2021; 11:200377. [PMID: 33561383 PMCID: PMC8061687 DOI: 10.1098/rsob.200377] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
The syndecans are the major family of transmembrane proteoglycans, usually bearing multiple heparan sulfate chains. They are present on virtually all nucleated cells of vertebrates and are also present in invertebrates, indicative of a long evolutionary history. Genetic models in both vertebrates and invertebrates have shown that syndecans link to the actin cytoskeleton and can fine-tune cell adhesion, migration, junction formation, polarity and differentiation. Although often associated as co-receptors with other classes of receptors (e.g. integrins, growth factor and morphogen receptors), syndecans can nonetheless signal to the cytoplasm in discrete ways. Syndecan expression levels are upregulated in development, tissue repair and an array of human diseases, which has led to the increased appreciation that they may be important in pathogenesis not only as diagnostic or prognostic agents, but also as potential targets. Here, their functions in development and inflammatory diseases are summarized, including their potential roles as conduits for viral pathogen entry into cells.
Collapse
Affiliation(s)
- Sandeep Gopal
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Samantha Arokiasamy
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Csilla Pataki
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - James R. Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - John R. Couchman
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
21
|
Salinas-Marín R, Villanueva-Cabello TM, Martínez-Duncker I. Biology of Proteoglycans and Associated Glycosaminoglycans. COMPREHENSIVE GLYCOSCIENCE 2021:63-102. [DOI: 10.1016/b978-0-12-819475-1.00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Iyoda T, Fujita M, Fukai F. Biologically Active TNIIIA2 Region in Tenascin-C Molecule: A Major Contributor to Elicit Aggressive Malignant Phenotypes From Tumors/Tumor Stroma. Front Immunol 2020; 11:610096. [PMID: 33362799 PMCID: PMC7755593 DOI: 10.3389/fimmu.2020.610096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
Tenascin (TN)-C is highly expressed specifically in the lesions of inflammation-related diseases, including tumors. The expression level of TN-C in tumors and the tumor stroma is positively correlated with poor prognosis. However, no drugs targeting TN-C are currently clinically available, partly because the role of TN-C in tumor progression remains controversial. TN-C harbors an alternative splicing site in its fibronectin type III repeat domain, and its splicing variants including the type III-A2 domain are frequently detected in malignant tumors. We previously identified a biologically active region termed TNIIIA2 in the fibronectin type III-A2 domain of TN-C molecule and showed that this region is involved in promoting firm and persistent cell adhesion to fibronectin. In the past decade, through the exposure of various cell lines to peptides containing the TNIIIA2 region, we have published reports demonstrating the ability of the TNIIIA2 region to modulate distinct cellular activities, including survival/growth, migration, and invasion. Recently, we reported that the signals derived from TNIIIA2-mediated β1 integrin activation might play a crucial role for inducing malignant behavior of glioblastoma (GBM). GBM cells exposed to the TNIIIA2 region showed not only exacerbation of PDGF-dependent proliferation, but also acceleration of disseminative migration. On the other hand, we also found that the pro-inflammatory phenotypic changes were promoted when macrophages are stimulated with TNIIIA2 region in relatively low concentration and resulting MMP-9 upregulation is needed to release of the TNIIIA2 region from TN-C molecule. With the contribution of TNIIIA2-stimulated macrophages, the positive feedback spiral loop, which consists of the expression of TN-C, PDGF, and β1 integrin, and TNIIIA2 release, seemed to be activated in GBM with aggressive malignancy. Actually, the growth of transplanted GBM grafts in mice was significantly suppressed via the attenuation of β1 integrin activation. In this review, we thus introduce that the TNIIIA2 region has a significant impact on malignant progression of tumors by regulating cell adhesion. Importantly, it has been demonstrated that the TNIIIA2 region exerts unique biological functions through the extremely strong activation of β1-integrins and their long-lasting duration. These findings prompt us to develop new therapeutic agents targeting the TNIIIA2 region.
Collapse
Affiliation(s)
- Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
23
|
Benito-Jardón M, Strohmeyer N, Ortega-Sanchís S, Bharadwaj M, Moser M, Müller DJ, Fässler R, Costell M. αv-Class integrin binding to fibronectin is solely mediated by RGD and unaffected by an RGE mutation. J Cell Biol 2020; 219:e202004198. [PMID: 33141174 PMCID: PMC7644020 DOI: 10.1083/jcb.202004198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/20/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Fibronectin (FN) is an essential glycoprotein of the extracellular matrix; binds integrins, syndecans, collagens, and growth factors; and is assembled by cells into complex fibrillar networks. The RGD motif in FN facilitates cell binding- and fibrillogenesis through binding to α5β1 and αv-class integrins. However, whether RGD is the sole binding site for αv-class integrins is unclear. Most notably, substituting aspartate with glutamate (RGE) was shown to eliminate integrin binding in vitro, while mouse genetics revealed that FNRGE preserves αv-class integrin binding and fibrillogenesis. To address this conflict, we employed single-cell force spectroscopy, engineered cells, and RGD motif-deficient mice (Fn1ΔRGD/ΔRGD) to search for additional αv-class integrin-binding sites. Our results demonstrate that α5β1 and αv-class integrins solely recognize the FN-RGD motif and that αv-class, but not α5β1, integrins retain FN-RGE binding. Furthermore, Fn1ΔRGD/ΔRGD tissues and cells assemble abnormal and dysfunctional FNΔRGD fibrils in a syndecan-dependent manner. Our data highlight the central role of FN-RGD and the functionality of FN-RGE for αv-class integrins.
Collapse
Affiliation(s)
- María Benito-Jardón
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Nico Strohmeyer
- Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - Sheila Ortega-Sanchís
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | | | - Markus Moser
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | - Mercedes Costell
- Department of Biochemistry and Molecular Biology, Universitat de València, Burjassot, Spain
- Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| |
Collapse
|
24
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Buwa N, Mazumdar D, Balasubramanian N. Caveolin1 Tyrosine-14 Phosphorylation: Role in Cellular Responsiveness to Mechanical Cues. J Membr Biol 2020; 253:509-534. [PMID: 33089394 DOI: 10.1007/s00232-020-00143-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The plasma membrane is a dynamic lipid bilayer that engages with the extracellular microenvironment and intracellular cytoskeleton. Caveolae are distinct plasma membrane invaginations lined by integral membrane proteins Caveolin1, 2, and 3. Caveolae formation and stability is further supported by additional proteins including Cavin1, EHD2, Pacsin2 and ROR1. The lipid composition of caveolar membranes, rich in cholesterol and phosphatidylserine, actively contributes to caveolae formation and function. Post-translational modifications of Cav1, including its phosphorylation of the tyrosine-14 residue (pY14Cav1) are vital to its function in and out of caveolae. Cells that experience significant mechanical stress are seen to have abundant caveolae. They play a vital role in regulating cellular signaling and endocytosis, which could further affect the abundance and distribution of caveolae at the PM, contributing to sensing and/or buffering mechanical stress. Changes in membrane tension in cells responding to multiple mechanical stimuli affects the organization and function of caveolae. These mechanical cues regulate pY14Cav1 levels and function in caveolae and focal adhesions. This review, along with looking at the mechanosensitive nature of caveolae, focuses on the role of pY14Cav1 in regulating cellular mechanotransduction.
Collapse
Affiliation(s)
- Natasha Buwa
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Debasmita Mazumdar
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India
| | - Nagaraj Balasubramanian
- Indian Institute of Science Education and Research, Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, India.
| |
Collapse
|
26
|
Valdivia A, Cárdenas A, Brenet M, Maldonado H, Kong M, Díaz J, Burridge K, Schneider P, San Martín A, García-Mata R, Quest AFG, Leyton L. Syndecan-4/PAR-3 signaling regulates focal adhesion dynamics in mesenchymal cells. Cell Commun Signal 2020; 18:129. [PMID: 32811537 PMCID: PMC7433185 DOI: 10.1186/s12964-020-00629-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/17/2020] [Indexed: 01/04/2023] Open
Abstract
Background Syndecans regulate cell migration thus having key roles in scarring and wound healing processes. Our previous results have shown that Thy-1/CD90 can engage both αvβ3 integrin and Syndecan-4 expressed on the surface of astrocytes to induce cell migration. Despite a well-described role of Syndecan-4 during cell movement, information is scarce regarding specific Syndecan-4 partners involved in Thy-1/CD90-stimulated cell migration. Methods Mass spectrometry (MS) analysis of complexes precipitated with the Syndecan-4 cytoplasmic tail peptide was used to identify potential Syndecan-4-binding partners. The interactions found by MS were validated by immunoprecipitation and proximity ligation assays. The conducted research employed an array of genetic, biochemical and pharmacological approaches, including: PAR-3, Syndecan-4 and Tiam1 silencing, active Rac1 GEFs affinity precipitation, and video microscopy. Results We identified PAR-3 as a Syndecan-4-binding protein. Its interaction depended on the carboxy-terminal EFYA sequence present on Syndecan-4. In astrocytes where PAR-3 expression was reduced, Thy-1-induced cell migration and focal adhesion disassembly was impaired. This effect was associated with a sustained Focal Adhesion Kinase activation in the siRNA-PAR-3 treated cells. Our data also show that Thy-1/CD90 activates Tiam1, a PAR-3 effector. Additionally, we found that after Syndecan-4 silencing, Tiam1 activation was decreased and it was no longer recruited to the membrane. Syndecan-4/PAR-3 interaction and the alteration in focal adhesion dynamics were validated in mouse embryonic fibroblast (MEF) cells, thereby identifying this novel Syndecan-4/PAR-3 signaling complex as a general mechanism for mesenchymal cell migration involved in Thy-1/CD90 stimulation. Conclusions The newly identified Syndecan-4/PAR-3 signaling complex participates in Thy-1/CD90-induced focal adhesion disassembly in mesenchymal cells. The mechanism involves focal adhesion kinase dephosphorylation and Tiam1 activation downstream of Syndecan-4/PAR-3 signaling complex formation. Additionally, PAR-3 is defined here as a novel adhesome-associated component with an essential role in focal adhesion disassembly during polarized cell migration. These novel findings uncover signaling mechanisms regulating cell migration, thereby opening up new avenues for future research on Syndecan-4/PAR-3 signaling in processes such as wound healing and scarring. Graphical abstract ![]()
Collapse
Affiliation(s)
- Alejandra Valdivia
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile. .,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile. .,Microscopy in Medicine (MiM) Core, Emory University, Atlanta, GA, 30322, USA. .,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,School of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA.
| | - Areli Cárdenas
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Marianne Brenet
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Horacio Maldonado
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Department of Pediatrics, Pulmonology Division, Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,UNC Catalyst for Rare Disease, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Milene Kong
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Keith Burridge
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Alejandra San Martín
- School of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA
| | - Rafael García-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - Andrew F G Quest
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile.,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia, 838-0453, Santiago, Chile. .,Center for studies on Exercise, Metabolism and Cancer (CEMC) and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.
| |
Collapse
|
27
|
Lakoduk AM, Kadlecova Z, Schmid SL. A functionally neutral single chain antibody to measure beta-1 integrin uptake and recycling. Traffic 2020; 21:590-602. [PMID: 32613646 PMCID: PMC7442622 DOI: 10.1111/tra.12754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
Integrin‐mediated cell adhesion and signaling are critical for many physiological processes. The dynamic turnover of integrins and their associated adhesion complexes through endocytic and recycling pathways has emerged as an important mechanism for controlling cell migration and invasion in cancer. Thus, the regulation of integrin trafficking and how this may be altered by disease‐specific molecular mechanisms has generated considerable interest. However, current tools available to study integrin trafficking may cause artifacts and/or do not provide adequate kinetic information. Here, we report the generation of a functionally neutral and monovalent single chain antibody to quantitatively and qualitatively measure β1 integrin trafficking in cells. Our novel probe can be used in a variety of assays and allows for the biochemical characterization of rapid recycling of endogenous integrins. We also demonstrate its potential utility in live cell imaging, providing proof of principle to guide future integrin probe design. The dynamic turnover of integrins through endocytic trafficking pathways has emerged as a key mechanism for cell migration and invasion. Lakoduk et al. report the generation of a functionally neutral and monovalent antibody‐based probe to track and measure endogenous beta‐1 integrin uptake and fast recycling in multiple cell types. Their tool, scFvK20, serves as proof of principle inspiration for future integrin probe design.
Collapse
Affiliation(s)
- Ashley M Lakoduk
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Zuzana Kadlecova
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sandra L Schmid
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
28
|
Adhesion and growth factor receptor crosstalk mechanisms controlling cell migration. Essays Biochem 2020; 63:553-567. [PMID: 31551325 DOI: 10.1042/ebc20190025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 12/30/2022]
Abstract
Cell migration requires cells to sense and interpret an array of extracellular signals to precisely co-ordinate adhesion dynamics, local application of mechanical force, polarity signalling and cytoskeletal dynamics. Adhesion receptors and growth factor receptors (GFRs) exhibit functional and signalling characteristics that individually contribute to cell migration. Integrins transmit bidirectional mechanical forces and transduce long-range intracellular signals. GFRs are fast acting and highly sensitive signalling machines that initiate signalling cascades to co-ordinate global cellular processes. Syndecans are microenvironment sensors that regulate GTPases to control receptor trafficking, cytoskeletal remodelling and adhesion dynamics. However, an array of crosstalk mechanisms exists, which co-ordinate and integrate the functions of the different receptor families. Here we discuss the nature of adhesion receptor and GFR crosstalk mechanisms. The unifying theme is that efficient cell migration requires precise spatial and temporal co-ordination of receptor crosstalk. However, a higher order of complexity emerges; whereby multiple crosstalk mechanisms are integrated and subject to both positive and negative feedbacks. Exquisite and sensitive control of these mechanisms ensures that mechanical forces and pro-migratory signals are triggered in the right place and at the right time during cell migration. Finally, we discuss the challenges, and potential therapeutic benefits, associated with deciphering this complexity.
Collapse
|
29
|
Godmann L, Bollmann M, Korb-Pap A, König U, Sherwood J, Beckmann D, Mühlenberg K, Echtermeyer F, Whiteford J, De Rossi G, Pap T, Bertrand J. Antibody-mediated inhibition of syndecan-4 dimerisation reduces interleukin (IL)-1 receptor trafficking and signalling. Ann Rheum Dis 2020; 79:481-489. [PMID: 32094158 DOI: 10.1136/annrheumdis-2019-216847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Syndecan-4 (sdc4) is a cell-anchored proteoglycan that consists of a transmembrane core protein and glucosaminoglycan (GAG) side chains. Binding of soluble factors to the GAG chains of sdc4 may result in the dimerisation of sdc4 and the initiation of downstream signalling cascades. However, the question of how sdc4 dimerisation and signalling affects the response of cells to inflammatory stimuli is unknown. METHODS Sdc4 immunostaining was performed on rheumatoid arthritis (RA) tissue sections. Interleukin (IL)-1 induced extracellular signal-regulated kinases (ERK) phosphorylation and matrix metalloproteinase-3 production was investigated. Il-1 binding to sdc4 was investigated using immunoprecipitation. IL-1 receptor (IL1R1) staining on wild-type, sdc4 and IL1R1 knockout fibroblasts was performed in fluorescence-activated cell sorting analyses. A blocking sdc4 antibody was used to investigate sdc4 dimerisation, IL1R1 expression and the histological paw destruction in the human tumour necrosis factor-alpha transgenic mouse. RESULTS We show that in fibroblasts, the loss of sdc4 or the antibody-mediated inhibition of sdc4 dimerisation reduces the cell surface expression of the IL-1R and regulates the sensitivity of fibroblasts to IL-1. We demonstrate that IL-1 directly binds to sdc4 and in an IL-1R-independent manner leads to its dimerisation. IL-1-induced dimerisation of sdc4 regulates caveolin vesicle-mediated trafficking of the IL1R1, which in turn determines the responsiveness to IL-1. Administration of antibodies (Ab) against the dimerisation domain of sdc4, thus, strongly reduces the expression IL1R1 on arthritic fibroblasts both in vitro and an animal model of human RA. CONCLUSION Collectively, our data suggest that Ab that specifically inhibit sdc4 dimerisation may support anti-IL-1 strategies in diseases such as inflammatory arthritis.
Collapse
Affiliation(s)
- Lars Godmann
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Miriam Bollmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Adelheid Korb-Pap
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Ulrich König
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Joanna Sherwood
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Denise Beckmann
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Katja Mühlenberg
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Frank Echtermeyer
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hanover, Germany
| | - James Whiteford
- Centre for Microvascular Research, Queen Mary University of London, London, UK
| | - Giulia De Rossi
- Centre for Microvascular Research, Queen Mary University of London, London, UK
| | - Thomas Pap
- Institute of Musculoskeletal Medicine (IMM), University Muenster, Muenster, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
30
|
Egger AN, Rajabi‐Estarabadi A, Williams NM, Resnik SR, Fox JD, Wong LL, Jozic I. The importance of caveolins and caveolae to dermatology: Lessons from the caves and beyond. Exp Dermatol 2020; 29:136-148. [PMID: 31845391 PMCID: PMC7028117 DOI: 10.1111/exd.14068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Caveolae are flask-shaped invaginations of the cell membrane rich in cholesterol and sphingomyelin, with caveolin proteins acting as their primary structural components that allow compartmentalization and orchestration of various signalling molecules. In this review, we discuss how pleiotropic functions of caveolin-1 (Cav1) and its intricate roles in numerous cellular functions including lipid trafficking, signalling, cell migration and proliferation, as well as cellular senescence, infection and inflammation, are integral for normal development and functioning of skin and its appendages. We then examine how disruption of the homeostatic levels of Cav1 can lead to development of various cutaneous pathophysiologies including skin cancers, cutaneous fibroses, psoriasis, alopecia, age-related changes in skin and aberrant wound healing and propose how levels of Cav1 may have theragnostic value in skin physiology/pathophysiology.
Collapse
Affiliation(s)
- Andjela N. Egger
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Ali Rajabi‐Estarabadi
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Natalie M. Williams
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Sydney R. Resnik
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Joshua D. Fox
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Lulu L. Wong
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| |
Collapse
|
31
|
Ge J, Cheng X, Yuan C, Qian J, Wu C, Cao C, Yang H, Zhou F, Zou J. Syndecan-4 is a Novel Therapeutic Target for Intervertebral Disc Degeneration via Suppressing JNK/p53 Pathway. Int J Biol Sci 2020; 16:766-776. [PMID: 32071547 PMCID: PMC7019137 DOI: 10.7150/ijbs.40189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/06/2019] [Indexed: 01/07/2023] Open
Abstract
Syndecan-4 is a member of the polysaccharide syndecan family and plays a vital role in intervertebral disc development. Several studies have demonstrated the positive relationship between syndecan-4 expression and intervertebral disc degeneration. However, the detailed molecular mechanism by which syndecan-4 affects the degeneration of nucleus pulposus cells (NPCs) remains unclear. In this study, cell viability was determined by CCK-8 assay, mRNA level was determined by qPCR, and protein expression was determined by western blot. Molecular interaction was determined by chromatin immunoprecipitation assay. A rabbit intervertebral disc degeneration model was established to test for syndecan in vivo. We found that the morphology and viability of NPCs were not affected by the expression of syndecan-4 in the long term. While the NPC function were affected, which results in the degeneration of intervertebral disc. Syndecan-4 overexpression promoted the degeneration of NPCs. Syndecan-4 also activated the JNK signaling pathway and downstream p53 pathways, and promoted degeneration. Inhibition of the JNK pathway, which down-regulated p53 expression, alleviated the degeneration. In an in vivo study, syndecan-4 siRNA injection stopped the development of rabbit disc degeneration, and even created a reverse effect, in which JNK/p53 played a role. Syndecan-4 may be a novel therapeutic target for intervertebral disc degeneration via suppressing the JNK/p53 pathway.
Collapse
Affiliation(s)
- Jun Ge
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaoqiang Cheng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Chenxi Yuan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiale Qian
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Chunshen Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Cheng Cao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Feng Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
32
|
Nikmanesh M, Cancel LM, Shi Z, Tarbell JM. Heparan sulfate proteoglycan, integrin, and syndecan‐4 are mechanosensors mediating cyclic strain‐modulated endothelial gene expression in mouse embryonic stem cell‐derived endothelial cells. Biotechnol Bioeng 2019; 116:2730-2741. [DOI: 10.1002/bit.27104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 02/18/2019] [Accepted: 06/26/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Maria Nikmanesh
- Department of Biomedical Engineering, The City College of New YorkCUNY New York New York
| | - Limary M. Cancel
- Department of Biomedical Engineering, The City College of New YorkCUNY New York New York
| | - Zhong‐Dong Shi
- Developmental Biology ProgramMemorial Sloan‐Kettering Cancer Center New York New York
| | - John M. Tarbell
- Department of Biomedical Engineering, The City College of New YorkCUNY New York New York
| |
Collapse
|
33
|
Role of the Endocytosis of Caveolae in Intracellular Signaling and Metabolism. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 57:203-234. [PMID: 30097777 DOI: 10.1007/978-3-319-96704-2_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Caveolae are 60-80 nm invaginated plasma membrane (PM) nanodomains, with a specific lipid and protein composition, which assist and regulate multiple processes in the plasma membrane-ranging from the organization of signalling complexes to the mechanical adaptation to changes in PM tension. However, since their initial descriptions, these structures have additionally been found tightly linked to internalization processes, mechanoadaptation, to the regulation of signalling events and of endosomal trafficking. Here, we review caveolae biology from this perspective, and its implications for cell physiology and disease.
Collapse
|
34
|
Moreno-Layseca P, Icha J, Hamidi H, Ivaska J. Integrin trafficking in cells and tissues. Nat Cell Biol 2019; 21:122-132. [PMID: 30602723 PMCID: PMC6597357 DOI: 10.1038/s41556-018-0223-z] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/25/2018] [Indexed: 12/28/2022]
Abstract
Cell adhesion to the extracellular matrix is fundamental to metazoan multicellularity and is accomplished primarily through the integrin family of cell-surface receptors. Integrins are internalized and enter the endocytic-exocytic pathway before being recycled back to the plasma membrane. The trafficking of this extensive protein family is regulated in multiple context-dependent ways to modulate integrin function in the cell. Here, we discuss recent advances in understanding the mechanisms and cellular roles of integrin endocytic trafficking.
Collapse
Affiliation(s)
- Paulina Moreno-Layseca
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jaroslav Icha
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
35
|
Furini G, Verderio EAM. Spotlight on the Transglutaminase 2-Heparan Sulfate Interaction. Med Sci (Basel) 2019; 7:E5. [PMID: 30621228 PMCID: PMC6359630 DOI: 10.3390/medsci7010005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs), syndecan-4 (Sdc4) especially, have been suggested as potential partners of transglutaminase-2 (TG2) in kidney and cardiac fibrosis, metastatic cancer, neurodegeneration and coeliac disease. The proposed role for HSPGs in the trafficking of TG2 at the cell surface and in the extracellular matrix (ECM) has been linked to the fibrogenic action of TG2 in experimental models of kidney fibrosis. As the TG2-HSPG interaction is largely mediated by the heparan sulfate (HS) chains of proteoglycans, in the past few years a number of studies have investigated the affinity of TG2 for HS, and the TG2 heparin binding site has been mapped with alternative outlooks. In this review, we aim to provide a compendium of the main literature available on the interaction of TG2 with HS, with reference to the pathological processes in which extracellular TG2 plays a role.
Collapse
Affiliation(s)
- Giulia Furini
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK.
| | - Elisabetta A M Verderio
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK.
- BiGeA, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
36
|
Fröhling M, Tepasse P, Intemann J, Sambale M, Sherwood J, Paruzel P, Tiemeyer NM, Nowacki TM, Brückner M, Mennigen R, Lügering A, Echtermeyer F, Pap T, Stratis A, Bettenworth D. Syndecan-4 Modulates Epithelial Gut Barrier Function and Epithelial Regeneration in Experimental Colitis. Inflamm Bowel Dis 2018; 24:2579-2589. [PMID: 30053064 DOI: 10.1093/ibd/izy248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND The transmembrane heparan sulfate proteoglycan Syndecan-4 (Sdc4) plays an important role in the regulation of various inflammatory disorders. However, the involvement of Sdc4 in intestinal inflammation remains unknown. Therefore, we assessed the impact of Sdc4 deficiency on experimental colitis and epithelial wound healing in vitro and in vivo. METHODS Dextran sulfate sodium (DSS)-induced colitis was monitored in wild type and Sdc4-deficient (Sdc4-/-) mice by assessment of body weight, histology, inflammatory cellular infiltration, and colon length. Syndecan-4 expression was measured by immunohistochemistry, Western blot, and quantitative real-time PCR. Epithelial permeability was evaluated by Evans blue measurements, Western blot, and immunohistological analysis of tight junction protein expression. Impact of Sdc4 on epithelial wound healing was determined by scratch assay in vitro and by colonoscopy following mechanical wounding in vivo. RESULTS In Sdc4-/- mice, colitis-like symptoms including severe weight loss, shortened colon length, histological damage, and invasion of macrophages and granulocytes were markedly aggravated compared with wild type (WT) animals. Moreover, colonic epithelial permeability in Sdc4-/- mice was enhanced, while tight junction protein expression decreased. Furthermore, Sdc4-/- colonic epithelial cells had lower cell proliferation and migration rates which presented in vivo as a prolonged intestinal wound healing phenotype. Strikingly, in WT animals, Sdc4 expression was reduced during colitis and was elevated during recovery. CONCLUSIONS The loss of Sdc4 aggravates the course of experimental colitis, potentially through impaired epithelial cell integrity and regeneration. In view of the development of current treatment approaches involving Sdc4 inhibition for inflammatory disorders like arthritis, particular caution should be taken in case of adverse gastrointestinal side-effects.
Collapse
Affiliation(s)
- Mareike Fröhling
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Phil Tepasse
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Johanna Intemann
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Meike Sambale
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Joanna Sherwood
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Peter Paruzel
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Nina-Marie Tiemeyer
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Tobias M Nowacki
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Markus Brückner
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| | - Rudolf Mennigen
- Department of General Surgery, University Hospital Münster, Münster, Germany
| | | | - Frank Echtermeyer
- Department of Anesthesiology and Intensive Care Medicine, Medical University Hannover, Hannover, Germany
| | - Thomas Pap
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Athanasios Stratis
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster, Germany
| |
Collapse
|
37
|
Abstract
Cell adhesion to components of the cellular microenvironment via cell-surface adhesion receptors controls many aspects of cell behavior in a range of physiological and pathological processes. Multimolecular complexes of scaffolding and signaling proteins are recruited to the intracellular domains of adhesion receptors such as integrins, and these adhesion complexes tether the cytoskeleton to the plasma membrane and compartmentalize cellular signaling events. Integrin adhesion complexes are highly dynamic, and their assembly is tightly regulated. Comprehensive, unbiased, quantitative analyses of the composition of different adhesion complexes over the course of their formation will enable better understanding of how the dynamics of adhesion protein recruitment influence the functions of adhesion complexes in fundamental cellular processes. Here, a pipeline is detailed integrating biochemical isolation of integrin adhesion complexes during a time course, quantitative proteomic analysis of isolated adhesion complexes, and computational analysis of temporal proteomic data. This approach enables the characterization of adhesion complex composition and dynamics during complex assembly.
Collapse
Affiliation(s)
- Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
38
|
Osmani N, Pontabry J, Comelles J, Fekonja N, Goetz JG, Riveline D, Georges-Labouesse E, Labouesse M. An Arf6- and caveolae-dependent pathway links hemidesmosome remodeling and mechanoresponse. Mol Biol Cell 2017; 29:435-451. [PMID: 29237817 PMCID: PMC6014169 DOI: 10.1091/mbc.e17-06-0356] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 01/08/2023] Open
Abstract
Hemidesmosomes are epithelial-specific cell-matrix adhesions stably anchoring the intracellular keratin network to the extracellular matrix and providing mechanical resilience to epithelia. The small GTPase Arf6 and caveolae are essential for their remodeling, notably in response to external mechanical cues. Hemidesmosomes (HDs) are epithelial-specific cell–matrix adhesions that stably anchor the intracellular keratin network to the extracellular matrix. Although their main role is to protect the epithelial sheet from external mechanical strain, how HDs respond to mechanical stress remains poorly understood. Here we identify a pathway essential for HD remodeling and outline its role with respect to α6β4 integrin recycling. We find that α6β4 integrin chains localize to the plasma membrane, caveolae, and ADP-ribosylation factor-6+ (Arf6+) endocytic compartments. Based on fluorescence recovery after photobleaching and endocytosis assays, integrin recycling between both sites requires the small GTPase Arf6 but neither caveolin1 (Cav1) nor Cavin1. Strikingly, when keratinocytes are stretched or hypo-osmotically shocked, α6β4 integrin accumulates at cell edges, whereas Cav1 disappears from it. This process, which is isotropic relative to the orientation of stretch, depends on Arf6, Cav1, and Cavin1. We propose that mechanically induced HD growth involves the isotropic flattening of caveolae (known for their mechanical buffering role) associated with integrin diffusion and turnover.
Collapse
Affiliation(s)
- Naël Osmani
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Julien Pontabry
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Jordi Comelles
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France.,Laboratory of Cell Physics, ISIS/IGBMC, CNRS UMR 7006, 67000 Strasbourg, France
| | - Nina Fekonja
- Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Jacky G Goetz
- Inserm U1109, MN3T, 67200 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Daniel Riveline
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France .,Université de Strasbourg, 67000 Strasbourg, France.,Laboratory of Cell Physics, ISIS/IGBMC, CNRS UMR 7006, 67000 Strasbourg, France
| | - Elisabeth Georges-Labouesse
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Michel Labouesse
- IGBMC, Development and Stem Cells Program, CNRS (UMR 7104)/INSERM (U964), 67400 Illkirch, France .,Université de Strasbourg, 67000 Strasbourg, France.,UMR7622-CNRS, IBPS, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
39
|
Keller-Pinter A, Ughy B, Domoki M, Pettko-Szandtner A, Letoha T, Tovari J, Timar J, Szilak L. The phosphomimetic mutation of syndecan-4 binds and inhibits Tiam1 modulating Rac1 activity in PDZ interaction-dependent manner. PLoS One 2017; 12:e0187094. [PMID: 29121646 PMCID: PMC5679609 DOI: 10.1371/journal.pone.0187094] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/15/2017] [Indexed: 11/19/2022] Open
Abstract
The small GTPases of the Rho family comprising RhoA, Rac1 and Cdc42 function as molecular switches controlling several essential biochemical pathways in eukaryotic cells. Their activity is cycling between an active GTP-bound and an inactive GDP-bound conformation. The exchange of GDP to GTP is catalyzed by guanine nucleotide exchange factors (GEFs). Here we report a novel regulatory mechanism of Rac1 activity, which is controlled by a phosphomimetic (Ser179Glu) mutant of syndecan-4 (SDC4). SDC4 is a ubiquitously expressed transmembrane, heparan sulfate proteoglycan. In this study we show that the Ser179Glu mutant binds strongly Tiam1, a Rac1-GEF reducing Rac1-GTP by 3-fold in MCF-7 breast adenocarcinoma cells. Mutational analysis unravels the PDZ interaction between SDC4 and Tiam1 is indispensable for the suppression of the Rac1 activity. Neither of the SDC4 interactions is effective alone to block the Rac1 activity, on the contrary, lack of either of interactions can increase the activity of Rac1, therefore the Rac1 activity is the resultant of the inhibitory and stimulatory effects. In addition, SDC4 can bind and tether RhoGDI1 (GDP-dissociation inhibitor 1) to the membrane. Expression of the phosphomimetic SDC4 results in the accumulation of the Rac1–RhoGDI1 complex. Co-immunoprecipitation assays (co-IP-s) reveal that SDC4 can form complexes with RhoGDI1. Together, the regulation of the basal activity of Rac1 is fine tuned and SDC4 is implicated in multiple ways.
Collapse
Affiliation(s)
- Aniko Keller-Pinter
- Department of Biochemistry, Faculty of General Medicine, University of Szeged, Szeged, Hungary
| | - Bettina Ughy
- Institute of Plant Biology, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Monika Domoki
- Institute of Plant Biology, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Aladar Pettko-Szandtner
- Institute of Plant Biology, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | | | - Jozsef Tovari
- Department of the Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Jozsef Timar
- II. Department of Pathology, Semmelweis University; MTA-SE Molecular Oncology Research Group, Budapest, Hungary
| | - Laszlo Szilak
- Institute of Biology, Savaria Campus, Eötvös Lorand University, Szombathely, Hungary
- Szilak Laboratories Bioinformatics and Molecule-Design Ltd., Szeged, Hungary
- * E-mail:
| |
Collapse
|
40
|
Fibronectin promotes directional persistence in fibroblast migration through interactions with both its cell-binding and heparin-binding domains. Sci Rep 2017. [PMID: 28623309 PMCID: PMC5473823 DOI: 10.1038/s41598-017-03701-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The precise mechanisms through which insoluble, cell-adhesive ligands induce and regulate directional cell migration remain obscure. We recently demonstrated that elevated surface density of physically adsorbed plasma fibronectin (FN) promotes high directional persistence in fibroblast migration. While cell-FN association through integrins α5β1 and αvβ3 was necessary, substrates that selectively engaged these integrins did not support the phenotype. We here show that high directional persistence necessitates a combination of the cell-binding and C-terminal heparin-binding domains of FN, but does not require the engagement of syndecan-4 or integrin α4β1. FN treatment with various fixation agents indicated that associated changes in fibroblast motility were due to biochemical changes, rather than alterations in its physical state. The nature of the coating determined the ability of fibroblasts to assemble endogenous or exogenous FN, while FN fibrillogenesis played a minor, but significant, role in regulating directionality. Interestingly, knockdown of cellular FN abolished cell motility altogether, demonstrating a requirement for intracellular processes in enabling fibroblast migration on FN. Lastly, kinase inhibition experiments revealed that regulation of cell speed and directional persistence are decoupled. Hence, we have identified factors that render full-length FN a promoter of directional migration and discuss the possible, relevant mechanisms.
Collapse
|
41
|
Osawa S, Kurachi M, Yamamoto H, Yoshimoto Y, Ishizaki Y. Fibronectin on extracellular vesicles from microvascular endothelial cells is involved in the vesicle uptake into oligodendrocyte precursor cells. Biochem Biophys Res Commun 2017; 488:232-238. [PMID: 28499870 DOI: 10.1016/j.bbrc.2017.05.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 11/25/2022]
Abstract
We previously reported transplantation of brain microvascular endothelial cells (MVECs) into cerebral white matter infarction model improved the animal's behavioral outcome by increasing the number of oligodendrocyte precursor cells (OPCs). We also revealed extracellular vesicles (EVs) derived from MVECs promoted survival and proliferation of OPCs in vitro. In this study, we investigated the mechanism how EVs derived from MVECs contribute to OPC survival and proliferation. Protein mass spectrometry and enzyme-linked immunosorbent assay revealed fibronectin was abundant on the surface of EVs from MVECs. As fibronectin has been reported to promote OPC survival and proliferation via integrin signaling pathway, we blocked the binding between fibronectin and integrins using RGD sequence mimics. Blocking the binding, however, did not attenuate the survival and proliferation promoting effect of EVs on OPCs. Flow cytometric and imaging analyses revealed fibronectin on EVs mediates their internalization into OPCs by its binding to heparan sulfate proteoglycan on OPCs. OPC survival and proliferation promoted by EVs were attenuated by blocking the internalization of EVs into OPCs. These lines of evidence suggest that fibronectin on EVs mediates their internalization into OPCs, and the cargo of EVs promotes survival and proliferation of OPCs, independent of integrin signaling pathway.
Collapse
Affiliation(s)
- Sho Osawa
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hanako Yamamoto
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
42
|
Valdivia A, Goicoechea SM, Awadia S, Zinn A, Garcia-Mata R. Regulation of circular dorsal ruffles, macropinocytosis, and cell migration by RhoG and its exchange factor, Trio. Mol Biol Cell 2017; 28:1768-1781. [PMID: 28468978 PMCID: PMC5491185 DOI: 10.1091/mbc.e16-06-0412] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 11/11/2022] Open
Abstract
The small GTPase RhoG and its exchange factor, Trio, regulate the formation and size of circular dorsal ruffles and associated functions, including macropinocytosis and cell migration. Circular dorsal ruffles (CDRs) are actin-rich structures that form on the dorsal surface of many mammalian cells in response to growth factor stimulation. CDRs represent a unique type of structure that forms transiently and only once upon stimulation. The formation of CDRs involves a drastic rearrangement of the cytoskeleton, which is regulated by the Rho family of GTPases. So far, only Rac1 has been consistently associated with CDR formation, whereas the role of other GTPases in this process is either lacking or inconclusive. Here we show that RhoG and its exchange factor, Trio, play a role in the regulation of CDR dynamics, particularly by modulating their size. RhoG is activated by Trio downstream of PDGF in a PI3K- and Src-dependent manner. Silencing RhoG expression decreases the number of cells that form CDRs, as well as the area of the CDRs. The regulation of CDR area by RhoG is independent of Rac1 function. In addition, our results show the RhoG plays a role in the cellular functions associated with CDR formation, including macropinocytosis, receptor internalization, and cell migration. Taken together, our results reveal a novel role for RhoG in the regulation of CDRs and the cellular processes associated with their formation.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606.,Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322
| | | | - Sahezeel Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Ashtyn Zinn
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| |
Collapse
|
43
|
A Key Regulator of Cell Adhesion: Identification and Characterization of Important N-Glycosylation Sites on Integrin α5 for Cell Migration. Mol Cell Biol 2017; 37:MCB.00558-16. [PMID: 28167607 DOI: 10.1128/mcb.00558-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/29/2017] [Indexed: 11/20/2022] Open
Abstract
The N-glycosylation of integrin α5β1 is thought to control many fundamental aspects of cell behavior, including cell adhesion and migration. However, the mechanism of how N-glycans function remains largely obscure. Here, we used a loss-of-function approach. Wild-type (WT) integrin α5 and N-glycosylation mutant S3-5 (sites 3 to 5) integrin α5, which contains fewer N-glycans, were stably reconstituted in α5 knockout cancer cells. We found that the migration ability of S3-5 cells was decreased in comparison with that of the WT. Interestingly, the levels of phosphorylated focal adhesion kinase and actin stress fiber formation were greatly enhanced in the S3-5 mutant. In a mechanistic manner, the internalization of active but not total integrin α5β1 was inhibited in S3-5 cells, which is a process that is related to the enhanced expression of active integrin α5β1 on the cell surface. Importantly, restoration of N-glycosylation on the β-propeller domain of α5 reinstated the cell migration ability, active α5β1 expression, and internalization. Moreover, these N-glycans are critical for α5-syndecan-4 complex formation. These findings indicate that N-glycosylation on the β-propeller domain functions as a molecular switch to control the dynamics of α5β1 on the cell surface that in turn is required for optimum adhesion for cell migration.
Collapse
|
44
|
Kang H, Liu J, Sun A, Liu X, Fan Y, Deng X. Vascular smooth muscle cell glycocalyx mediates shear stress-induced contractile responses via a Rho kinase (ROCK)-myosin light chain phosphatase (MLCP) pathway. Sci Rep 2017; 7:42092. [PMID: 28191820 PMCID: PMC5304191 DOI: 10.1038/srep42092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/04/2017] [Indexed: 11/09/2022] Open
Abstract
The vascular smooth muscle cells (VSMCs) are exposed to interstitial flow induced shear stress that may be sensed by the surface glycocalyx, a surface layer composed primarily of proteoglycans and glycoproteins, to mediate cell contraction during the myogenic response. We, therefore, attempted to elucidate the signal pathway of the glycocalyx mechanotransduction in shear stress regulated SMC contraction. Human umbilical vein SMCs (HUVSMCs) deprived of serum for 3–4 days were exposed to a step increase (0 to 20 dyn/cm2) in shear stress in a parallel plate flow chamber, and reduction in the cell area was quantified as contraction. The expressions of Rho kinase (ROCK) and its downstream signal molecules, the myosin-binding subunit of myosin phosphatase (MYPT) and the myosin light chain 2 (MLC2), were evaluated. Results showed that the exposure of HUVSMCs to shear stress for 30 min induced cell contraction significantly, which was accompanied by ROCK1 up-regulation, re-distribution, as well as MYPT1 and MLC activation. However, these shear induced phenomenon could be completely abolished by heparinase III or Y-27632 pre-treatment. These results indicate shear stress induced VSMC contraction was mediated by cell surface glycocalyx via a ROCK-MLC phosphatase (MLCP) pathway, providing evidence of the glycocalyx mechanotransduction in myogenic response.
Collapse
Affiliation(s)
- Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Jiajia Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Anqiang Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Xiao Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| | - Xiaoyan Deng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191 China
| |
Collapse
|
45
|
Russell-Puleri S, Dela Paz NG, Adams D, Chattopadhyay M, Cancel L, Ebong E, Orr AW, Frangos JA, Tarbell JM. Fluid shear stress induces upregulation of COX-2 and PGI 2 release in endothelial cells via a pathway involving PECAM-1, PI3K, FAK, and p38. Am J Physiol Heart Circ Physiol 2016; 312:H485-H500. [PMID: 28011582 PMCID: PMC5402016 DOI: 10.1152/ajpheart.00035.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 12/16/2022]
Abstract
Vascular endothelial cells play an important role in the regulation of vascular function in response to mechanical stimuli in both healthy and diseased states. Prostaglandin I2 (PGI2) is an important antiatherogenic prostanoid and vasodilator produced in endothelial cells through the action of the cyclooxygenase (COX) isoenzymes COX-1 and COX-2. However, the mechanisms involved in sustained, shear-induced production of COX-2 and PGI2 have not been elucidated but are determined in the present study. We used cultured endothelial cells exposed to steady fluid shear stress (FSS) of 10 dyn/cm2 for 5 h to examine shear stress-induced induction of COX-2/PGI2 Our results demonstrate the relationship between the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1) and the intracellular mechanoresponsive molecules phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK), and mitogen-activated protein kinase p38 in the FSS induction of COX-2 expression and PGI2 release. Knockdown of PECAM-1 (small interference RNA) expression inhibited FSS-induced activation of α5β1-integrin, upregulation of COX-2, and release of PGI2 in both bovine aortic endothelial cells (BAECs) and human umbilical vein endothelial cells (HUVECs). Furthermore, inhibition of the PI3K pathway (LY294002) substantially inhibited FSS activation of α5β1-integrin, upregulation of COX-2 gene and protein expression, and release of PGI2 in BAECs. Inhibition of integrin-associated FAK (PF573228) and MAPK p38 (SB203580) also inhibited the shear-induced upregulation of COX-2. Finally, a PECAM-1-/- mouse model was characterized by reduced COX-2 immunostaining in the aorta and reduced plasma PGI2 levels compared with wild-type mice, as well as complete inhibition of acute flow-induced PGI2 release compared with wild-type animals.NEW & NOTEWORTHY In this study we determined the major mechanotransduction pathway by which blood flow-driven shear stress activates cyclooxygenase-2 (COX-2) and prostaglandin I2 (PGI2) release in endothelial cells. Our work has demonstrated for the first time that COX-2/PGI2 mechanotransduction is mediated by the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1).
Collapse
Affiliation(s)
| | | | - Diana Adams
- La Jolla Bioengineering Institute, La Jolla, California
| | | | - Limary Cancel
- Department of Biomedical Engineering, City College of New York, New York, New York
| | - Eno Ebong
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts; and
| | - A Wayne Orr
- Department of Pathology, Louisiana State University, Shreveport, Louisiana
| | | | - John M Tarbell
- Department of Biomedical Engineering, City College of New York, New York, New York;
| |
Collapse
|
46
|
Xiong N, Li S, Tang K, Bai H, Peng Y, Yang H, Wu C, Liu Y. Involvement of caveolin-1 in low shear stress-induced breast cancer cell motility and adhesion: Roles of FAK/Src and ROCK/p-MLC pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:12-22. [PMID: 27773611 DOI: 10.1016/j.bbamcr.2016.10.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/07/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
Abstract
Tumor cells translocating to distant sites are subjected to hemodynamic shear forces during their passage in the blood vessels. Low shear stress (LSS) plays a critical role in the regulation of various aspects of tumor cells functions, including motility and adhesion. Beyond its structural role, caveolin-1 (Cav-1), the important component of caveolae, represents a modulator of several cancer-associated functions as tumor progression and metastasis. However, the role of Cav-1 in regulating tumor cells response to shear stress remains poorly explored. Here, we characterized the role of LSS and Cav-1 in mediating cell motility and adhesion on human breast carcinoma MDA-MB-231 cells. We first showed that LSS exposure promoted cell polarity and focal adhesion (FA) dynamics, thus indicating elevated cell migration. Silencing of Cav-1 leaded to a significantly lower formation of stress fibers. However, LSS exposure was able to rescue it via the alteration of actin-associated proteins expression, including ROCK, p-MLC, cofilin and filamin A. Time-lapse migration assay indicated that Cav-1 expression fostered MDA-MB-231 cells motility and LSS triggered cells to rapidly generate new lamellipodia. Furthermore, Cav-1 and LSS significantly influenced cell adhesion. Taken together, our findings provide insights into mechanisms underlying LSS triggered events mediated by downstream Cav-1, including FAK/Src and ROCK/p-MLC pathways, involved in the reorganization of the cytoskeleton, cell motility, FA dynamics and breast cancer cell adhesion.
Collapse
Affiliation(s)
- Niya Xiong
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Kai Tang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hongxia Bai
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yueting Peng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| |
Collapse
|
47
|
Abstract
Integrins are a family of heterodimeric receptors that bind to components of the extracellular matrix and influence cellular processes as varied as proliferation and migration. These effects are achieved by tight spatiotemporal control over intracellular signalling pathways, including those that mediate cytoskeletal reorganisation. The ability of integrins to bind to ligands is governed by integrin conformation, or activity, and this is widely acknowledged to be an important route to the regulation of integrin function. Over the last 15 years, however, the pathways that regulate endocytosis and recycling of integrins have emerged as major players in controlling integrin action, and studying integrin trafficking has revealed fresh insight into the function of this fascinating class of extracellular matrix receptors, in particular in the context of cell migration and invasion. Here, we review our current understanding of the contribution of integrin trafficking to cell motility.
Collapse
Affiliation(s)
- Nikki R Paul
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, M13 9PT, UK
| | - Guillaume Jacquemet
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, M13 9PT, UK
| | - Patrick T Caswell
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, M13 9PT, UK.
| |
Collapse
|
48
|
Das S, Majid M, Baker AB. Syndecan-4 enhances PDGF-BB activity in diabetic wound healing. Acta Biomater 2016; 42:56-65. [PMID: 27381525 DOI: 10.1016/j.actbio.2016.07.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 01/13/2023]
Abstract
UNLABELLED Non-healing ulcers are a common consequence of long-term diabetes and severe peripheral vascular disease. These non-healing wounds are a major source of morbidity in patients with diabetes and place a heavy financial burden on the healthcare system. Growth factor therapies are an attractive strategy for enhancing wound closure in non-healing wounds but have only achieved mixed results in clinical trials. Platelet derived growth factor-BB (PDGF-BB) is the only currently approved growth factor therapy for non-healing wounds. However, PDGF-BB therapy is not effective in many patients and requires high doses that increase the potential for side effects. In this work, we demonstrate that syndecan-4 delivered in a proteoliposomal formulation enhances PDGF-BB activity in diabetic wound healing. In particular, syndecan-4 proteoliposomes enhance the migration of keratinocytes derived from patients with diabetes. In addition, syndecan-4 proteoliposomes sensitize keratinocytes to PDGF-BB stimulation, enhancing the intracellular signaling response to PDGF-BB. We further demonstrated that co-therapy with syndecan-4 proteoliposomes enhanced wound closure in diabetic, hyperlipidemic ob/ob mice. Wounds treated with both syndecan-4 proteoliposomes and PDGF-BB had increased re-epithelization and angiogenesis in comparison to wounds treated with PDGF-BB alone. Moreover, the wounds treated with syndecan-4 proteoliposomes and PDGF-BB also had increased M2 macrophages and reduced M1 macrophages, suggesting syndecan-4 delivery induces immunomodulation within the healing wounds. Together our findings support that syndecan-4 proteoliposomes markedly improve PDGF-BB efficacy for wound healing and may be useful in enhancing treatments for non-healing wounds. STATEMENT OF SIGNIFICANCE Non-healing wounds are major healthcare issue for patients with diabetes and peripheral vascular disease. Growth factor therapies have potential for healing chronic wounds but have not been effective for many patients. PDGF-BB is currently the only approved growth factor for enhancing wound healing. However, it has not seen widespread adoption due to limited efficacy and high cost. In this work, we have developed an enhancing agent that improves the activity of PDGF-BB in promoting wound healing in animals with diabetes. This co-therapy may be useful in improving the efficacy of PDGFBB and enhance its safety through lowering the dose of growth factor needed to improve wound healing.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States
| | - Marjan Majid
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, United States; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
49
|
Das S, Singh G, Majid M, Sherman MB, Mukhopadhyay S, Wright CS, Martin PE, Dunn AK, Baker AB. Syndesome Therapeutics for Enhancing Diabetic Wound Healing. Adv Healthc Mater 2016; 5:2248-60. [PMID: 27385307 PMCID: PMC5228475 DOI: 10.1002/adhm.201600285] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/24/2016] [Indexed: 12/19/2022]
Abstract
Chronic wounds represent a major healthcare and economic problem worldwide. Advanced wound dressings that incorporate bioactive compounds have great potential for improving outcomes in patients with chronic wounds but significant challenges in designing treatments that are effective in long-standing, nonhealing wounds. Here, an optimized wound healing gel was developed that delivers syndecan-4 proteoliposomes ("syndesomes") with fibroblast growth factor-2 (FGF-2) to enhance diabetic wound healing. In vitro studies demonstrate that syndesomes markedly increase migration of keratinocytes and fibroblasts isolated from both nondiabetic and diabetic donors. In addition, syndesome treatment leads to increased endocytic processing of FGF-2 that includes enhanced recycling of FGF-2 to the cell surface after uptake. The optimized syndesome formulation was incorporated into an alginate wound dressing and tested in a splinted wound model in diabetic, ob/ob mice. It was found that wounds treated with syndesomes and FGF-2 have markedly enhanced wound closure in comparison to wounds treated with only FGF-2. Moreover, syndesomes have an immunomodulatory effect on wound macrophages, leading to a shift toward the M2 macrophage phenotype and alterations in the wound cytokine profile. Together, these studies show that delivery of exogenous syndecan-4 is an effective method for enhancing wound healing in the long-term diabetic diseased state.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78731, USA
| | - Gunjan Singh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78731, USA
| | - Marjan Majid
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78731, USA
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, 78731, USA
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, 78731, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78731, USA
| | - Catherine S Wright
- Diabetes Research Group, Department of Life Sciences and Institute for Applied Health Research, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Patricia E Martin
- Diabetes Research Group, Department of Life Sciences and Institute for Applied Health Research, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Andrew K Dunn
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78731, USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78731, USA.
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78731, USA.
- The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, 78731, USA.
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, 78731, USA.
| |
Collapse
|
50
|
Alvarez A, Lagos-Cabré R, Kong M, Cárdenas A, Burgos-Bravo F, Schneider P, Quest AFG, Leyton L. Integrin-mediated transactivation of P2X7R via hemichannel-dependent ATP release stimulates astrocyte migration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2175-88. [PMID: 27235833 DOI: 10.1016/j.bbamcr.2016.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/29/2016] [Accepted: 05/23/2016] [Indexed: 01/09/2023]
Abstract
Our previous reports indicate that ligand-induced αVβ3 integrin and Syndecan-4 engagement increases focal adhesion formation and migration of astrocytes. Additionally, ligated integrins trigger ATP release through unknown mechanisms, activating P2X7 receptors (P2X7R), and the uptake of Ca(2+) to promote cell adhesion. However, whether the activation of P2X7R and ATP release are required for astrocyte migration and whether αVβ3 integrin and Syndecan-4 receptors communicate with P2X7R via ATP remains unknown. Here, cells were stimulated with Thy-1, a reported αVβ3 integrin and Syndecan-4 ligand. Results obtained indicate that ATP was released by Thy-1 upon integrin engagement and required the participation of phosphatidylinositol-3-kinase (PI3K), phospholipase-C gamma (PLCγ) and inositol trisphosphate (IP3) receptors (IP3R). IP3R activation leads to increased intracellular Ca(2+), hemichannel (Connexin-43 and Pannexin-1) opening, and ATP release. Moreover, silencing of the P2X7R or addition of hemichannel blockers precluded Thy-1-induced astrocyte migration. Finally, Thy-1 lacking the integrin-binding site did not stimulate ATP release, whereas Thy-1 mutated in the Syndecan-4-binding domain increased ATP release, albeit to a lesser extent and with delayed kinetics compared to wild-type Thy-1. Thus, hemichannels activated downstream of an αVβ3 integrin-PI3K-PLCγ-IP3R pathway are responsible for Thy-1-induced, hemichannel-mediated and Syndecan-4-modulated ATP release that transactivates P2X7Rs to induce Ca(2+) entry. These findings uncover a hitherto unrecognized role for hemichannels in the regulation of astrocyte migration via P2X7R transactivation induced by integrin-mediated ATP release.
Collapse
Affiliation(s)
- Alvaro Alvarez
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Raúl Lagos-Cabré
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Milene Kong
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Areli Cárdenas
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Francesca Burgos-Bravo
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Andrew F G Quest
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Advanced Center for Chronic Diseases, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Advanced Center for Chronic Diseases, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile; Biomedical Neuroscience Institute, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453 Santiago, Chile.
| |
Collapse
|