1
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2024:S1534-5807(24)00545-8. [PMID: 39413783 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
2
|
Rekad Z, Ruff M, Radwanska A, Grall D, Ciais D, Van Obberghen-Schilling E. Coalescent RNA-localizing and transcriptional activities of SAM68 modulate adhesion and subendothelial basement membrane assembly. eLife 2023; 12:e85165. [PMID: 37585334 PMCID: PMC10431919 DOI: 10.7554/elife.85165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
Endothelial cell interactions with their extracellular matrix are essential for vascular homeostasis and expansion. Large-scale proteomic analyses aimed at identifying components of integrin adhesion complexes have revealed the presence of several RNA binding proteins (RBPs) of which the functions at these sites remain poorly understood. Here, we explored the role of the RBP SAM68 (Src associated in mitosis, of 68 kDa) in endothelial cells. We found that SAM68 is transiently localized at the edge of spreading cells where it participates in membrane protrusive activity and the conversion of nascent adhesions to mechanically loaded focal adhesions by modulation of integrin signaling and local delivery of β-actin mRNA. Furthermore, SAM68 depletion impacts cell-matrix interactions and motility through induction of key matrix genes involved in vascular matrix assembly. In a 3D environment SAM68-dependent functions in both tip and stalk cells contribute to the process of sprouting angiogenesis. Altogether, our results identify the RBP SAM68 as a novel actor in the dynamic regulation of blood vessel networks.
Collapse
Affiliation(s)
- Zeinab Rekad
- Université Côte d'Azur, CNRS, INSERM, iBVNiceFrance
| | - Michaël Ruff
- Université Côte d'Azur, CNRS, INSERM, iBVNiceFrance
| | | | | | | | | |
Collapse
|
3
|
Zhang Z, Liu C, Wang M, Sun R, Yang Z, Hua Z, Wu Y, Wu M, Wang H, Qiu W, Yin H, Yang M. Treating solid tumors with TCR-based chimeric antigen receptor targeting extra domain B-containing fibronectin. J Immunother Cancer 2023; 11:e007199. [PMID: 37586774 PMCID: PMC10432677 DOI: 10.1136/jitc-2023-007199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND The suppression of chimeric antigen receptor (CAR) T cells by the tumor microenvironment (TME) is a crucial obstacle in the T-cell-based treatment of solid tumors. Extra domain B (EDB)-fibronectin is an oncofetal antigen expressed on the endothelium layer of the neovasculature and cancer cells. Though recognized as a T cell therapy target, engineered CAR T cells thus far have failed to demonstrate satisfactory in vivo efficacy. In this study, we report that targeting EDB-fibronectin by redirected TCR-CAR T cells (rTCR-CAR) bypasses the suppressive TME for solid tumor treatment and sufficiently suppressed tumor growth.We generated EDB-targeting CAR by fusing single-chain variable fragment to CD3ε, resulting in rTCR-CAR. Human primary T cells and Jurkat cells were used to study the EDB-targeting T cells. Differences to the traditional second-generation CAR T cell in signaling, immune synapse formation, and T cell exhaustion were characterized. Cytotoxicity of the rTCR-CAR T cells was tested in vitro, and therapeutic efficacies were demonstrated using xenograft models. METHODS RESULTS: In the xenograft models, the rTCR-CAR T cells demonstrated in vivo efficacies superior to that based on traditional CAR design. A significant reduction in tumor vessel density was observed alongside tumor growth inhibition, extending even to tumor models established with EDB-negative cancer cells. The rTCR-CAR bound to immobilized EDB, and the binding led to immune synapse structures superior to that formed by second-generation CARs. By a mechanism similar to that for the conventional TCR complex, EDB-fibronectin activated the rTCR-CAR, resulting in rTCR-CAR T cells with low basal activation levels and increased in vivo expansion. CONCLUSION Our study has demonstrated the potential of rTCR-CAR T cells targeting the EDB-fibronectin as an anticancer therapeutic. Engineered to possess antiangiogenic and cytotoxic activities, the rTCR-CAR T cells showed therapeutic efficacies not impacted by the suppressive TMEs. These combined characteristics of a single therapeutic agent point to its potential to achieve sustained control of solid tumors.
Collapse
Affiliation(s)
- Zhijie Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chang Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Muhan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rongcheng Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Zhe Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhen Hua
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yushuang Wu
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Mengting Wu
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| | - Hang Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen Qiu
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meijia Yang
- Jiangsu Cell Tech Medical Research Institute, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Lickert S, Kenny M, Selcuk K, Mehl JL, Bender M, Früh SM, Burkhardt MA, Studt JD, Nieswandt B, Schoen I, Vogel V. Platelets drive fibronectin fibrillogenesis using integrin αIIbβ3. SCIENCE ADVANCES 2022; 8:eabj8331. [PMID: 35275711 PMCID: PMC8916723 DOI: 10.1126/sciadv.abj8331] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Platelets interact with multiple adhesion proteins during thrombogenesis, yet little is known about their ability to assemble fibronectin matrix. In vitro three-dimensional superresolution microscopy complemented by biophysical and biochemical methods revealed fundamental insights into how platelet contractility drives fibronectin fibrillogenesis. Platelets adhering to thrombus proteins (fibronectin and fibrin) versus basement membrane components (laminin and collagen IV) pull fibronectin fibrils along their apical membrane versus underneath their basal membrane, respectively. In contrast to other cell types, platelets assemble fibronectin nanofibrils using αIIbβ3 rather than α5β1 integrins. Apical fibrillogenesis correlated with a stronger activation of integrin-linked kinase, higher platelet traction forces, and a larger tension in fibrillar-like adhesions compared to basal fibrillogenesis. Our findings have potential implications for how mechanical thrombus integrity might be maintained during remodeling and vascular repair.
Collapse
Affiliation(s)
- Sebastian Lickert
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Martin Kenny
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Kateryna Selcuk
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Johanna L. Mehl
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Markus Bender
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Susanna M. Früh
- Hahn-Schickard, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
- Laboratory for MEMS Applications, IMTEK Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | - Melanie A. Burkhardt
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Jan-Dirk Studt
- Division of Hematology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine – Chair I, University Hospital, and Rudolf Virchow Center, Julius Maximilian University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
- Corresponding author. (V.V.); (I.S.)
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
- Corresponding author. (V.V.); (I.S.)
| |
Collapse
|
5
|
Sun R, Kim AH. The multifaceted mechanisms of malignant glioblastoma progression and clinical implications. Cancer Metastasis Rev 2022; 41:871-898. [PMID: 35920986 PMCID: PMC9758111 DOI: 10.1007/s10555-022-10051-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023]
Abstract
With the application of high throughput sequencing technologies at single-cell resolution, studies of the tumor microenvironment in glioblastoma, one of the most aggressive and invasive of all cancers, have revealed immense cellular and tissue heterogeneity. A unique extracellular scaffold system adapts to and supports progressive infiltration and migration of tumor cells, which is characterized by altered composition, effector delivery, and mechanical properties. The spatiotemporal interactions between malignant and immune cells generate an immunosuppressive microenvironment, contributing to the failure of effective anti-tumor immune attack. Among the heterogeneous tumor cell subpopulations of glioblastoma, glioma stem cells (GSCs), which exhibit tumorigenic properties and strong invasive capacity, are critical for tumor growth and are believed to contribute to therapeutic resistance and tumor recurrence. Here we discuss the role of extracellular matrix and immune cell populations, major components of the tumor ecosystem in glioblastoma, as well as signaling pathways that regulate GSC maintenance and invasion. We also highlight emerging advances in therapeutic targeting of these components.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Albert H. Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110 USA ,The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
6
|
Mechanical Aspects of Angiogenesis. Cancers (Basel) 2021; 13:cancers13194987. [PMID: 34638470 PMCID: PMC8508205 DOI: 10.3390/cancers13194987] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The formation of new blood vessels from already existing ones is a process of high clinical relevance, since it is of great importance for both physiological and pathological processes. In regard to tumors, the process is crucial, since it ensures the supply with nutrients and the growth of the tumor. The influence of mechanical factors on this biological process is an emerging field. Until now, the shear force of the blood flow has been considered the main mechanical parameter during angiogenesis. This review article provides an overview of further mechanical cues, with particular focus on the surrounding extracellular matrix impacting the cell behavior and, thus, regulating angiogenesis. This underlines the enormous importance of the mechanical properties of the extracellular matrix on cell biological processes and shows how changing the mechanics of the extracellular matrix could be used as a possible therapeutic approach in cancer therapy. Abstract Angiogenesis is of high clinical relevance as it plays a crucial role in physiological (e.g., tissue regeneration) and pathological processes (e.g., tumor growth). Besides chemical signals, such as VEGF, the relationship between cells and the extracellular matrix (ECM) can influence endothelial cell behavior during angiogenesis. Previously, in terms of the connection between angiogenesis and mechanical factors, researchers have focused on shear forces due to blood flow. However, it is becoming increasingly important to include the direct influence of the ECM on biological processes, such as angiogenesis. In this context, we focus on the stiffness of the surrounding ECM and the adhesion of cells to the ECM. Furthermore, we highlight the mechanical cues during the main stages of angiogenesis: cell migration, tip and stalk cells, and vessel stabilization. It becomes clear that the different stages of angiogenesis require various chemical and mechanical cues to be modulated by/modulate the stiffness of the ECM. Thus, changes of the ECM during tumor growth represent additional potential dysregulations of angiogenesis in addition to erroneous biochemical signals. This awareness could be the basis of therapeutic approaches to counteract specific processes in tumor angiogenesis.
Collapse
|
7
|
Liao KC, Chuo V, Fagg WS, Modahl CM, Widen S, Garcia-Blanco MA. The RNA binding protein Quaking represses splicing of the Fibronectin EDA exon and downregulates the interferon response. Nucleic Acids Res 2021; 49:10034-10045. [PMID: 34428287 PMCID: PMC8464043 DOI: 10.1093/nar/gkab732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023] Open
Abstract
Quaking (QKI) controls RNA metabolism in many biological processes including innate immunity, where its roles remain incompletely understood. To illuminate these roles, we performed genome scale transcriptome profiling in QKI knockout cells with or without poly(I:C) transfection, a double-stranded RNA analog that mimics viral infection. Analysis of RNA-sequencing data shows that QKI knockout upregulates genes induced by interferons, suggesting that QKI is an immune suppressor. Furthermore, differential splicing analysis shows that QKI primarily controls cassette exons, and among these events, we noted that QKI silences splicing of the extra domain A (EDA) exon in fibronectin (FN1) transcripts. QKI knockout results in elevated production and secretion of FN1-EDA protein, which is a known activator of interferons. Consistent with an upregulation of the interferon response in QKI knockout cells, our results show reduced production of dengue virus-2 and Japanese encephalitis virus in these cells. In conclusion, we demonstrate that QKI downregulates the interferon system and attenuates the antiviral state.
Collapse
Affiliation(s)
- Kuo-Chieh Liao
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Vanessa Chuo
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - W Samuel Fagg
- Transplant Division, Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cassandra M Modahl
- Department of Biological Sciences, National University of Singapore, Singapore 119077, Singapore
| | - Steven Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mariano A Garcia-Blanco
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
8
|
Han HJ, Sung JY, Kim SH, Yun UJ, Kim H, Jang EJ, Yoo HE, Hong EK, Goh SH, Moon A, Lee JS, Ye SK, Shim J, Kim YN. Fibronectin regulates anoikis resistance via cell aggregate formation. Cancer Lett 2021; 508:59-72. [PMID: 33771684 DOI: 10.1016/j.canlet.2021.03.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
The loss of cell-matrix interactions induces apoptosis, known as anoikis. For successful distant metastasis, circulating tumor cells (CTCs) that have lost matrix attachment need to acquire anoikis resistance in order to survive. Cell aggregate formation confers anoikis resistance, and CTC clusters are more highly metastatic compared to single cells; however, the molecular mechanisms underlying this aggregation are not well understood. In this study, we demonstrated that cell detachment increased cell aggregation and upregulated fibronectin (FN) levels in lung and breast cancer cells, but not in their normal counterparts. FN knockdown decreased cell aggregation and increased anoikis. In addition, cell detachment induced cell-cell adhesion proteins, including E-cadherin, desmoglein-2, desmocollin-2/3, and plakoglobin. Interestingly, FN knockdown decreased the levels of desmoglein-2, desmocollin-2/3, and plakoglobin, but not E-cadherin, suggesting the involvement of desmosomal junction in cell aggregation. Accordingly, knockdown of desmoglein-2, desmocollin-2, or plakoglobin reduced cell aggregation and increased cell sensitivity to anoikis. Previously, we reported that NADPH oxidase 4 (Nox4) upregulation is important for anoikis resistance. Nox4 inhibition by siRNA or apocynin decreased cell aggregation and increased anoikis with the downregulation of FN, and, consequently, decreased desmoglein-2, desmocollin-2/3, or plakoglobin. The coexpression of Nox4 and FN was found to be significant in lung and breast cancer patients, based on cBioPortal data. In vivo mouse lung metastasis model showed that FN knockdown suppressed lung metastasis and thus enhanced survival. FN staining of micro tissue array revealed that FN expression was positive for human lung cancer (61%) and breast cancer (58%) patients. Furthermore, the expression levels of FN, desmoglein-2, desmocollin-2, and plakoglobin were significantly correlated with the poor survival of lung and breast cancer patients, as per the Kaplan-Meier plotter analysis. Altogether, our data suggest that FN upregulation and enhanced desmosomal interactions are critical for cell aggregation and anoikis resistance upon cell detachment.
Collapse
Affiliation(s)
- Hyeong-Jun Han
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Jee Young Sung
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Su-Hyeon Kim
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Un-Jung Yun
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Hyeryeong Kim
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Eun-Ju Jang
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Ha-Eun Yoo
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Eun Kyung Hong
- Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Sung-Ho Goh
- Division of Precision Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Aree Moon
- Pharmacy, Duksung Women's University, Seoul, Republic of Korea
| | - Jae-Seon Lee
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sang-Ku Ye
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jaegal Shim
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea
| | - Yong-Nyun Kim
- Division of Translational Science, National Cancer Center, Goyang, Republic of Korea.
| |
Collapse
|
9
|
Efthymiou G, Radwanska A, Grapa AI, Beghelli-de la Forest Divonne S, Grall D, Schaub S, Hattab M, Pisano S, Poet M, Pisani DF, Counillon L, Descombes X, Blanc-Féraud L, Van Obberghen-Schilling E. Fibronectin Extra Domains tune cellular responses and confer topographically distinct features to fibril networks. J Cell Sci 2021; 134:jcs.252957. [PMID: 33526715 DOI: 10.1242/jcs.252957] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular fibronectin (FN; also known as FN1) variants harboring one or two alternatively spliced so-called extra domains (EDB and EDA) play a central bioregulatory role during development, repair processes and fibrosis. Yet, how the extra domains impact fibrillar assembly and function of the molecule remains unclear. Leveraging a unique biological toolset and image analysis pipeline for direct comparison of the variants, we demonstrate that the presence of one or both extra domains impacts FN assembly, function and physical properties of the matrix. When presented to FN-null fibroblasts, extra domain-containing variants differentially regulate pH homeostasis, survival and TGF-β signaling by tuning the magnitude of cellular responses, rather than triggering independent molecular switches. Numerical analyses of fiber topologies highlight significant differences in variant-specific structural features and provide a first step for the development of a generative model of FN networks to unravel assembly mechanisms and investigate the physical and functional versatility of extracellular matrix landscapes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Agata Radwanska
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Anca-Ioana Grapa
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France.,Université Côte d'Azur, Inria, CNRS, i3S, Nice 06902, France
| | | | - Dominique Grall
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | | | - Maurice Hattab
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Sabrina Pisano
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | - Mallorie Poet
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | | | | | | | | | | |
Collapse
|
10
|
Clapero F, Tortarolo D, Valdembri D, Serini G. Quantifying Polarized Extracellular Matrix Secretion in Cultured Endothelial Cells. Methods Mol Biol 2021; 2217:301-311. [PMID: 33215388 DOI: 10.1007/978-1-0716-0962-0_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
In endothelial cells (ECs), the onset of apicobasal polarity is primarily regulated by the interaction of integrins with the surrounding extracellular matrix (ECM). ECs secrete and polymerize fibronectin (FN), a unique, permissive substrate that allows for vascular morphogenesis and lumen formation. We previously identified a signaling pathway that, under the control of the adhesion site adaptor protein PPFIA1, integrates the polarized secretion of freshly synthesized FN with the recycling of conformationally active α5β1 integrin, the main FN receptor in ECs. To characterize the functional role of PPFIA1-dependent signaling in ECs, we set up a Transwell-based assay to quantify the polarized secretion of ECM proteins. To this aim, we allowed ECs to form a confluent monolayer on the Transwell membrane and checked its integrity by measuring transendothelial electric resistance and controlling the stability of tight junctions over time by fluorescent confocal microscope analysis. Finally, we quantified apical and basolateral FN secretion in control and PPFIA1-silenced EC culture medium by western blot analysis coupled to spike-in normalization.
Collapse
Affiliation(s)
- Fabiana Clapero
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Torino School of Medicine, Torino, Italy
| | - Dora Tortarolo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Torino School of Medicine, Torino, Italy
| | - Donatella Valdembri
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy. .,Department of Oncology, University of Torino School of Medicine, Torino, Italy.
| | - Guido Serini
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy. .,Department of Oncology, University of Torino School of Medicine, Torino, Italy.
| |
Collapse
|
11
|
Dolhikh HV, Maslak HS, Didenko VI, Klenina IA, Dolhikh АО. Levels of isoforms of fibronectin and α5/CD49e integrin on lymphocytes and in blood plasma in the conditions of chronic diffuse liver diseases. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Chronic diffuse liver diseases are characterized by accumulation of complex inflammatory infiltrate in the liver tissues, blood, and lympha, and activation of the immune system. Leukocytes become involved in the area of inflammation after the activation of receptors of blood adhesia, particularly integrins and their ligands. Plasma lymphocytes quickly activate the function of integrins by changing their conformation, leading to high affinity and underlying the formation of strong stable connection between the components of extracellular matrix. A vitally important role in the process of liver fibrogenesis is performed by a pro-fibrogenicic protein fibronectin which induces the expresson of collagen genes and precedes the deposition of other components of matrix. The studies were conducted in the group of patients suffering from chronic diffuse liver diseases of non-viral etiology aged 28–60 years, n = 36 and in the group of 15 practically healthy volunteer donors aged 25 to 52 years without a history of liver diseases using the methods of flow cytofluorometry, immunoenzymatic analysis, and quantitative real-time polymerase chain reaction. The patients of the group with chronic diffuse liver diseases were observed to have statistically significant decrease in the concentration of plasmatic form of fibronectin measuring 27.6% compared with the control group. We determined increase in the concentration of cellular fibronectin in blood plasma of patients with the diseases on average accounting for 63.8% compared with the norm, and the highest increase in this parameter equaling 77.2% was seen in patients suffering from drug-induced hepatitis. Significant increase in the level of exposure of cellular FN on blood lymphocytes was determined in patients with chronic diffuse liver diseases, measuring 231.8%, whereas the level of plasmatic form of fibronectin in these cells was decreased (statistically unreliable). For α5-integrin subunit, we determined a 390.8% increase in the level of its exposure in blood lymphocytes in the surveyed groups compared with the control. Level of blood lymphocytes that express the cellular fibronectin significantly decreased by 140.1%. Statistical characteristics of diagnostic possibility of the parameters of the level of plasmatic and cellular fibronectin in blood, determined over the analysis of ROC-curves, demonstrated excellent informativeness of these tests. Analysis of the possibility of predicting the presence of pathology using the model of logistic regression revealed zero error of prediction and maximum efficiency of the tests: intensity of exposure of α5-integrin receptor on the surface of lymphocytes, intensity of exposure of plasmatic fibronectin on the surface of lymphocytes, intensity of exposure of cellular fibronectin on the surface of lymphocytes, concentration of plasmatic fibronectin in blood, concentration of cellular fibronectin in blood plasma. These parameters may be proposed for further surveys for developing serologic biomarkers based on the parameters for diagnostics of chronic diffuse liver diseases.
Collapse
|
12
|
Cui J, Dean D, Hornicek FJ, Chen Z, Duan Z. The role of extracelluar matrix in osteosarcoma progression and metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:178. [PMID: 32887645 PMCID: PMC7650219 DOI: 10.1186/s13046-020-01685-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy and responsible for considerable morbidity and mortality due to its high rates of pulmonary metastasis. Although neoadjuvant chemotherapy has improved 5-year survival rates for patients with localized OS from 20% to over 65%, outcomes for those with metastasis remain dismal. In addition, therapeutic regimens have not significantly improved patient outcomes over the past four decades, and metastases remains a primary cause of death and obstacle in curative therapy. These limitations in care have given rise to numerous works focused on mechanisms and novel targets of OS pathogenesis, including tumor niche factors. OS is notable for its hallmark production of rich extracellular matrix (ECM) of osteoid that goes beyond simple physiological growth support. The aberrant signaling and structural components of the ECM are rich promoters of OS development, and very recent works have shown the specific pathogenic phenotypes induced by these macromolecules. Here we summarize the current developments outlining how the ECM contributes to OS progression and metastasis with supporting mechanisms. We also illustrate the potential of tumorigenic ECM elements as prognostic biomarkers and therapeutic targets in the evolving clinical management of OS.
Collapse
Affiliation(s)
- Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, 69 Chuanshan Road, Hengyang, 421001, Hunan, China.,Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Dylan Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA
| | - Zhiwei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, 69 Chuanshan Road, Hengyang, 421001, Hunan, China.
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. South, Los Angeles, CA, 90095, USA.
| |
Collapse
|
13
|
Roberts AL, Mavlyutov TA, Perlmutter TE, Curry SM, Harris SL, Chauhan AK, McDowell CM. Fibronectin extra domain A (FN-EDA) elevates intraocular pressure through Toll-like receptor 4 signaling. Sci Rep 2020; 10:9815. [PMID: 32555351 PMCID: PMC7299944 DOI: 10.1038/s41598-020-66756-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/27/2020] [Indexed: 02/08/2023] Open
Abstract
Elevated intraocular pressure (IOP) is a major risk factor for the development and progression of primary open angle glaucoma and is due to trabecular meshwork (TM) damage, which leads to impaired aqueous humor outflow. Here, we explore a novel molecular mechanism involved in glaucomatous TM damage. We investigated the role of an endogenous Toll-like receptor 4 (TLR4) ligand, fibronectin-EDA (FN-EDA), in TGFβ2-induced ocular hypertension in mice. We utilized transgenic mouse strains that either constitutively express only FN containing the EDA isoform or contain an EDA-null allele and express only FN lacking EDA, with or without a mutation in Tlr4, in our inducible mouse model of ocular hypertension by injection of Ad5.TGFβ2. IOP was measured over time and eyes accessed by immunohistochemistry for total FN and FN-EDA expression. Constitutively active EDA caused elevated IOP starting at 14 weeks of age. Ad5.TGFβ2 induced ocular hypertension in wildtype C57BL/6J mice and further amplified the IOP in constitutively active EDA mice. TLR4 null and EDA null mice blocked Ad5.TGFβ-induced ocular hypertension. Total FN and FN-EDA isoform expression increased in response to Ad5.TGFβ2. These data suggest that both TLR4 and FN-EDA contribute to TGFβ2 induced ocular hypertension.
Collapse
Affiliation(s)
- Amanda L Roberts
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Timur A Mavlyutov
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Tanisha E Perlmutter
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Stacy M Curry
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Sherri L Harris
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Anil K Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Colleen M McDowell
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
14
|
Schwefel K, Spiegler S, Kirchmaier BC, Dellweg PKE, Much CD, Pané-Farré J, Strom TM, Riedel K, Felbor U, Rath M. Fibronectin rescues aberrant phenotype of endothelial cells lacking either CCM1, CCM2 or CCM3. FASEB J 2020; 34:9018-9033. [PMID: 32515053 DOI: 10.1096/fj.201902888r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Loss-of-function variants in CCM1/KRIT1, CCM2, and CCM3/PDCD10 are associated with autosomal dominant cerebral cavernous malformations (CCMs). CRISPR/Cas9-mediated CCM3 inactivation in human endothelial cells (ECs) has been shown to induce profound defects in cell-cell interaction as well as actin cytoskeleton organization. We here show that CCM3 inactivation impairs fibronectin expression and consequently leads to reduced fibers in the extracellular matrix. Despite the complexity and high molecular weight of fibronectin fibrils, our in vitro model allowed us to reveal that fibronectin supplementation restored aberrant spheroid formation as well as altered EC morphology, and suppressed actin stress fiber formation. Yet, fibronectin replacement neither enhanced the stability of tube-like structures nor inhibited the survival advantage of CCM3-/- ECs. Importantly, CRISPR/Cas9-mediated introduction of biallelic loss-of-function variants into either CCM1 or CCM2 demonstrated that the impaired production of a functional fibronectin matrix is a common feature of CCM1-, CCM2-, and CCM3-deficient ECs.
Collapse
Affiliation(s)
- Konrad Schwefel
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefanie Spiegler
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Bettina C Kirchmaier
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, University of Frankfurt, Frankfurt am Main, Germany
| | - Patricia K E Dellweg
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Christiane D Much
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Jan Pané-Farré
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Katharina Riedel
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Ute Felbor
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Matthias Rath
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
15
|
Umana-Diaz C, Pichol-Thievend C, Marchand MF, Atlas Y, Salza R, Malbouyres M, Barret A, Teillon J, Ardidie-Robouant C, Ruggiero F, Monnot C, Girard P, Guilluy C, Ricard-Blum S, Germain S, Muller L. Scavenger Receptor Cysteine-Rich domains of Lysyl Oxidase-Like2 regulate endothelial ECM and angiogenesis through non-catalytic scaffolding mechanisms. Matrix Biol 2020; 88:33-52. [DOI: 10.1016/j.matbio.2019.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
|
16
|
Efthymiou G, Saint A, Ruff M, Rekad Z, Ciais D, Van Obberghen-Schilling E. Shaping Up the Tumor Microenvironment With Cellular Fibronectin. Front Oncol 2020; 10:641. [PMID: 32426283 PMCID: PMC7203475 DOI: 10.3389/fonc.2020.00641] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022] Open
Abstract
Normal tissue homeostasis and architecture restrain tumor growth. Thus, for a tumor to develop and spread, malignant cells must overcome growth-repressive inputs from surrounding tissue and escape immune surveillance mechanisms that curb cancer progression. This is achieved by promoting the conversion of a physiological microenvironment to a pro-tumoral state and it requires a constant dialog between malignant cells and ostensibly normal cells of adjacent tissue. Pro-tumoral reprogramming of the stroma is accompanied by an upregulation of certain extracellular matrix (ECM) proteins and their cognate receptors. Fibronectin (FN) is one such component of the tumor matrisome. This large multidomain glycoprotein dimer expressed over a wide range of human cancers is assembled by cell-driven forces into a fibrillar array that provides an obligate scaffold for the deposition of other matrix proteins and binding sites for functionalization by soluble factors in the tumor microenvironment. Encoded by a single gene, FN regulates the proliferation, motile behavior and fate of multiple cell types, largely through mechanisms that involve integrin-mediated signaling. These processes are coordinated by distinct isoforms of FN, collectively known as cellular FN (as opposed to circulating plasma FN) that arise through alternative splicing of the FN1 gene. Cellular FN isoforms differ in their solubility, receptor binding ability and spatiotemporal expression, and functions that have yet to be fully defined. FN induction at tumor sites constitutes an important step in the acquisition of biological capabilities required for several cancer hallmarks such as sustaining proliferative signaling, promoting angiogenesis, facilitating invasion and metastasis, modulating growth suppressor activity and regulating anti-tumoral immunity. In this review, we will first provide an overview of ECM reprogramming through tumor-stroma crosstalk, then focus on the role of cellular FN in tumor progression with respect to these hallmarks. Last, we will discuss the impact of dysregulated ECM on clinical efficacy of classical (radio-/chemo-) therapies and emerging treatments that target immune checkpoints and explore how our expanding knowledge of the tumor ECM and the central role of FN can be leveraged for therapeutic benefit.
Collapse
Affiliation(s)
| | - Angélique Saint
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.,Centre Antoine Lacassagne, Nice, France
| | - Michaël Ruff
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | - Zeinab Rekad
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | | | | |
Collapse
|
17
|
Gagné D, Benoit YD, Groulx JF, Vachon PH, Beaulieu JF. ILK supports RhoA/ROCK-mediated contractility of human intestinal epithelial crypt cells by inducing the fibrillogenesis of endogenous soluble fibronectin during the spreading process. BMC Mol Cell Biol 2020; 21:14. [PMID: 32183701 PMCID: PMC7079544 DOI: 10.1186/s12860-020-00259-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/05/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Fibronectin (FN) assembly into an insoluble fibrillar matrix is a crucial step in many cell responses to extracellular matrix (ECM) properties, especially with regards to the integrin-related mechanosensitive signaling pathway. We have previously reported that the silencing of expression of integrin-linked kinase (ILK) in human intestinal epithelial crypt (HIEC) cells causes significant reductions in proliferation and spreading through concomitantly acquired impairment of soluble FN deposition. These defects in ILK-depleted cells are rescued by growth on exogenous FN. In the present study we investigated the contribution of ILK in the fibrillogenesis of FN and its relation to integrin-actin axis signaling and organization. RESULTS We show that de novo fibrillogenesis of endogenous soluble FN is ILK-dependent. This function seemingly induces the assembly of an ECM that supports increased cytoskeletal tension and the development of a fully spread contractile cell phenotype. We observed that HIEC cell adhesion to exogenous FN or collagen-I (Col-I) is sufficient to restore fibrillogenesis of endogenous FN in ILK-depleted cells. We also found that optimal engagement of the Ras homolog gene family member A (RhoA) GTPase/Rho-associated kinase (ROCK-1, ROCK-2)/myosin light chain (MLC) pathway, actin ventral stress fiber formation, and integrin adhesion complex (IAC) maturation rely primarily upon the cell's capacity to execute FN fibrillogenesis, independent of any significant ILK input. Lastly, we confirm the integrin α5β1 as the main integrin responsible for FN assembly, although in ILK-depleted cells αV-class integrins expression is needed to allow the rescue of FN fibrillogenesis on exogenous substrate. CONCLUSION Our study demonstrates that ILK specifically induces the initiation of FN fibrillogenesis during cell spreading, which promotes RhoA/ROCK-dependent cell contractility and maturation of the integrin-actin axis structures. However, the fibrillogenesis process and its downstream effect on RhoA signaling, cell contractility and spreading are ILK-independent in human intestinal epithelial crypt cells.
Collapse
Affiliation(s)
- David Gagné
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4 Canada
| | - Yannick D. Benoit
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5 Canada
| | - Jean-François Groulx
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, 92093 USA
| | - Pierre H. Vachon
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4 Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, and Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4 Canada
| |
Collapse
|
18
|
Jordahl S, Solorio L, Neale DB, McDermott S, Jordahl JH, Fox A, Dunlay C, Xiao A, Brown M, Wicha M, Luker GD, Lahann J. Engineered Fibrillar Fibronectin Networks as Three-Dimensional Tissue Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904580. [PMID: 31565823 PMCID: PMC6851443 DOI: 10.1002/adma.201904580] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Indexed: 05/19/2023]
Abstract
Extracellular matrix (ECM) proteins, and most prominently, fibronectin (Fn), are routinely used in the form of adsorbed pre-coatings in an attempt to create a cell-supporting environment in both two- and three-dimensional cell culture systems. However, these protein coatings are typically deposited in a form which is structurally and functionally distinct from the ECM-constituting fibrillar protein networks naturally deposited by cells. Here, the cell-free and scalable synthesis of freely suspended and mechanically robust three-dimensional (3D) networks of fibrillar fibronectin (fFn) supported by tessellated polymer scaffolds is reported. Hydrodynamically induced Fn fibrillogenesis at the three-phase contact line between air, an Fn solution, and a tessellated scaffold microstructure yields extended protein networks. Importantly, engineered fFn networks promote cell invasion and proliferation, enable in vitro expansion of primary cancer cells, and induce an epithelial-to-mesenchymal transition in cancer cells. Engineered fFn networks support the formation of multicellular cancer structures cells from plural effusions of cancer patients. With further work, engineered fFn networks can have a transformative impact on fundamental cell studies, precision medicine, pharmaceutical testing, and pre-clinical diagnostics.
Collapse
Affiliation(s)
- Stacy Jordahl
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Luis Solorio
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Dylan B Neale
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Sean McDermott
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Jacob H Jordahl
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Alexandra Fox
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Christopher Dunlay
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Annie Xiao
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Martha Brown
- Department of Internal Medicine, University of Michigan, 1500 E Medical Center Dr SPC 5916, Ann Arbor, MI, 48109, USA
| | - Max Wicha
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Biointerfaces Institute, Departments of Chemical Engineering, Materials Science and Engineering, Biomedical Engineering, and Macromolecular Science and Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| |
Collapse
|
19
|
Hanouna G, Tang E, Perez J, Vandermeersch S, Haymann JP, Baud L, Letavernier E. Preventing Calpain Externalization by Reducing ABCA1 Activity with Probenecid Limits Melanoma Angiogenesis and Development. J Invest Dermatol 2019; 140:445-454. [PMID: 31425704 DOI: 10.1016/j.jid.2019.06.148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/23/2019] [Accepted: 06/17/2019] [Indexed: 11/17/2022]
Abstract
Calpains, intracellular proteases specifically inhibited by calpastatin, play a major role in neoangiogenesis involved in tumor invasiveness and metastasis. They are partly exteriorized via the ATP-binding cassette transporter A1(ABCA1) transporter, but the importance of this process in tumor growth is still unknown. The aim of our study was to investigate the role of extracellular calpains in a model of melanoma by blocking their extracellular activity or exteriorization. In the first approach, a B16-F10 model of melanoma was developed in transgenic mice expressing high extracellular levels of calpastatin. In these mice, tumor growth was inhibited by ∼ 3-fold compared with wild-type animals. In vitro cytotoxicity assays and in vivo tumor studies have demonstrated that this protection was associated with a defect in tumor neoangiogenesis. Similarly, in wild-type animals given probenecid to blunt ABCA1 activity, melanoma tumor growth was inhibited by ∼ 3-fold. Again, this response was associated with a defect in neoangiogenesis. In vitro studies confirmed that probenecid limited endothelial cell migration and capillary formation from vascular explants. The observed reduction in fibronectin cleavage under these conditions is potentially involved in the response. Collectively, these studies demonstrate that probenecid, by blunting ABCA1 activity and thereby calpain exteriorization, limits melanoma tumor neoangiogenesis and invasiveness.
Collapse
Affiliation(s)
- Guillaume Hanouna
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France
| | - Ellie Tang
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France
| | - Joëlle Perez
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France
| | - Sophie Vandermeersch
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France
| | - Jean-Philippe Haymann
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France; Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Laurent Baud
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France; Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Emmanuel Letavernier
- Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UMR_S 1155 and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Paris, France; Institut National de la Santé et de la Recherche Médicale, UMR_S 1155, Paris, France; Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France.
| |
Collapse
|
20
|
Al-Yafeai Z, Yurdagul A, Peretik JM, Alfaidi M, Murphy PA, Orr AW. Endothelial FN (Fibronectin) Deposition by α5β1 Integrins Drives Atherogenic Inflammation. Arterioscler Thromb Vasc Biol 2019; 38:2601-2614. [PMID: 30354234 DOI: 10.1161/atvbaha.118.311705] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Alterations in extracellular matrix quantity and composition contribute to atherosclerosis, with remodeling of the subendothelial basement membrane to an FN (fibronectin)-rich matrix preceding lesion development. Endothelial cell interactions with FN prime inflammatory responses to a variety of atherogenic stimuli; however, the mechanisms regulating early atherogenic FN accumulation remain unknown. We previously demonstrated that oxLDL (oxidized low-density lipoprotein) promotes endothelial proinflammatory gene expression by activating the integrin α5β1, a classic mediator of FN fibrillogenesis. Approach and Results- We now show that oxLDL drives robust endothelial FN deposition and inhibiting α5β1 (blocking antibodies, α5 knockout cells) completely inhibits oxLDL-induced FN deposition. Consistent with this, inducible endothelial-specific α5 integrin deletion in ApoE knockout mice significantly reduces atherosclerotic plaque formation, associated with reduced early atherogenic inflammation. Unlike TGFβ (transforming growth factor β)-induced FN deposition, oxLDL does not induce FN expression (mRNA, protein) or the endothelial-to-mesenchymal transition phenotype. In addition, we show that cell-derived and plasma-derived FN differentially affect endothelial function, with only cell-derived FN capable of supporting oxLDL-induced VCAM-1 (vascular cell adhesion molecule 1) expression, despite plasma FN deposition by oxLDL. The inclusion of alternative exon EIIIA (EDA) of FN (EIIIA) and alternative exon EIIIB (EDB) of FN (EIIIB) domains in cell-derived FN mediates this effect, as EIIIA/EIIIB knockout endothelial cells show diminished oxLDL-induced inflammation. Furthermore, our data suggest that EIIIA/EIIIB-positive cellular FN is required for maximal α5β1 recruitment to focal adhesions and FN fibrillogenesis. Conclusions- Taken together, our data demonstrate that endothelial α5 integrins drive oxLDL-induced FN deposition and early atherogenic inflammation. Additionally, we show that α5β1-dependent endothelial FN deposition mediates oxLDL-dependent endothelial inflammation and FN fibrillogenesis.
Collapse
Affiliation(s)
- Zaki Al-Yafeai
- From the Department of Cellular and Molecular Physiology (Z.A.-Y., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Arif Yurdagul
- Department of Cell Biology and Anatomy (A.Y., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Jonette M Peretik
- Department of Pathology and Translational Pathobiology (J.M.P., M.A., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Mabruka Alfaidi
- Department of Pathology and Translational Pathobiology (J.M.P., M.A., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| | - Patrick A Murphy
- Center for Vascular Biology, UConn Health, Farmington, CT (P.A.M.)
| | - A Wayne Orr
- From the Department of Cellular and Molecular Physiology (Z.A.-Y., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Department of Cell Biology and Anatomy (A.Y., A.W.O.), LSU Health Sciences Center, Shreveport, LA.,Department of Pathology and Translational Pathobiology (J.M.P., M.A., A.W.O.), LSU Health Sciences Center, Shreveport, LA
| |
Collapse
|
21
|
Fibronectin Promotes Cell Growth and Migration in Human Renal Cell Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20112792. [PMID: 31181623 PMCID: PMC6600362 DOI: 10.3390/ijms20112792] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Abstract
The prognostic and therapeutic values of fibronectin have been reported in patients with renal cell carcinoma (RCC). However, the underlying mechanisms of malignancy in RCC are not completely understood. We found that silencing of fibronectin expression attenuated human RCC 786-O and Caki-1 cell growth and migration. Silencing of potential fibronectin receptor integrin α5 and integrin β1 decreased 786-O cell ability in movement and chemotactic migration. Biochemical examination revealed a reduction of cyclin D1 and vimentin expression, transforming growth factor-β1 (TGF-β1) production, as well as Src and Smad phosphorylation in fibronectin-silenced 786-O and Caki-1 cells. Pharmacological inhibition of Src decreased 786-O cell growth and migration accompanied by a reduction of cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. In 786-O cells, higher activities in cell growth and migration than in Caki-1 cells were noted, along with elevated fibronectin and TGF-β1 expression. The additions of exogenous fibronectin and TGF-β1 promoted Caki-1 cell growth and migration, and increased cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. These findings highlight the role of fibronectin in RCC cell growth and migration involving Src and TGF-β1 signaling.
Collapse
|
22
|
Santos Rizzo Zuttion MS, Dias Câmara DA, Dariolli R, Takimura C, Wenceslau C, Kerkis I. In vitro heterogeneity of porcine adipose tissue-derived stem cells. Tissue Cell 2019; 58:51-60. [PMID: 31133246 DOI: 10.1016/j.tice.2019.04.001] [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: 12/26/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 01/27/2023]
Abstract
Tissue-specific adult stem cells (ASC) are heterogeneous and characterized by a mix of progenitor cells that produce cells at various stages of differentiation, and ultimately different terminally differentiated cells. Understanding the heterogeneity of ASCs may lead to the development of improved protocols of cell isolation and optimized cell therapy clinical protocols. Using a combination of enzymatic and explant culture protocols, we obtained pADSC population, which is composed by two distinct morphologies: fibroblast-like cells (FLCs) and endothelial-like cells (ELCs). Both cell sub-types efficiently formed colonies, expressed CD90+/CD105+/CD44+, and differentially expressed such markers such as Nestin, Vimentin, Fibronectin, Cytokeratin, Connexin 43, CD31, CD34 and CD146 as well as the pluripotent stem cell markers Oct-4, Nanog and Sox2. Mixed populations of pADSCs did not lose their multipotentiality and the cells were able to undergo osteogenic, chondrogenic, adipogenic and myogenic differentiation. Furthermore, the mixed population spontaneously formed capillary tube structures. Our findings suggest that different subpopulations can be isolated from adipose tissue and that the ADSCs need to be better evaluated using a wide panel of different markers related to cell differentiation, which is important for stem cell therapy and regenerative medicine, particularly for advanced stem cells therapies - products that are currently under investigation or even use.
Collapse
Affiliation(s)
- Marilia Sanches Santos Rizzo Zuttion
- Laboratory of Genetics, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, SP, 05503-900, Brazil; Federal University of São Paulo, R. Sena Madureira, 1500 - Vila Clementino, São Paulo, SP, 04021-001, Brazil.
| | - Diana Aparecida Dias Câmara
- Laboratory of Genetics, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, SP, 05503-900, Brazil; Federal University of São Paulo, R. Sena Madureira, 1500 - Vila Clementino, São Paulo, SP, 04021-001, Brazil.
| | - Rafael Dariolli
- Heart Institute (InCor), University of São Paulo Medical School, Brazil: Av. Dr. Enéas de Carvalho Aguiar, 44 - Pinheiros, São Paulo, SP, 05403-900, Brazil.
| | - Celso Takimura
- Heart Institute (InCor), University of São Paulo Medical School, Brazil: Av. Dr. Enéas de Carvalho Aguiar, 44 - Pinheiros, São Paulo, SP, 05403-900, Brazil.
| | - Cristiane Wenceslau
- Laboratory of Genetics, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, SP, 05503-900, Brazil.
| | - Irina Kerkis
- Laboratory of Genetics, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, SP, 05503-900, Brazil; Federal University of São Paulo, R. Sena Madureira, 1500 - Vila Clementino, São Paulo, SP, 04021-001, Brazil.
| |
Collapse
|
23
|
Soubies E, Radwanska A, Grall D, Blanc-Féraud L, Van Obberghen-Schilling E, Schaub S. Nanometric axial resolution of fibronectin assembly units achieved with an efficient reconstruction approach for multi-angle-TIRF microscopy. Sci Rep 2019; 9:1926. [PMID: 30760745 PMCID: PMC6374485 DOI: 10.1038/s41598-018-36119-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 02/01/2023] Open
Abstract
High resolution imaging of molecules at the cell-substrate interface is required for understanding key biological processes. Here we propose a complete pipeline for multi-angle total internal reflection fluorescence microscopy (MA-TIRF) going from instrument design and calibration procedures to numerical reconstruction. Our custom setup is endowed with a homogeneous field illumination and precise excitation beam angle. Given a set of MA-TIRF acquisitions, we deploy an efficient joint deconvolution/reconstruction algorithm based on a variational formulation of the inverse problem. This algorithm offers the possibility of using various regularizations and can run on graphics processing unit (GPU) for rapid reconstruction. Moreover, it can be easily used with other MA-TIRF devices and we provide it as an open-source software. This ensemble has enabled us to visualize and measure with unprecedented nanometric resolution, the depth of molecular components of the fibronectin assembly machinery at the basal surface of endothelial cells.
Collapse
Affiliation(s)
- Emmanuel Soubies
- Université Côte d'Azur, CNRS, Inria, I3S, France. .,Biomedical Imaging Group, EPFL, Lausanne, Switzerland.
| | | | | | | | | | - Sébastien Schaub
- Université Côte d'Azur, CNRS, Inria, I3S, France. .,Université Côte d'Azur, CNRS, Inserm, iBV, France.
| |
Collapse
|
24
|
Ravindran K, Salem MM, Alturki AY, Thomas AJ, Ogilvy CS, Moore JM. Endothelialization following Flow Diversion for Intracranial Aneurysms: A Systematic Review. AJNR Am J Neuroradiol 2019; 40:295-301. [PMID: 30679207 DOI: 10.3174/ajnr.a5955] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/08/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND The underlying mechanism of action of flow diverters is believed to be the induction of aneurysm thrombosis and simultaneous endothelial cell growth along the device struts, thereby facilitating aneurysm exclusion from the circulation. Although extensive attention has been paid to the role of altered cerebrovascular hemodynamics using computational fluid dynamics analyses, relatively less emphasis has been placed on the role of the vascular endothelium in promoting aneurysm healing. PURPOSE Our aim was to systematically review all available literature investigating the mechanism of action of flow diverters in both human patients and preclinical models. DATA SOURCES A systematic search of PubMed, Cochrane Central Register of Controlled Trials MEDLINE, EMBASE, and the Web of Science electronic data bases was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. STUDY SELECTION We selected articles assessing the role of endothelialization in flow-diverter treatment of cerebral aneurysms, including both preclinical and clinical studies. DATA ANALYSIS Ten articles were eligible for inclusion in this review. Two assessed endothelialization in human patients, while the other 8 used preclinical models (either rabbits or pigs). DATA SYNTHESIS Methods used to assess endothelialization included optical coherence tomography and scanning electron microscopy. LIMITATIONS A limitation was the heterogeneity of studies. CONCLUSIONS Current data regarding the temporal relationship to flow-diverter placement has largely been derived from work in preclinical animal models. Whether these cells along the device struts originate from adjacent endothelial cells or are the result of homing of circulating endothelial progenitor cells is equivocal.
Collapse
Affiliation(s)
- K Ravindran
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - M M Salem
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - A Y Alturki
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - A J Thomas
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - C S Ogilvy
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - J M Moore
- From the Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
Hassanisaber H, Jafari L, Campeau MA, Drevelle O, Lauzon MA, Langelier E, Faucheux N, Rouleau L. The effect of substrate bulk stiffness on focal and fibrillar adhesion formation in human abdominal aortic endothelial cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 98:572-583. [PMID: 30813060 DOI: 10.1016/j.msec.2018.12.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/07/2018] [Accepted: 12/28/2018] [Indexed: 01/01/2023]
Abstract
Endothelial cell (EC) dysfunction contributes to atherosclerosis, which is associated with arterial stiffening and fibronectin (FN) deposition, by ECs and smooth muscle cells (SMCs). The effect of stiffness on the EC/FN interaction and fibrillar adhesion formation has been poorly studied. An in vitro model was prepared that included FN-coated polydimethylsiloxane (PDMS) films with similar hydrophobicity and roughness but distinct Young's modulus values, mimicking healthy (1.0 MPa) and atherosclerotic (2.8 MPa) arteries. Human aortic abdominal endothelial cells (HAAECs) seeded on 1.0 MPa PDMS films spread over time and reached their maximum surface area faster than on 2.8 MPa PDMS films. In addition, HAAECs appeared to organize focal adhesion more rapidly on 1.0 MPa PDMS films, despite the similar cell binding domain accessibility to adsorbed FN. Interestingly, we also observed up to a ~5-fold increase in the percentage of HAAECs that had a well-developed fibrillar adhesion on 1.0 MPa compared to 2.8 MPa PDMS films as verified by integrin α5 subunits, tensin, and FN staining. This variation did not affect EC migration. These results suggest that there are favourable conditions for FN matrix assembly by ECs in early atherosclerosis rather than at advanced stages. Our in vitro model will therefore be helpful to understand the influence of bulk stiffness on cells involved in atherosclerosis.
Collapse
Affiliation(s)
- H Hassanisaber
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - L Jafari
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| | - M A Campeau
- Department of Chemical Engineering, McGill University, 3610 Rue University, Montréal, Québec, H3A 0C5, Canada.
| | - O Drevelle
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Thermal Plasma and Nanomaterial Synthesis Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - M-A Lauzon
- 3D Cell Culture Systems Laboratory, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec J1K 2R1, Canada.
| | - E Langelier
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - N Faucheux
- Cell-biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec, J1K 2R1, Canada; Clinical Research Centre of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| | - L Rouleau
- Department of Mechanical Engineering, Université de Sherbrooke, 2500 boul. Université, Sherbrooke, Québec, J1K 2R1, Canada.
| |
Collapse
|
26
|
Wang WY, Twu CW, Liu YC, Lin HH, Chen CJ, Lin JC. Fibronectin promotes nasopharyngeal cancer cell motility and proliferation. Biomed Pharmacother 2018; 109:1772-1784. [PMID: 30551431 DOI: 10.1016/j.biopha.2018.11.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Nasopharyngeal cancer (NPC) is an Epstein-Barr virus (EBV)-associated carcinoma. Fibronectin is regarded as a prognosticator in NPC and its involvement in cell motility has been reported in EBV infection and viral latent membrane protein 1 (LMP1) overexpression NPC cell lines. However, its malignant potential in NPC cell lines without harbouring the EBV genome has not been investigated. We investigatd and compared among four NPC cell lines, and the results revealed a positive association between fibronectin levels and NPC cell motility as well as proliferation. Studies of antibody neutralization, exogenous addition, overexpression, and RNA interference confirmed a migration role of fibronectin in NPC cells involving integrin α5, Src, Rac1, and Cdc42, implying a mesenchymal-like cell movement. Furthermore, hypoxia-inducible factor-1α (HIF-1α) and transforming growth factor-β1 (TGF-β1) were identified as alternative activators of fibronectin expression and NPC cell migration. Besides cell migration, studies of RNA interference also showed a stimulatory effect of fibronectin in NPC cell proliferation. Mechanistic studies further revealed a subsequent reduction of HIF-1α, TGF-β1, cyclin D1, β-catenin, vimentin, and Slug together with decreased Src and Akt phosphorylation after fibronectin knockdown. Parallel studies in a xenograft tumor mice model further showed that tumor growth correlated well with elevation of circulating fibronectin and activation of the identified intracellular signaling molecules. The results of our study highlight a role of fibronectin in NPC cell motility and proliferation in concerted action with HIF-1α and TGF-β1 possibly through linking molecules Src and Akt. Fibronectin overexpression and autoantibody are known to have potential prognostic value in patients with NPC. Our findings shed light on the biochemical and molecular mechanisms underlying the pathogenic role of fibronectin in this disease.
Collapse
Affiliation(s)
- Wen-Yi Wang
- Department of Nursing, Hung-Kuang University, Taichung, Taiwan
| | - Chih-Wen Twu
- Department of Otorhinolaryngology, Taichung Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chun Liu
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Hua Lin
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Jin-Ching Lin
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
27
|
Marchand M, Monnot C, Muller L, Germain S. Extracellular matrix scaffolding in angiogenesis and capillary homeostasis. Semin Cell Dev Biol 2018; 89:147-156. [PMID: 30165150 DOI: 10.1016/j.semcdb.2018.08.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/31/2018] [Accepted: 08/14/2018] [Indexed: 01/03/2023]
Abstract
The extracellular matrix (ECM) of blood vessels, which is composed of both the vascular basement membrane (BM) and the interstitial ECM is identified as a crucial component of the vasculature. We here focus on the unique molecular composition and scaffolding of the capillary ECM, which provides structural support to blood vessels and regulates properties of endothelial cells and pericytes. The major components of the BM are collagen IV, laminins, heparan sulfate proteoglycans and nidogen and also associated proteins such as collagen XVIII and fibronectin. Their organization and scaffolding in the BM is required for proper capillary morphogenesis and maintenance of vascular homeostasis. The BM also regulates vascular mechanosensing. A better understanding of the mechanical and structural properties of the vascular BM and interstitial ECM therefore opens new perspectives to control physiological and pathological angiogenesis and vascular homeostasis. The overall aim of this review is to explain how ECM scaffolding influences angiogenesis and capillary integrity.
Collapse
Affiliation(s)
- Marion Marchand
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France.
| |
Collapse
|
28
|
Kryza T, Parent C, Pardessus J, Petit A, Burlaud-Gaillard J, Reverdiau P, Iochmann S, Labas V, Courty Y, Heuzé-Vourc'h N. Human kallikrein-related peptidase 12 stimulates endothelial cell migration by remodeling the fibronectin matrix. Sci Rep 2018; 8:6331. [PMID: 29679011 PMCID: PMC5910384 DOI: 10.1038/s41598-018-24576-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/05/2018] [Indexed: 12/30/2022] Open
Abstract
Kallikrein-related peptidase 12 (KLK12) is a kallikrein family peptidase involved in angiogenesis - a complex biological process in which the sprouting, migration and stabilization of endothelial cells requires extracellular matrix remodeling. To characterize the molecular mechanisms associated with KLK12's proangiogenic activity, we evaluated its ability to hydrolyze various matrix proteins. Our results show that KLK12 efficiently cleaved the human extracellular matrix proteins fibronectin and tenascin, both of which are involved in the regulation of endothelial cell adhesion and migration. For fibronectin, the major proteolytic product generated by KLK12 was a 29 kDa fragment containing the amino-terminal domain and the first five type I fibronectin-domains, which are essential for regulating fibronectin assembly. We also demonstrated that KLK12-mediated fibronectin proteolysis antagonizes fibronectin polymerization and fibronectin fibril formation by endothelial cells, leading to an increase in cell migration. Furthermore, a polyclonal antibody raised against KLK12's proteolytic cleavage site on fibronectin prevented the KLK12-dependent inhibition of fibronectin polymerization and the KLK12-mediated pro-migratory effect on endothelial cells. Taken as a whole, our results indicate that KLK12's proangiogenic effect is mediated through several molecular mechanisms.
Collapse
Affiliation(s)
- T Kryza
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France.,Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
| | - C Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Pardessus
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - A Petit
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - J Burlaud-Gaillard
- Université François Rabelais de Tours, F-37032, Tours, France.,Plateforme IBiSA de Microscopie Electronique, Université François Rabelais de Tours, F-37032, Tours, France
| | - P Reverdiau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - S Iochmann
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - V Labas
- PRC, INRA, CNRS, Université François Rabelais de Tours, IFCE, F-37380, Nouzilly, France.,PAIB, CIRE, INRA, CHRU de Tours, Université François Rabelais de Tours, F-37380, Nouzilly, France
| | - Y Courty
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France.,Université François Rabelais de Tours, F-37032, Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032, Tours, France. .,Université François Rabelais de Tours, F-37032, Tours, France.
| |
Collapse
|
29
|
Langer EM, Kendsersky ND, Daniel CJ, Kuziel GM, Pelz C, Murphy KM, Capecchi MR, Sears RC. ZEB1-repressed microRNAs inhibit autocrine signaling that promotes vascular mimicry of breast cancer cells. Oncogene 2018; 37:1005-1019. [PMID: 29084210 PMCID: PMC5823716 DOI: 10.1038/onc.2017.356] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/13/2022]
Abstract
During normal tumor growth and in response to some therapies, tumor cells experience acute or chronic deprivation of nutrients and oxygen and induce tumor vascularization. While this occurs predominately through sprouting angiogenesis, tumor cells have also been shown to directly contribute to vessel formation through vascular mimicry (VM) and/or endothelial transdifferentiation. The extrinsic and intrinsic mechanisms underlying tumor cell adoption of endothelial phenotypes, however, are not well understood. Here we show that serum withdrawal induces mesenchymal breast cancer cells to undergo VM and that knockdown of the epithelial-to-mesenchymal transition (EMT) regulator, Zinc finger E-box binding homeobox 1 (ZEB1), or overexpression of the ZEB1-repressed microRNAs (miRNAs), miR-200c, miR-183, miR-96 and miR-182 inhibits this process. We find that secreted proteins Fibronectin 1 (FN1) and serine protease inhibitor (serpin) family E member 2 (SERPINE2) are essential for VM in this system. These secreted factors are upregulated in mesenchymal cells in response to serum withdrawal, and overexpression of VM-inhibiting miRNAs abrogates this upregulation. Intriguingly, the receptors for these secreted proteins, low-density lipoprotein receptor-related protein 1 (LRP1) and Integrin beta 1 (ITGB1), are also targets of the VM-inhibiting miRNAs, suggesting that autocrine signaling stimulating VM is regulated by ZEB1-repressed miRNA clusters. Together, these data provide mechanistic insight into the regulation of VM and suggest that miRNAs repressed during EMT, in addition to suppressing migratory and stem-like properties of tumor cells, also inhibit endothelial phenotypes of breast cancer cells adopted in response to a nutrient-deficient microenvironment.
Collapse
Affiliation(s)
- E M Langer
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - N D Kendsersky
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - C J Daniel
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - G M Kuziel
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - C Pelz
- Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, OR, USA
| | - K M Murphy
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Pathology & Immunology, Washington University, St. Louis, MO, USA
| | - M R Capecchi
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - R C Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
30
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
31
|
Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis 2017; 20:519-531. [PMID: 28667352 PMCID: PMC5660148 DOI: 10.1007/s10456-017-9563-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/21/2017] [Indexed: 12/02/2022]
Abstract
Fibronectin containing alternatively spliced EIIIA and EIIIB domains is largely absent from mature quiescent vessels in adults, but is highly expressed around blood vessels during developmental and pathological angiogenesis. The precise functions of fibronectin and its splice variants during developmental angiogenesis however remain unclear due to the presence of cardiac, somitic, mesodermal and neural defects in existing global fibronectin KO mouse models. Using a rare family of surviving EIIIA EIIIB double KO mice, as well as inducible endothelial-specific fibronectin-deficient mutant mice, we show that vascular development in the neonatal retina is regulated in an autocrine manner by endothelium-derived fibronectin, and requires both EIIIA and EIIIB domains and the RGD-binding α5 and αv integrins for its function. Exogenous sources of fibronectin do not fully substitute for the autocrine function of endothelial fibronectin, demonstrating that fibronectins from different sources contribute differentially to specific aspects of angiogenesis.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- University of Suffolk, James Hehir Building, University Avenue, Ipswich, Suffolk, IP3 0FS, UK
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- Duke University Medical Center, 307 Research Drive, Durham, NC, 27710, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA.
| |
Collapse
|
32
|
Counterbalancing anti-adhesive effects of Tenascin-C through fibronectin expression in endothelial cells. Sci Rep 2017; 7:12762. [PMID: 28986537 PMCID: PMC5630602 DOI: 10.1038/s41598-017-13008-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/12/2017] [Indexed: 12/22/2022] Open
Abstract
Cellular fibronectin (FN) and tenascin-C (TNC) are prominent development- and disease-associated matrix components with pro- and anti-adhesive activity, respectively. Whereas both are present in the tumour vasculature, their functional interplay on vascular endothelial cells remains unclear. We have previously shown that basally-oriented deposition of a FN matrix restricts motility and promotes junctional stability in cultured endothelial cells and that this effect is tightly coupled to expression of FN. Here we report that TNC induces FN expression in endothelial cells. This effect counteracts the potent anti-adhesive activity of TNC and leads to the assembly of a dense highly-branched subendothelial matrix that enhances tubulogenic activity. These findings suggest that pro-angiogenic remodelling of the perivascular matrix may involve TNC-induced upregulation of FN in endothelial cells.
Collapse
|
33
|
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: 3.9] [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
|
34
|
Guo X, Li W, Ma M, Lu X, Zhang H. Endothelial cell-derived matrix promotes the metabolic functional maturation of hepatocyte via integrin-Src signalling. J Cell Mol Med 2017; 21:2809-2822. [PMID: 28470937 PMCID: PMC5661128 DOI: 10.1111/jcmm.13195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/19/2017] [Indexed: 01/07/2023] Open
Abstract
The extracellular matrix (ECM) microenvironment is involved in the regulation of hepatocyte phenotype and function. Recently, the cell‐derived extracellular matrix has been proposed to represent the bioactive and biocompatible materials of the native ECM. Here, we show that the endothelial cell‐derived matrix (EC matrix) promotes the metabolic maturation of human adipose stem cell‐derived hepatocyte‐like cells (hASC‐HLCs) through the activation of the transcription factor forkhead box protein A2 (FOXA2) and the nuclear receptors hepatocyte nuclear factor 4 alpha (HNF4α) and pregnane X receptor (PXR). Reducing the fibronectin content in the EC matrix or silencing the expression of α5 integrin in the hASC‐HLCs inhibited the effect of the EC matrix on Src phosphorylation and hepatocyte maturation. The inhibition of Src phosphorylation using the inhibitor PP2 or silencing the expression of Src in hASC‐HLCs also attenuated the up‐regulation of the metabolic function of hASC‐HLCs in a nuclear receptor‐dependent manner. These data elucidate integrin‐Src signalling linking the extrinsic EC matrix signals and metabolic functional maturation of hepatocyte. This study provides a model for studying the interaction between hepatocytes and non‐parenchymal cell‐derived matrix.
Collapse
Affiliation(s)
- Xinyue Guo
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Weihong Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Minghui Ma
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Xin Lu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Haiyan Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Sens C, Huck K, Pettera S, Uebel S, Wabnitz G, Moser M, Nakchbandi IA. Fibronectins containing extradomain A or B enhance osteoblast differentiation via distinct integrins. J Biol Chem 2017; 292:7745-7760. [PMID: 28325836 DOI: 10.1074/jbc.m116.739987] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
Fibronectin is a multidomain protein secreted by various cell types. It forms a network of fibers within the extracellular matrix and impacts intracellular processes by binding to various molecules, primarily integrin receptors on the cells. Both the presence of several isoforms and the ability of the various domains and isoforms to bind to a variety of integrins result in a wide range of effects. In vivo findings suggest that fibronectin isoforms produced by the osteoblasts enhance their differentiation. Here we report that the isoform characterized by the presence of extradomain A activates α4β1 integrin and augments osteoblast differentiation. In addition, the isoform containing extradomain B enhances the binding of fibronectin through the RGD sequence to β3-containing integrin, resulting in increased mineralization by and differentiation of osteoblasts. Our study thus reveals novel functions for two fibronectin isoforms and the mediating receptors in osteoblast differentiation.
Collapse
Affiliation(s)
- Carla Sens
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and.,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Katrin Huck
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and.,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Pettera
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Stephan Uebel
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Guido Wabnitz
- the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Markus Moser
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and
| | - Inaam A Nakchbandi
- From the Max-Planck Institute of Biochemistry, 82152 Martinsried and .,the Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
36
|
|
37
|
Abstract
During cancer progression, the extracellular matrix (ECM) undergoes dramatic changes, which promote cancer cell migration and invasion. In the remodeled tumor ECM, fibronectin (FN) level is upregulated to assist tumor growth, progression, and invasion. FN serves as a central organizer of ECM molecules and mediates the crosstalk between the tumor microenvironment and cancer cells. Its upregulation is correlated with angiogenesis, cancer progression, metastasis, and drug resistance. A number of FN-targeting ligands have been developed for cancer imaging and therapy. Thus far, FN-targeting imaging agents have been tested for nuclear imaging, MRI, and fluorescence imaging, for tumor detection and localization. FN-targeting therapeutics, including nuclear medicine, chemotherapy drugs, cytokines, and photothermal moieties, were also developed in cancer therapy. Because of the prevalence of FN overexpression in cancer, FN targeting imaging agents and therapeutics have the promise of broad applications in the diagnosis, treatment, and image-guided interventions of many types of cancers. This review will summarize current understanding on the role of FN in cancer, discuss the design and development of FN-targeting agents, and highlight the applications of these FN-targeting agents in cancer imaging and therapy.
Collapse
Affiliation(s)
- Zheng Han
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
38
|
Gopal S, Veracini L, Grall D, Butori C, Schaub S, Audebert S, Camoin L, Baudelet E, Radwanska A, Beghelli-de la Forest Divonne S, Violette SM, Weinreb PH, Rekima S, Ilie M, Sudaka A, Hofman P, Van Obberghen-Schilling E. Fibronectin-guided migration of carcinoma collectives. Nat Commun 2017; 8:14105. [PMID: 28102238 PMCID: PMC5253696 DOI: 10.1038/ncomms14105] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 11/29/2016] [Indexed: 12/14/2022] Open
Abstract
Functional interplay between tumour cells and their neoplastic extracellular matrix plays a decisive role in malignant progression of carcinomas. Here we provide a comprehensive data set of the human HNSCC-associated fibroblast matrisome. Although much attention has been paid to the deposit of collagen, we identify oncofetal fibronectin (FN) as a major and obligate component of the matrix assembled by stromal fibroblasts from head and neck squamous cell carcinomas (HNSCC). FN overexpression in tumours from 435 patients corresponds to an independent unfavourable prognostic indicator. We show that migration of carcinoma collectives on fibrillar FN-rich matrices is achieved through αvβ6 and α9β1 engagement, rather than α5β1. Moreover, αvβ6-driven migration occurs independently of latent TGF-β activation and Smad-dependent signalling in tumour epithelial cells. These results provide insights into the adhesion-dependent events at the tumour–stroma interface that govern the collective mode of migration adopted by carcinoma cells to invade surrounding stroma in HNSCC. Tumour microenvironment influences the migration of cancer cells. Here the authors analyse the proteomic constitution of the extracellular matrix and identify a role for fibronectin in regulating the collective migration of squamous cell carcinoma cells.
Collapse
Affiliation(s)
- Sandeep Gopal
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | - Laurence Veracini
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | - Dominique Grall
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | - Catherine Butori
- Université Côte d'Azur, Laboratoire de Pathologie Clinique et Expérimentale, Biobank [BB-0033-00025] CHU Nice-Pasteur, 06001 Nice, France
| | - Sébastien Schaub
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | - Stéphane Audebert
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Luc Camoin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Emilie Baudelet
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Agata Radwanska
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | | | | | | | - Samah Rekima
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France
| | - Marius Ilie
- Université Côte d'Azur, Laboratoire de Pathologie Clinique et Expérimentale, Biobank [BB-0033-00025] CHU Nice-Pasteur, 06001 Nice, France
| | - Anne Sudaka
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France.,Centre Antoine Lacassagne, 06189 Nice, France
| | - Paul Hofman
- Université Côte d'Azur, Laboratoire de Pathologie Clinique et Expérimentale, Biobank [BB-0033-00025] CHU Nice-Pasteur, 06001 Nice, France
| | - Ellen Van Obberghen-Schilling
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose (iBV), Parc Valrose, 06100 Nice, France.,Centre Antoine Lacassagne, 06189 Nice, France
| |
Collapse
|
39
|
3-Hydroxyflavone inhibits human osteosarcoma U2OS and 143B cells metastasis by affecting EMT and repressing u-PA/MMP-2 via FAK-Src to MEK/ERK and RhoA/MLC2 pathways and reduces 143B tumor growth in vivo. Food Chem Toxicol 2016; 97:177-186. [PMID: 27600294 DOI: 10.1016/j.fct.2016.09.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 11/22/2022]
Abstract
Many natural flavonoids have cytostatic and apoptotic properties; however, we little know whether the effect of synthetic 3-hydroxyflavone on metastasis and tumor growth of human osteosarcoma. Here, we tested the hypothesis that 3-hydroxyflavone suppresses human osteosarcoma cells metastasis and tumor growth. 3-hydroxyflavone, up to 50 μM without cytotoxicity, inhibited U2OS and 143B cells motility, invasiveness and migration by reducing matrix metalloproteinase (MMP)-2 and urokinase-type plasminogen activator (u-PA) and also impaired cell adhesion to gelatin. 3-hydroxyflavone significantly reduced p-focal adhesion kinase (FAK) Tyr397, p-FAK Tyr925, p-steroid receptor coactivator (Src), p-mitogen/extracellular signal-regulated kinase (MEK)1/2, p-myosin light chain (MLC)2 Ser19, epithelial cell adhesion molecule, Ras homolog gene family (Rho)A and fibronectin expressions. 3-hydroxyflavone also affected the epithelial-mesenchymal transition (EMT) by down-regulating expressions of Vimentin and α-catenin with activation of the transcription factor Slug. In nude mice xenograft model and tail vein injection model showed that 3-hydroxyflavone reduced 143B tumor growth and lung metastasis. 3-hydroxyflavone possesses the anti-metastatic activity of U2OS and 143B cells by affecting EMT and repressing u-PA/MMP-2 via FAK-Src to MEK/ERK and RhoA/MLC2 pathways and suppresses 143B tumor growth in vivo. This may lead to clinical trials of osteosarcoma chemotherapy to confirm the promising result in the future.
Collapse
|
40
|
Huang SF, Horng CT, Hsieh YS, Hsieh YH, Chu SC, Chen PN. Epicatechin-3-gallate reverses TGF-β1-induced epithelial-to-mesenchymal transition and inhibits cell invasion and protease activities in human lung cancer cells. Food Chem Toxicol 2016; 94:1-10. [DOI: 10.1016/j.fct.2016.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/21/2016] [Accepted: 05/12/2016] [Indexed: 12/25/2022]
|
41
|
Veracini L, Grall D, Schaub S, Beghelli-de la Forest Divonne S, Etienne-Grimaldi MC, Milano G, Bozec A, Babin E, Sudaka A, Thariat J, Van Obberghen-Schilling E. Elevated Src family kinase activity stabilizes E-cadherin-based junctions and collective movement of head and neck squamous cell carcinomas. Oncotarget 2016; 6:7570-83. [PMID: 25779657 PMCID: PMC4480700 DOI: 10.18632/oncotarget.3071] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/23/2014] [Indexed: 11/25/2022] Open
Abstract
EGF receptor (EGFR) overexpression is thought to drive head and neck carcinogenesis however clinical responses to EGFR-targeting agents have been modest and alternate targets are actively sought to improve results. Src family kinases (SFKs), reported to act downstream of EGFR are among the alternative targets for which increased expression or activity in epithelial tumors is commonly associated to the dissolution of E-cadherin-based junctions and acquisition of a mesenchymal-like phenotype. Robust expression of total and activated Src was observed in advanced stage head and neck tumors (N=60) and in head and neck squamous cell carcinoma lines. In cultured cancer cells Src co-localized with E-cadherin in cell-cell junctions and its phosphorylation on Y419 was both constitutive and independent of EGFR activation. Selective inhibition of SFKs with SU6656 delocalized E-cadherin and disrupted cellular junctions without affecting E-cadherin expression and this effect was phenocopied by knockdown of Src or Yes. These findings reveal an EGFR-independent role for SFKs in the maintenance of intercellular junctions, which likely contributes to the cohesive invasion E-cadherin-positive cells in advanced tumors. Further, they highlight the need for a deeper comprehension of molecular pathways that drive collective cell invasion, in absence of mesenchymal transition, in order to combat tumor spread.
Collapse
Affiliation(s)
- Laurence Veracini
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France
| | - Dominique Grall
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France
| | - Sébastien Schaub
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France
| | - Stéphanie Beghelli-de la Forest Divonne
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France.,Department of Pathology, Centre Antoine Lacassagne, Nice, France
| | | | - Gérard Milano
- Laboratory of Oncopharmacology, Centre Antoine Lacassagne, Nice, France
| | - Alexandre Bozec
- Department of Otorhinolaryngology, Centre Antoine Lacassagne, Nice, France
| | - Emmanuel Babin
- Department of Otorhinolaryngology and Cervicofacial Surgery, CHU, Caen, France
| | - Anne Sudaka
- Department of Pathology, Centre Antoine Lacassagne, Nice, France
| | - Juliette Thariat
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France.,Laboratory of Oncopharmacology, Centre Antoine Lacassagne, Nice, France
| | - Ellen Van Obberghen-Schilling
- University of Nice Sophia Antipolis, UFR Sciences, Nice, France.,CNRS, UMR7277, Nice, France.,Inserm, U1091, Nice, France.,Department of Pathology, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
42
|
Brennan JR, Hocking DC. Cooperative effects of fibronectin matrix assembly and initial cell-substrate adhesion strength in cellular self-assembly. Acta Biomater 2016; 32:198-209. [PMID: 26712598 PMCID: PMC4754160 DOI: 10.1016/j.actbio.2015.12.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/24/2015] [Accepted: 12/18/2015] [Indexed: 11/17/2022]
Abstract
The cell-dependent polymerization of intercellular fibronectin fibrils can stimulate cells to self-assemble into multicellular structures. The local physical cues that support fibronectin-mediated cellular self-assembly are largely unknown. Here, fibronectin matrix analogs were used as synthetic adhesive substrates to model cell-matrix fibronectin fibrils having different integrin-binding specificity, affinity, and/or density. We utilized this model to quantitatively assess the relationship between adhesive forces derived from cell-substrate interactions and the ability of fibronectin fibril assembly to induce cellular self-assembly. Results indicate that the strength of initial, rather than mature, cell-substrate attachments correlates with the ability of substrates to support fibronectin-mediated cellular self-assembly. The cellular response to soluble fibronectin was bimodal and independent of the integrin-binding specificity of the substrate; increasing soluble fibronectin levels above a critical threshold increased aggregate cohesion on permissive substrates. Once aggregates formed, continuous fibronectin polymerization was necessary to maintain cohesion. During self-assembly, soluble fibronectin decreased cell-substrate adhesion strength and induced aggregate cohesion via a Rho-dependent mechanism, suggesting that the balance of contractile forces derived from fibronectin fibrils within cell-cell versus cell-substrate adhesions controls self-assembly and aggregate cohesion. Thus, initial cell-substrate attachment strength may provide a quantitative basis with which to build predictive models of fibronectin-mediated microtissue fabrication on a variety of substrates. STATEMENT OF SIGNIFICANCE Cellular self-assembly is a process by which cells and extracellular matrix (ECM) proteins spontaneously organize into three-dimensional (3D) tissues in the absence of external forces. Cellular self-assembly can be initiated in vitro, and represents a potential tool for tissue engineers to organize cells into modular building blocks for artificial tissue fabrication. Fibronectin is an ECM protein that plays a key role in tissue formation during embryonic development. Additionally, the cell-mediated process of converting soluble fibronectin into insoluble, ECM-associated fibrils has been shown to initiate cellular self-assembly in vitro. In this study, we examine the relationship between the strength of cell-substrate adhesions and the ability of fibronectin fibril assembly to induce cellular self-assembly. Our results indicate that substrate composition and density play cooperative roles with cell-mediated fibronectin matrix assembly to control the transition of cells from 2D monolayers into 3D multicellular aggregates. Results of this study provide a quantitative approach to build predictive models of cellular self-assembly, as well as a simple cell-culture platform to produce biomimetic units for modular tissue engineering.
Collapse
Affiliation(s)
- James R Brennan
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Denise C Hocking
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
43
|
Fernandez CE, Yen RW, Perez SM, Bedell HW, Povsic TJ, Reichert WM, Truskey GA. Human Vascular Microphysiological System for in vitro Drug Screening. Sci Rep 2016; 6:21579. [PMID: 26888719 PMCID: PMC4757887 DOI: 10.1038/srep21579] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/27/2016] [Indexed: 01/03/2023] Open
Abstract
In vitro human tissue engineered human blood vessels (TEBV) that exhibit vasoactivity can be used to test human toxicity of pharmaceutical drug candidates prior to pre-clinical animal studies. TEBVs with 400–800 μM diameters were made by embedding human neonatal dermal fibroblasts or human bone marrow-derived mesenchymal stem cells in dense collagen gel. TEBVs were mechanically strong enough to allow endothelialization and perfusion at physiological shear stresses within 3 hours after fabrication. After 1 week of perfusion, TEBVs exhibited endothelial release of nitric oxide, phenylephrine-induced vasoconstriction, and acetylcholine-induced vasodilation, all of which were maintained up to 5 weeks in culture. Vasodilation was blocked with the addition of the nitric oxide synthase inhibitor L-NG-Nitroarginine methyl ester (L-NAME). TEBVs elicited reversible activation to acute inflammatory stimulation by TNF-α which had a transient effect upon acetylcholine-induced relaxation, and exhibited dose-dependent vasodilation in response to caffeine and theophylline. Treatment of TEBVs with 1 μM lovastatin for three days prior to addition of Tumor necrosis factor – α (TNF-α) blocked the injury response and maintained vasodilation. These results indicate the potential to develop a rapidly-producible, endothelialized TEBV for microphysiological systems capable of producing physiological responses to both pharmaceutical and immunological stimuli.
Collapse
Affiliation(s)
- C E Fernandez
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - R W Yen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - S M Perez
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - H W Bedell
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - T J Povsic
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC 27708
| | - W M Reichert
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - G A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| |
Collapse
|
44
|
Vidak S, Kubben N, Dechat T, Foisner R. Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2α (LAP2α) through expression of extracellular matrix proteins. Genes Dev 2016; 29:2022-36. [PMID: 26443848 PMCID: PMC4604344 DOI: 10.1101/gad.263939.115] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A single heterozygous mutation of LMNA generates the lamin A/C variant progerin and causes Hutchinson-Gilford progeria syndrome (HGPS). Vidak et al. show that this mutation leads to loss of LAP2α and nucleoplasmic lamins A/C, impaired proliferation, and down-regulation of extracellular matrix components. Ectopic expression of LAP2α in cells expressing progerin restores proliferation and extracellular matrix expression but not the levels of nucleoplasmic lamins A/C. Lamina-associated polypeptide 2α (LAP2α) localizes throughout the nucleoplasm and interacts with the fraction of lamins A/C that is not associated with the peripheral nuclear lamina. The LAP2α–lamin A/C complex negatively affects cell proliferation. Lamins A/C are encoded by LMNA, a single heterozygous mutation of which causes Hutchinson-Gilford progeria syndrome (HGPS). This mutation generates the lamin A variant progerin, which we show here leads to loss of LAP2α and nucleoplasmic lamins A/C, impaired proliferation, and down-regulation of extracellular matrix components. Surprisingly, contrary to wild-type cells, ectopic expression of LAP2α in cells expressing progerin restores proliferation and extracellular matrix expression but not the levels of nucleoplasmic lamins A/C. We conclude that, in addition to its cell cycle-inhibiting function with lamins A/C, LAP2α can also regulate extracellular matrix components independently of lamins A/C, which may help explain the proliferation-promoting function of LAP2α in cells expressing progerin.
Collapse
Affiliation(s)
- Sandra Vidak
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Nard Kubben
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Thomas Dechat
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Roland Foisner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| |
Collapse
|
45
|
Yasuda N, Sekine H, Bise R, Okano T, Shimizu T. Tracing behavior of endothelial cells promotes vascular network formation. Microvasc Res 2015; 105:125-31. [PMID: 26687561 DOI: 10.1016/j.mvr.2015.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 12/08/2015] [Accepted: 12/08/2015] [Indexed: 12/12/2022]
Abstract
The in vitro formation of network structures derived from endothelial cells in grafts before transplantation contributes to earlier engraftment. In a previous study, endothelial cells migrated to form a net-shaped structure in co-culture. However, the specific network formation behavior of endothelial cells during migration remains unclear. In this study, we demonstrated the tracing behavior and cell cycle of endothelial cells using Fucci-labeled (Fluorescent Ubiquitination-based Cell Cycle Indicator) endothelial cells. Here, we observed the co-culture of Fucci-labeled human umbilical vein endothelial cells (HUVECs) together with normal human dermal fibroblasts (NHDFs) using time-lapse imaging and analyzed by multicellular concurrent tracking. In the G0/G1 period, HUVECs migrate faster than in the S/G2/M period, because G0/G1 is the mobile phase and S/G2/M is the proliferation phase in the cell cycle. When HUVECs are co-cultured, they tend to move randomly until they find existing tracks that they then follow to form clusters. Extracellular matrix (ECM) staining showed that collagen IV, laminin and thrombospondin deposited in accordance with endothelial cell networks. Therefore the HUVECs may migrate on the secreted ECM and exhibit tracing behavior, where the HUVECs migrate toward each other. These results suggested that ECM and a cell phase contributed to form a network by accelerating cell migration.
Collapse
Affiliation(s)
- Noriko Yasuda
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| | - Ryoma Bise
- Advanced Business Center, Dai Nippon Printing Co., Ltd., Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan.
| |
Collapse
|
46
|
Wang X, Astrof S. Neural crest cell-autonomous roles of fibronectin in cardiovascular development. Development 2015; 143:88-100. [PMID: 26552887 DOI: 10.1242/dev.125286] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022]
Abstract
The chemical and mechanical properties of extracellular matrices (ECMs) modulate diverse aspects of cellular fates; however, how regional heterogeneity in ECM composition regulates developmental programs is not well understood. We discovered that fibronectin 1 (Fn1) is expressed in strikingly non-uniform patterns during mouse development, suggesting that regionalized synthesis of the ECM plays cell-specific regulatory roles during embryogenesis. To test this hypothesis, we ablated Fn1 in the neural crest (NC), a population of multi-potent progenitors expressing high levels of Fn1. We found that Fn1 synthesized by the NC mediated morphogenesis of the aortic arch artery and differentiation of NC cells into vascular smooth muscle cells (VSMCs) by regulating Notch signaling. We show that NC Fn1 signals in an NC cell-autonomous manner through integrin α5β1 expressed by the NC, leading to activation of Notch and differentiation of VSMCs. Our data demonstrate an essential role of the localized synthesis of Fn1 in cardiovascular development and spatial regulation of Notch signaling.
Collapse
Affiliation(s)
- Xia Wang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Sophie Astrof
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| |
Collapse
|
47
|
Nätynki M, Kangas J, Miinalainen I, Sormunen R, Pietilä R, Soblet J, Boon LM, Vikkula M, Limaye N, Eklund L. Common and specific effects of TIE2 mutations causing venous malformations. Hum Mol Genet 2015; 24:6374-89. [PMID: 26319232 DOI: 10.1093/hmg/ddv349] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 08/24/2015] [Indexed: 01/09/2023] Open
Abstract
Venous malformations (VMs) are localized defects in vascular morphogenesis frequently caused by mutations in the gene for the endothelial tyrosine kinase receptor TIE2. Here, we report the analysis of a comprehensive collection of 22 TIE2 mutations identified in patients with VM, either as single amino acid substitutions or as double-mutations on the same allele. Using endothelial cell (EC) cultures, mouse models and ultrastructural analysis of tissue biopsies from patients, we demonstrate common as well as mutation-specific cellular and molecular features, on the basis of which mutations cluster into categories that correlate with data from genetic studies. Comparisons of double-mutants with their constituent single-mutant forms identified the pathogenic contributions of individual changes, and their compound effects. We find that defective receptor trafficking and subcellular localization of different TIE2 mutant forms occur via a variety of mechanisms, resulting in attenuated response to ligand. We also demonstrate, for the first time, that TIE2 mutations cause chronic activation of the MAPK pathway resulting in loss of normal EC monolayer due to extracellular matrix (ECM) fibronectin deficiency and leading to upregulation of plasminogen/plasmin proteolytic pathway. Corresponding EC and ECM irregularities are observed in affected tissues from mouse models and patients. Importantly, an imbalance between plasminogen activators versus inhibitors would also account for high d-dimer levels, a major feature of unknown cause that distinguishes VMs from other vascular anomalies.
Collapse
Affiliation(s)
- Marjut Nätynki
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jaakko Kangas
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | - Raija Sormunen
- Biocenter Oulu, University of Oulu, Oulu, Finland, Department of Pathology and Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Riikka Pietilä
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Julie Soblet
- Human Molecular Genetics, de Duve Institute, and
| | - Laurence M Boon
- Human Molecular Genetics, de Duve Institute, and Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | - Nisha Limaye
- Human Molecular Genetics, de Duve Institute, and
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland,
| |
Collapse
|
48
|
Sun Y, Kim HS, Saw PE, Jon S, Moon WK. Targeted Therapy for Breast Cancer Stem Cells by Liposomal Delivery of siRNA against Fibronectin EDB. Adv Healthc Mater 2015; 4:1675-80. [PMID: 26097122 DOI: 10.1002/adhm.201500190] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/20/2015] [Indexed: 12/20/2022]
Abstract
Targeted therapy for breast cancer stem cell (BCSC): A novel liposomal system (APTEDB -LS-siRNA(EDB) ) that enables simultaneous targeting and knockdown of extra domain B of fibronectin (EDB-FN) shows potent therapeutic efficacy in the BCSC-derived tumors in vivo.
Collapse
Affiliation(s)
- Yujin Sun
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro, Jongno-gu Seoul 110-744 South Korea
- Department of Radiology; Yanbian University Hospital; 1327 JuZi Street Yanji City JiLin Province 133000 China
| | - Hoe Suk Kim
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro, Jongno-gu Seoul 110-744 South Korea
| | - Phei Er Saw
- Korea Advanced Institute of Science and Technology (KAIST); 291 Daehak-ro, Yuseong-gu Daejeon 305-701 South Korea
| | - Sangyong Jon
- Korea Advanced Institute of Science and Technology (KAIST); 291 Daehak-ro, Yuseong-gu Daejeon 305-701 South Korea
| | - Woo Kyung Moon
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro, Jongno-gu Seoul 110-744 South Korea
| |
Collapse
|
49
|
Tissue stiffness regulates serine/arginine-rich protein-mediated splicing of the extra domain B-fibronectin isoform in tumors. Proc Natl Acad Sci U S A 2015; 112:8314-9. [PMID: 26106154 DOI: 10.1073/pnas.1505421112] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alternative splicing of proteins gives rise to different isoforms that play a crucial role in regulating several cellular processes. Notably, splicing profiles are altered in several cancer types, and these profiles are believed to be involved in driving the oncogenic process. Although the importance of alternative splicing alterations occurring during cancer is increasingly appreciated, the underlying regulatory mechanisms remain poorly understood. In this study, we use both biochemical and physical tools coupled with engineered models, patient samples, and a murine model to investigate the role of the mechanical properties of the tumor microenvironment in regulating the production of the extra domain-B (EDB) splice variant of fibronectin (FN), a hallmark of tumor angiogenesis. Specifically, we show that the amount of EDB-FN produced by endothelial cells increases with matrix stiffness both in vitro and within mouse mammary tumors. Matrix stiffness regulates splicing through the activation of serine/arginine rich (SR) proteins, the splicing factors involved in the production of FN isoforms. Activation of the SR proteins by matrix stiffness and the subsequent production of EDB-FN are dependent on intracellular contractility and PI3K-AKT signaling. Notably, matrix stiffness-mediated splicing is not limited to EDB-FN, but also affects splicing in the production of PKC βII and the VEGF 165b splice variant. Together, these results demonstrate that the mechanical properties of the microenvironment regulate alternative splicing and establish a previously unidentified mechanism by which cells can adapt to their microenvironment.
Collapse
|
50
|
Yoneda A. Fibronectin Matrix Assembly and Its Significant Role in Cancer Progression and Treatment. TRENDS GLYCOSCI GLYC 2015. [DOI: 10.4052/tigg.1421.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Atsuko Yoneda
- Laboratory of Genome and Biosignals, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|