1
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Sun Y, Hamlin AJ, Schwarzbauer JE. Fibronectin matrix assembly at a glance. J Cell Sci 2025; 138:jcs263834. [PMID: 40130407 PMCID: PMC12050093 DOI: 10.1242/jcs.263834] [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] [Indexed: 03/26/2025] Open
Abstract
The organization and mechanics of extracellular matrix (ECM) protein polymers determine tissue structure and function. Secreted ECM components are assembled into polymers via a cell-mediated process. The specific mechanisms that cells use for assembly are crucial for generating tissue-appropriate matrices. Fibronectin (FN) is a ubiquitous and abundant ECM protein that is assembled into a fibrillar matrix by a receptor-mediated process, and the FN matrix provides a foundation for incorporation of many other proteins into the ECM. In this Cell Science at a Glance article and the accompanying poster, we describe the domain organization of FN and the events that initiate and propagate a stable insoluble network of FN fibrils. We also discuss intracellular pathways that regulate FN assembly and the impact of changes in assembly on disease progression.
Collapse
Affiliation(s)
- Yu Sun
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Aaron J. Hamlin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
3
|
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 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
4
|
Loreti M, Cecchini A, Kaufman CD, Stamenkovic C, Renero A, Nicoletti C, Kervadec A, Guarnaccia G, Mayer D, Colas A, Lorenzo Puri P, Sacco A. Tenascin-C from the tissue microenvironment promotes muscle stem cell self-renewal through Annexin A2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620732. [PMID: 39554125 PMCID: PMC11565721 DOI: 10.1101/2024.10.29.620732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Skeletal muscle tissue self-repair occurs through the finely timed activation of resident muscle stem cells (MuSC). Following perturbation, MuSC exit quiescence, undergo myogenic commitment, and differentiate to regenerate the injured muscle. This process is coordinated by signals present in the tissue microenvironment, however the precise mechanisms by which the microenvironment regulates MuSC activation are still poorly understood. Here, we identified Tenascin-C (TnC), an extracellular matrix (ECM) glycoprotein, as a key player in promoting of MuSC self-renewal and function. We show that fibro-adipogenic progenitors (FAPs) are the primary cellular source of TnC during muscle repair, and that MuSC sense TnC signaling through cell the surface receptor Annexin A2. We provide in vivo evidence that TnC is required for efficient muscle repair, as mice lacking TnC exhibit a regeneration phenotype of premature aging. We propose that the decline of TnC in physiological aging contributes to inefficient muscle regeneration in aged muscle. Taken together, our results highlight the pivotal role of TnC signaling during muscle repair in healthy and aging skeletal muscle.
Collapse
Affiliation(s)
- Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: J&J, 3880 Murphy Canyon Rd, San Diego, CA 92123, USA
| | - Alessandra Cecchini
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Collin D. Kaufman
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Cedomir Stamenkovic
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alma Renero
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Chiara Nicoletti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Anais Kervadec
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Current affiliation: Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, CA 92121, USA
| | - Gabriele Guarnaccia
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Daphne Mayer
- Rice University, 6100 Main St, Huston, TX 77005, USA
| | - Alexandre Colas
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Sun Y, King B, Hamlin AJ, Saniepay M, Gorshkov K, Barker G, Ziegler M, Mukundan S, Cvijic ME, Schwarzbauer JE. Identification of a fibronectin-binding protein signature associated with idiopathic pulmonary fibrosis. Cells Dev 2024; 179:203941. [PMID: 39038657 PMCID: PMC11344656 DOI: 10.1016/j.cdev.2024.203941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
The extracellular matrix (ECM) is a critical component of tissue where it provides structural and signaling support to cells. Its dysregulation and accumulation lead to fibrosis, a major clinical challenge underlying many diseases that currently has little effective treatment. An understanding of the key molecular initiators of fibrosis would be both diagnostically useful and provide potential targets for therapeutics. The ECM protein fibronectin (FN) is upregulated in fibrotic conditions and other ECM proteins depend on assembly of a FN foundational ECM for their matrix incorporation. We used cell culture and in vivo models to investigate the role of FN in the progression of lung fibrosis. We confirmed that normal human lung fibroblasts (NHLFs) treated with transforming growth factor-beta (TGF-β) to stimulate fibrotic gene expression significantly increased both FN expression and its assembly into a matrix. We found that levels of alternatively spliced EDA and EDB exons were proportional to the increase in total FN RNA and protein showing that inclusion of these exons is not enhanced by TGF-β stimulation. RNA-sequencing identified 43 core matrisome genes that were significantly up- or down-regulated by TGF-β treatment and a Luminex immunoassay demonstrated increased levels of ECM proteins in conditioned medium of TGF-β-treated NHLFs. Interestingly, among the regulated core matrisome genes, 16 encode known FN-binding proteins and, of these, insulin-like growth factor binding protein 3 (IGFBP3) was most highly up-regulated. To link the NHLF results with in vivo disease, we analyzed lung tissue and bronchoalveolar lavage fluid from bleomycin-treated mice and found dramatically higher levels of FN and the FN-binding proteins IGFBP3, tenascin-C, and type I collagen in fibrotic conditions compared to controls. Altogether, our data identify a set of FN-binding proteins whose upregulation is characteristic of IPF and suggest that FN provides the foundational matrix for deposition of these proteins as fibrosis develops.
Collapse
Affiliation(s)
- Yu Sun
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA 08544
| | - Benjamin King
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Aaron J. Hamlin
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA 08544
| | - Mersedeh Saniepay
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Kirill Gorshkov
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Gregory Barker
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Milinda Ziegler
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Shilpaa Mukundan
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Mary Ellen Cvijic
- Leads Discovery and Optimization, Bristol Myers Squibb, Lawrenceville, New Jersey, USA 08648
| | - Jean E. Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA 08544
| |
Collapse
|
6
|
De Magalhães CG, Cvekl A, Jaeger RG, Yan CYI. Lens placode modulates extracellular matrix formation during early eye development. Differentiation 2024; 138:100792. [PMID: 38935992 PMCID: PMC11247415 DOI: 10.1016/j.diff.2024.100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
The role extracellular matrix (ECM) in multiple events of morphogenesis has been well described, little is known about its specific role in early eye development. One of the first morphogenic events in lens development is placodal thickening, which converts the presumptive lens ectoderm from cuboidal to pseudostratified epithelium. This process occurs in the anterior pre-placodal ectoderm when the optic vesicle approaches the cephalic ectoderm and is regulated by transcription factor Pax6 and secreted BMP4. Since cells and ECM have a dynamic relationship of interdependence and modulation, we hypothesized that the ECM evolves with cell shape changes during lens placode formation. This study investigates changes in optic ECM including both protein distribution deposition, extracellular gelatinase activity and gene expression patterns during early optic development using chicken and mouse models. In particular, the expression of Timp2, a metalloprotease inhibitor, corresponds with a decrease in gelatinase activity within the optic ECM. Furthermore, we demonstrate that optic ECM remodeling depends on BMP signaling in the placode. Together, our findings suggest that the lens placode plays an active role in remodeling the optic ECM during early eye development.
Collapse
Affiliation(s)
- Cecília G De Magalhães
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - Ales Cvekl
- Department of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruy G Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil
| | - C Y Irene Yan
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
7
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M. Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E. Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
8
|
De Magalhães CG, Cvekl A, Jaeger RG, Yan CYI. Lens Placode Modulates Extracellular Matrix Formation During Early Eye Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.30.569417. [PMID: 38076974 PMCID: PMC10705410 DOI: 10.1101/2023.11.30.569417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The role extracellular matrix (ECM) in multiple events of morphogenesis has been well described, little is known about its specific role in early eye development. One of the first morphogenic events in lens development is placodal thickening, which converts the presumptive lens ectoderm from cuboidal to pseudostratified epithelium. This process occurs in the anterior pre-placodal ectoderm when the optic vesicle approaches the cephalic ectoderm. Since cells and ECM have a dynamic relationship of interdependence and modulation, we hypothesized that the ECM evolves with cell shape changes during lens placode formation. This study investigates changes in optic ECM including both protein distribution deposition, extracellular gelatinase activity and gene expression patterns during early optic development using chicken and mouse models. In particular, the expression of Timp2 , a metalloprotease inhibitor, corresponds with a decrease in gelatinase activity within the optic ECM. Furthermore, we demonstrate that optic ECM remodeling depends on BMP signaling in the placode. Together, our findings suggest that the lens placode plays an active role in remodeling the optic ECM during early eye development.
Collapse
|
9
|
Abedsaeidi M, Hojjati F, Tavassoli A, Sahebkar A. Biology of Tenascin C and its Role in Physiology and Pathology. Curr Med Chem 2024; 31:2706-2731. [PMID: 37021423 DOI: 10.2174/0929867330666230404124229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 01/25/2023] [Accepted: 02/10/2023] [Indexed: 04/07/2023]
Abstract
Tenascin-C (TNC) is a multimodular extracellular matrix (ECM) protein hexameric with several molecular forms (180-250 kDa) produced by alternative splicing at the pre-mRNA level and protein modifications. The molecular phylogeny indicates that the amino acid sequence of TNC is a well-conserved protein among vertebrates. TNC has binding partners, including fibronectin, collagen, fibrillin-2, periostin, proteoglycans, and pathogens. Various transcription factors and intracellular regulators tightly regulate TNC expression. TNC plays an essential role in cell proliferation and migration. Unlike embryonic tissues, TNC protein is distributed over a few tissues in adults. However, higher TNC expression is observed in inflammation, wound healing, cancer, and other pathological conditions. It is widely expressed in a variety of human malignancies and is recognized as a pivotal factor in cancer progression and metastasis. Moreover, TNC increases both pro-and anti-inflammatory signaling pathways. It has been identified as an essential factor in tissue injuries such as damaged skeletal muscle, heart disease, and kidney fibrosis. This multimodular hexameric glycoprotein modulates both innate and adaptive immune responses regulating the expression of numerous cytokines. Moreover, TNC is an important regulatory molecule that affects the onset and progression of neuronal disorders through many signaling pathways. We provide a comprehensive overview of the structural and expression properties of TNC and its potential functions in physiological and pathological conditions.
Collapse
Affiliation(s)
- Malihehsadat Abedsaeidi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Farzaneh Hojjati
- Division of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Amin Tavassoli
- Division of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Sharma A, Hill KE, Schwarzbauer JE. Extracellular matrix composition affects outgrowth of dendrites and dendritic spines on cortical neurons. Front Cell Neurosci 2023; 17:1177663. [PMID: 37388410 PMCID: PMC10300442 DOI: 10.3389/fncel.2023.1177663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
The composition of the extracellular matrix (ECM) in nervous tissue plays an important role in controlling neuronal outgrowth and synapse development. Changes in both protein and glycosaminoglycan components of the ECM occur with tissue injury and may affect neuron growth. To investigate neuron responses to alterations in fibronectin (FN), a major component of the wound ECM, we grew cortical neurons on cell-derived decellularized matrices composed of wild type FN (FN+/+) or of a mutant form of FN (FNΔ/+) from which the III13 heparin-binding site had been deleted by CRISPR-Cas 9 gene editing. The most significant effect of the mutant FN was a reduction in dendrite outgrowth. Not only were dendrites shorter on mutant FNΔ/+-collagen (COL) matrix than on wild type (FN+/+-COL) matrix, but the number of dendrites and dendritic spines per neuron and the spine densities were also dramatically reduced on FNΔ/+-COL matrices. Mass spectrometry and immunostaining identified a reduction in tenascin-C (TN-C) levels in the mutant matrix. TN-C is an ECM protein that binds to the III13 site of FN and modulates cell-matrix interactions and has been linked to dendrite development. We propose that TN-C binding to FN in the wound matrix supports dendrite and spine development during repair of damaged neural tissue. Overall, these results show that changes in ECM composition can dramatically affect elaboration of neurites and support the idea that the ECM microenvironment controls neuron morphology and connectivity.
Collapse
Affiliation(s)
| | | | - Jean E. Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
11
|
Benn MC, Pot SA, Moeller J, Yamashita T, Fonta CM, Orend G, Kollmannsberger P, Vogel V. How the mechanobiology orchestrates the iterative and reciprocal ECM-cell cross-talk that drives microtissue growth. SCIENCE ADVANCES 2023; 9:eadd9275. [PMID: 36989370 PMCID: PMC10058249 DOI: 10.1126/sciadv.add9275] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Controlled tissue growth is essential for multicellular life and requires tight spatiotemporal control over cell proliferation and differentiation until reaching homeostasis. As cells synthesize and remodel extracellular matrix, tissue growth processes can only be understood if the reciprocal feedback between cells and their environment is revealed. Using de novo-grown microtissues, we identified crucial actors of the mechanoregulated events, which iteratively orchestrate a sharp transition from tissue growth to maturation, requiring a myofibroblast-to-fibroblast transition. Cellular decision-making occurs when fibronectin fiber tension switches from highly stretched to relaxed, and it requires the transiently up-regulated appearance of tenascin-C and tissue transglutaminase, matrix metalloprotease activity, as well as a switch from α5β1 to α2β1 integrin engagement and epidermal growth factor receptor signaling. As myofibroblasts are associated with wound healing and inflammatory or fibrotic diseases, crucial knowledge needed to advance regenerative strategies or to counter fibrosis and cancer progression has been gained.
Collapse
Affiliation(s)
- Mario C. Benn
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Simon A. Pot
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Tadahiro Yamashita
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Charlotte M. Fonta
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France
- Université Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Philip Kollmannsberger
- Biomedical Physics, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| |
Collapse
|
12
|
Bijelić D, Adžić M, Perić M, Reiss G, Milošević M, Andjus PR, Jakovčevski I. Tenascin-C fibronectin D domain is involved in the fine-tuning of glial response to CNS injury in vitro. Front Cell Dev Biol 2022; 10:952208. [PMID: 36092707 PMCID: PMC9462431 DOI: 10.3389/fcell.2022.952208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding processes that occur after injuries to the central nervous system is essential in order to gain insight into how the restoration of function can be improved. Extracellular glycoprotein tenascin-C (TnC) has numerous functions in wound healing process depending on the expression time, location, isoform and binding partners which makes it interesting to study in this context. We used an in vitro injury model, the mixed culture of cortical astrocytes and microglia, and observed that without TnC microglial cells tend to populate gap area in greater numbers and proliferate more, whereas astrocytes build up in the border region to promote faster gap closure. Alternatively spliced domain of TnC, fibronectin type III-like repeat D (FnD) strongly affected physiological properties and morphology of both astrocytes and microglia in this injury model. The rate of microglial proliferation in the injury region decreased significantly with the addition of FnD. Additionally, density of microglia also decreased, in part due to reduced proliferation, and possibly due to reduced migration and increased contact inhibition between enlarged FnD-treated cells. Overall morphology of FnD-treated microglia resembled the activated pro-inflammatory cells, and elevated expression of iNOS was in accordance with this phenotype. The effect of FnD on astrocytes was different, as it did not affect their proliferation, but stimulated migration of reactivated astrocytes into the scratched area 48 h after the lesion. Elevated expression and secretion of TNF-α and IL-1β upon FnD treatment indicated the onset of inflammation. Furthermore, on Western blots we observed increased intensity of precursor bands of β1 integrin and appearance of monomeric bands of P2Y12R after FnD treatment which substantiates and clarifies its role in cellular shape and motility changes. Our results show versatile functions of TnC and in particular FnD after injury, mostly contributing to ongoing inflammation in the injury region. Based on our findings, FnD might be instrumental in limiting immune cell infiltration, and promoting astrocyte migration within the injury region, thus influencing spaciotemporal organization of the wound and surrounding area.
Collapse
Affiliation(s)
- Dunja Bijelić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Dunja Bijelić, ; Igor Jakovčevski,
| | - Marija Adžić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Mina Perić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| | - Milena Milošević
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Igor Jakovčevski
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| |
Collapse
|
13
|
Hammond NL, Dixon MJ. Revisiting the embryogenesis of lip and palate development. Oral Dis 2022; 28:1306-1326. [PMID: 35226783 PMCID: PMC10234451 DOI: 10.1111/odi.14174] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022]
Abstract
Clefts of the lip and palate (CLP), the major causes of congenital facial malformation globally, result from failure of fusion of the facial processes during embryogenesis. With a prevalence of 1 in 500-2500 live births, CLP causes major morbidity throughout life as a result of problems with facial appearance, feeding, speaking, obstructive apnoea, hearing and social adjustment and requires complex, multi-disciplinary care at considerable cost to healthcare systems worldwide. Long-term outcomes for affected individuals include increased mortality compared with their unaffected siblings. The frequent occurrence and major healthcare burden imposed by CLP highlight the importance of dissecting the molecular mechanisms driving facial development. Identification of the genetic mutations underlying syndromic forms of CLP, where CLP occurs in association with non-cleft clinical features, allied to developmental studies using appropriate animal models is central to our understanding of the molecular events underlying development of the lip and palate and, ultimately, how these are disturbed in CLP.
Collapse
Affiliation(s)
- Nigel L. Hammond
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Michael J. Dixon
- Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
14
|
Zhang Q, Wang P, Fang X, Lin F, Fang J, Xiong C. Collagen gel contraction assays: From modelling wound healing to quantifying cellular interactions with three-dimensional extracellular matrices. Eur J Cell Biol 2022; 101:151253. [PMID: 35785635 DOI: 10.1016/j.ejcb.2022.151253] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 06/06/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022] Open
Abstract
Cells respond to and actively remodel the extracellular matrix (ECM). The dynamic and bidirectional interaction between cells and ECM, especially their mechanical interactions, has been found to play an essential role in triggering a series of complex biochemical and biomechanical signal pathways and in regulating cellular functions and behaviours. The collagen gel contraction assay (CGCA) is a widely used method to investigate cell-ECM interactions in 3D environments and provides a mechanically associated readout reflecting 3D cellular contractility. In this review, we summarize various versions of CGCA, with an emphasis on recent high-throughput and low-consumption CGCA techniques. More importantly, we focus on the technique of force monitoring during the contraction of collagen gel, which provides a quantitative characterization of the overall forces generated by all the resident cells in the collagen hydrogel. Accordingly, we present recent biological applications of the CGCA, which have expanded from the initial wound healing model to other studies concerning cell-ECM interactions, including fibrosis, cancer, tissue repair and the preparation of biomimetic microtissues.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Pudi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Xu Fang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feng Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Jing Fang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
15
|
Rogers JD, Richardson WJ. Fibroblast mechanotransduction network predicts targets for mechano-adaptive infarct therapies. eLife 2022; 11:e62856. [PMID: 35138248 PMCID: PMC8849334 DOI: 10.7554/elife.62856] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Regional control of fibrosis after myocardial infarction is critical for maintaining structural integrity in the infarct while preventing collagen accumulation in non-infarcted areas. Cardiac fibroblasts modulate matrix turnover in response to biochemical and biomechanical cues, but the complex interactions between signaling pathways confound efforts to develop therapies for regional scar formation. We employed a logic-based ordinary differential equation model of fibroblast mechano-chemo signal transduction to predict matrix protein expression in response to canonical biochemical stimuli and mechanical tension. Functional analysis of mechano-chemo interactions showed extensive pathway crosstalk with tension amplifying, dampening, or reversing responses to biochemical stimuli. Comprehensive drug target screens identified 13 mechano-adaptive therapies that promote matrix accumulation in regions where it is needed and reduce matrix levels in regions where it is not needed. Our predictions suggest that mechano-chemo interactions likely mediate cell behavior across many tissues and demonstrate the utility of multi-pathway signaling networks in discovering therapies for context-specific disease states.
Collapse
Affiliation(s)
- Jesse D Rogers
- Department of Bioengineering; Clemson UniversityClemsonUnited States
| | | |
Collapse
|
16
|
Atia L, Fredberg JJ, Gov NS, Pegoraro AF. Are cell jamming and unjamming essential in tissue development? Cells Dev 2021; 168:203727. [PMID: 34363993 PMCID: PMC8935248 DOI: 10.1016/j.cdev.2021.203727] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/07/2021] [Accepted: 07/28/2021] [Indexed: 11/25/2022]
Abstract
The last decade has seen a surge of evidence supporting the existence of the transition of the multicellular tissue from a collective material phase that is regarded as being jammed to a collective material phase that is regarded as being unjammed. The jammed phase is solid-like and effectively 'frozen', and therefore is associated with tissue homeostasis, rigidity, and mechanical stability. The unjammed phase, by contrast, is fluid-like and effectively 'melted', and therefore is associated with mechanical fluidity, plasticity and malleability that are required in dynamic multicellular processes that sculpt organ microstructure. Such multicellular sculpturing, for example, occurs during embryogenesis, growth and remodeling. Although unjamming and jamming events in the multicellular collective are reminiscent of those that occur in the inert granular collective, such as grain in a hopper that can flow or clog, the analogy is instructive but limited, and the implications for cell biology remain unclear. Here we ask, are the cellular jamming transition and its inverse --the unjamming transition-- mere epiphenomena? That is, are they dispensable downstream events that accompany but neither cause nor quench these core multicellular processes? Drawing from selected examples in developmental biology, here we suggest the hypothesis that, to the contrary, the graded departure from a jammed phase enables controlled degrees of malleability as might be required in developmental dynamics. We further suggest that the coordinated approach to a jammed phase progressively slows those dynamics and ultimately enables long-term mechanical stability as might be required in the mature homeostatic multicellular tissue.
Collapse
Affiliation(s)
- Lior Atia
- Department of Mechanical Engineering, Ben Gurion University, Beer-Sheva, Israel
| | - Jeffrey J Fredberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Nir S Gov
- Department of Chemical and Biological Physics, Weizmann Institute, Israel
| | | |
Collapse
|
17
|
Halper J. Basic Components of Connective Tissues and Extracellular Matrix: Fibronectin, Fibrinogen, Laminin, Elastin, Fibrillins, Fibulins, Matrilins, Tenascins and Thrombospondins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:105-126. [PMID: 34807416 DOI: 10.1007/978-3-030-80614-9_4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Collagens are the most abundant components of the extracellular matrix (ECM) and many types of soft tissues. Elastin is another major component of certain soft tissues, such as arterial walls and ligaments. It is an insoluble polymer of the monomeric soluble precursor tropoelastin, and the main component of elastic fibers in matrix tissue where it provides elastic recoil and resilience to a variety of connective tissues, e.g., aorta and ligaments. Elastic fibers regulate activity of transforming growth factors β (TGFβ) through their association with fibrillin microfibrils. Elastin also plays a role in cell adhesion, cell migration, and has the ability to participate in cell signaling. Mutations in the elastin gene lead to cutis laxa. Many other molecules, though lower in quantity, function as essential, structural and/or functional components of the extracellular matrix in soft tissues. Some of these are reviewed in this chapter. Besides their basic structure, biochemistry and physiology, their roles in disorders of soft tissues are discussed only briefly as most chapters in this volume deal with relevant individual compounds. Fibronectin with its multidomain structure plays a role of "master organizer" in matrix assembly as it forms a bridge between cell surface receptors, e.g., integrins, and compounds such collagen, proteoglycans and other focal adhesion molecules. It also plays an essential role in the assembly of fibrillin-1 into a structured network. Though the primary role of fibrinogen is in clot formation, after conversion to fibrin by thrombin it also binds to a variety of compounds, particularly to various growth factors, and as such, fibrinogen is a player in cardiovascular and extracellular matrix physiology. Laminins contribute to the structure of the ECM and modulate cellular functions such as adhesion, differentiation, migration, stability of phenotype, and resistance towards apoptosis. Fibrillins represent the predominant core of microfibrils in elastic as well as non-elastic extracellular matrixes, and interact closely with tropoelastin and integrins. Not only do microfibrils provide structural integrity of specific organ systems, but they also provide basis for elastogenesis in elastic tissues. Fibrillin is important for the assembly of elastin into elastic fibers. Mutations in the fibrillin-1 gene are closely associated with Marfan syndrome. Latent TGFβ binding proteins (LTBPs) are included here as their structure is similar to fibrillins. Several categories of ECM components described after fibrillins are sub-classified as matricellular proteins, i.e., they are secreted into ECM, but do not provide structure. Rather they interact with cell membrane receptors, collagens, proteases, hormones and growth factors, communicating and directing cell-ECM traffic. Fibulins are tightly connected with basement membranes, elastic fibers and other components of extracellular matrix and participate in formation of elastic fibers. Matrilins have been emerging as a new group of supporting actors, and their role in connective tissue physiology and pathophysiology has not been fully characterized. Tenascins are ECM polymorphic glycoproteins found in many connective tissues in the body. Their expression is regulated by mechanical stress both during development and in adulthood. Tenascins mediate both inflammatory and fibrotic processes to enable effective tissue repair and play roles in pathogenesis of Ehlers-Danlos, heart disease, and regeneration and recovery of musculo-tendinous tissue. One of the roles of thrombospondin 1 is activation of TGFβ. Increased expression of thrombospondin and TGFβ activity was observed in fibrotic skin disorders such as keloids and scleroderma. Cartilage oligomeric matrix protein (COMP) or thrombospondin-5 is primarily present in the cartilage. High levels of COMP are present in fibrotic scars and systemic sclerosis of the skin, and in tendon, especially with physical activity, loading and post-injury. It plays a role in vascular wall remodeling and has been found in atherosclerotic plaques as well.
Collapse
Affiliation(s)
- Jaroslava Halper
- Department of Pathology, College of Veterinary Medicine, and Department of Basic Sciences, AU/UGA Medical Partnership, The University of Georgia, Athens, GA, USA.
| |
Collapse
|
18
|
Structural and Functional Modulation of Perineuronal Nets: In Search of Important Players with Highlight on Tenascins. Cells 2021; 10:cells10061345. [PMID: 34072323 PMCID: PMC8230358 DOI: 10.3390/cells10061345] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the brain plays a crucial role in providing optimal conditions for neuronal function. Interactions between neurons and a specialized form of ECM, perineuronal nets (PNN), are considered a key mechanism for the regulation of brain plasticity. Such an assembly of interconnected structural and regulatory molecules has a prominent role in the control of synaptic plasticity. In this review, we discuss novel ways of studying the interplay between PNN and its regulatory components, particularly tenascins, in the processes of synaptic plasticity, mechanotransduction, and neurogenesis. Since enhanced neuronal activity promotes PNN degradation, it is possible to study PNN remodeling as a dynamical change in the expression and organization of its constituents that is reflected in its ultrastructure. The discovery of these subtle modifications is enabled by the development of super-resolution microscopy and advanced methods of image analysis.
Collapse
|
19
|
Tucić M, Stamenković V, Andjus P. The Extracellular Matrix Glycoprotein Tenascin C and Adult Neurogenesis. Front Cell Dev Biol 2021; 9:674199. [PMID: 33996833 PMCID: PMC8117239 DOI: 10.3389/fcell.2021.674199] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Tenascin C (TnC) is a glycoprotein highly expressed in the extracellular matrix (ECM) during development and in the adult central nervous system (CNS) in regions of active neurogenesis, where neuron development is a tightly regulated process orchestrated by extracellular matrix components. In addition, newborn cells also communicate with glial cells, astrocytes and microglia, indicating the importance of signal integration in adult neurogenesis. Although TnC has been recognized as an important molecule in the regulation of cell proliferation and migration, complete regulatory pathways still need to be elucidated. In this review we discuss the formation of new neurons in the adult hippocampus and the olfactory system with specific reference to TnC and its regulating functions in this process. Better understanding of the ECM signaling in the niche of the CNS will have significant implications for regenerative therapies.
Collapse
Affiliation(s)
- Milena Tucić
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vera Stamenković
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry "Jean Giaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
20
|
Li W, Sancho A, Chung WL, Vinik Y, Groll J, Zick Y, Medalia O, Bershadsky AD, Geiger B. Differential cellular responses to adhesive interactions with galectin-8- and fibronectin-coated substrates. J Cell Sci 2021; 134:jcs252221. [PMID: 33722978 PMCID: PMC8106957 DOI: 10.1242/jcs.252221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
The mechanisms underlying the cellular response to extracellular matrices (ECMs) that consist of multiple adhesive ligands are still poorly understood. Here, we address this topic by monitoring specific cellular responses to two different extracellular adhesion molecules - the main integrin ligand fibronectin and galectin-8, a lectin that binds β-galactoside residues - as well as to mixtures of the two proteins. Compared with cell spreading on fibronectin, cell spreading on galectin-8-coated substrates resulted in increased projected cell area, more-pronounced extension of filopodia and, yet, the inability to form focal adhesions and stress fibers. These differences can be partially reversed by experimental manipulations of small G-proteins of the Rho family and their downstream targets, such as formins, the Arp2/3 complex and Rho kinase. We also show that the physical adhesion of cells to galectin-8 was stronger than adhesion to fibronectin. Notably, galectin-8 and fibronectin differently regulate cell spreading and focal adhesion formation, yet act synergistically to upregulate the number and length of filopodia. The physiological significance of the coherent cellular response to a molecularly complex matrix is discussed. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Wenhong Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ana Sancho
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Würzburg, 97070, Germany
- Department of Automatic Control and Systems Engineering, University of the Basque Country UPV/EHU, San Sebastian, 20018, Spain
| | - Wen-Lu Chung
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Yaron Vinik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Würzburg, 97070, Germany
| | - Yehiel Zick
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Alexander D. Bershadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| |
Collapse
|
21
|
Garrison CM, Schwarzbauer JE. Fibronectin fibril alignment is established upon initiation of extracellular matrix assembly. Mol Biol Cell 2021; 32:739-752. [PMID: 33625865 PMCID: PMC8108514 DOI: 10.1091/mbc.e20-08-0533] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The physical structure of the extracellular matrix (ECM) is tissue-specific and fundamental to normal tissue function. Proper alignment of ECM fibers is essential for the functioning of a variety of tissues. While matrix assembly in general has been intensively investigated, little is known about the mechanisms required for formation of aligned ECM fibrils. We investigated the initiation of fibronectin (FN) matrix assembly using fibroblasts that assemble parallel ECM fibrils and found that matrix assembly sites, where FN fibrillogenesis is initiated, were oriented in parallel at the cell poles. We show that these polarized matrix assembly sites progress into fibrillar adhesions and ultimately into aligned FN fibrils. Cells that assemble an unaligned meshwork matrix form matrix assembly sites around the cell periphery, but the distribution of matrix assembly sites in these cells could be modulated through micropatterning or mechanical stretch. While an elongated cell shape corresponds with a polarized matrix assembly site distribution, these two features are not absolutely linked, since we discovered that transforming growth factor beta (TGF-β1) enhances matrix assembly site polarity and assembly of aligned fibrils independent of cell elongation. We conclude that the ultimate orientation of FN fibrils is determined by the alignment and distribution of matrix assembly sites that form during the initial stages of cell–FN interactions.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
22
|
Vega ME, Finlay JB, Vasishtha M, Schwarzbauer JE. Elevated glucose alters global gene expression and tenascin-C alternative splicing in mesangial cells. Matrix Biol Plus 2021; 8:100048. [PMID: 33543041 PMCID: PMC7852322 DOI: 10.1016/j.mbplus.2020.100048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/19/2020] [Indexed: 12/25/2022] Open
Abstract
Mesangial cells are the major extracellular matrix (ECM)-producing cells in the kidney glomerulus and, when exposed to elevated glucose levels, they up-regulate assembly of fibronectin (FN) and other ECM proteins. Increases in glucose concentration are known to alter gene expression; here we investigated the connection between increased ECM production and changes in gene expression in mesangial cells. Comparison of mesangial cells grown in normal or high glucose conditions by RNA-sequencing showed significant expression changes in over 6000 genes and, when grouped by KEGG pathway analysis, identified the ECM-receptor interaction and focal adhesion pathways among the top 5 upregulated pathways. Of note was the significant increase in expression of tenascin-C (TN-C), a known regulator of FN matrix assembly. Mouse TN-C has multiple isoforms due to alternative splicing of 6 FNIII repeat exons. In addition to the transcriptional increase with high glucose, exon inclusion via alternative splicing was also changed resulting in production of higher molecular weight isoforms of TN-C. Mesangial cells grown in normal glucose secreted small isoforms with 1–2 variable repeats included whereas in high glucose large isoforms estimated to include 5 repeats were secreted. Unlike the smaller isoforms, the larger TN-C was not detected in the FN matrix. This change in TN-C isoforms may affect the regulation of FN matrix assembly and in this way may contribute to increased ECM accumulation under high glucose conditions. Elevated glucose alters gene expression in cultured mesangial cells. RNA-sequencing identifies increased expression of ECM proteins and receptors. High glucose changes tenascin-C isoform expression by alternative splicing. Differential ECM localization is detected for large vs small tenascin-C isoforms. Switch in tenascin-C may contribute to ECM accumulation in the diabetic glomerulus.
Collapse
Affiliation(s)
- Maria E Vega
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - John B Finlay
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Mansi Vasishtha
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
23
|
Hanmin C, Xiangyue Z, Lenahan C, Ling W, Yibo O, Yue H. Pleiotropic Role of Tenascin-C in Central Nervous System Diseases: From Basic to Clinical Applications. Front Neurol 2020; 11:576230. [PMID: 33281711 PMCID: PMC7691598 DOI: 10.3389/fneur.2020.576230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
The extracellular matrix is composed of a variety of macromolecular substances secreted by cells, which form a complex network that supports and connects tissue structures, regulates the morphogenesis of tissues, and maintains the physiological activities of cells. Tenascin-C, a secreted extracellular matrix glycoprotein, is abundantly expressed after exposure to pathological stimuli. It plays an important regulatory role in brain tumors, vascular diseases, and neurodegenerative diseases by mediating inflammatory responses, inducing brain damage, and promoting cell proliferation, migration, and angiogenesis through multiple signaling pathways. Therefore, tenascin-C may become a potential therapeutic target for intracranial diseases. Here, we review and discuss the latest literature regarding tenascin-C, and we comprehensively explain the role and clinical significance of tenascin-C in intracranial diseases.
Collapse
Affiliation(s)
- Chen Hanmin
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Xiangyue
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Wang Ling
- Department of Operating Room, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ou Yibo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - He Yue
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Tenascin-C Function in Glioma: Immunomodulation and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:149-172. [PMID: 32845507 DOI: 10.1007/978-3-030-48457-6_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
First identified in the 1980s, tenascin-C (TNC) is a multi-domain extracellular matrix glycoprotein abundantly expressed during the development of multicellular organisms. TNC level is undetectable in most adult tissues but rapidly and transiently induced by a handful of pro-inflammatory cytokines in a variety of pathological conditions including infection, inflammation, fibrosis, and wound healing. Persistent TNC expression is associated with chronic inflammation and many malignancies, including glioma. By interacting with its receptor integrin and a myriad of other binding partners, TNC elicits context- and cell type-dependent function to regulate cell adhesion, migration, proliferation, and angiogenesis. TNC operates as an endogenous activator of toll-like receptor 4 and promotes inflammatory response by inducing the expression of multiple pro-inflammatory factors in innate immune cells such as microglia and macrophages. In addition, TNC drives macrophage differentiation and polarization predominantly towards an M1-like phenotype. In contrast, TNC shows immunosuppressive function in T cells. In glioma, TNC is expressed by tumor cells and stromal cells; high expression of TNC is correlated with tumor progression and poor prognosis. Besides promoting glioma invasion and angiogenesis, TNC has been found to affect the morphology and function of tumor-associated microglia/macrophages in glioma. Clinically, TNC can serve as a biomarker for tumor progression; and TNC antibodies have been utilized as an adjuvant agent to deliver anti-tumor drugs to target glioma. A better mechanistic understanding of how TNC impacts innate and adaptive immunity during tumorigenesis and tumor progression will open new therapeutic avenues to treat brain tumors and other malignancies.
Collapse
|
25
|
Bugg D, Bretherton R, Kim P, Olszewski E, Nagle A, Schumacher AE, Chu N, Gunaje J, DeForest CA, Stevens K, Kim DH, Davis J. Infarct Collagen Topography Regulates Fibroblast Fate via p38-Yes-Associated Protein Transcriptional Enhanced Associate Domain Signals. Circ Res 2020; 127:1306-1322. [PMID: 32883176 DOI: 10.1161/circresaha.119.316162] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Myocardial infarction causes spatial variation in collagen organization and phenotypic diversity in fibroblasts, which regulate the heart's ECM (extracellular matrix). The relationship between collagen structure and fibroblast phenotype is poorly understood but could provide insights regarding the mechanistic basis for myofibroblast heterogeneity in the injured heart. OBJECTIVE To investigate the role of collagen organization in cardiac fibroblast fate determination. METHODS AND RESULTS Biomimetic topographies were nanofabricated to recapitulate differential collagen organization in the infarcted mouse heart. Here, adult cardiac fibroblasts were freshly isolated and cultured on ECM topographical mimetics for 72 hours. Aligned mimetics caused cardiac fibroblasts to elongate while randomly organized topographies induced circular morphology similar to the disparate myofibroblast morphologies measured in vivo. Alignment cues also induced myofibroblast differentiation, as >60% of fibroblasts formed αSMA (α-smooth muscle actin) stress fibers and expressed myofibroblast-specific ECM genes like Postn (periostin). By contrast, random organization caused 38% of cardiac fibroblasts to express αSMA albeit with downregulated myofibroblast-specific ECM genes. Coupling topographical cues with the profibrotic agonist, TGFβ (transforming growth factor beta), additively upregulated myofibroblast-specific ECM genes independent of topography, but only fibroblasts on flat and randomly oriented mimetics had increased percentages of fibroblasts with αSMA stress fibers. Increased tension sensation at focal adhesions induced myofibroblast differentiation on aligned mimetics. These signals were transduced by p38-YAP (yes-associated protein)-TEAD (transcriptional enhanced associate domain) interactions, in which both p38 and YAP-TEAD (yes-associated protein transcriptional enhanced associate domain) binding were required for myofibroblast differentiation. By contrast, randomly oriented mimetics did not change focal adhesion tension sensation or enrich for p38-YAP-TEAD interactions, which explains the topography-dependent diversity in fibroblast phenotypes observed here. CONCLUSIONS Spatial variations in collagen organization regulate cardiac fibroblast phenotype through mechanical activation of p38-YAP-TEAD signaling, which likely contribute to myofibroblast heterogeneity in the infarcted myocardium.
Collapse
Affiliation(s)
- Darrian Bugg
- Pathology (D.B., J.G., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | - Ross Bretherton
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | - Peter Kim
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle
| | - Emily Olszewski
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | - Abigail Nagle
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | | | - Nick Chu
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle
| | - Jagadambika Gunaje
- Pathology (D.B., J.G., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | - Cole A DeForest
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle.,Institute for Stem Cell and Regenerative Medicine (C.A.D., K.S., J.D.), University of Washington, Seattle.,Chemical Engineering (C.A.D.), University of Washington, Seattle
| | - Kelly Stevens
- Bioengineering (R.B., P.K., E.O., A.N., N.C., C.A.D., K.S., J.D.), University of Washington, Seattle.,Pathology (D.B., J.G., K.S., J.D.), University of Washington, Seattle.,Institute for Stem Cell and Regenerative Medicine (C.A.D., K.S., J.D.), University of Washington, Seattle.,Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| | - Deok-Ho Kim
- Biomedical Engineering, Johns Hopkins University, Baltimore, MD (D.-H.K.).,Medicine, Johns Hopkins School of Medicine, Baltimore, MD (D.-H.K.)
| | - Jennifer Davis
- Center for Cardiovascular Biology (D.B., R.B., E.O., A.N., J.G., K.S., J.D.), University of Washington, Seattle
| |
Collapse
|
26
|
Garrison CM, Singh-Varma A, Pastino AK, Steele JAM, Kohn J, Murthy NS, Schwarzbauer JE. A multilayered scaffold for regeneration of smooth muscle and connective tissue layers. J Biomed Mater Res A 2020; 109:733-744. [PMID: 32654327 DOI: 10.1002/jbm.a.37058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 01/26/2023]
Abstract
Tissue regeneration often requires recruitment of different cell types and rebuilding of two or more tissue layers to restore function. Here, we describe the creation of a novel multilayered scaffold with distinct fiber organizations-aligned to unaligned and dense to porous-to template common architectures found in adjacent tissue layers. Electrospun scaffolds were fabricated using a biodegradable, tyrosine-derived terpolymer, yielding densely-packed, aligned fibers that transition into randomly-oriented fibers of increasing diameter and porosity. We demonstrate that differently-oriented scaffold fibers direct cell and extracellular matrix (ECM) organization, and that scaffold fibers and ECM protein networks are maintained after decellularization. Smooth muscle and connective tissue layers are frequently adjacent in vivo; we show that within a single scaffold, the architecture supports alignment of contractile smooth muscle cells and deposition by fibroblasts of a meshwork of ECM fibrils. We rolled a flat scaffold into a tubular construct and, after culture, showed cell viability, orientation, and tissue-specific protein expression in the tube were similar to the flat-sheet scaffold. This scaffold design not only has translational potential for reparation of flat and tubular tissue layers but can also be customized for alternative applications by introducing two or more cell types in different combinations.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Anya Singh-Varma
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Alexandra K Pastino
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Joseph A M Steele
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - N Sanjeeva Murthy
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
27
|
Teo BSX, Gan RY, Abdul Aziz S, Sirirak T, Mohd Asmani MF, Yusuf E. In vitro evaluation of antioxidant and antibacterial activities of Eucheuma cottonii extract and its in vivo evaluation of the wound-healing activity in mice. J Cosmet Dermatol 2020; 20:993-1001. [PMID: 32659861 DOI: 10.1111/jocd.13624] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/11/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Eucheuma Cottonii is a type of red algae obtained from Sabah with main active component, sulfated polysaccharide or k-carrageenan. AIMS The objective of this research was to evaluate the antioxidant, antibacterial and potential wound-healing properties in aqueous extraction of E cottonii in order to meet the increasing demand for halal and natural cosmeceutical products. METHODS AND RESULTS Aqueous extract of E cottonii was investigated for active compounds by phytochemical screening and IR spectroscopy. Antioxidant activity was carried out using DPPH method, and the IC50 value was 1.99 mg/mL. Antibacterial activity was examined against Staphylococcus Aureus using Kirby-Bauer disk diffusion method and showed 10.03 ± 0.06 mm zone of inhibition, achieved by 200 mg/mL of extracts. A wound was made by skin excision of area around 100 mm2 on each mouse. Test group was treated with aqueous extract gel (10% w/w); meanwhile, the mice that were treated with honey acted as the positive control group and the untreated mice as negative control group. Results showed that the wound contraction rate inclined to aqueous extracts as compared to untreated group (P < .05). Percentage of wound healing for aqueous extracts and untreated group were 87.7% ± 2.0% and 57.6% ± 5.3%, respectively. CONCLUSION Aqueous extract was found to be comparable to the honey in wound healing.
Collapse
Affiliation(s)
- Brian Sheng Xian Teo
- Pharmaceutical Chemistry Unit, School of Pharmacy, Management & Science University, Shah Alam, Malaysia
| | - Rui Yi Gan
- Pharmaceutical Chemistry Unit, School of Pharmacy, Management & Science University, Shah Alam, Malaysia
| | - Sarah Abdul Aziz
- Pharmaceutical Chemistry Unit, School of Pharmacy, Management & Science University, Shah Alam, Malaysia
| | - Thanchanok Sirirak
- Faculty of Pharmaceutical Science, Burapha University, Chon Buri, Thailand
| | - Mohd Fadli Mohd Asmani
- Pharmaceutical Chemistry Unit, School of Pharmacy, Management & Science University, Shah Alam, Malaysia
| | - Eddy Yusuf
- The International Centre for Halal Studies (ICHLAS), Management & Science University, Shah Alam, Malaysia
| |
Collapse
|
28
|
Giblin SP, Schwenzer A, Midwood KS. Alternative splicing controls cell lineage-specific responses to endogenous innate immune triggers within the extracellular matrix. Matrix Biol 2020; 93:95-114. [PMID: 32599145 DOI: 10.1016/j.matbio.2020.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/15/2020] [Accepted: 06/21/2020] [Indexed: 01/08/2023]
Abstract
The identification of barely more than 20,000 human genes was amongst the most surprising outcomes of the human genome project. Alternative splicing provides an essential means of expanding the proteome, enabling a single gene to encode multiple, distinct isoforms by selective inclusion or exclusion of exons from mature mRNA. However, mis-regulation of this process is associated with most human diseases. Here, we examine the impact of post-transcriptional processing on extracellular matrix function, focusing on the complex alternative splicing patterns of tenascin-C, a molecule that can exist in as many as 500 different isoforms. We demonstrate that the pro-inflammatory activity of this endogenous innate immune trigger is controlled by inclusion or exclusion of a novel immunomodulatory site located within domains AD2AD1, identifying this as a mechanism that prevents unnecessary inflammation in healthy tissues but enables rapid immune cell mobilization and activation upon tissue damage, and defining how this goes awry in autoimmune disease.
Collapse
Affiliation(s)
- Sean P Giblin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Anja Schwenzer
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Kim S Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
29
|
Huang Y, Kyriakides TR. The role of extracellular matrix in the pathophysiology of diabetic wounds. Matrix Biol Plus 2020; 6-7:100037. [PMID: 33543031 PMCID: PMC7852307 DOI: 10.1016/j.mbplus.2020.100037] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/29/2022] Open
Abstract
Impaired healing leading to the formation of ulcerated wounds is a critical concern in patients with diabetes. Abnormalities in extracellular matrix (ECM) production and remodeling contribute to tissue dysfunction and delayed healing. Specifically, diabetes-induced changes in the expression and/or activity of structural proteins, ECM-modifying enzymes, proteoglycans, and matricellular proteins have been reported. In this review, we provide a summary of the key ECM molecules and associated changes in skin and diabetic wounds. Such information should allow for new insights in the understanding of impaired wound healing and lead to the development of ECM-based therapeutic strategies.
Collapse
Affiliation(s)
- Yaqing Huang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA.,Department of Pathology, Yale University, New Haven, CT 06519, USA
| | - Themis R Kyriakides
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA.,Department of Pathology, Yale University, New Haven, CT 06519, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| |
Collapse
|
30
|
Gruber BL, Mienaltowski MJ, MacLeod JN, Schittny J, Kasper S, Flück M. Tenascin-C expression controls the maturation of articular cartilage in mice. BMC Res Notes 2020; 13:78. [PMID: 32066496 PMCID: PMC7027060 DOI: 10.1186/s13104-020-4906-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Expression of the de-adhesive extracellular matrix protein tenascin-C (TNC) is associated with the early postnatal development of articular cartilage which is both load-dependent and associated with chondrocyte differentiation. We assessed morphological changes in the articular cartilage of TNC deficient mice at postnatal ages of 1, 4 and 8 weeks compared to age-matched wildtype mice. RESULTS Cartilage integrity was assessed based on hematoxylin and eosin stained-sections from the tibial bone using a modified Mankin score. Chondrocyte density and cartilage thickness were assessed morphometrically. TNC expression was localized based on immunostaining. At 8 weeks of age, the formed tangential/transitional zone of the articular cartilage was 27% thicker and the density of chondrocytes in the articular cartilage was 55% lower in wildtype than the TNC-deficient mice. TNC protein expression was associated with chondrocytes. No relevant changes were found in mice at 1 and 4 weeks of age. The findings indicate a role of tenascin-C in the post-natal maturation of the extracellular matrix in articular cartilage. This might be a compensatory mechanism to strengthen resilience against mechanical stress.
Collapse
Affiliation(s)
- Bastian L Gruber
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Lengghalde 5, 8008, Zurich, Switzerland
| | - Michael J Mienaltowski
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA.,Department of Animal Science, University of California Davis, Davis, CA, USA
| | - James N MacLeod
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | | | - Stephanie Kasper
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Lengghalde 5, 8008, Zurich, Switzerland
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Lengghalde 5, 8008, Zurich, Switzerland. .,Institute of Anatomy, University of Berne, Berne, Switzerland.
| |
Collapse
|
31
|
Hawkins AG, Julian CM, Konzen S, Treichel S, Lawlor ER, Bailey KM. Microenvironmental Factors Drive Tenascin C and Src Cooperation to Promote Invadopodia Formation in Ewing Sarcoma. Neoplasia 2019; 21:1063-1072. [PMID: 31521948 PMCID: PMC6745492 DOI: 10.1016/j.neo.2019.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 01/03/2023] Open
Abstract
Ewing sarcoma is a bone tumor most commonly diagnosed in adolescents and young adults. Survival for patients with recurrent or metastatic Ewing sarcoma is dismal and there is a dire need to better understand the mechanisms of cell metastasis specific to this disease. Our recent work demonstrated that microenvironmental stress leads to increased Ewing sarcoma cell invasion through Src activation. Additionally, we have shown that the matricellular protein tenascin C (TNC) promotes metastasis in Ewing sarcoma. A major role of both TNC and Src is mediation of cell-cell and cell-matrix interactions resulting in changes in cell motility, invasion, and adhesion. However, it remains largely unknown, if and how, TNC and Src are linked in these processes. We hypothesized that TNC is a positive regulator of invadopodia formation in Ewing sarcoma through its ability to activate Src. We demonstrate here that both tumor cell endogenous and exogenous TNC can enhance Src activation and invadopodia formation in Ewing sarcoma. We found that microenvironmental stress upregulates TNC expression and this is dampened with application of the Src inhibitor dasatinib, suggesting that TNC expression and Src activation cooperate to promote the invasive phenotype. This work reports the impact of stress-induced TNC expression on enhancing cell invadopodia formation, provides evidence for a feed forward loop between TNC and Src to promote cell metastatic behavior, and highlights a pathway by which microenvironment-driven TNC expression could be therapeutically targeted in Ewing sarcoma.
Collapse
Affiliation(s)
- Allegra G Hawkins
- Department of Pediatrics, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Claire M Julian
- Department of Pediatrics, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15224
| | - Sonja Konzen
- Department of Pediatrics, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Sydney Treichel
- Department of Pediatrics, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Elizabeth R Lawlor
- Department of Pediatrics, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA, 48109
| | - Kelly M Bailey
- Department of Pediatrics, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA 15224.
| |
Collapse
|
32
|
Ferrié C, Kasper S, Wanivenhaus F, Flück M. Time course of costamere-related alterations in focal adhesion signaling and composition of rat soleus muscle after achilles tenotomy. Data Brief 2019; 25:103999. [PMID: 31463339 PMCID: PMC6706776 DOI: 10.1016/j.dib.2019.103999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 11/25/2022] Open
Abstract
Sarcolemma-based focal adhesions (costameres) are a central hub for the cytoskeletal anchoring of myofibrils and mechano-regulated signaling. Here we report the time course of alterations in focal adhesion-associated signaling and fiber composition in rat soleus muscle after Achilles tenotomy. The report includes data from tenotomized muscles and contralateral mock controls to expose whether muscle degeneration after tenotomy is due to the transection of the Achilles tendon, or circumjacent surgical manipulations of the tendon. With respect to the interpretation of the data regarding mechanistic implications of costamere-associated processes for surgical repair of the detached muscle-tendon complex the reader is referred to the accompanying research article 'Focal adhesion kinase coordinates costamere-related JNK signaling with muscle fiber transformation after Achilles tenotomy and tendon reconstruction' Ferrié et al., 2019.
Collapse
Affiliation(s)
- Céline Ferrié
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Stephanie Kasper
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Florian Wanivenhaus
- Department of Orthopedic Surgery, Balgrist University Hospital, Zurich, Switzerland
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland
| |
Collapse
|
33
|
Spurlin JW, Siedlik MJ, Nerger BA, Pang MF, Jayaraman S, Zhang R, Nelson CM. Mesenchymal proteases and tissue fluidity remodel the extracellular matrix during airway epithelial branching in the embryonic avian lung. Development 2019; 146:dev.175257. [PMID: 31371376 DOI: 10.1242/dev.175257] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 07/16/2019] [Indexed: 12/31/2022]
Abstract
Reciprocal epithelial-mesenchymal signaling is essential for morphogenesis, including branching of the lung. In the mouse, mesenchymal cells differentiate into airway smooth muscle that wraps around epithelial branches, but this contractile tissue is absent from the early avian lung. Here, we have found that branching morphogenesis in the embryonic chicken lung requires extracellular matrix (ECM) remodeling driven by reciprocal interactions between the epithelium and mesenchyme. Before branching, the basement membrane wraps the airway epithelium as a spatially uniform sheath. After branch initiation, however, the basement membrane thins at branch tips; this remodeling requires mesenchymal expression of matrix metalloproteinase 2, which is necessary for branch extension but for not branch initiation. As branches extend, tenascin C (TNC) accumulates in the mesenchyme several cell diameters away from the epithelium. Despite its pattern of accumulation, TNC is expressed exclusively by epithelial cells. Branch extension coincides with deformation of adjacent mesenchymal cells, which correlates with an increase in mesenchymal fluidity at branch tips that may transport TNC away from the epithelium. These data reveal novel epithelial-mesenchymal interactions that direct ECM remodeling during airway branching morphogenesis.
Collapse
Affiliation(s)
- James W Spurlin
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Michael J Siedlik
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Bryan A Nerger
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Mei-Fong Pang
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Sahana Jayaraman
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Rawlison Zhang
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Departments of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA .,Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
34
|
Qi J, Esfahani DR, Huang T, Ozark P, Bartom E, Hashizume R, Bonner ER, An S, Horbinski CM, James CD, Saratsis AM. Tenascin-C expression contributes to pediatric brainstem glioma tumor phenotype and represents a novel biomarker of disease. Acta Neuropathol Commun 2019; 7:75. [PMID: 31092287 PMCID: PMC6518697 DOI: 10.1186/s40478-019-0727-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/22/2019] [Indexed: 12/27/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG), an infiltrative, high grade glioma (HGG) affecting young children, has the highest mortality rate of all pediatric cancers. Despite treatment, average survival is less than twelve months, and five-year survival under 5%. We previously detected increased expression of Tenascin-C (TNC) protein in DIPG cerebrospinal fluid and tumor tissue relative to normal specimens. TNC is an extracellular matrix (ECM) glycoprotein that mediates cell-matrix interactions, guides migrating neurons during normal brain development and is thought to maintain the periventricular stem cell niche in the developing brain. Tumor TNC expression is reported in adult glioma and other cancers. However, the pattern and effects of TNC expression in DIPG has not been previously explored. Here, we characterize TNC expression in patient derived pediatric supratentorial HGG (n = 3) and DIPG (n = 6) cell lines, as well as pediatric glioma tumor (n = 50) and normal brain tissue specimens (n = 3). We found tumor specific TNC gene and protein overexpression that directly correlated with higher tumor grade (WHO III and IV, p = 0.05), H3K27 M mutation (p = 0.012), shorter progression free survival (p = 0.034), and poorer overall survival (0.041) in association with these factors. TNC knockdown via lentiviral shRNA transfection of HGG (n = 1) and DIPG (n = 3) cell lines resulted in decreased cell proliferation, migration, and invasion in vitro (p < 0.01), while TNC cDNA transfection resulted in increased cell migration, invasion and proliferation (p < 0.01) as well as altered cell morphology in H3K27 M mutant DIPG lines. Whole transcriptome sequencing analysis (RNA-Seq) on DIPG (n = 3) and HGG (n = 2) cell lines after TNC cDNA, shRNA, and empty vector control transfection revealed the effects of TNC expression level on global gene expression profiles. Together, our findings reveal TNC expression in DIPG in association with H3K27 M mutation and VEGF signaling, and suggest that TNC may contribute to DIPG tumor phenotype, and serve as a clinically detectable biomarker for DIPG.
Collapse
Affiliation(s)
- J. Qi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - D. R. Esfahani
- Department of Neurological Surgery, University of Illinois at Chicago, Chicago, IL USA
| | - T. Huang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - P. Ozark
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - E. Bartom
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - R. Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - E. R. Bonner
- Center for Genetic Medicine, Children’s National Health System, Washington, DC 20010 USA
- Institute for Biomedical Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC USA
| | - S. An
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - C. M. Horbinski
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - C. D. James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - A. M. Saratsis
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL USA
- Division of Pediatric Neurosurgery, Department of Surgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL USA
| |
Collapse
|
35
|
Ferrié C, Kasper S, Wanivenhaus F, Flück M. Focal adhesion kinase coordinates costamere-related JNK signaling with muscle fiber transformation after Achilles tenotomy and tendon reconstruction. Exp Mol Pathol 2019; 108:42-56. [PMID: 30879953 DOI: 10.1016/j.yexmp.2019.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
Achilles tendon rupture necessitates rapid tendon reattachment to reinstate plantar flexion before affected muscles deteriorate through muscle fiber atrophy and transformation. The implicated process may involve alterations in sarcolemmal sites of myofibril attachment (costameres), which control myofibrillogenesis via a mechano-regulated mechanism through integrin-associated focal adhesion kinase (FAK). We assessed the contribution of FAK to alterations in fiber type composition and expression of costamere-associated structural proteins, the phosphorylation status of Y397-FAK and downstream mTOR/JNK-P70S6K hypertrophy signaling in rat soleus muscle after Achilles tenotomy and tendon repair. Achilles tenotomy induced a profound deterioration of muscle composition 14 days, but not 4 days, following tendon release, comprising specifically increased area percentages of fast type fibers, fibers with internal nuclei, and connective tissue. Concomitantly, expression of costameric proteins FAK and meta-vinculin, and phosphorylation of T421/S424-P70S6K and T183/Y185-JNK was elevated, all of which was mitigated by tendon reattachment immediately after release. Overexpression of FAK in soleus muscle fibers and reattachment corrected the expression of meta- and gamma-vinculin isoforms to the lower levels in mock controls while further enhancing T183/Y185-JNK phosphorylation and levels of FAK C-terminus-related inhibitory proteins. Co-overexpression of the FAK inhibitor, FRNK, lowered FAK-overexpression driven Y397-FAK phosphorylation and T183/Y185-JNK phosphorylation. FAK levels correlated to molecular and cellular hallmarks of fiber degeneration. The findings demarcate the window between 4 and 14 days after tenotomy as costamere-dependent muscle transformation process, and expose that FAK overexpression prevents molecular aspects of the pathology which within the study limitations does not result in the mitigation of muscle fiber degeneration.250 words.
Collapse
Affiliation(s)
- Céline Ferrié
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Stephanie Kasper
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland
| | - Florian Wanivenhaus
- Department of Orthopedic Surgery, Balgrist University Hospital, Zurich, Switzerland
| | - Martin Flück
- Laboratory for Muscle Plasticity, Department of Orthopedics, University of Zurich, Balgrist Campus, Zurich, Switzerland.
| |
Collapse
|
36
|
Cooper J, Giancotti FG. Integrin Signaling in Cancer: Mechanotransduction, Stemness, Epithelial Plasticity, and Therapeutic Resistance. Cancer Cell 2019; 35:347-367. [PMID: 30889378 PMCID: PMC6684107 DOI: 10.1016/j.ccell.2019.01.007] [Citation(s) in RCA: 607] [Impact Index Per Article: 101.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/10/2018] [Accepted: 01/14/2019] [Indexed: 12/16/2022]
Abstract
Integrins mediate cell adhesion and transmit mechanical and chemical signals to the cell interior. Various mechanisms deregulate integrin signaling in cancer, empowering tumor cells with the ability to proliferate without restraint, to invade through tissue boundaries, and to survive in foreign microenvironments. Recent studies have revealed that integrin signaling drives multiple stem cell functions, including tumor initiation, epithelial plasticity, metastatic reactivation, and resistance to oncogene- and immune-targeted therapies. Here, we discuss the mechanisms leading to the deregulation of integrin signaling in cancer and its various consequences. We place emphasis on novel functions, determinants of context dependency, and mechanism-based therapeutic opportunities.
Collapse
Affiliation(s)
- Jonathan Cooper
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Filippo G Giancotti
- Department of Cancer Biology and David H. Koch Center for Applied Research of Genitourinary Cancers, UT MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
37
|
Imanaka-Yoshida K, Matsumoto KI. Multiple Roles of Tenascins in Homeostasis and Pathophysiology of Aorta. Ann Vasc Dis 2018; 11:169-180. [PMID: 30116408 PMCID: PMC6094038 DOI: 10.3400/avd.ra.17-00118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tenascins are a family of large extracellular matrix (ECM) glycoproteins. Four family members (tenascin-C, -R, -X, and -W) have been identified to date. Each member consists of the same types of structural domains and exhibits time- and tissue-specific expression patterns, suggesting their specific roles in embryonic development and tissue remodeling. Among them, the significant involvement of tenascin-C (TNC) and tenascin-X (TNX) in the progression of vascular diseases has been examined in detail. TNC is strongly up-regulated under pathological conditions, induced by a number of inflammatory mediators and mechanical stress. TNC has diverse functions, particularly in the regulation of inflammatory responses. Recent studies suggest that TNC is involved in the pathophysiology of aneurysmal and dissecting lesions, in part by protecting the vascular wall from destructive mechanical stress. TNX is strongly expressed in vascular walls, and its distribution is often reciprocal to that of TNC. TNX is involved in the stability and maintenance of the collagen network and elastin fibers. A deficiency in TNX results in a form of Ehlers–Danlos syndrome (EDS). Although their exact roles in vascular diseases have not yet been elucidated, TNC and TNX are now being recognized as promising biomarkers for diagnosis and risk stratification of vascular diseases.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie, Japan.,Mie University Research Center for Matrix Biology, Tsu, Mie, Japan
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Shimane, Japan
| |
Collapse
|
38
|
Abstract
Tenascin-C (TN-C) is a glycoprotein component of the extracellular matrix (ECM). TN-C consists of four distinct domains, including the tenascin assembly domain, epidermal growth factor-like repeats, fibronectin type III-like repeats, and the fibrinogen-like globe (FBG) domain. This review summarizes the role of TN-C in articular cartilage. Expression of TN-C is associated with the development of articular cartilage but markedly decreases during maturation of chondrocytes and disappears almost completely in adult articular cartilage. Increased expression of TN-C has been found at diseased cartilage and synovial sites in osteoarthritis (OA) and rheumatoid arthritis (RA). TN-C is increased in the synovial fluid in patients with OA and RA. In addition, serum TN-C is elevated in RA patients. TN-C could be a useful biochemical marker for joint disease. The addition of TN-C results in different effects among TN-C domains. TN-C fragments might be endogenous inducers of cartilage matrix degradation; however, full-length TN-C could promote cartilage repair and prevent cartilage degeneration. The deficiency of TN-C enhanced cartilage degeneration in the spontaneous OA in aged joints and surgical OA model. The clinical significance of TN-C effects on cartilage is not straightforward.
Collapse
Affiliation(s)
- Masahiro Hasegawa
- a Department of Orthopaedic Surgery , Mie University Graduate School of Medicine , Mie , Japan
| | - Toshimichi Yoshida
- b Department of Pathology & Matrix Biology , Mie University Graduate School of Medicine , Mie , Japan
| | - Akihiro Sudo
- a Department of Orthopaedic Surgery , Mie University Graduate School of Medicine , Mie , Japan
| |
Collapse
|
39
|
Abstract
Cells dynamically assemble and organize into complex tissues during development, and the resulting three-dimensional (3D) arrangement of cells and their surrounding extracellular matrix in turn feeds back to regulate cell and tissue function. Recent advances in engineered cultures of cells to model 3D tissues or organoids have begun to capture this dynamic reciprocity between form and function. Here, we describe the underlying principles that have advanced the field, focusing in particular on recent progress in using mechanical constraints to recapitulate the structure and function of musculoskeletal tissues.
Collapse
Affiliation(s)
- Jeroen Eyckmans
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA .,The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Christopher S Chen
- Department of Biomedical Engineering and the Biological Design Center, Boston University, Boston, MA 02215, USA .,The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
40
|
Chiquet M, Blumer S, Angelini M, Mitsiadis TA, Katsaros C. Mesenchymal Remodeling during Palatal Shelf Elevation Revealed by Extracellular Matrix and F-Actin Expression Patterns. Front Physiol 2016; 7:392. [PMID: 27656150 PMCID: PMC5013070 DOI: 10.3389/fphys.2016.00392] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/23/2016] [Indexed: 12/17/2022] Open
Abstract
During formation of the secondary palate in mammalian embryos, two vertically oriented palatal shelves rapidly elevate into a horizontal position above the tongue, meet at the midline, and fuse to form a single entity. Previous observations suggested that elevation occurs by a simple 90° rotation of the palatal shelves. More recent findings showed that the presumptive midline epithelial cells are not located at the tips of palatal shelves before elevation, but mostly toward their medial/lingual part. This implied extensive tissue remodeling during shelf elevation. Nevertheless, it is still not known how the shelf mesenchyme reorganizes during this process, and what mechanism drives it. To address this question, we mapped the distinct and restricted expression domains of certain extracellular matrix components within the developing palatal shelves. This procedure allowed to monitor movements of entire mesenchymal regions relative to each other during shelf elevation. Consistent with previous notions, our results confirm a flipping movement of the palatal shelves anteriorly, whereas extensive mesenchymal reorganization is observed more posteriorly. There, the entire lingual portion of the vertical shelves moves close to the midline after elevation, whereas the mesenchyme at the original tip of the shelves ends up ventrolaterally. Moreover, we observed that the mesenchymal cells of elevating palatal shelves substantially align their actin cytoskeleton, their extracellular matrix, and their nuclei in a ventral to medial direction. This indicates that, like in other morphogenetic processes, actin-dependent cell contractility is a major driving force for mesenchymal tissue remodeling during palatogenesis.
Collapse
Affiliation(s)
- Matthias Chiquet
- Department of Orthodontics and Dentofacial Orthopedics, Medical Faculty, School of Dental Medicine, University of Bern Bern, Switzerland
| | - Susan Blumer
- Department of Orthodontics and Dentofacial Orthopedics, Medical Faculty, School of Dental Medicine, University of Bern Bern, Switzerland
| | - Manuela Angelini
- Department of Orthodontics and Dentofacial Orthopedics, Medical Faculty, School of Dental Medicine, University of Bern Bern, Switzerland
| | - Thimios A Mitsiadis
- Orofacial Development and Regeneration, Center for Dental Medicine, Institute for Oral Biology, University of Zurich Zurich, Switzerland
| | - Christos Katsaros
- Department of Orthodontics and Dentofacial Orthopedics, Medical Faculty, School of Dental Medicine, University of Bern Bern, Switzerland
| |
Collapse
|
41
|
Abstract
ABSTRACT
Tenascin-C (TNC) is a hexameric, multimodular extracellular matrix protein with several molecular forms that are created through alternative splicing and protein modifications. It is highly conserved amongst vertebrates, and molecular phylogeny indicates that it evolved before fibronectin. Tenascin-C has many extracellular binding partners, including matrix components, soluble factors and pathogens; it also influences cell phenotype directly through interactions with cell surface receptors. Tenascin-C protein synthesis is tightly regulated, with widespread protein distribution in embryonic tissues, but restricted distribution of tenascin-C in adult tissues. Tenascin-C is also expressed de novo during wound healing or in pathological conditions, including chronic inflammation and cancer. First described as a modulator of cell adhesion, tenascin-C also directs a plethora of cell signaling and gene expression programs by shaping mechanical and biochemical cues within the cellular microenvironment. Exploitment of the pathological expression and function of tenascin-C is emerging as a promising strategy to develop new diagnostic, therapeutic and bioengineering tools. In this Cell Science at a Glance article and the accompanying poster we provide a succinct and comprehensive overview of the structural and functional features of tenascin-C and its potential roles in developing embryos and under pathological conditions.
Collapse
Affiliation(s)
- Kim S. Midwood
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Matthias Chiquet
- Department of Orthodontics and Dentofacial Orthopedics, Medical Faculty, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| | - Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California at Davis, Davis CA 95616-8643, USA
| | - Gertraud Orend
- Inserm U1109, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy (MN3T) team, 3 av. Molière, Strasbourg 67200, France
- Université de Strasbourg, Strasbourg 67000, France
- LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| |
Collapse
|
42
|
Abstract
Tenascins are a family of extracellular matrix molecules that are mainly expressed in embryonic development and down-regulated in adulthood. A re-expression in the adult occurs under pathological conditions such as inflammation, regeneration or neoplasia. As the most prominent member of the tenascin family, TN-C, is highly expressed in glioma tissue and rising evidence suggests that TN-C plays a crucial role in cell migration or invasion - the most fatal characteristics of glioma - also the other members of this protein family have been investigated with regard to their impact on glioma biology. For all tenascins correlations between the expression levels of the different family members and the degree of malignancy and invasiveness of glial tumors could be detected. Overall, the former and recent results in the research on glioma and tenascins point at distinct roles of each of the molecules in glioma biology and the devastating properties of these tumors.
Collapse
Affiliation(s)
- Nicole Brösicke
- a Department of Cell Morphology and Molecular Neurobiology ; Ruhr-University Bochum ; Bochum , Germany
| | | |
Collapse
|
43
|
James JL, Hurley DG, Gamage TKJB, Zhang T, Vather R, Pantham P, Murthi P, Chamley LW. Isolation and characterisation of a novel trophoblast side-population from first trimester placentae. Reproduction 2015; 150:449-62. [DOI: 10.1530/rep-14-0646] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 08/06/2015] [Indexed: 12/18/2022]
Abstract
The placenta is responsible for all nutrient and gas exchange between mother and baby during pregnancy. The differentiation of specialised placental epithelial cells called trophoblasts is essential for placental function, but we understand little about how these populations arise. Mouse trophoblast stem cells have allowed us to understand many of the factors that regulate murine trophoblast lineage development, but the human placenta is anatomically very different from the mouse, and it is imperative to isolate a human trophoblast stem cell to understand human placental development. Here we have developed a novel methodology to isolate a Hoechst side-population of trophoblasts from early gestation placentae and compared their transcriptome to differentiated trophoblast populations (cytotrophoblasts and extravillous trophoblasts) using microarray technology. Side-population trophoblasts clustered as a transcriptomically distinct population but were more closely related to cytotrophoblasts than extravillous trophoblasts. Side-population trophoblasts up-regulated a number of genes characteristic of trophectoderm and murine trophoblast stem cells in comparison to cytotrophoblasts or extravillous trophoblasts and could be distinguished from both of these more mature populations by a unique set of 22 up-regulated genes, which were enriched for morphogenesis and organ development and the regulation of growth functions. Cells expressing two of these genes (LAMA2 and COL6A3) were distributed throughout the cytotrophoblast layer at the trophoblast/mesenchymal interface. Comparisons to previously published trophoblast progenitor populations suggest that the side-population trophoblasts isolated in this work are a novel human trophoblast population. Future work will determine whether these cells exhibit functional progenitor/stem cell attributes.
Collapse
|
44
|
Kular JK, Basu S, Sharma RI. The extracellular matrix: Structure, composition, age-related differences, tools for analysis and applications for tissue engineering. J Tissue Eng 2014; 5:2041731414557112. [PMID: 25610589 PMCID: PMC4883592 DOI: 10.1177/2041731414557112] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/28/2014] [Indexed: 12/23/2022] Open
Abstract
The extracellular matrix is a structural support network made up of diverse proteins, sugars and other components. It influences a wide number of cellular processes including migration, wound healing and differentiation, all of which is of particular interest to researchers in the field of tissue engineering. Understanding the composition and structure of the extracellular matrix will aid in exploring the ways the extracellular matrix can be utilised in tissue engineering applications especially as a scaffold. This review summarises the current knowledge of the composition, structure and functions of the extracellular matrix and introduces the effect of ageing on extracellular matrix remodelling and its contribution to cellular functions. Additionally, the current analytical technologies to study the extracellular matrix and extracellular matrix–related cellular processes are also reviewed.
Collapse
Affiliation(s)
- Jaspreet K Kular
- Department of Chemical Engineering, University of Bath, Bath, UK ; Centre for Regenerative Medicine, University of Bath, Bath, UK
| | - Shouvik Basu
- Department of Chemical Engineering, University of Bath, Bath, UK
| | - Ram I Sharma
- Department of Chemical Engineering, University of Bath, Bath, UK ; Centre for Regenerative Medicine, University of Bath, Bath, UK ; Centre for Sustainable Chemical Technologies, University of Bath, Bath, UK
| |
Collapse
|
45
|
Chiquet-Ehrismann R, Orend G, Chiquet M, Tucker RP, Midwood KS. Tenascins in stem cell niches. Matrix Biol 2014; 37:112-23. [DOI: 10.1016/j.matbio.2014.01.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 12/16/2022]
|
46
|
Reversible modulation of myofibroblast differentiation in adipose-derived mesenchymal stem cells. PLoS One 2014; 9:e86865. [PMID: 24466271 PMCID: PMC3900664 DOI: 10.1371/journal.pone.0086865] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 12/18/2013] [Indexed: 12/21/2022] Open
Abstract
Unregulated activity of myofibroblasts, highly contractile cells that deposit abundant extracellular matrix (ECM), leads to fibrosis. To study the modulation of myofibroblast activity, we used human adipose-derived mesenchymal stem cells (ADSCs), which have much potential in regenerative medicine. We found that ADSCs treated with TGF-β developed a myofibroblastic phenotype with increases in α-smooth muscle actin (α-SMA), a myofibroblast marker, and ECM proteins type I collagen and fibronectin. In contrast, treatment with bFGF had the opposite effect. bFGF-differentiated ADSCs showed marked down-regulation of α-SMA expression, collagen I, and fibronectin, and loss of focal adhesions and stress fibers. Functionally, bFGF-differentiated ADSCs were significantly more migratory, which correlated with up-regulation of tenascin-C, an anti-adhesive ECM protein, and vimentin, a pro-migratory cytoskeletal protein. On the other hand, TGF-β-differentiated ADSCs were significantly more contractile than bFGF-differentiated cells. Interestingly, cells completely reversed their morphologies, marker expression, signaling pathways, and contractility versus migratory profiles when switched from culture with one growth factor to the other, demonstrating that the myofibroblast differentiation process is not terminal. Cell differentiation was associated with activation of Smad2 downstream of TGF-β and of ERK/MAP kinase downstream of bFGF. Reversibility of the TGF-β-induced myofibroblastic phenotype depends, in part, on bFGF-induced ERK/MAP kinase signaling. These findings show that ADSC differentiation into myofibroblasts and re-differentiation into fibroblast-like cells can be manipulated with growth factors, which may have implications in the development of novel therapeutic strategies to reduce the risk of fibrosis.
Collapse
|
47
|
Reynolds AB, Kanner SB, Bouton AH, Schaller MD, Weed SA, Flynn DC, Parsons JT. SRChing for the substrates of Src. Oncogene 2013; 33:4537-47. [PMID: 24121272 DOI: 10.1038/onc.2013.416] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/16/2013] [Accepted: 08/17/2013] [Indexed: 12/12/2022]
Abstract
By the mid 1980's, it was clear that the transforming activity of oncogenic Src was linked to the activity of its tyrosine kinase domain and attention turned to identifying substrates, the putative next level of control in the pathway to transformation. Among the first to recognize the potential of phosphotyrosine-specific antibodies, Parsons and colleagues launched a risky shotgun-based approach that led ultimately to the cDNA cloning and functional characterization of many of today's best-known Src substrates (for example, p85-Cortactin, p110-AFAP1, p130Cas, p125FAK and p120-catenin). Two decades and over 6000 citations later, the original goals of the project may be seen as secondary to the enormous impact of these protein substrates in many areas of biology. At the request of the editors, this review is not restricted to the current status of the substrates, but reflects also on the anatomy of the project itself and some of the challenges and decisions encountered along the way.
Collapse
Affiliation(s)
- A B Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - S B Kanner
- Arrowhead Research Corporation, Madison, WI, USA
| | - A H Bouton
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - M D Schaller
- Department of Biochemistry, 3124 HSN, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - S A Weed
- Department of Neurobiology and Anatomy, 1833 Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV, USA
| | - D C Flynn
- Department of Medical Lab Sciences, College of Health Sciences, University of Delaware, Newark, DE, USA
| | - J T Parsons
- Departments of Microbiology, Immunology and Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA, USA
| |
Collapse
|
48
|
Grogan SP, Chen X, Sovani S, Taniguchi N, Colwell CW, Lotz MK, D'Lima DD. Influence of cartilage extracellular matrix molecules on cell phenotype and neocartilage formation. Tissue Eng Part A 2013; 20:264-74. [PMID: 23962090 DOI: 10.1089/ten.tea.2012.0618] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interaction between chondrocytes and the cartilage extracellular matrix (ECM) is essential for maintaining the cartilage's role as a low-friction and load-bearing tissue. In this study, we examined the influence of cartilage zone-specific ECM on human articular chondrocytes (HAC) in two-dimensional and three-dimensional (3D) environments. Two culture systems were used. SYSTEM 1: HAC were cultured on cell-culture plates that had been precoated with the following ECM molecules for 7 days: decorin, biglycan, tenascin C (superficial zone), collagen type II, hyaluronan (HA) (middle and deep zones), and osteopontin (deep zone). Uncoated standard culture plates were used as controls. Expanded cells were examined for phenotypic changes using real-time polymerase chain reaction. In addition, expanded cells were placed into high-density pellet cultures for 14 days. Neocartilage formation was assessed via gene expression and histology evaluations. SYSTEM 2: HAC that were cultured on untreated plates and encapsulated in a 3D alginate scaffold were mixed with one of the zone-specific ECM molecules. Cell viability, gene expression, and histology assessments were conducted on 14-day-old tissues. In HAC monolayer culture, exposure to decorin, HA, and osteopontin increased COL2A1 and aggrecan messenger RNA (mRNA) levels compared with controls. Biglycan up-regulated aggrecan without a significant impact on COL2A1 expression; Tenascin C reduced COL2A1 expression. Neocartilage formed after preculture on tenascin C and collagen type II expressed higher COL2A1 mRNA compared with control pellets. Preculture of HAC on HA decreased both COL2A1 and aggrecan expression levels compared with controls, which was consistent with histology. Reduced proteoglycan 4 (PRG4) mRNA levels were observed in HAC pellets that had been precultured with biglycan and collagen type II. Exposing HAC to HA directly in 3D-alginate culture most effectively induced neocartilage formation, showing increased COL2A1 and aggrecan, and reduced COL1A1 compared with controls. Decorin treatments increased HAC COL2A1 mRNA levels. These data indicate that an appropriate exposure to cartilage-specific ECM proteins could be used to enhance cartilage formation and to even induce the formation of zone-specific phenotypes to improve cartilage regeneration.
Collapse
Affiliation(s)
- Shawn P Grogan
- 1 Shiley Center for Orthopaedic Research and Education , Scripps Clinic, La Jolla, California
| | | | | | | | | | | | | |
Collapse
|
49
|
Keller KE, Vranka JA, Haddadin RI, Kang MH, Oh DJ, Rhee DJ, Yang YF, Sun YY, Kelley MJ, Acott TS. The effects of tenascin C knockdown on trabecular meshwork outflow resistance. Invest Ophthalmol Vis Sci 2013; 54:5613-23. [PMID: 23882691 DOI: 10.1167/iovs.13-11620] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Tenascin C (TNC) is a matricellular glycoprotein whose expression in adult tissue is indicative of tissue remodeling. The purpose of the current study was to determine the localization of TNC in trabecular meshwork (TM) tissue and to analyze the effects of TNC on intraocular pressure (IOP). METHODS Human TM frontal sections were immunostained with anti-TNC and imaged by confocal microscopy. TNC mRNA and protein levels were quantitated in anterior segments perfused at physiological and elevated pressure. Short, hairpin RNA (shRNA) silencing lentivirus targeting full-length TNC (shTNC) was applied to anterior segment perfusion organ cultures. The IOPs and central corneal thickness (CCT) of wild-type, TNC(-/-), and tenascin X (TNX(-/-)) knockout mice were measured. RESULTS TNC was distributed in the juxtacanalicular (JCT) region of adult human TM, predominantly in the basement membrane underlying the inner wall of Schlemm's canal. Application of shTNC lentivirus to human and porcine anterior segments in perfusion culture did not significantly affect outflow rate. Although TNC was upregulated in response to pressure, there was no difference in outflow rate when shTNC-silenced anterior segments were subjected to elevated pressure. Furthermore, IOPs and CCTs were not significantly different between TNC(-/-) or TNX(-/-) and wild-type mice. CONCLUSIONS TNC does not appear to contribute directly to outflow resistance. However, TNC immunolocalization in the JCT of adult human eyes suggests that certain areas of the TM are being continuously remodeled with or without an IOP increase.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rodrigues M, Yates CC, Nuschke A, Griffith L, Wells A. The matrikine tenascin-C protects multipotential stromal cells/mesenchymal stem cells from death cytokines such as FasL. Tissue Eng Part A 2013; 19:1972-83. [PMID: 23541003 DOI: 10.1089/ten.tea.2012.0568] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multipotential stromal cells/mesenchymal stem cells (MSCs) are attractive candidates for regenerative therapy due to the ability of these cells to differentiate and positively influence neighboring cells. However, on implantation for wound reconstruction, these cells are lost as they are challenged by nonspecific inflammation signals generated in the wound environment and in response to any implanted foreign body. We have previously shown that sustained and surface-restricted epidermal growth factor receptor (EGFR) signaling by a tethered form of its prototypal ligand EGF enhances survival of MSC in the presence of death cytokines such as FasL, serum deprivation, and low oxygen in vitro. This was proposed to be due to the plasma membrane restriction of EGFR signaling. Interestingly, during wound repair, an extracellular matrix (ECM) component Tenascin-C (TNC) containing EGF-like repeats (EGFL) and fibronectin-like repeats (FNL) is upregulated. A few of the 14 EGFL on each of the 6 arms, especially the 14th, bind as low-affinity/high-avidity ligands to EGFR causing sustained surface-restricted EGFR signaling. We queried whether signaling by this physiologically relevant EGFR matrikine also protects MSCs from FasL-induced death. MSCs grown on TNC and Collagen I (as TNC by itself is antiadhesive) displayed a survival advantage in the presence of FasL. TNC neither sequestered nor neutralized FasL; rather, the effects of survival were via cell signaling. This survival was dependent on TNC activating EGFR and downstream pathways of Erk and Akt through EGFL; to a much lesser extent, the FNL of TNC also contributed to survival. Taken together, these results suggest that providing MSCs with a nonimmunogenic naturally occurring ECM moiety such as TNC enhances their survival in the presence of death factors, and this advantage occurs via signaling through EGFR primarily and integrins only to a minor extent. This matrix component is proposed to supplement MSC delivery on the scaffolds to provide a survival advantage against death upon in vivo implantation.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|