1
|
Longmate WM. The epidermal integrin-mediated secretome regulates the skin microenvironment during tumorigenesis and repair. Matrix Biol 2024:S0945-053X(24)00130-6. [PMID: 39491760 DOI: 10.1016/j.matbio.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Integrins are cellular transmembrane receptors that physically connect the cytoskeleton with the extracellular matrix. As such, they are positioned to mediate cellular responses to microenvironmental cues. Importantly, integrins also regulate their own microenvironment through secreted factors, also known as the integrin-mediated secretome. Epidermal integrins, or integrins expressed by keratinocytes of the skin epidermis, regulate the cutaneous microenvironment through the contribution of matrix components, via proteolytic matrix remodeling, or by mediating factors like cytokines and growth factors that can promote support for nearby but distinct cells of the stroma, such as immune cells, endothelial cells, and fibroblasts. This role for integrins is enhanced during both pathological and repair tissue remodeling processes, such as tumor growth and progression and wound healing. This review will discuss examples of how the epithelial integrin-mediated secretome can regulate the tissue microenvironment. Although different epithelial integrins in various contexts will be explored, emphasis will be given to epidermal integrins that regulate the secretome during wound healing and cutaneous tumor progression. Epidermal integrin α3β1 is of particular focus as well, since this integrin has been revealed as a key regulator of the keratinocyte secretome.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
2
|
Grenier C, Lin IH, Peters D, Pozzi A, Lennon R, Naylor RW. Integrin alpha1 beta1 promotes interstitial fibrosis in a mouse model of polycystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619080. [PMID: 39484448 PMCID: PMC11526950 DOI: 10.1101/2024.10.18.619080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Fibrosis is the cause of end-stage kidney failure in patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD). The molecular and cellular mechanisms involved in fibrosis are complex and anti-fibrotic therapies have so far failed to make an impact on patient welfare. Using unbiased proteomics analysis on the Pkd1 nl/nl mouse, we found that expression of the integrin α1 subunit is increased in this model of ADPKD. In human ADPKD tissue and two single cell RNA kidney disease datasets, ITGA1 was also upregulated. To investigate the functional role of this integrin subunit in ADPKD, we generated a Pkd1 nl/nl Itga1 -/- mouse. We observed a significant reduction in kidney volume and kidney dysfunction in mice lacking the integrin α1 subunit. Kidneys from Pkd1 nl/nl Itga1 -/- mice had smaller cysts and reduced interstitial expansion and tubular atrophy. Picrosirius red staining identified a restriction in collagen staining in the interstitium and the myofibroblast marker α smooth muscle actin was also downregulated. Myofibroblast cell proliferation was reduced in Pkd1 nl/nl Itga1 -/- mice and primary fibroblast cultures demonstrated an abrogated fibrogenic phenotype in integrin α1-depleted fibroblasts. These results highlight a previously unrecognised role for the integrin α1 subunit in kidney fibrosis.
Collapse
|
3
|
Coelho NM, Riahi P, Wang Y, Ali A, Norouzi M, Kotlyar M, Jurisica I, McCulloch CA. The major vault protein integrates adhesion-driven signals to regulate collagen remodeling. Cell Signal 2024; 124:111447. [PMID: 39368789 DOI: 10.1016/j.cellsig.2024.111447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
DDR1 interacts with fibrillar collagen and can affect β1 integrin-dependent signaling, but the mechanism that mediates functional interactions between these two different receptors is not defined. We searched for molecules that link DDR1 and β1 integrin-dependent signaling in response to collagen binding. The activation of DDR1 by binding to fibrillar collagen reduced by 5-fold, β1 integrin-dependent ERK phosphorylation that leads to MMP1 expression. In contrast, pharmacological inhibition of DDR1 or culturing cells on fibronectin restored ERK phosphorylation and MMP1 expression mediated by the β1 integrin. A phospho-site screen indicated that collagen-induced DDR1 activation inhibited β1 integrin-dependent ERK signaling by regulating autophosphorylation of focal adhesion kinase (FAK). Immunoprecipitation, mass spectrometry, and protein-protein interaction mapping showed that while DDR1 and FAK do not interact directly, the major vault protein (MVP) binds DDR1 and FAK depending on the substrate. MVP associated with DDR1 in cells expressing β1 integrin that were cultured on collagen. Knockdown of MVP restored ERK activation and MMP1 expression in DDR1-expressing cells cultured on collagen. Immunostaining of invasive cancers in human colon showed colocalization of DDR1 with MVP. These data indicate that MVP interactions with DDR1 and FAK contribute to the regulation of β1 integrin-dependent signaling pathways that drive collagen degradation.
Collapse
Affiliation(s)
- Nuno M Coelho
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Pardis Riahi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Aiman Ali
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Max Kotlyar
- Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada; Osteoarthritis Research Program, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, ON, Canada
| | | |
Collapse
|
4
|
Yu D, Lu Z, Nie F, Chong Y. Integrins regulation of wound healing processes: insights for chronic skin wound therapeutics. Front Cell Infect Microbiol 2024; 14:1324441. [PMID: 38505290 PMCID: PMC10949986 DOI: 10.3389/fcimb.2024.1324441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Integrins are heterodimers composed of non-covalently associated alpha and beta subunits that mediate the dynamic linkage between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs and are involved in different physiological and pathological molecular responses in vivo. Wound healing is an important process in the recovery from traumatic diseases and consists of three overlapping phases: inflammation, proliferation, and remodeling. Integrin regulation acts throughout the wound healing process to promote wound healing. Prolonged inflammation may lead to failure of wound healing, such as wound chronicity. One of the main causes of chronic wound formation is bacterial colonization of the wound. In this review, we review the role of integrins in the regulation of wound healing processes such as angiogenesis and re-epithelialization, as well as the role of integrins in mediating bacterial infections during wound chronicity, and the challenges and prospects of integrins as therapeutic targets for infected wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fengsong Nie
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
5
|
Hunter EJ, Hamaia SW, Kim PSK, Malcor JDM, Farndale RW. The effects of inhibition and siRNA knockdown of collagen-binding integrins on human umbilical vein endothelial cell migration and tube formation. Sci Rep 2022; 12:21601. [PMID: 36517525 PMCID: PMC9751114 DOI: 10.1038/s41598-022-25937-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Blood vessels in the body are lined with endothelial cells which have vital roles in numerous physiological and pathological processes. Collagens are major constituents of the extracellular matrix, and many adherent cells express several collagen-binding adhesion receptors. Here, we study the endothelium-collagen interactions mediated by the collagen-binding integrins, α1β1, α2β1, α10β1 and α11β1 expressed in human umbilical vein endothelial cells (HUVECs). Using qPCR, we found expression of the α10 transcript of the chondrocyte integrin, α10β1, along with the more abundant α2, and low-level expression of α1. The α11 transcript was not detected. Inhibition or siRNA knockdown of the α2-subunit resulted in impaired HUVEC adhesion, spreading and migration on collagen-coated surfaces, whereas inhibition or siRNA knockdown of α1 had no effect on these processes. In tube formation assays, inhibition of either α1 or α2 subunits impaired the network complexity, whereas siRNA knockdown of these integrins had no such effect. Knockdown of α10 had no effect on cell spreading, migration or tube formation in these conditions. Overall, our results indicate that the collagen-binding integrins, α1β1 and α2β1 play a central role in endothelial cell motility and self-organisation.
Collapse
Affiliation(s)
- Emma J Hunter
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
- Stem Cell and Brain Research Institute, Université Lyon 1, INSERM U1208, 18 Avenue Doyen Lépine, 69500, Bron, France
| | - Samir W Hamaia
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
| | - Peter S-K Kim
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
| | - Jean-Daniel M Malcor
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMS3444 BioSciences Gerland-Lyon Sud, UMR5305, CNRS/Université Lyon 1, Lyon, France
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK.
- CambCol Laboratories Ltd, 18 Oak Lane, Littleport, Ely, CB6 1QZ, UK.
| |
Collapse
|
6
|
Mohammad Omar J, Hai Y, Jin S. Hypoxia-induced factor and its role in liver fibrosis. PeerJ 2022; 10:e14299. [PMID: 36523459 PMCID: PMC9745792 DOI: 10.7717/peerj.14299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis develops as a result of severe liver damage and is considered a major clinical concern throughout the world. Many factors are crucial for liver fibrosis progression. While advancements have been made to understand this disease, no effective pharmacological drug and treatment strategies have been established that can effectively prevent liver fibrosis or even could halt the fibrotic process. Most of those advances in curing liver fibrosis have been aimed towards mitigating the causes of fibrosis, including the development of potent antivirals to inhibit the hepatitis virus. It is not practicable for many individuals; however, a liver transplant becomes the only suitable alternative. A liver transplant is an expensive procedure. Thus, there is a significant need to identify potential targets of liver fibrosis and the development of such agents that can effectively treat or reverse liver fibrosis by targeting them. Researchers have identified hypoxia-inducible factors (HIFs) in the last 16 years as important transcription factors driving several facets of liver fibrosis, making them possible therapeutic targets. The latest knowledge on HIFs and their possible role in liver fibrosis, along with the cell-specific activities of such transcription factors that how they play role in liver fibrosis progression, is discussed in this review.
Collapse
Affiliation(s)
- Jan Mohammad Omar
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| | - Yang Hai
- College of International Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
8
|
Zeltz C, Alam J, Liu H, Erusappan PM, Hoschuetzky H, Molven A, Parajuli H, Cukierman E, Costea DE, Lu N, Gullberg D. α11β1 Integrin is Induced in a Subset of Cancer-Associated Fibroblasts in Desmoplastic Tumor Stroma and Mediates In Vitro Cell Migration. Cancers (Basel) 2019; 11:E765. [PMID: 31159419 PMCID: PMC6627481 DOI: 10.3390/cancers11060765] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/26/2019] [Accepted: 05/29/2019] [Indexed: 01/06/2023] Open
Abstract
Integrin α11β1 is a collagen receptor that has been reported to be overexpressed in the stroma of non-small cell lung cancer (NSCLC) and of head and neck squamous cell carcinoma (HNSCC). In the current study, we further analyzed integrin α11 expression in 14 tumor types by screening a tumor tissue array while using mAb 203E3, a newly developed monoclonal antibody to human α11. Different degrees of expression of integrin α11 were observed in the stroma of breast, ovary, skin, lung, uterus, stomach, and pancreatic ductal adenocarcinoma (PDAC) tumors. Co-expression queries with the myofibroblastic cancer-associated fibroblast (myCAF) marker, alpha smooth muscle actin (αSMA), demonstrated a moderate level of α11+ in myCAFs associated with PDAC and HNSCC tumors, and a lack of α11 expression in additional stromal cells (i.e., cells positive for fibroblast-specific protein 1 (FSP1) and NG2). The new function-blocking α11 antibody, mAb 203E1, inhibited cell adhesion to collagen I, partially hindered fibroblast-mediated collagen remodeling and obstructed the three-dimensional (3D) migration rates of PDAC myCAFs. Our data demonstrate that integrin α11 is expressed in a subset of non-pericyte-derived CAFs in a range of cancers and suggest that α11β1 constitutes an important receptor for collagen remodeling and CAF migration in the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Jahedul Alam
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Hengshuo Liu
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Pugazendhi M Erusappan
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Heinz Hoschuetzky
- nanoTools Antikörpertechnik, Tscheulinstr. 21, 79331 Teningen, Germany.
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, NO-5020 Bergen, Norway.
- Department of Pathology, Haukeland University Hospital, NO-5020 Bergen, Norway.
| | - Himalaya Parajuli
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Edna Cukierman
- Cancer Biology Department, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA.
| | - Daniela-Elena Costea
- Department of Pathology, Haukeland University Hospital, NO-5020 Bergen, Norway.
- Department of Clinical Medicine, Center for Cancer Biomarkers CCBIO and Gade Laboratory for Pathology, University of Bergen, NO-5020 Bergen, Norway.
| | - Ning Lu
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, NO-5009 Bergen, Norway.
| |
Collapse
|
9
|
Sipilä KH, Drushinin K, Rappu P, Jokinen J, Salminen TA, Salo AM, Käpylä J, Myllyharju J, Heino J. Proline hydroxylation in collagen supports integrin binding by two distinct mechanisms. J Biol Chem 2018; 293:7645-7658. [PMID: 29615493 DOI: 10.1074/jbc.ra118.002200] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/23/2018] [Indexed: 11/06/2022] Open
Abstract
Collagens are the most abundant extracellular matrix proteins in vertebrates and have a characteristic triple-helix structure. Hydroxylation of proline residues is critical for helix stability, and diminished prolyl hydroxylase activity causes wide-spread defects in connective tissues. Still, the role of proline hydroxylation in the binding of collagen receptors such as integrins is unclear. Here, we isolated skin collagen from genetically modified mice having reduced prolyl 4-hydroxylase activity. At room temperature, the reduced proline hydroxylation did not affect interactions with the recombinant integrin α2I domain, but at 37 °C, collagen hydroxylation correlated with the avidity of α2I domain binding. Of note, LC-MS/MS analysis of isolated skin collagens revealed no major changes in the hydroxyproline content of the main integrin-binding sites. Thus, the disrupted α2I domain binding at physiological temperatures was most likely due to structural destabilization of the collagenous helix. Integrin α2I binding to the triple-helical GFPGER motif was slightly weaker than to GFOGER (O = hydroxyproline). This phenomenon was more prominent when α1 integrin was tested. Integrin α1β1 expressed on CHO cells and recombinant α1I domain showed remarkably slower binding velocity and weaker avidity to GFPGER when compared with GFOGER. Structural modeling revealed the critical interaction between Arg-218 in α1I and the hydroxyproline residue in the integrin-binding motif. The role of Arg-218 was further validated by testing a variant R218D α1I domain in solid-phase binding assays. Thus, our results show that the lack of proline hydroxylation in collagen can affect integrin binding by a direct mechanism and via structural destabilization of the triple helix.
Collapse
Affiliation(s)
- Kalle H Sipilä
- From the Department of Biochemistry, University of Turku, FI-20014 Turku, Finland
| | - Kati Drushinin
- the Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland, and
| | - Pekka Rappu
- From the Department of Biochemistry, University of Turku, FI-20014 Turku, Finland
| | - Johanna Jokinen
- From the Department of Biochemistry, University of Turku, FI-20014 Turku, Finland
| | - Tiina A Salminen
- the Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland
| | - Antti M Salo
- the Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland, and
| | - Jarmo Käpylä
- From the Department of Biochemistry, University of Turku, FI-20014 Turku, Finland
| | - Johanna Myllyharju
- the Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland, and
| | - Jyrki Heino
- From the Department of Biochemistry, University of Turku, FI-20014 Turku, Finland,
| |
Collapse
|
10
|
Vining KH, Scherba JC, Bever AM, Alexander MR, Celiz AD, Mooney DJ. Synthetic Light-Curable Polymeric Materials Provide a Supportive Niche for Dental Pulp Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:10.1002/adma.201704486. [PMID: 29215170 PMCID: PMC5788014 DOI: 10.1002/adma.201704486] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/19/2017] [Indexed: 05/08/2023]
Abstract
Dental disease annually affects billions of patients, and while regenerative dentistry aims to heal dental tissue after injury, existing polymeric restorative materials, or fillings, do not directly participate in the healing process in a bioinstructive manner. There is a need for restorative materials that can support native functions of dental pulp stem cells (DPSCs), which are capable of regenerating dentin. A polymer microarray formed from commercially available monomers to rapidly identify materials that support DPSC adhesion is used. Based on these findings, thiol-ene chemistry is employed to achieve rapid light-curing and minimize residual monomer of the lead materials. Several triacrylate bulk polymers support DPSC adhesion, proliferation, and differentiation in vitro, and exhibit stiffness and tensile strength similar to existing dental materials. Conversely, materials composed of a trimethacrylate monomer or bisphenol A glycidyl methacrylate, which is a monomer standard in dental materials, do not support stem cell adhesion and negatively impact matrix and signaling pathways. Furthermore, thiol-ene polymerized triacrylates are used as permanent filling materials at the dentin-pulp interface in direct contact with irreversibly injured pulp tissue. These novel triacrylate-based biomaterials have potential to enable novel regenerative dental therapies in the clinic by both restoring teeth and providing a supportive niche for DPSCs.
Collapse
Affiliation(s)
- Kyle H Vining
- Wyss Institute for Biologically Inspired Engineering and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Jacob C Scherba
- Wyss Institute for Biologically Inspired Engineering and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Alaina M Bever
- Wyss Institute for Biologically Inspired Engineering and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Adam D Celiz
- Wyss Institute for Biologically Inspired Engineering and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering and Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
11
|
Minchin JEN, Rawls JF. Elucidating the role of plexin D1 in body fat distribution and susceptibility to metabolic disease using a zebrafish model system. Adipocyte 2017; 6:277-283. [PMID: 28792859 DOI: 10.1080/21623945.2017.1356504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular disease, diabetes and cancer were responsible for 68% of all deaths worldwide in 2012. The regional distribution of lipid deposited within adipose tissue (AT) - so called body fat distribution (BFD) - is a strong risk factor for NCDs. BFD is highly heritable; however, the genetic basis of BFD is almost entirely unknown. Genome-wide association studies have identified several loci associated with BFD, including at Plexin D1 (PLXND1) - a gene known to modulate angiogenesis. We recently demonstrated that zebrafish homozygous for a null mutation in plxnd1 had a reduced capacity to store lipid in visceral AT (VAT) leading to altered BFD. Moreover, we found that type V collagens were upregulated in plxnd1 mutants, and mediated the inhibitory effect of Plxnd1 on VAT growth. These results strengthen evidence that Plxnd1 influences BFD in human populations, and validate zebrafish as a model to study BFD. However, many pertinent questions remain unanswered. Here we outline potential Plxnd1 mechanisms of action in AT, and describe the genetic architecture at human PLXND1 that is associated with BFD and NCD susceptibility.
Collapse
Affiliation(s)
- James E. N. Minchin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
12
|
Russo C, Lazzaro V, Gazzaruso C, Maurotti S, Ferro Y, Pingitore P, Fumo F, Coppola A, Gallotti P, Zambianchi V, Fodaro M, Galliera E, Marazzi MG, Corsi Romanelli MM, Giannini S, Romeo S, Pujia A, Montalcini T. Proinsulin C-peptide modulates the expression of ERK1/2, type I collagen and RANKL in human osteoblast-like cells (Saos-2). Mol Cell Endocrinol 2017; 442:134-141. [PMID: 28007656 DOI: 10.1016/j.mce.2016.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 12/29/2022]
Abstract
A lower bone mass accompanied by a higher bone fragility with increased risk of fracture are observed in individuals with type 1 diabetes mellitus. Low C-peptide levels are associated with low lumbar mineral density in postmenopausal woman. In this work, we investigated the role of C-peptide on the osteoblast cell biology in vitro. We examined intracellular pathways and we found that C peptide activates ERK1/2 in human osteoblast-like cells (Saos-2). We also observed that proinsulin C-peptide prevents a reduction of type I collagen expression and decreases, in combination with insulin, receptor activator of nuclear factor-κB (RANKL) levels. In this work we show for the first time that Cpeptide activates a specific intracellular pathway in osteoblasts and it modulates the expression of protein involved in bone remodeling. Our results suggest that both C-peptide may have a role in bone metabolism. Further studies are needing to fully clarify its role.
Collapse
Affiliation(s)
- Cristina Russo
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Veronica Lazzaro
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Carmine Gazzaruso
- Internal and Emergency Medicine, and Ce.R.C.A. Clinical Institute "Beato Matteo", Vigevano, Italy
| | - Samantha Maurotti
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Yvelise Ferro
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Piero Pingitore
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden
| | - Francesca Fumo
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Adriana Coppola
- Internal and Emergency Medicine, and Ce.R.C.A. Clinical Institute "Beato Matteo", Vigevano, Italy
| | - Pietro Gallotti
- Internal and Emergency Medicine, and Ce.R.C.A. Clinical Institute "Beato Matteo", Vigevano, Italy
| | - Valentina Zambianchi
- Internal and Emergency Medicine, and Ce.R.C.A. Clinical Institute "Beato Matteo", Vigevano, Italy
| | - Mariangela Fodaro
- Internal and Emergency Medicine, and Ce.R.C.A. Clinical Institute "Beato Matteo", Vigevano, Italy
| | - Emanuela Galliera
- Department of Biomedical, Surgical and Dental Science, University of Milan, Italy
| | | | | | - Sandro Giannini
- Department of Medical and Surgical Sciences, University of Padova, Italy
| | - Stefano Romeo
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy; Department of Molecular and Clinical Medicine, University of Gothenburg, Sweden
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, Italy.
| |
Collapse
|
13
|
Abstract
Reactive astrogliosis occurs after central nervous system (CNS) injuries whereby resident astrocytes form rapid responses along a graded continuum. Following CNS lesions, naïve astrocytes are converted into reactive astrocytes and eventually into scar-forming astrocytes that block axon regeneration and neural repair. It has been known for decades that scarring development and its related extracellular matrix molecules interfere with regeneration of injured axons after CNS injury, but the cellular and molecular mechanisms for controlling astrocytic scar formation and maintenance are not well known. Recent use of various genetic tools has made tremendous progress in better understanding genesis of reactive astrogliosis. Especially, the latest experiments demonstrate environment-dependent plasticity of reactive astrogliosis because reactive astrocytes isolated from injured spinal cord form scarring astrocytes when transplanted into injured spinal cord, but revert in retrograde to naive astrocytes when transplanted into naive spinal cord. The interactions between upregulated type I collagen and its receptor integrin β1 and the N-cadherin-mediated cell adhesion appear to play major roles for local astrogliosis around the lesion. This review centers on the environment-dependent plasticity of reactive astrogliosis after spinal cord injury and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Fatima M Nathan
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
14
|
Ge C, Wang Z, Zhao G, Li B, Liao J, Sun H, Franceschi RT. Discoidin Receptor 2 Controls Bone Formation and Marrow Adipogenesis. J Bone Miner Res 2016; 31:2193-2203. [PMID: 27341689 PMCID: PMC5135576 DOI: 10.1002/jbmr.2893] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 06/16/2016] [Accepted: 06/22/2016] [Indexed: 01/09/2023]
Abstract
Cell-extracellular matrix (ECM) interactions play major roles in controlling progenitor cell fate and differentiation. The receptor tyrosine kinase, discoidin domain receptor 2 (DDR2), is an important mediator of interactions between cells and fibrillar collagens. DDR2 signals through both ERK1/2 and p38 MAP kinase, which stimulate osteoblast differentiation and bone formation. Here we show that DDR2 is critical for skeletal development and differentiation of marrow progenitor cells to osteoblasts while suppressing marrow adipogenesis. Smallie mice (Ddr2slie/slie ), which contain a nonfunctional Ddr2 allele, have multiple skeletal defects. A progressive decrease in tibial trabecular bone volume/total volume (BV/TV) was observed when wild-type (WT), Ddr2wt/slie , and Ddr2slie/slie mice were compared. These changes were associated with reduced trabecular number (Tb.N) and trabecular thickness (Tb.Th) and increased trabecular spacing (Tb.Sp) in both males and females, but reduced cortical thickness only in Ddr2slie/slie females. Bone changes were attributed to decreased bone formation rather than increased osteoclast activity. Significantly, marrow fat and adipocyte-specific mRNA expression were significantly elevated in Ddr2slie/slie animals. Additional skeletal defects include widened calvarial sutures and reduced vertebral trabecular bone. To examine the role of DDR2 signaling in cell differentiation, bone marrow stromal cells (BMSCs) were grown under osteogenic and adipogenic conditions. Ddr2slie/slie cells exhibited defective osteoblast differentiation and accelerated adipogenesis. Changes in differentiation were related to activity of runt-related transcription factor 2 (RUNX2) and PPARγ, transcription factors that are both controlled by MAPK-dependent phosphorylation. Specifically, the defective osteoblast differentiation in calvarial cells from Ddr2slie/slie mice was associated with reduced ERK/MAP kinase and RUNX2-S319 phosphorylation and could be rescued with a constitutively active phosphomimetic RUNX2 mutant. Also, DDR2 was shown to increase RUNX2-S319 phosphorylation and transcriptional activity while also increasing PPARγ-S112 phosphorylation, but reducing its activity. DDR2 is, therefore, important for maintenance of osteoblast activity and suppression of marrow adipogenesis in vivo and these actions are related to changes in MAPK-dependent RUNX2 and PPARγ phosphorylation. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
| | - Zhengyan Wang
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
| | - Guisheng Zhao
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
| | - Binbin Li
- Department of Oral Pathology, Peking University School of Stomatology, Beijing 10081, P.R.China
| | - Jinhui Liao
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
| | - Hanshi Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
| | - Renny T. Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078
- Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-0600
| |
Collapse
|
15
|
The role of biophysical properties of provisional matrix proteins in wound repair. Matrix Biol 2016; 60-61:124-140. [PMID: 27534610 DOI: 10.1016/j.matbio.2016.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/15/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022]
Abstract
Wound healing is a complex, dynamic process required for maintaining homeostasis in an organism. Along with being controlled biochemically, wound healing is also controlled through the transduction of biophysical stimuli through cell interactions with the extracellular matrix (ECM). This review provides an overview of the ECM's role in the wound healing process and subsequently expands on the variety of roles biophysical phenomenon play.
Collapse
|
16
|
Baumann S, Hennet T. Collagen Accumulation in Osteosarcoma Cells lacking GLT25D1 Collagen Galactosyltransferase. J Biol Chem 2016; 291:18514-24. [PMID: 27402836 DOI: 10.1074/jbc.m116.723379] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Indexed: 01/22/2023] Open
Abstract
Collagen is post-translationally modified by prolyl and lysyl hydroxylation and subsequently by glycosylation of hydroxylysine. Despite the widespread occurrence of the glycan structure Glc(α1-2)Gal linked to hydroxylysine in animals, the functional significance of collagen glycosylation remains elusive. To address the role of glycosylation in collagen expression, folding, and secretion, we used the CRISPR/Cas9 system to inactivate the collagen galactosyltransferase GLT25D1 and GLT25D2 genes in osteosarcoma cells. Loss of GLT25D1 led to increased expression and intracellular accumulation of collagen type I, whereas loss of GLT25D2 had no effect on collagen secretion. Inactivation of the GLT25D1 gene resulted in a compensatory induction of GLT25D2 expression. Loss of GLT25D1 decreased collagen glycosylation by up to 60% but did not alter collagen folding and thermal stability. Whereas cells harboring individually inactivated GLT25D1 and GLT25D2 genes could be recovered and maintained in culture, cell clones with simultaneously inactive GLT25D1 and GLT25D2 genes could be not grown and studied, suggesting that a complete loss of collagen glycosylation impairs osteosarcoma cell proliferation and viability.
Collapse
Affiliation(s)
- Stephan Baumann
- From the Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland
| | - Thierry Hennet
- From the Institute of Physiology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
17
|
Contribution of collagen adhesion receptors to tissue fibrosis. Cell Tissue Res 2016; 365:521-38. [DOI: 10.1007/s00441-016-2440-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
|
18
|
Zeltz C, Gullberg D. The integrin-collagen connection--a glue for tissue repair? J Cell Sci 2016; 129:653-64. [PMID: 26857815 DOI: 10.1242/jcs.180992] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The α1β1, α2β1, α10β1 and α11β1 integrins constitute a subset of the integrin family with affinity for GFOGER-like sequences in collagens. Integrins α1β1 and α2β1 were originally identified on a subset of activated T-cells, and have since been found to be expressed on a number of cell types including platelets (α2β1), vascular cells (α1β1, α2β1), epithelial cells (α1β1, α2β1) and fibroblasts (α1β1, α2β1). Integrin α10β1 shows a distribution that is restricted to mesenchymal stem cells and chondrocytes, whereas integrin α11β1 appears restricted to mesenchymal stem cells and subsets of fibroblasts. The bulk of the current literature suggests that collagen-binding integrins only have a limited role in adult connective tissue homeostasis, partly due to a limited availability of cell-binding sites in the mature fibrillar collagen matrices. However, some recent data suggest that, instead, they are more crucial for dynamic connective tissue remodeling events--such as wound healing--where they might act specifically to remodel and restore the tissue architecture. This Commentary discusses the recent development in the field of collagen-binding integrins, their roles in physiological and pathological settings with special emphasis on wound healing, fibrosis and tumor-stroma interactions, and include a discussion of the most recently identified newcomers to this subfamily--integrins α10β1 and α11β1.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, Bergen N-5009, Norway
| |
Collapse
|
19
|
Roth KJ, Copple BL. Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell Mol Gastroenterol Hepatol 2015; 1:589-597. [PMID: 28210703 PMCID: PMC5301877 DOI: 10.1016/j.jcmgh.2015.09.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Liver fibrosis remains a significant clinical problem in the United States and throughout the world. Although important advances in the understanding of this disease have been made, no effective pharmacologic agents have been developed that directly prevent or reverse the fibrotic process. Many of the successes in liver fibrosis treatment have been targeted toward treating the cause of fibrosis, such as the development of new antivirals that eradicate hepatitis virus. For many patients, however, this is not feasible, so a liver transplant remains the only viable option. Thus, there is a critical need to identify new therapeutic targets that will slow or reverse the progression of fibrosis in such patients. Research over the last 16 years has identified hypoxia-inducible factors (HIFs) as key transcription factors that drive many aspects of liver fibrosis, making them potential targets of therapy. In this review, we discuss the latest work on HIFs and liver fibrosis, including the cell-specific functions of these transcription factors in the development of liver fibrosis.
Collapse
Key Words
- BDL, bile duct ligation
- CCl4, carbon tetrachloride
- Ccr, C-C chemokine receptor
- FGF, fibroblast growth factor
- HGF, hepatocyte growth factor
- HIFs, hypoxia-inducible factors
- HSC, hepatic stellate cell
- Hepatic Stellate Cells
- Hypoxia-Inducible Factors
- Jmjd, Jumonji domain-containing
- Kupffer Cells
- Liver Fibrosis
- PAI-1, plasminogen activator inhibitor-1
- PDGF, platelet-derived growth factor
- Rgs, regulator of G-protein signaling
- TGF-β, transforming growth factor β
- VEGF, vascular endothelial growth factor
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
| | - Bryan L. Copple
- Correspondence Address correspondence to: Bryan L. Copple, PhD, Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, B403 Life Sciences Building, East Lansing, Michigan 48824.Department of Pharmacology and ToxicologyMichigan State University1355 Bogue Street, B403 Life Sciences BuildingEast LansingMichigan 48824
| |
Collapse
|
20
|
Cereceres S, Touchet T, Browning MB, Smith C, Rivera J, Höök M, Whitfield-Cargile C, Russell B, Cosgriff-Hernandez E. Chronic Wound Dressings Based on Collagen-Mimetic Proteins. Adv Wound Care (New Rochelle) 2015; 4:444-456. [PMID: 26244101 DOI: 10.1089/wound.2014.0614] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/27/2015] [Indexed: 12/26/2022] Open
Abstract
Objective: Chronic wounds are projected to reach epidemic proportions due to the aging population and the increasing incidence of diabetes. There is a strong clinical need for an improved wound dressing that can balance wound moisture, promote cell migration and proliferation, and degrade at an appropriate rate to minimize the need for dressing changes. Approach: To this end, we have developed a bioactive, hydrogel microsphere wound dressing that incorporates a collagen-mimetic protein, Scl2GFPGER, to promote active wound healing. A redesigned Scl2GFPGER, engineered collagen (eColGFPGER), was created to reduce steric hindrance of integrin-binding motifs and increase overall stability of the triple helical backbone, thereby resulting in increased cell adhesion to substrates. Results: This study demonstrates the successful modification of the Scl2GFPGER protein to eColGFPGER, which displayed enhanced stability and integrin interactions. Fabrication of hydrogel microspheres provided a matrix with adaptive moisture technology, and degradation rates have potential for use in human wounds. Innovation: This collagen-mimetic wound dressing was designed to permit controlled modulation of cellular interactions and degradation rate without impact on other physical properties. Its fabrication into uniform hydrogel microspheres provides a bioactive dressing that can readily conform to irregular wounds. Conclusion: Overall, this new eColGFPGER shows strong promise in the generation of bioactive hydrogels for wound healing as well as a variety of tissue scaffolds.
Collapse
Affiliation(s)
- Stacy Cereceres
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Tyler Touchet
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Mary Beth Browning
- Institute for Bioscience and Technology, Texas A&M Health Science Center, Houston, Texas
| | - Clayton Smith
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - Jose Rivera
- Institute for Bioscience and Technology, Texas A&M Health Science Center, Houston, Texas
| | - Magnus Höök
- Institute for Bioscience and Technology, Texas A&M Health Science Center, Houston, Texas
| | | | - Brooke Russell
- Institute for Bioscience and Technology, Texas A&M Health Science Center, Houston, Texas
| | | |
Collapse
|
21
|
Abstract
Type I collagen is a fibrillar protein, a member of a large family of collagen proteins. It is present in most body tissues, usually in combination with other collagens and other components of extracellular matrix. Its synthesis is increased in various pathological situations, in healing wounds, in fibrotic tissues and in many tumors. After extraction from collagen-rich tissues it is widely used in studies of cell behavior, especially those of fibroblasts and myofibroblasts. Cells cultured in a classical way, on planar plastic dishes, lack the third dimension that is characteristic of body tissues. Collagen I forms gel at neutral pH and may become a basis of a 3D matrix that better mimics conditions in tissue than plastic dishes.
Collapse
Affiliation(s)
- Jiří Kanta
- a Department of Medical Biochemistry; Medical Faculty in Hradec Králové; Charles University ; Prague , Czech Republic
| |
Collapse
|
22
|
Olivares-Navarrete R, Rodil SE, Hyzy SL, Dunn GR, Almaguer-Flores A, Schwartz Z, Boyan BD. Role of integrin subunits in mesenchymal stem cell differentiation and osteoblast maturation on graphitic carbon-coated microstructured surfaces. Biomaterials 2015; 51:69-79. [PMID: 25770999 DOI: 10.1016/j.biomaterials.2015.01.035] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/26/2014] [Accepted: 01/20/2015] [Indexed: 12/20/2022]
Abstract
Surface roughness, topography, chemistry, and energy promote osteoblast differentiation and increase osteogenic local factor production in vitro and bone-to-implant contact in vivo, but the mechanisms involved are not well understood. Knockdown of integrin heterodimer alpha2beta1 (α2β1) blocks the osteogenic effects of the surface, suggesting signaling by this integrin homodimer is required. The purpose of the present study was to separate effects of surface chemistry and surface structure on integrin expression by coating smooth or rough titanium (Ti) substrates with graphitic carbon, retaining surface morphology but altering surface chemistry. Ti surfaces (smooth [Ra < 0.4 μm], rough [Ra ≥ 3.4 μm]) were sputter-coated using a magnetron sputtering system with an ultrapure graphite target, producing a graphitic carbon thin film. Human mesenchymal stem cells and MG63 osteoblast-like cells had higher mRNA for integrin subunits α1, α2, αv, and β1 on rough surfaces in comparison to smooth, and integrin αv on graphitic-carbon-coated rough surfaces in comparison to Ti. Osteogenic differentiation was greater on rough surfaces in comparison to smooth, regardless of chemistry. Silencing integrins β1, α1, or α2 decreased osteoblast maturation on rough surfaces independent of surface chemistry. Silencing integrin αv decreased maturation only on graphitic carbon-coated surfaces, not on Ti. These results suggest a major role of the integrin β1 subunit in roughness recognition, and that integrin alpha subunits play a major role in surface chemistry recognition.
Collapse
Affiliation(s)
- Rene Olivares-Navarrete
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Sandra E Rodil
- Instituto de Investigaciones en Materiales, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Sharon L Hyzy
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Ginger R Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Zvi Schwartz
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
23
|
Schulz JN, Zeltz C, Sørensen IW, Barczyk M, Carracedo S, Hallinger R, Niehoff A, Eckes B, Gullberg D. Reduced granulation tissue and wound strength in the absence of α11β1 integrin. J Invest Dermatol 2015; 135:1435-1444. [PMID: 25634355 PMCID: PMC4407012 DOI: 10.1038/jid.2015.24] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/24/2022]
Abstract
Previous wound healing studies have failed to define a role for either α1β1 or α2β1 integrin in fibroblast-mediated wound contraction, suggesting the involvement of another collagen receptor in this process. Our previous work demonstrated that the integrin subunit α11 is highly induced during wound healing both at the mRNA and protein level, prompting us to investigate and dissect the role of the integrin α11β1 during this process. Therefore, we used mice with a global ablation of either α2 or α11 or both integrin subunits and investigated the repair of excisional wounds. Analyses of wounds demonstrated that α11β1 deficiency results in reduced granulation tissue formation and impaired wound contraction, independently of the presence of α2β1. Our combined in vivo and in vitro data further demonstrate that dermal fibroblasts lacking α11β1 are unable to efficiently convert to myofibroblasts, resulting in scar tissue with compromised tensile strength. Moreover, we suggest that the reduced stability of the scar is a consequence of poor collagen remodeling in α11−/− wounds associated with defective transforming growth factor-β–dependent JNK signaling.
Collapse
Affiliation(s)
| | - Cédric Zeltz
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Ida W Sørensen
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Malgorzata Barczyk
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Sergio Carracedo
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Ralf Hallinger
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopedics, German Sport University, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics, University of Cologne, Cologne, Germany
| | - Beate Eckes
- Department of Dermatology, University of Cologne, Cologne, Germany.
| | - Donald Gullberg
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway.
| |
Collapse
|
24
|
Borza CM, Chen X, Zent R, Pozzi A. Cell Receptor-Basement Membrane Interactions in Health and Disease: A Kidney-Centric View. CURRENT TOPICS IN MEMBRANES 2015; 76:231-53. [PMID: 26610916 PMCID: PMC4913201 DOI: 10.1016/bs.ctm.2015.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-extracellular matrix (ECM) interactions are essential for tissue development, homeostasis, and response to injury. Basement membranes (BMs) are specialized ECMs that separate epithelial or endothelial cells from stromal components and interact with cells via cellular receptors, including integrins and discoidin domain receptors. Disruption of cell-BM interactions due to either injury or genetic defects in either the ECM components or cellular receptors often lead to irreversible tissue injury and loss of organ function. Animal models that lack specific BM components or receptors either globally or in selective tissues have been used to help with our understanding of the molecular mechanisms whereby cell-BM interactions regulate organ function in physiological and pathological conditions. We review recently published works on animal models that explore how cell-BM interactions regulate kidney homeostasis in both health and disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Xiwu Chen
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| |
Collapse
|
25
|
Integrin-mediated adhesion and mechano-sensing in cutaneous wound healing. Cell Tissue Res 2014; 360:571-82. [DOI: 10.1007/s00441-014-2064-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/11/2014] [Indexed: 12/30/2022]
|
26
|
Koivisto L, Heino J, Häkkinen L, Larjava H. Integrins in Wound Healing. Adv Wound Care (New Rochelle) 2014; 3:762-783. [PMID: 25493210 DOI: 10.1089/wound.2013.0436] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Indexed: 01/06/2023] Open
Abstract
Significance: Regulation of cell adhesions during tissue repair is fundamentally important for cell migration, proliferation, and protein production. All cells interact with extracellular matrix proteins with cell surface integrin receptors that convey signals from the environment into the nucleus, regulating gene expression and cell behavior. Integrins also interact with a variety of other proteins, such as growth factors, their receptors, and proteolytic enzymes. Re-epithelialization and granulation tissue formation are crucially dependent on the temporospatial function of multiple integrins. This review explains how integrins function in wound repair. Recent Advances: Certain integrins can activate latent transforming growth factor beta-1 (TGF-β1) that modulates wound inflammation and granulation tissue formation. Dysregulation of TGF-β1 function is associated with scarring and fibrotic disorders. Therefore, these integrins represent targets for therapeutic intervention in fibrosis. Critical Issues: Integrins have multifaceted functions and extensive crosstalk with other cell surface receptors and molecules. Moreover, in aberrant healing, integrins may assume different functions, further increasing the complexity of their functionality. Discovering and understanding the role that integrins play in wound healing provides an opportunity to identify the mechanisms for medical conditions, such as excessive scarring, chronic wounds, and even cancer. Future Directions: Integrin functions in acute and chronic wounds should be further addressed in models better mimicking human wounds. Application of any products in acute or chronic wounds will potentially alter integrin functions that need to be carefully considered in the design.
Collapse
Affiliation(s)
- Leeni Koivisto
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Lari Häkkinen
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| |
Collapse
|
27
|
Olivares-Navarrete R, Hyzy SL, Berg ME, Schneider JM, Hotchkiss K, Schwartz Z, Boyan BD. Osteoblast lineage cells can discriminate microscale topographic features on titanium-aluminum-vanadium surfaces. Ann Biomed Eng 2014; 42:2551-61. [PMID: 25227453 PMCID: PMC4239824 DOI: 10.1007/s10439-014-1108-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/03/2014] [Indexed: 01/23/2023]
Abstract
Titanium (Ti) and Ti alloys are used in orthopaedic/spine applications where biological implant fixation, or osseointegration, is required for long-term stability. These implants employ macro-scale features to provide mechanical stability until arthrodesis, features that are too large to influence healing at the cellular level. Micron-scale rough Ti alloy (Ti–6Al–4V) increases osteoblastic differentiation and osteogenic factor production in vitro and increases in vivo bone formation; however, effects of overall topography, including sub-micron scale and nanoscale features, on osteoblast lineage cells are less well appreciated. To address this, Ti6Al4V surfaces with macro/micro/nano-textures were generated using sand blasting and acid etching that had comparable average roughness values but differed in other roughness parameters (total roughness, profile roughness, maximum peak height, maximum valley depth, root-mean-squared roughness, kurtosis, skewness) (#5, #9, and #12). Human mesenchymal stem cells (HMSCs) and normal human osteoblasts (NHOst) were cultured for 7 days on the substrates and then analyzed for alkaline phosphatase activity and osteocalcin content, production of osteogenic local factors, and integrin subunit expression. All three surfaces supported osteoblastic differentiation of HMSCs and further maturation of NHOst cells, but the greatest response was seen on the #9 substrate, which had the lowest skewness and kurtosis. The #9 surface also induced highest expression of α2 and β1 integrin mRNA. HMSCs produced highest levels of ITGAV on #9, suggesting this integrin may play a role for early lineage cells. These results indicate that osteoblast lineage cells are sensitive to specific micro/nanostructures, even when overall macro roughness is comparable and suggest that skewness and kurtosis are important variables.
Collapse
|
28
|
Reduced FOXO1 expression accelerates skin wound healing and attenuates scarring. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2465-79. [PMID: 25010393 DOI: 10.1016/j.ajpath.2014.05.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 04/21/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022]
Abstract
The forkhead box O (FOXO) family has been extensively investigated in aging and metabolism, but its role in tissue-repair processes remains largely unknown. Herein, we clarify the molecular aspect of the FOXO family in skin wound healing. We demonstrated that Foxo1 and Foxo3a were both up-regulated during murine skin wound healing. Partial knockout of Foxo1 in Foxo1(+/-) mice throughout the body led to accelerated skin wound healing with enhanced keratinocyte migration, reduced granulation tissue formation, and decreased collagen density, accompanied by an attenuated inflammatory response, but we observed no wound phenotype in Foxo3a(-/-) mice. Fibroblast growth factor 2, adiponectin, and notch1 genes were significantly increased at wound sites in Foxo1(+/-) mice, along with markedly altered extracellular signal-regulated kinase 1/2 and AKT phosphorylation. Similarly, transient knockdown of Foxo1 at the wound site by local delivery of antisense oligodeoxynucleotides enhanced skin wound healing. The link between FOXO1 and scarring extends to patients, in particular keloid scars, where we see FOXO1 expression markedly increased in fibroblasts and inflammatory cells within the otherwise normal dermis. This occurs in the immediate vicinity of the keloid by comparison to the center of the mature keloid, indicating that FOXO1 is associated with the overgrowth of this fibrotic response into adjacent normal skin. Overall, our data indicate that molecular targeting of FOXO1 may improve the quality of healing and reduce pathological scarring.
Collapse
|
29
|
Chen X, Wang H, Liao HJ, Hu W, Gewin L, Mernaugh G, Zhang S, Zhang ZY, Vega-Montoto L, Vanacore RM, Fässler R, Zent R, Pozzi A. Integrin-mediated type II TGF-β receptor tyrosine dephosphorylation controls SMAD-dependent profibrotic signaling. J Clin Invest 2014; 124:3295-310. [PMID: 24983314 DOI: 10.1172/jci71668] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 05/21/2014] [Indexed: 12/20/2022] Open
Abstract
Tubulointerstitial fibrosis underlies all forms of end-stage kidney disease. TGF-β mediates both the development and the progression of kidney fibrosis through binding and activation of the serine/threonine kinase type II TGF-β receptor (TβRII), which in turn promotes a TβRI-mediated SMAD-dependent fibrotic signaling cascade. Autophosphorylation of serine residues within TβRII is considered the principal regulatory mechanism of TβRII-induced signaling; however, there are 5 tyrosine residues within the cytoplasmic tail that could potentially mediate TβRII-dependent SMAD activation. Here, we determined that phosphorylation of tyrosines within the TβRII tail was essential for SMAD-dependent fibrotic signaling within cells of the kidney collecting duct. Conversely, the T cell protein tyrosine phosphatase (TCPTP) dephosphorylated TβRII tail tyrosine residues, resulting in inhibition of TβR-dependent fibrotic signaling. The collagen-binding receptor integrin α1β1 was required for recruitment of TCPTP to the TβRII tail, as mice lacking this integrin exhibited impaired TCPTP-mediated tyrosine dephosphorylation of TβRII that led to severe fibrosis in a unilateral ureteral obstruction model of renal fibrosis. Together, these findings uncover a crosstalk between integrin α1β1 and TβRII that is essential for TβRII-mediated SMAD activation and fibrotic signaling pathways.
Collapse
|
30
|
Miller CG, Pozzi A, Zent R, Schwarzbauer JE. Effects of high glucose on integrin activity and fibronectin matrix assembly by mesangial cells. Mol Biol Cell 2014; 25:2342-50. [PMID: 24943838 PMCID: PMC4142608 DOI: 10.1091/mbc.e14-03-0800] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aberrant accumulation of collagen IV defines diabetic nephropathy. It is shown here that high glucose increases fibronectin matrix assembly by activating integrin receptors on kidney cells. Collagen IV accumulation depends on this fibronectin matrix. Targeting fibronectin matrix may be a useful therapy to stem matrix accumulation in the diabetic kidney. The filtration unit of the kidney is the glomerulus, a capillary network supported by mesangial cells and extracellular matrix (ECM). Glomerular function is compromised in diabetic nephropathy (DN) by uncontrolled buildup of ECM, especially type IV collagen, which progressively occludes the capillaries. Increased levels of the ECM protein fibronectin (FN) are also present; however, its role in DN is unknown. Mesangial cells cultured under high glucose conditions provide a model system for studying the effect of elevated glucose on deposition of FN and collagen IV. Imaging of mesangial cell cultures and analysis of detergent-insoluble matrix show that, under high glucose conditions, mesangial cells assembled significantly more FN matrix, independent of FN protein levels. High glucose conditions induced protein kinase C–dependent β1 integrin activation, and FN assembly in normal glucose was increased by stimulation of integrin activity with Mn2+. Collagen IV incorporation into the matrix was also increased under high glucose conditions and colocalized with FN fibrils. An inhibitor of FN matrix assembly prevented collagen IV deposition, demonstrating dependence of collagen IV on FN matrix. We conclude that high glucose induces FN assembly, which contributes to collagen IV accumulation. Enhanced assembly of FN might facilitate dysregulated ECM accumulation in DN.
Collapse
Affiliation(s)
- Charles G Miller
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232Department of Medicine, Veterans Affairs Medical Center, Nashville, TN 37212
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232Department of Medicine, Veterans Affairs Medical Center, Nashville, TN 37212Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | | |
Collapse
|
31
|
Endothelial cells enhance the in vivo bone-forming ability of osteogenic cell sheets. J Transl Med 2014; 94:663-73. [PMID: 24709778 DOI: 10.1038/labinvest.2014.55] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/30/2014] [Accepted: 03/06/2014] [Indexed: 01/20/2023] Open
Abstract
Addressing the problem of vascularization is of vital importance when engineering three-dimensional (3D) tissues. Endothelial cells are increasingly used in tissue-engineered constructs to obtain prevascularization and to enhance in vivo neovascularization. Rat bone marrow stromal cells were cultured in thermoresponsive dishes under osteogenic conditions with human umbilical vein endothelial cells (HUVECs) to obtain homotypic or heterotypic cell sheets (CSs). Cells were retrieved as sheets from the dishes after incubation at 20 °C. Monoculture osteogenic CSs were stacked on top of homotypic or heterotypic CSs, and subcutaneously implanted in the dorsal flap of nude mice for 7 days. The implants showed mineralized tissue formation under both conditions. Transplanted osteogenic cells were found at the new tissue site, demonstrating CS bone-inductive effect. Perfused vessels, positive for human CD31, confirmed the contribution of HUVECs for the neovascularization of coculture CS constructs. Furthermore, calcium quantification and expression of osteocalcin and osterix genes were higher for the CS constructs, with HUVECs demonstrating the more robust osteogenic potential of these constructs. This work demonstrates the potential of using endothelial cells, combined with osteogenic CSs, to increase the in vivo vascularization of CS-based 3D constructs for bone tissue engineering purposes.
Collapse
|
32
|
Sipilä K, Haag S, Denessiouk K, Käpylä J, Peters EC, Denesyuk A, Hansen U, Konttinen Y, Johnson MS, Holmdahl R, Heino J. Citrullination of collagen II affects integrin‐mediated cell adhesion in a receptor‐specific manner. FASEB J 2014; 28:3758-68. [DOI: 10.1096/fj.13-247767] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Kalle Sipilä
- Department of BiochemistryUniversity of TurkuTurkuFinland
| | - Sabrina Haag
- Division of Medical Inflammation Research, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, Department of BioscienceÅbo Akademi UniversityTurkuFinland
| | - Jarmo Käpylä
- Department of BiochemistryUniversity of TurkuTurkuFinland
| | - Eric C. Peters
- Genomics Institute of the Novartis Research FoundationSan DiegoCaliforniaUSA
| | - Alexander Denesyuk
- Structural Bioinformatics Laboratory, Biochemistry, Department of BioscienceÅbo Akademi UniversityTurkuFinland
| | - Uwe Hansen
- Department of Physiology, Chemistry, and PathobiochemistryMuenster University HospitalMuensterGermany
| | - Yrjö Konttinen
- Department of MedicineInstitute of Clinical Medicine, University of HelsinkiHelsinkiFinland
| | - Mark S. Johnson
- Structural Bioinformatics Laboratory, Biochemistry, Department of BioscienceÅbo Akademi UniversityTurkuFinland
| | - Rikard Holmdahl
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
- Division of Medical Inflammation Research, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Jyrki Heino
- Department of BiochemistryUniversity of TurkuTurkuFinland
| |
Collapse
|
33
|
Longmate WM, DiPersio CM. Integrin Regulation of Epidermal Functions in Wounds. Adv Wound Care (New Rochelle) 2014; 3:229-246. [PMID: 24669359 DOI: 10.1089/wound.2013.0516] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/13/2014] [Indexed: 12/12/2022] Open
Abstract
Significance: Integrins are bidirectional signaling receptors for extracellular matrix that regulate both inside-out signaling that controls keratinocyte-mediated changes to the wound microenvironment and outside-in signaling that controls keratinocyte responses to microenvironmental changes. As such, integrins represent attractive therapeutic targets for treatment of chronic wounds or general promotion of wound healing. Advances in wound management are particularly important as the elderly and diabetic populations within the United States continue to grow. Recent Advances: Although integrins are best known for mediating cell adhesion and migration, integrins in wound epidermis also control cell survival, proliferation, matrix remodeling, and paracrine crosstalk to other cellular compartments of the wound. Importantly, the concept of targeting integrins in the clinic has been established for treatment of certain cancers and other diseases, laying the groundwork for similar exploitation of integrins as targets to treat chronic wounds. Critical Issues: Despite their attractiveness as therapeutic targets, integrins have complex roles in wound healing that are impacted by both their own expression and a highly dynamic wound microenvironment that determines ligand availability. Therefore, identifying relevant integrin ligands in the wound and understanding both distinct and overlapping functions that different integrins play in the epidermis will be critical to determine their precise roles in wound healing. Future Directions: Future research should focus on gaining a thorough understanding of the highly coordinated functions of different integrins in wound epidermis, and on determining which of these functions go awry in pathological wounds. This focus should facilitate development of integrin-targeting therapeutics for treating chronic wounds.
Collapse
Affiliation(s)
- Whitney M. Longmate
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York
| | - C. Michael DiPersio
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York
| |
Collapse
|
34
|
Heino J. Cellular signaling by collagen-binding integrins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:143-55. [PMID: 25023173 DOI: 10.1007/978-94-017-9153-3_10] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The four collagen-binding αI domain integrins form their own subgroup among cell adhesion receptors. The signaling functions of α1β1 and α2β1 integrins have been analyzed in many experimental models, whereas less studies are available about the more recently found α10β1 and α11β1 heterodimers. Interestingly, collagen binding by α1β1 and α2β1 often generates opposite cellular responses. For example α1β1 has often been reported to promote cell proliferation and to suppress collagen synthesis, whereas α2β1 can in many model systems inhibit growth and promote collagen synthesis. There are obviously cell type dependent factors modifying the signaling. Additionally the structure and the organization of collagenous matrix play a critic role. Many recent studies have also stressed the importance of the crosstalk between the integrins and other cell surface receptors.
Collapse
Affiliation(s)
- Jyrki Heino
- Department of Biochemistry, University of Turku, 20014, Turku, Finland,
| |
Collapse
|
35
|
Abstract
Integrin α1β1 is widely expressed in mesenchyme and the immune system, as well as a minority of epithelial tissues. Signaling through α1 contributes to the regulation of extracellular matrix composition, in addition to supplying in some tissues a proliferative and survival signal that appears to be unique among the collagen binding integrins. α1 provides a tissue retention function for cells of the immune system including monocytes and T cells, where it also contributes to their long-term survival, providing for peripheral T cell memory, and contributing to diseases of autoimmunity. The viability of α1 null mice, as well as the generation of therapeutic monoclonal antibodies against this molecule, have enabled studies of the role of α1 in a wide range of pathophysiological circumstances. The immune functions of α1 make it a rational therapeutic target.
Collapse
|
36
|
Zeltz C, Orgel J, Gullberg D. Molecular composition and function of integrin-based collagen glues-introducing COLINBRIs. Biochim Biophys Acta Gen Subj 2013; 1840:2533-48. [PMID: 24361615 DOI: 10.1016/j.bbagen.2013.12.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/13/2013] [Accepted: 12/14/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Despite detailed knowledge about the structure and signaling properties of individual collagen receptors, much remains to be learned about how these receptors participate in linking cells to fibrillar collagen matrices in tissues. In addition to collagen-binding integrins, a group of proteins with affinity both for fibrillar collagens and integrins link these two protein families together. We have introduced the name COLINBRI (COLlagen INtegrin BRIdging) for this set of molecules. Whereas collagens are the major building blocks in tissues and defects in these structural proteins have severe consequences for tissue integrity, the mild phenotypes of the integrin type of collagen receptors have raised questions about their importance in tissue biology and pathology. SCOPE OF REVIEW We will discuss the two types of cell linkages to fibrillar collagen (direct- versus indirect COLINBRI-mediated) and discuss how the parallel existence of direct and indirect linkages to collagens may ensure tissue integrity. MAJOR CONCLUSIONS The observed mild phenotypes of mice deficient in collagen-binding integrins and the relatively restricted availability of integrin-binding sequences in mature fibrillar collagen matrices support the existence of indirect collagen-binding mechanisms in parallel with direct collagen binding in vivo. GENERAL SIGNIFICANCE A continued focus on understanding the molecular details of cell adhesion mechanisms to collagens will be important and will benefit our understanding of diseases like tissue- and tumor fibrosis where collagen dynamics are disturbed. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | - Joseph Orgel
- Departments of Biology, Physics and Biomedical Engineering, Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 3440 S. Dearborn Ave, Chicago, IL 60616, USA
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| |
Collapse
|
37
|
Abstract
A major hallmark of chronic kidney injury is fibrosis, which is characterized by increased accumulation of extracellular matrix components that replace the damaged tissue. Normally, the synthesis and degradation of extracellular matrix components are finely regulated; however, when matrix replacement goes unchecked, there is unwanted and irreversible tissue scarring with consequent organ damage, organ failure, and, in certain cases, death. Many factors, including cell-matrix interactions, play a role in the development of renal fibrosis. Cell-matrix interactions are made possible by integrins, a family of transmembrane receptors that, upon binding to the extracellular matrix, activate intracellular signaling. Thus, they control various cell functions, including survival, proliferation, migration, and matrix homeostasis. Genetic mutations in humans and the development of animal models lacking integrins in selective parts of the kidney have improved our understanding of molecular mechanisms and pathways controlling matrix remodeling in kidney disease. Here we outline the major integrins involved in kidney disease and some of the major molecular mechanisms whereby integrins contribute to kidney fibrosis.
Collapse
Affiliation(s)
- Ambra Pozzi
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 27232, USA.
| | | |
Collapse
|
38
|
Widgerow AD. Chronic wounds - is cellular 'reception' at fault? Examining integrins and intracellular signalling. Int Wound J 2013; 10:185-92. [PMID: 22494436 PMCID: PMC7950432 DOI: 10.1111/j.1742-481x.2012.00967.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
As with all physiologic processes, chronic wounds are associated with unique intracellular and cellular/extracellular matrix (ECM) receptor types and signalling messages. These cellular receptors mediate responses of the epidermis to provisional wound matrix and change in form and number in cases of impaired wound healing. Integrins are the major cell-surface receptors for cell adhesion and migration and epidermal keratinocytes express several integrins that bind ECM ligands in provisional wound ECM. Integrin receptors and more particularly integrin clusters and focal adhesion points appear to influence epidermal and dermal cell matrix interactions, cell motility, cell phenotype and ultimate healing trajectory. In chronic wounds, a variety of changes in receptors have been identified: decreased integrin α5β1 receptors affect the integration of fibronectin and subsequent keratinocyte migration; integrin αvβ6 stimulate transforming growth factor (TGF)-β and may increase the susceptibility to ulceration and fibrosis; however, TGF-β signal receptors have been found to be dysfunctional in many chronic wounds; additionally receptor interactions result in increased senescent cells including fibroblasts, myofibroblasts and even keratinocytes - this produces a degradative ECM and wound bed and corrosive chronic wound fluid. The activation or inhibition of integrin receptors by various agents may provide an excellent means of influencing wound healing. This process offers an earlier intervention into the wound healing cascade promoting intrinsic healing and elaboration of growth factors and ECM proteins, which may be more cost effective than the traditional attempts at extrinsic addition of these agents.
Collapse
Affiliation(s)
- Alan D Widgerow
- Faculty of Health Sciences, Plastic Surgery Department, University of the Witwatersrand, Johannnesburg, South Africa and Adar science Inc., Irvine, CA, USA.
| |
Collapse
|
39
|
Zeisberg EM, Zeisberg M. The role of promoter hypermethylation in fibroblast activation and fibrogenesis. J Pathol 2012; 229:264-73. [DOI: 10.1002/path.4120] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 01/26/2023]
Affiliation(s)
- Elisabeth M Zeisberg
- Department of Cardiology and Pneumology; Göttingen University Medical Centre, Georg August University; Göttingen Germany
- German Centre for Cardiovascular Research (DZHK); Göttingen Germany
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology; Göttingen University Medical Centre, Georg August University; Göttingen Germany
| |
Collapse
|
40
|
Abstract
Integrins are heterodimers that mediate cell attachment to the extracellular matrix. Previously, we used fibroblast-specific knockout mice to show that the integrin β1 subunit was required in vivo for dermal fibrogenesis and cutaneous tissue repair. Here, we show that integrin β1 expression by fibroblasts is required for dermal homeostasis; at 8 weeks after deletion of integrin β1, the dermis is significantly thinner, expressing less collagen and displaying reduced generation of reactive oxygen species (ROS). Cultured integrin β1-deficient fibroblasts show reduced rac1 activation and ROS generation. Overexpressing rac1 in integrin β1-deficient fibroblasts restored ROS generation; adding hydrogen peroxide to integrin β1-deficient fibroblasts restored Col1a2 (collagen, type I, α2) and α-smooth muscle actin (α-SMA) mRNA expression and α-SMA protein expression and stress fiber formation. Thus integrin β1 expression by fibroblasts is required for the maintenance of dermal connective tissue via a rac/ROS pathway.
Collapse
Affiliation(s)
- Shangxi Liu
- Department of Dentistry, University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
41
|
Abstract
Initial efforts at biologic skin replacement strategies were mainly directed toward keratinocyte regeneration and epithelial replacement. It soon became evident that without a good dermal scaffold, the long-term efficacy of epithelial replacement was very limited. Further studies have focused on matrix replacement predominantly involving collagen frameworks with or without cellular additions. The fibroblast is central to the process of dermal regeneration and to the success of biologic matrix design. The sequence of cellular focal adhesion, integrin phosphorylated activation, intracellular and extracellular signaling, cytoskeletal activation, changes in cell morphology, and cytokine growth factor interaction are all important in influencing cell proliferation, cell spreading, neocollagenesis, and collagen translocation. A basic acellular matrix with chemical composition and correct physical structure (pore size and resistance) that takes cognizance of this sequence of matrix deposition and fibroblast functionality should be successful in promoting intrinsic healing and dermal replacement.
Collapse
|
42
|
Shi M, Pedchenko V, Greer BH, Van Horn WD, Santoro SA, Sanders CR, Hudson BG, Eichman BF, Zent R, Pozzi A. Enhancing integrin α1 inserted (I) domain affinity to ligand potentiates integrin α1β1-mediated down-regulation of collagen synthesis. J Biol Chem 2012; 287:35139-35152. [PMID: 22888006 DOI: 10.1074/jbc.m112.358648] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin α1β1 binding to collagen IV, which is mediated by the α1-inserted (I) domain, down-regulates collagen synthesis. When unligated, a salt bridge between Arg(287) and Glu(317) is thought to keep this domain in a low affinity conformation. Ligand binding opens the salt bridge leading to a high-affinity conformation. How modulating integrin α1β1 affinity alters collagen homeostasis is unknown. To address this question, we utilized a thermolysin-derived product of the α1α2α1 network of collagen IV (α1α2α1(IV) truncated protomer) that selectively binds integrin α1β1. We show that an E317A substitution enhanced binding to the truncated protomer, consistent with a previous finding that this substitution eliminates the salt bridge. Surprisingly, we show that an R287A substitution did not alter binding, whereas R287E/E317R substitutions enhanced binding to the truncated protomer. NMR spectroscopy and molecular modeling suggested that eliminating the Glu(317) negative charge is sufficient to induce a conformational change toward the open state. Thus, the role played by Glu(317) is largely independent of the salt bridge. We further show that cells expressing E317A or R287E/E317R substitutions have enhanced down-regulation of collagen IV synthesis, which is mediated by the ERK/MAPK pathway. In conclusion, we have demonstrated that modulating the affinity of the extracellular α1 I domain to collagen IV enhances outside-in signaling by potentiating ERK activation and enhancing the down-regulation of collagen synthesis.
Collapse
Affiliation(s)
- Mingjian Shi
- Department of Medicine (Nephrology), Vanderbilt University, Nashville, Tennessee 37232
| | - Vadim Pedchenko
- Department of Medicine (Nephrology), Vanderbilt University, Nashville, Tennessee 37232; Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232
| | - Briana H Greer
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232
| | - Wade D Van Horn
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| | - Samuel A Santoro
- Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee 37232
| | - Charles R Sanders
- Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| | - Billy G Hudson
- Department of Medicine (Nephrology), Vanderbilt University, Nashville, Tennessee 37232; Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| | - Brandt F Eichman
- Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| | - Roy Zent
- Department of Medicine (Nephrology), Vanderbilt University, Nashville, Tennessee 37232; Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Medicine, Veterans Affairs Medical Center, Nashville, Tennessee 37212
| | - Ambra Pozzi
- Department of Medicine (Nephrology), Vanderbilt University, Nashville, Tennessee 37232; Center for Matrix Biology, Vanderbilt University, Nashville, Tennessee 37232; Department of Medicine, Veterans Affairs Medical Center, Nashville, Tennessee 37212.
| |
Collapse
|
43
|
Borza CM, Pozzi A. The role of cell-extracellular matrix interactions in glomerular injury. Exp Cell Res 2012; 318:1001-10. [PMID: 22417893 DOI: 10.1016/j.yexcr.2012.02.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/24/2012] [Indexed: 01/09/2023]
Abstract
Glomerulosclerosis is characterized by excessive deposition of extracellular matrix within the glomeruli of the kidney, glomerular cell death, and subsequent loss of functional glomeruli. While in physiological situations the levels of extracellular matrix components are kept constant by a tight balance between formation and degradation, in the case of injury that results in fibrosis there is increased matrix deposition relative to its breakdown. Multiple factors control matrix synthesis and degradation, thus contributing to the development of glomerulosclerosis. This review focuses primarily on the role of cell-matrix interactions, which play a critical role in governing glomerular cell cues in both healthy and diseased kidneys. Cell-extracellular matrix interactions are made possible by various cellular receptors including integrins, discoidin domain receptors, and dystroglycan. Upon binding to a selective extracellular matrix protein, these receptors activate intracellular signaling pathways that can either downregulate or upregulate matrix synthesis and deposition. This, together with the observation that changes in the expression levels of matrix receptors have been documented in glomerular disease, clearly emphasizes the contribution of cell-matrix interactions in glomerular injury. Understanding the molecular mechanisms whereby extracellular matrix receptors regulate matrix homeostasis in the course of glomerular injury is therefore critical for devising more effective therapies to treat and ideally prevent glomerulosclerosis.
Collapse
Affiliation(s)
- Corina M Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, TN 37232, USA.
| | | |
Collapse
|
44
|
Yu L, Su Y, Paueksakon P, Cheng H, Chen X, Wang H, Harris RC, Zent R, Pozzi A. Integrin α1/Akita double-knockout mice on a Balb/c background develop advanced features of human diabetic nephropathy. Kidney Int 2012; 81:1086-97. [PMID: 22297672 PMCID: PMC3345314 DOI: 10.1038/ki.2011.474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Animal models that mimic human diabetic nephropathy are useful to identify key factors in pathogenesis of this disease, as well as the development of new therapies. Several mouse models of diabetes have features of human diabetic nephropathy, yet none of these completely fulfill the Animal Models of Diabetes Complications Consortium criteria and completely reproduce pathological and functional features of the human disease. The Akita mouse carries a mutation in the insulin-2 gene and, to date, only survives as heterozygotes that develop spontaneous type 1 diabetes. Here we show that Akita mice with mutation of both insulin-2 alleles (Akita knockout (KO)) survive if crossed onto the Balb/c background. These mice develop hyperglycemia, more severe albuminuria, and mesangial sclerosis compared with heterozygous mice on the same genetic background. Interestingly, crossing these AkitaKO mice with integrin α1KO mice, a model of exacerbated glomerulosclerosis after injury and also on the Balb/c background, resulted in a 16-fold increase in albuminuria, significant mesangial matrix expansion, nodular and diffuse glomerulosclerosis, and a 2-fold increase in glomerular basement membrane thickening when compared with nondiabetic mice. Moreover, a significant decline in glomerular filtration was evident in the α1KOAkitaKO mice at 6 months of age. Thus, the integrin α1KOAkitaKO Balb/c mouse represents a promising model presenting with most features of human diabetic nephropathy.
Collapse
Affiliation(s)
- Ling Yu
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lahti M, Bligt E, Niskanen H, Parkash V, Brandt AM, Jokinen J, Patrikainen P, Käpylä J, Heino J, Salminen TA. Structure of collagen receptor integrin α(1)I domain carrying the activating mutation E317A. J Biol Chem 2011; 286:43343-51. [PMID: 22030389 PMCID: PMC3234817 DOI: 10.1074/jbc.m111.261909] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 10/07/2011] [Indexed: 11/06/2022] Open
Abstract
We have analyzed the structure and function of the integrin α(1)I domain harboring a gain-of-function mutation E317A. To promote protein crystallization, a double variant with an additional C139S mutation was used. In cell adhesion assays, the E317A mutation promoted binding to collagen. Similarly, the double mutation C139S/E317A increased adhesion compared with C139S alone. Furthermore, soluble α(1)I C139S/E317A was a higher avidity collagen binder than α(1)I C139S, indicating that the double variant represents an activated form. The crystal structure of the activated variant of α(1)I was solved at 1.9 Å resolution. The E317A mutation results in the unwinding of the αC helix, but the metal ion has moved toward loop 1, instead of loop 2 in the open α(2)I. Furthermore, unlike in the closed αI domains, the metal ion is pentacoordinated and, thus, prepared for ligand binding. Helix 7, which has moved downward in the open α(2)I structure, has not changed its position in the activated α(1)I variant. During the integrin activation, Glu(335) on helix 7 binds to the metal ion at the metal ion-dependent adhesion site (MIDAS) of the β(1) subunit. Interestingly, in our cell adhesion assays E317A could activate collagen binding even after mutating Glu(335). This indicates that the stabilization of helix 7 into its downward position is not required if the α(1) MIDAS is already open. To conclude, the activated α(1)I domain represents a novel conformation of the αI domain, mimicking the structural state where the Arg(287)-Glu(317) ion pair has just broken during the integrin activation.
Collapse
Affiliation(s)
- Matti Lahti
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Eva Bligt
- the Structural Bioinformatics Laboratory, Department of Biosciences, Åbo Akademi University, Turku FI-20520, Finland
| | - Henri Niskanen
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Vimal Parkash
- the Structural Bioinformatics Laboratory, Department of Biosciences, Åbo Akademi University, Turku FI-20520, Finland
| | - Anna-Maria Brandt
- the Structural Bioinformatics Laboratory, Department of Biosciences, Åbo Akademi University, Turku FI-20520, Finland
| | - Johanna Jokinen
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Pekka Patrikainen
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Jarmo Käpylä
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Jyrki Heino
- From the Department of Biochemistry and Food Chemistry, University of Turku, Turku FI-20014, Finland and
| | - Tiina A. Salminen
- the Structural Bioinformatics Laboratory, Department of Biosciences, Åbo Akademi University, Turku FI-20520, Finland
| |
Collapse
|
46
|
Integrin-mediated cell-matrix interaction in physiological and pathological blood vessel formation. JOURNAL OF ONCOLOGY 2011; 2012:125278. [PMID: 21941547 PMCID: PMC3175391 DOI: 10.1155/2012/125278] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 07/15/2011] [Indexed: 02/07/2023]
Abstract
Physiological as well as pathological blood vessel formation are fundamentally dependent on cell-matrix interaction. Integrins, a family of major cell adhesion receptors, play a pivotal role in development, maintenance, and remodeling of the vasculature. Cell migration, invasion, and remodeling of the extracellular matrix (ECM) are integrin-regulated processes, and the expression of certain integrins also correlates with tumor progression. Recent advances in the understanding of how integrins are involved in the regulation of blood vessel formation and remodeling during tumor progression are highlighted. The increasing knowledge of integrin function at the molecular level, together with the growing repertoire of integrin inhibitors which allow their selective pharmacological manipulation, makes integrins suited as potential diagnostic markers and therapeutic targets.
Collapse
|
47
|
Pradeepkumar Singh L, Vivek Sharma A, Swarnakar S. Upregulation of collagenase-1 and -3 in indomethacin-induced gastric ulcer in diabetic rats: role of melatonin. J Pineal Res 2011; 51:61-74. [PMID: 21342246 DOI: 10.1111/j.1600-079x.2010.00845.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Collagenases are key proteases involved in inflammation and injury. We addressed whether collagenases have an association with the susceptibility of gastric injury under diabetes as well as the effect of melatonin on collagenases in ulcerated gastric tissues. Diabetes was induced in rats by a single dose of streptozotocin (STZ) followed by gastric ulceration using indomethacin, and melatonin's action was studied by its application prior to indomethacin exposure. Ulcer indices and damage were elevated significantly in gastric tissues of diabetic compared with nondiabetic rats. Melatonin reversed the effect of indomethacin during protection of gastric ulcers in diabetic rats. Matrix metalloproteinase (MMP)-13 (i.e., collagenase-3) was upregulated in diabetic gastric mucosa and enhanced further upon ulceration while melatonin ameliorated their activity. In addition, gastric tissues showed enhanced expression of both MMP-1 (i.e., collagenases-1) and -13 significantly in diabetic rats compared with nondiabetic animals and more so during ulceration while tissue inhibitors of metalloproteinase-1 (TIMP-1) showed an opposite trend. MMP-2 activities exhibited a ∼50% downregulation during gastric ulceration which were rescued by melatonin. Moreover, increased expression of both MMP-1 and -13 was mediated by activator protein-1 activation via extracellular signal-regulated kinase 1/2 which were parallel to upregulation of tumor necrosis factor-α, interleukin-1β, and heat shock protein-70 during ulceration. Melatonin arrested collagenase expression by downregulation of these signaling molecules thereby halting the progression of the disease. We conclude that diabetic gastric tissues are susceptible to ulceration and associated with MMP-1 and -13 upregulation in indomethacin-induced injury. Additionally, melatonin protects the gastric damage under diabetes via regulation of both MMP-1 and -13.
Collapse
Affiliation(s)
- Laishram Pradeepkumar Singh
- Department of Physiology, Drug Development Diagnostics and Biotechnology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | | | | |
Collapse
|
48
|
Suen RS, Rampersad SN, Stewart DJ, Courtman DW. Differential roles of endothelin-1 in angiotensin II-induced atherosclerosis and aortic aneurysms in apolipoprotein E-null mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:1549-59. [PMID: 21718678 DOI: 10.1016/j.ajpath.2011.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 04/29/2011] [Accepted: 05/17/2011] [Indexed: 12/16/2022]
Abstract
Because both endothelin-1 (ET-1) and angiotensin II (AngII) are independent mediators of arterial remodeling, we sought to determine the role of ET receptor inhibition in AngII-accelerated atherosclerosis and aortic aneurysm formation. We administered saline or AngII and/or bosentan, an endothelin receptor antagonist (ERA) for 7, 14, or 28 days to 6-week- and 6-month-old apolipoprotein E-knockout mice. AngII treatment increased aortic atherosclerosis, which was reduced by ERA. ET-1 immunostaining was localized to macrophage-rich regions in aneurysmal vessels. ERA did not prevent AngII-induced aneurysm formation but instead may have increased aneurysm incidence. In AngII-treated animals with aneurysms, ERA had a profound effect on the non-aneurysmal thoracic aorta via increasing wall thickness, collagen/elastin ratio, wall stiffness, and viscous responses. These observations were confirmed in acute in vitro collagen sheet production models in which ERA inhibited AngII's dose-dependent effect on collagen type 1 α 1 (COL1A1) gene transcription. However, chronic treatment reduced matrix metalloproteinase 2 mRNA expression but enhanced COL3A1, tissue inhibitor of metalloproteinase 1 (TIMP-1), and TIMP-2 mRNA expressions. These data confirm a role for the ET system in AngII-accelerated atherosclerosis but suggest that ERA therapy is not protective against the formation of AngII-induced aneurysms and can paradoxically stimulate a chronic arterial matrix remodeling response.
Collapse
Affiliation(s)
- Renée S Suen
- Terrence Donnelly Research Laboratories, Division of Cardiology, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
49
|
Chang EH, Pezzulo AA, Zabner J. Do cell junction protein mutations cause an airway phenotype in mice or humans? Am J Respir Cell Mol Biol 2011; 45:202-20. [PMID: 21297078 DOI: 10.1165/rcmb.2010-0498tr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cell junction proteins connect epithelial cells to each other and to the basement membrane. Genetic mutations of these proteins can cause alterations in some epithelia leading to varied phenotypes such as deafness, renal disease, skin disorders, and cancer. This review examines if genetic mutations in these proteins affect the function of lung airway epithelia. We review cell junction proteins with examples of disease mutation phenotypes in humans and in mouse knockout models. We also review which of these genes are expressed in airway epithelium by microarray expression profiling and immunocytochemistry. Last, we present a comprehensive literature review to find the lung phenotype when cell junction and adhesion genes are mutated or subject to targeted deletion. We found that in murine models, targeted deletion of cell junction and adhesion genes rarely result in a lung phenotype. Moreover, mutations in these genes in humans have no obvious lung phenotype. Our research suggests that simply because a cell junction or adhesion protein is expressed in an organ does not imply that it will exhibit a drastic phenotype when mutated. One explanation is that because a functioning lung is critical to survival, redundancy in the system is expected. Therefore mutations in a single gene might be compensated by a related function of a similar gene product. Further studies in human and animal models will help us understand the overlap in the function of cell junction gene products. Finally, it is possible that the human lung phenotype is subtle and has not yet been described.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology–Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, USA
| | | | | |
Collapse
|
50
|
Girgert R, Martin M, Kruegel J, Miosge N, Temme J, Eckes B, Müller GA, Gross O. Integrin α2-deficient mice provide insights into specific functions of collagen receptors in the kidney. FIBROGENESIS & TISSUE REPAIR 2010; 3:19. [PMID: 20860797 PMCID: PMC2954872 DOI: 10.1186/1755-1536-3-19] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Accepted: 09/22/2010] [Indexed: 11/19/2022]
Abstract
Background Integrins are important cellular receptors for collagens. Within the glomerulus, podocytes regulate the integrity of the glomerular basement membrane (GBM) by sensing the presence of collagen and regulating collagen IV synthesis. The present study evaluates the role of integrin α2 (ITGA2) in cell-matrix interaction. Methods and Results ITGA2-deficient mice had normal renal function but moderate proteinuria and enhanced glomerular and tubulointerstitial matrix deposition. Electron microscopy demonstrated irregular podocyte-matrix interaction, causing pathological protrusions towards the urinary (podocyte) side of the GBM. These characteristic subepithelial bulges mimic the renal phenotype of mice, which are deficient in another collagen receptor, discoidin domain receptor (DDR)1. Using immunogold staining, ITGA2 expression was found to localize to the basolateral site of the podocyte foot processes. ITGA2-deficient mice overexpressed transforming growth factor (TGF)β and connective tissue growth factor (CTGF) compared with wild-type mice. Using in situ hybridization, tubular cells were found to be the primary site of TGFβ synthesis and podocytes the source of CTGF in ITGA2-deficient mice. Conclusion These findings support our hypothesis that both these collagen receptors (ITGA2 and DDR1) play a similar role within the kidney. Further, cell-matrix interaction via collagen receptors seems to be crucial for maintenance of normal GBM architecture and function. Targeting collagen receptors such as ITGA2 might be a new form of treatment for progressive fibrotic diseases.
Collapse
Affiliation(s)
- Rainer Girgert
- Department of Nephrology and Rheumatology, Georg-August-University Goettingen, Goettingen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|