1
|
Michalska Z, Ostaszewska A, Fularczyk M, Dzierżyńska M, Bielak K, Morytz J, Sieradzan AK, Archacka K, Brzoska E, Rodziewicz-Motowidło S, Ciemerych MA. In Vitro Bioactivity Evaluation of IL-4 and SDF-1 Mimicking Peptides Engineered to Enhance Skeletal Muscle Reconstruction. J Biomed Mater Res A 2025; 113:e37898. [PMID: 40087853 DOI: 10.1002/jbm.a.37898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Skeletal muscle regeneration depends on satellite cells, which, in response to injury, activate, proliferate, and reconstruct damaged tissue. However, under certain conditions, such as large injuries or myopathies, this process may not be properly executed, and muscle function may be affected. Thus, pro-regenerative actions, such as the use of various factors or cells, are widely tested as a tool to improve muscle regeneration. In the current study, we designed peptides derived from the IL-4 and SDF-1 proteins, namely IL-4-X, IL-4-Y, SDF-1-X, and SDF-1-Y. We showed that these peptides can bind to appropriate receptors and can adopt proper structure in solution. Importantly, we documented, using in vitro culture, that they do not negatively affect the cells that are present and active in skeletal muscles, such as myoblasts and fibroblasts, bone marrow stromal cells, as well as induced pluripotent stem cells, which can serve as a source of myoblasts. The presence of peptides did not affect cell proliferation compared to untreated cells. In vitro culture and differentiation protocols documented that selected IL-4 and SDF-1 peptides increased cell migration and inhibited undesirable adipogenic differentiation. Thus, we proved that these peptides are safe to use in in vivo studies aimed at improving skeletal muscle regeneration.
Collapse
Affiliation(s)
- Zuzanna Michalska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Anna Ostaszewska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Martyna Fularczyk
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Maria Dzierżyńska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kacper Bielak
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Justyna Morytz
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Adam K Sieradzan
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
2
|
Hu J, Zhu Y, Li X, Pang Z, Li X, Zhang H, Wang Y, Li P, Zhou Q. AQP3-liposome@GelMA promotes overloaded-induced degenerated disc regeneration via IBSP/ITG αVβ3/AKT pathway. Int J Biol Macromol 2025; 292:139238. [PMID: 39733899 DOI: 10.1016/j.ijbiomac.2024.139238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Medical and conservative treatments for intervertebral disc degeneration (IDD) primarily focus on alleviating symptoms. However, effective curative therapies that promote disc regeneration remain lacking. Recent advancements in disc repair materials offer a potential solution, but identifying effective cytokines for regeneration and developing efficient drug delivery systems are crucial for success. This study demonstrated a negative correlation between AQP3 expression levels and the extent of disc degeneration induced by mechanical overload, as evidenced in both in vivo and in vitro models, suggesting that AQP3 is a key regulator of intervertebral disc (IVD) homeostasis. Moreover, the overexpression of AQP3 or exogenous AQP3 protein significantly repaired degenerated IVDs. As a membrane protein, exogenous AQP3 is challenging for cells to recognize and internalize. To address this issue, we designed liposomes to encapsulate AQP3 and incorporated them into GelMA (AQP3-lipo@GelMA) for targeted repair of IDD resulting from high mechanical pressure. The encapsulation of AQP3 in liposomes improved cellular recognition and uptake, thereby enhancing its functionality at the cell membrane. Additionally, AQP3 within this material inhibited the binding of integrins to sialic acid-binding proteins (IBSP), which subsequently reduced the expression of the downstream integrin αVβ3. This cascade effect indirectly activated the AKT pathway, promoting the survival of NP cells. In vivo experiments, we found that AQP3-lipo@GelMA had an efficient function of repairing degenerated intervertebral disc. This study introduced AQP3-lipo@GelMA as a promising material for clinical applications in IVD repair.
Collapse
Affiliation(s)
- Junxian Hu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yibo Zhu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xiaoxiao Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Zeyu Pang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Xiangwei Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Huilin Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yiyang Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Pei Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Qiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China; Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| |
Collapse
|
3
|
Roth CE, Niederau C, Radermacher C, Rizk M, Neuss S, Jankowski J, Apel C, Craveiro RB, Wolf M. Knockout of Bone Sialoprotein in Cementoblasts Cell Lines Affects Specific Gene Expression in Unstimulated and Mechanically Stimulated Conditions. Ann Anat 2023; 249:152102. [PMID: 37150306 DOI: 10.1016/j.aanat.2023.152102] [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: 12/22/2022] [Revised: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
One of the major components in cementum extracellular matrix is bone sialoprotein (BSP). BSP knockout (Ibsp) mice were reported to have a nonfunctional hypo-mineralized cementum, as well as detachment and disorganization of the periodontal ligament tissue. However, studies investigating the influence of Ibsp in cementoblasts are missing yet. This study investigates the influences of Bsp in three cementoblasts cell lines (OCCM.30-WT,IbspΔNterm, and IbspKAE). The mRNA expression of cementoblast and osteoclast markers (Col1a1, Alpl, Ocn, Runx2, Ctsk, Rankl and Opg) and the cell morphology were compared. Additionally, a functional monocyte adhesion assay was performed. To understand the influence of external stimuli, the effect of Ibsp was investigated under static compressive force, mimicking the compression side of orthodontic tooth movement. Cementoblasts with genotype IbspΔNterm and IbspKAE showed slight differences in cell morphology compared to OCCM.30-WT, as well as different gene expression. Under compressive force, the Ibsp cell lines presented expression pattern markers similar to the OCCM.30-WT cell line. However, Cathepsin K was strongly upregulated in IbspΔNterm cementoblasts under compressive force. This study provides insight into the role of BSP in cementoblasts and explores the influence of BSP on periodontal ligament tissues. BSP markers in cementoblasts seem to be involved in the regulation of cementum organization as an important factor for a functional periodontium. In summary, our findings provide a basis for investigations regarding molecular biology interactions of BSP in cementoblasts, and a supporting input for understanding the periodontal and cellular cementum remodeling.
Collapse
Affiliation(s)
- Charlotte E Roth
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Correspondence to: Pauwelsstr. 30, 52074 Aachen, Germany. E-mail:
| | - Christian Niederau
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| | - Chloé Radermacher
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Helmholtz Institute for Biomedical Engineering, Bionterface Group, RWTH Aachen University, 52056 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074, Germany
| | - Marta Rizk
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, Bionterface Group, RWTH Aachen University, 52056 Aachen, Germany; Institute of Pathology, RWTH Aachen University Hospital, 52074, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Germany
| | - Christian Apel
- Department of Biohybrid & Medical Textiles (BioTex), Institut of Applied Medical Engineering, RWTH Aachen University & Hospital, Germany
| | - Rogerio B Craveiro
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany; Correspondence to: Pauwelsstr. 30, 52074 Aachen, Germany. E-mail:
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, University of Aachen, 52074 Aachen, Germany
| |
Collapse
|
4
|
Mao L, Wang L, Xu J, Zou J. The role of integrin family in bone metabolism and tumor bone metastasis. Cell Death Discov 2023; 9:119. [PMID: 37037822 PMCID: PMC10086008 DOI: 10.1038/s41420-023-01417-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Integrins have been the research focus of cell-extracellular matrix adhesion (ECM) and cytokine receptor signal transduction. They are involved in the regulation of bone metabolism of bone precursor cells, mesenchymal stem cells (MSCs), osteoblasts (OBs), osteoclasts (OCs), and osteocytes. Recent studies expanded and updated the role of integrin in bone metabolism, and a large number of novel cytokines were found to activate bone metabolism pathways through interaction with integrin receptors. Integrins act as transducers that mediate the regulation of bone-related cells by mechanical stress, fluid shear stress (FSS), microgravity, hypergravity, extracellular pressure, and a variety of physical factors. Integrins mediate bone metastasis of breast, prostate, and lung cancer by promoting cancer cell adhesion, migration, and survival. Integrin-mediated targeted therapy showed promising prospects in bone metabolic diseases. This review emphasizes the latest research results of integrins in bone metabolism and bone metastasis and provides a vision for treatment strategies.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Lian Wang
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, WA, 6009, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China.
| |
Collapse
|
5
|
Nagasaki K, Chavez M, Nagasaki A, Taylor J, Tan M, Ma M, Ralston E, Thew M, Kim DG, Somerman M, Foster B. The Bone Sialoprotein RGD Domain Modulates and Maintains Periodontal Development. J Dent Res 2022; 101:1238-1247. [PMID: 35686360 PMCID: PMC9403724 DOI: 10.1177/00220345221100794] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Bone sialoprotein (gene: Ibsp; protein: BSP) is a multifunctional extracellular matrix protein present in bone, cementum, and dentin. Accumulating evidence supports BSP as a key regulator of mineralized tissue formation via evolutionarily conserved functional domains, including a C-terminal integrin-binding Arg-Gly-Asp (RGD) domain implicated in extracellular matrix-cell signaling. Ablation of Ibsp in mice (Ibsp-/-) results in impaired bone growth and mineralization and defective osteoclastogenesis, with effects in the craniofacial region including reduced acellular cementum formation, detachment of the periodontal ligament (PDL), alveolar bone hypomineralization, and severe periodontal breakdown. We hypothesized that BSP-RGD plays an important role in cementum and alveolar bone formation and mineralization, as well as periodontal function. This hypothesis was tested by replacing the RGD motif with a nonfunctional Lys-Ala-Glu (KAE) sequence in (IbspKAE/KAE) mice and OCCM.30 murine (IbspKAE) cementoblasts. The RGD domain was not critical for acellular or cellular cementum formation in IbspKAE/KAE mice. However, PDL volume and thickness were increased, and significantly more tartrate-resistant acid phosphatase-positive osteoclasts were found on alveolar bone surfaces of IbspKAE/KAE mice versus wild type mice. PDL organization was disrupted as indicated by picrosirius red stain, second harmonic generation imaging, dynamic mechanical analysis, and decreased asporin proteoglycan localization. In vitro studies implicated RGD functions in cell migration, adhesion, and mineralization, and this was confirmed by an ossicle implant model where cells lacking BSP-RGD showed substantial defects as compared with controls. In total, the BSP-RGD domain is implicated in periodontal development, though the scale and scope of changes indicated by in vitro studies indicate that other factors may partially compensate for and reduce the phenotypic severity of mice lacking BSP-RGD in vivo.
Collapse
Affiliation(s)
- K. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.B. Chavez
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.M. Taylor
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.H. Tan
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M. Ma
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - E. Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.E. Thew
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - D.-G. Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M.J. Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B.L. Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
Chen J, Hua J, Song W. Screen Key Genes Associated with Distraction-Induced Osteogenesis of Stem Cells Using Bioinformatics Methods. Int J Mol Sci 2021; 22:ijms22126505. [PMID: 34204414 PMCID: PMC8235684 DOI: 10.3390/ijms22126505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 01/07/2023] Open
Abstract
Background: Applying mesenchymal stem cells (MSCs), together with the distraction osteogenesis (DO) process, displayed enhanced bone quality and shorter treatment periods. The DO guides the differentiation of MSCs by providing mechanical clues. However, the underlying key genes and pathways are largely unknown. The aim of this study was to screen and identify hub genes involved in distraction-induced osteogenesis of MSCs and potential molecular mechanisms. Material and Methods: The datasets were downloaded from the ArrayExpress database. Three samples of negative control and two samples subjected to 5% cyclic sinusoidal distraction at 0.25 Hz for 6 h were selected for screening differentially expressed genes (DEGs) and then analysed via bioinformatics methods. The Gene Ontology (GO) terms and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment were investigated. The protein–protein interaction (PPI) network was visualised through the Cytoscape software. Gene set enrichment analysis (GSEA) was conducted to verify the enrichment of a self-defined osteogenic gene sets collection and identify osteogenic hub genes. Results: Three hub genes (IL6, MMP2, and EP300) that were highly associated with distraction-induced osteogenesis of MSCs were identified via the Venn diagram. These hub genes could provide a new understanding of distraction-induced osteogenic differentiation of MSCs and serve as potential gene targets for optimising DO via targeted therapies.
Collapse
Affiliation(s)
- Jishizhan Chen
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK;
| | - Jia Hua
- UCL Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Stanmore, London HA7 4AP, UK;
- The Griffin Institute (Northwick Park Institute for Medical Research), Harrow, London HA1 3UJ, UK
- Faculty of Science and Technology, Middlesex University, London NW4 4BT, UK
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK;
- Correspondence:
| |
Collapse
|
7
|
Abstract
BACKGROUND In spite of advances in the treatment of cartilage defects using cell and scaffold-based therapeutic strategies, the long-term outcome is still not satisfying since clinical scores decline years after treatment. Scaffold materials currently used in clinical settings have shown limitations in providing suitable biomechanical properties and an authentic and protective environment for regenerative cells. To tackle this problem, we developed a scaffold material based on decellularised human articular cartilage. METHODS Human articular cartilage matrix was engraved using a CO2 laser and treated for decellularisation and glycosaminoglycan removal. Characterisation of the resulting scaffold was performed via mechanical testing, DNA and GAG quantification and in vitro cultivation with adipose-derived stromal cells (ASC). Cell vitality, adhesion and chondrogenic differentiation were assessed. An ectopic, unloaded mouse model was used for the assessment of the in vivo performance of the scaffold in combination with ASC and human as well as bovine chondrocytes. The novel scaffold was compared to a commercial collagen type I/III scaffold. FINDINGS Crossed line engravings of the matrix allowed for a most regular and ubiquitous distribution of cells and chemical as well as enzymatic matrix treatment was performed to increase cell adhesion. The biomechanical characteristics of this novel scaffold that we term CartiScaff were found to be superior to those of commercially available materials. Neo-tissue was integrated excellently into the scaffold matrix and new collagen fibres were guided by the laser incisions towards a vertical alignment, a typical feature of native cartilage important for nutrition and biomechanics. In an ectopic, unloaded in vivo model, chondrocytes and mesenchymal stromal cells differentiated within the incisions despite the lack of growth factors and load, indicating a strong chondrogenic microenvironment within the scaffold incisions. Cells, most noticeably bone marrow-derived cells, were able to repopulate the empty chondrocyte lacunae inside the scaffold matrix. INTERPRETATION Due to the better load-bearing, its chondrogenic effect and the ability to guide matrix-deposition, CartiScaff is a promising biomaterial to accelerate rehabilitation and to improve long term clinical success of cartilage defect treatment. FUNDING Austrian Research Promotion Agency FFG ("CartiScaff" #842455), Lorenz Böhler Fonds (16/13), City of Vienna Competence Team Project Signaltissue (MA23, #18-08).
Collapse
|
8
|
Repopulation of decellularised articular cartilage by laser-based matrix engraving. EBioMedicine 2021; 64:103196. [PMID: 33483297 PMCID: PMC7910698 DOI: 10.1016/j.ebiom.2020.103196] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/25/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Background In spite of advances in the treatment of cartilage defects using cell and scaffold-based therapeutic strategies, the long-term outcome is still not satisfying since clinical scores decline years after treatment. Scaffold materials currently used in clinical settings have shown limitations in providing suitable biomechanical properties and an authentic and protective environment for regenerative cells. To tackle this problem, we developed a scaffold material based on decellularised human articular cartilage. Methods Human articular cartilage matrix was engraved using a CO2 laser and treated for decellularisation and glycosaminoglycan removal. Characterisation of the resulting scaffold was performed via mechanical testing, DNA and GAG quantification and in vitro cultivation with adipose-derived stromal cells (ASC). Cell vitality, adhesion and chondrogenic differentiation were assessed. An ectopic, unloaded mouse model was used for the assessment of the in vivo performance of the scaffold in combination with ASC and human as well as bovine chondrocytes. The novel scaffold was compared to a commercial collagen type I/III scaffold. Findings Crossed line engravings of the matrix allowed for a most regular and ubiquitous distribution of cells and chemical as well as enzymatic matrix treatment was performed to increase cell adhesion. The biomechanical characteristics of this novel scaffold that we term CartiScaff were found to be superior to those of commercially available materials. Neo-tissue was integrated excellently into the scaffold matrix and new collagen fibres were guided by the laser incisions towards a vertical alignment, a typical feature of native cartilage important for nutrition and biomechanics. In an ectopic, unloaded in vivo model, chondrocytes and mesenchymal stromal cells differentiated within the incisions despite the lack of growth factors and load, indicating a strong chondrogenic microenvironment within the scaffold incisions. Cells, most noticeably bone marrow-derived cells, were able to repopulate the empty chondrocyte lacunae inside the scaffold matrix. Interpretation Due to the better load-bearing, its chondrogenic effect and the ability to guide matrix-deposition, CartiScaff is a promising biomaterial to accelerate rehabilitation and to improve long term clinical success of cartilage defect treatment. Funding Austrian Research Promotion Agency FFG (“CartiScaff” #842455), Lorenz Böhler Fonds (16/13), City of Vienna Competence Team Project Signaltissue (MA23, #18-08)
Collapse
|
9
|
Giannoni P, Muraglia A, Giordano C, Narcisi R, Cancedda R, Quarto R, Chiesa R. Osteogenic Differentiation of Human Mesenchymal Stromal Cells on Surface-Modified Titanium Alloys for Orthopedic and Dental Implants. Int J Artif Organs 2018; 32:811-20. [DOI: 10.1177/039139880903201107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose Surface properties of titanium alloys, used for orthopedic and dental applications, are known to affect implant interactions with host tissues. Osteointegration, bone growth and remodeling in the area surrounding the implants can be implemented by specific biomimetic treatments; these allow the preparation of micro/nanostructured titanium surfaces with a thickened oxide layer, doped with calcium and phosphorus ions. We have challenged these experimental titanium alloys with primary human bone marrow stromal cells to compare the osteogenic differentiation outcomes of the cells once they are seeded onto the modified surfaces, thus simulating a prosthetic device-biological interface of clinical relevance. Methods A specific anodic spark discharge was the biomimetic treatment of choice, providing experimental titanium disks treated with different alkali etching approaches. The disks, checked by electron microscopy and spectroscopy, were subsequently used as substrates for the proliferation and osteogenic differentiation of human cells. Expression of markers of the osteogenic lineage was assessed by means of qualitative and quantitative PCR, by cytochemistry, immunohistochemistry Western blot and matrix metalloprotease activity analyses. Results Metal surfaces were initially less permissive for cell growth. Untreated control substrates were less efficient in sustaining mineralized matrix deposition upon osteogenic induction of the cells. Interestingly, bone sialo protein and matrix metalloprotease 2 levels were enhanced on experimental metals compared to control surfaces, particularly for titanium oxide coatings etched with KOH. Discussion As a whole, the KOH-modification of titanium surfaces seems to allow the best osteogenic differentiation of human mesenchymal stromal cells, representing a possible plus for future clinical prosthetic applications.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | | | - Carmen Giordano
- Giulio Natta Department of Industrial Chemistry and Chemical Engineering, Milan Polytechnic University, Milan - Italy
| | - Roberto Narcisi
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | - Ranieri Cancedda
- Laboratory of Regenerative Medicine, National Cancer Research Institute, University of Genoa - Italy
| | - Rodolfo Quarto
- Stem Cell Laboratory, Advanced Biotechnology Center, Genoa - Italy
| | - Roberto Chiesa
- Giulio Natta Department of Industrial Chemistry and Chemical Engineering, Milan Polytechnic University, Milan - Italy
| |
Collapse
|
10
|
Zepp M, Kovacheva M, Altankhuyag M, Westphal G, Berger I, Gather KS, Hilbig H, Neuhaus J, Hänsch GM, Armbruster FP, Berger MR. IDK1 is a rat monoclonal antibody against hypoglycosylated bone sialoprotein with application as biomarker and therapeutic agent in breast cancer skeletal metastasis. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2017; 4:55-68. [PMID: 29416877 PMCID: PMC5783975 DOI: 10.1002/cjp2.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 12/24/2022]
Abstract
Changes in glycosylation are salient features of cancer cells. Here, we report on the diagnostic and therapeutic properties of IDK1, an antibody against tumour associated, hypoglycosylated bone sialoprotein (hypo‐BSP). The affinity of the rat monoclonal antibody IDK1 for hypo‐BSP, as determined by microscale thermophoresis, was three orders of magnitude higher than for mature BSP, whereas the mouse monoclonal antibody used had similar affinity for both BSP forms. IDK1 showed no activity against the proliferation or migration of normal or cancer cells growing in vitro. In vivo, however, IDK1 caused dose‐dependent regression of soft tissue and skeletal lesions in nude rats harbouring human MDA‐MB‐231 cells. At optimal dose, 80% of the treated rats showed complete remission of all tumour lesions. Analysis of BSP expression in vitro by fluorescence‐activated cell sorting (FACS) and immunocytochemistry showed basal levels of this protein, which were visible only in a fraction of these cells. Cells of the metastatic cell lines MDA‐MB‐231 and PC‐3 were more often positive for hypo‐BSP. In addition, there was co‐expression of both forms in some cells, but almost no co‐localization; rather, hypo‐BSP was present in the nucleus, and mature BSP was detected extra‐cellularly. Normal osteoblasts and osteoclasts were negative for hypo‐BSP. Breast cancer tissue, however, showed strong expression of mature BSP, which was present intra‐cellularly as well as in vesicles outside cells. Hypo‐BSP was present mainly in lesions from skeletal sites, thus explaining the antineoplastic activity of IDK1, which was high in lesions growing in the vicinity of the skeleton but low in lesions growing subcutaneously. Finally, hypo‐BSP was detected in specimens from breast cancer patients, with a significantly greater intensity in skeletal metastases as compared to the respective primary cancers. In conclusion, IDK‐1 is an antibody with diagnostic and therapeutic applications in skeletal metastases of breast cancer.
Collapse
Affiliation(s)
- Michael Zepp
- Toxicology and Chemotherapy UnitGerman Cancer Research CenterHeidelbergGermany
| | - Marineta Kovacheva
- Toxicology and Chemotherapy UnitGerman Cancer Research CenterHeidelbergGermany
| | | | | | - Irina Berger
- Institute of Pathology, Klinikum KasselKasselGermany
| | | | | | | | - Gertrud M Hänsch
- Institute of ImmunologyUniversity Hospital HeidelbergHeidelbergGermany
| | | | - Martin R Berger
- Toxicology and Chemotherapy UnitGerman Cancer Research CenterHeidelbergGermany
| |
Collapse
|
11
|
Paiva KBS, Granjeiro JM. Matrix Metalloproteinases in Bone Resorption, Remodeling, and Repair. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:203-303. [PMID: 28662823 DOI: 10.1016/bs.pmbts.2017.05.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are the major protease family responsible for the cleavage of the matrisome (global composition of the extracellular matrix (ECM) proteome) and proteins unrelated to the ECM, generating bioactive molecules. These proteins drive ECM remodeling, in association with tissue-specific and cell-anchored inhibitors (TIMPs and RECK, respectively). In the bone, the ECM mediates cell adhesion, mechanotransduction, nucleation of mineralization, and the immobilization of growth factors to protect them from damage or degradation. Since the first description of an MMP in bone tissue, many other MMPs have been identified, as well as their inhibitors. Numerous functions have been assigned to these proteins, including osteoblast/osteocyte differentiation, bone formation, solubilization of the osteoid during bone resorption, osteoclast recruitment and migration, and as a coupling factor in bone remodeling under physiological conditions. In turn, a number of pathologies, associated with imbalanced bone remodeling, arise mainly from MMP overexpression and abnormalities of the ECM, leading to bone osteolysis or bone formation. In this review, we will discuss the functions of MMPs and their inhibitors in bone cells, during bone remodeling, pathological bone resorption (osteoporosis and bone metastasis), bone repair/regeneration, and emergent roles in bone bioengineering.
Collapse
Affiliation(s)
- Katiucia B S Paiva
- Laboratory of Extracellular Matrix Biology and Cellular Interaction (LabMec), Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| | - José M Granjeiro
- National Institute of Metrology, Quality and Technology (InMetro), Bioengineering Laboratory, Duque de Caxias, RJ, Brazil; Fluminense Federal University, Dental School, Niterói, RJ, Brazil
| |
Collapse
|
12
|
Kusuyama J, Bandow K, Ohnishi T, Hisadome M, Shima K, Semba I, Matsuguchi T. Osteopontin inhibits osteoblast responsiveness through the down-regulation of focal adhesion kinase mediated by the induction of low-molecular weight protein tyrosine phosphatase. Mol Biol Cell 2017; 28:1326-1336. [PMID: 28331074 PMCID: PMC5426847 DOI: 10.1091/mbc.e16-10-0716] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/23/2022] Open
Abstract
Osteopontin (OPN), a major marker of osteogenic differentiation, suppresses osteoblast responses to mechanical stress and cytokines, including HGF and PDGF. These OPN-induced effects are mediated through focal adhesion kinase inactivation by the induction of low–molecular weight protein tyrosine phosphatase. Osteopontin (OPN) is an osteogenic marker protein. Osteoblast functions are affected by inflammatory cytokines and pathological conditions. OPN is highly expressed in bone lesions such as those in rheumatoid arthritis. However, local regulatory effects of OPN on osteoblasts remain ambiguous. Here we examined how OPN influences osteoblast responses to mechanical stress and growth factors. Expression of NO synthase 1 (Nos1) and Nos2 was increased by low-intensity pulsed ultrasound (LIPUS) in MC3T3-E1 cells and primary osteoblasts. The increase of Nos1/2 expression was abrogated by both exogenous OPN overexpression and recombinant OPN treatment, whereas it was promoted by OPN-specific siRNA and OPN antibody. Moreover, LIPUS-induced phosphorylation of focal adhesion kinase (FAK), a crucial regulator of mechanoresponses, was down-regulated by OPN treatments. OPN also attenuated hepatocyte growth factor–induced vitamin D receptor (Vdr) expression and platelet-derived growth factor–induced cell mobility through the repression of FAK activity. Of note, the expression of low–molecular weight protein tyrosine phosphatase (LMW-PTP), a FAK phosphatase, was increased in both OPN-treated and differentiated osteoblasts. CD44 was a specific OPN receptor for LWW-PTP induction. Consistently, the suppressive influence of OPN on osteoblast responsiveness was abrogated by LMW-PTP knockdown. Taken together, these results reveal novel functions of OPN in osteoblast physiology.
Collapse
Affiliation(s)
- Joji Kusuyama
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Kenjiro Bandow
- Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakato 350-0283, Japan
| | - Tomokazu Ohnishi
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Mitsuhiro Hisadome
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan.,Department of Dermatology, Field of Advanced Therapeutics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Kaori Shima
- Department of Oral Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Ichiro Semba
- Department of Oral Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Tetsuya Matsuguchi
- Department of Oral Biochemistry, Field of Developmental Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| |
Collapse
|
13
|
Riehl BD, Lee JS, Ha L, Kwon IK, Lim JY. Flowtaxis of osteoblast migration under fluid shear and the effect of RhoA kinase silencing. PLoS One 2017; 12:e0171857. [PMID: 28199362 PMCID: PMC5310897 DOI: 10.1371/journal.pone.0171857] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023] Open
Abstract
Despite the important role of mechanical signals in bone remodeling, relatively little is known about how fluid shear affects osteoblastic cell migration behavior. Here we demonstrated that MC3T3-E1 osteoblast migration could be activated by physiologically-relevant levels of fluid shear in a shear stress-dependent manner. Interestingly, shear-sensitive osteoblast migration behavior was prominent only during the initial period after the onset of the steady flow (for about 30 min), exhibiting shear stress-dependent migration speed, displacement, arrest coefficient, and motility coefficient. For example, cell speed at 1 min was 0.28, 0.47, 0.51, and 0.84 μm min-1 for static, 2, 15, and 25 dyne cm-2 shear stress, respectively. Arrest coefficient (measuring how often cells are paused during migration) assessed for the first 30 min was 0.40, 0.26, 0.24, and 0.12 respectively for static, 2, 15, and 25 dyne cm-2. After this initial period, osteoblasts under steady flow showed decreased migration capacity and diminished shear stress dependency. Molecular interference of RhoA kinase (ROCK), a regulator of cytoskeletal tension signaling, was found to increase the shear-sensitive window beyond the initial period. Cells with ROCK-shRNA had increased migration in the flow direction and continued shear sensitivity, resulting in greater root mean square displacement at the end of 120 min of measurement. It is notable that the transient osteoblast migration behavior was in sharp contrast to mesenchymal stem cells that exhibited sustained shear sensitivity (as we recently reported, J. R. Soc. Interface. 2015; 12:20141351). The study of fluid shear as a driving force for cell migration, i.e., "flowtaxis", and investigation of molecular mechanosensors governing such behavior (e.g., ROCK as tested in this study) may provide new and improved insights into the fundamental understanding of cell migration-based homeostasis.
Collapse
Affiliation(s)
- Brandon D. Riehl
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Jeong Soon Lee
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Ligyeom Ha
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Il Keun Kwon
- The Graduate School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- The Graduate School of Dentistry, Kyung Hee University, Seoul, Korea
- * E-mail:
| |
Collapse
|
14
|
Yang Z, Wu B, Jia S, Zhao Y, Hou R, Liu X, Wang X, Chen L, Yang X, Lei D, Wang L. The mechanically activated p38/MMP-2 signaling pathway promotes bone marrow mesenchymal stem cell migration in rats. Arch Oral Biol 2017; 76:55-60. [PMID: 28126687 DOI: 10.1016/j.archoralbio.2017.01.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 11/01/2016] [Accepted: 01/18/2017] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The aim of the present study was to investigate the effect of static strain on bone marrow mesenchymal stem cell (BMMSC) migration and whether the p38/matrix metalloproteinase-2 (MMP-2) axis plays a role in induction of BMMSC migration under mechanical strain. DESIGN Both in vivo and in vitro investigations were performed. Twelve adult male Sprague-Dawley rats were randomly divided into 2 groups (n=6 per group). Rats in the experimental group underwent right mandibular distraction osteogenesis, whereas rats in the control group were subjected to osteotomy in the mandible without distraction. Immunohistochemistry and immunofluorescence were performed to evaluate phospho-p38 (p-p38) and Nestin expression. BMMSCs were isolated from rat mandibles. BMMSCs in the experimental group were subjected to static mechanical strain for 2h, whereas those in the control group underwent no strain. The biological roles of static strain and the p38/MMP-2 axis in BMMSC migration were evaluated by Transwell assays and western blotting by inhibiting p38 phosphorylation. RESULTS There were significantly more Nestin+ cells in the bone calluses of the experimental group than in those of the control group. In addition, Nestin+/p-p38+ cell numbers were significantly higher in the experimental group than in the control group, indicating that static strain activated p38 signaling in BMMSCs in vivo. In accordance with in vivo results, static strain in vitro stimulated phosphorylation of p38 in BMMSCs. Furthermore, expression of MMP-2 was elevated in BMMSCs under static strain compared with the control, and strain-induced MMP-2 expression was abolished by inhibition of p38 phosphorylation in BMMSCs. Moreover, Transwell assay results showed that static strain promoted BMMSC migration, which was abolished by inhibition of p38 phosphorylation. CONCLUSIONS The present study demonstrated that static strain can promote the migration ability of BMMSCs via p38/MMP-2 signaling. To the best of our knowledge, this study is the first report demonstrating that the p38/MMP-2 axis governs BMMSC migration under static mechanical strain.
Collapse
Affiliation(s)
- Zihui Yang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Baolei Wu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Sen Jia
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yinghua Zhao
- Department of Prosthodontics, Stomatology Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Hou
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xiaochang Liu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xinge Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Litong Chen
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Delin Lei
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| | - Lei Wang
- Department of Oral & Maxillofacial-Head and Neck Oncology, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China; State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
15
|
Özdal-Kurt F, Tuğlu I, Vatansever HS, Tong S, Şen BH, Deliloğlu-Gürhan SI. The effect of different implant biomaterials on the behavior of canine bone marrow stromal cells during their differentiation into osteoblasts. Biotech Histochem 2016; 91:412-22. [DOI: 10.1080/10520295.2016.1183819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
16
|
Bouleftour W, Juignet L, Bouet G, Granito RN, Vanden-Bossche A, Laroche N, Aubin JE, Lafage-Proust MH, Vico L, Malaval L. The role of the SIBLING, Bone Sialoprotein in skeletal biology - Contribution of mouse experimental genetics. Matrix Biol 2016; 52-54:60-77. [PMID: 26763578 DOI: 10.1016/j.matbio.2015.12.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/23/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
Bone Sialoprotein (BSP) is a member of the "Small Integrin-Binding Ligand N-linked Glycoproteins" (SIBLING) extracellular matrix protein family of mineralized tissues. BSP has been less studied than other SIBLING proteins such as Osteopontin (OPN), which is coexpressed with it in several skeletal cell types. Here we review the contribution of genetically engineered mice (BSP gene knockout and overexpression) to the understanding of the role of BSP in the bone organ. The studies made so far highlight the role of BSP in skeletal mineralization, as well as its importance for proper osteoblast and osteoclast differentiation and activity, most prominently in primary/repair bone. The absence of BSP also affects the local environment of the bone tissue, in particular hematopoiesis and vascularization. Interestingly, lack of BSP induces an overexpression of OPN, and the cognate protein could be responsible for some aspects of the BSP gene knockout skeletal phenotype, while replacing BSP for some of its functions. Such interplay between the partly overlapping functions of SIBLING proteins, as well as the network of cross-regulations in which they are involved should now be the focus of further work.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laura Juignet
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Guenaelle Bouet
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, UK
| | | | - Arnaud Vanden-Bossche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Norbert Laroche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laurence Vico
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Luc Malaval
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France.
| |
Collapse
|
17
|
Skodje A, Idris SBM, Sun Y, Bartaula S, Mustafa K, Finne-Wistrand A, Wikesjö UME, Leknes KN. Biodegradable polymer scaffolds loaded with low-dose BMP-2 stimulate periodontal ligament cell differentiation. J Biomed Mater Res A 2014; 103:1991-8. [DOI: 10.1002/jbm.a.35334] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 08/22/2014] [Accepted: 09/10/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Anders Skodje
- Faculty of Medicine and Dentistry; Department of Clinical Dentistry - Periodontics; University of Bergen; Bergen Norway
| | - Shaza Bushra M. Idris
- Faculty of Medicine and Dentistry; Department of Clinical Dentistry - Center for Clinical Dental Research; University of Bergen; Bergen Norway
| | - Yang Sun
- Department of Fibre and Polymer Technology; KTH Royal Institute of Technology; Stockholm Sweden
| | - Sushma Bartaula
- Faculty of Medicine and Dentistry; Department of Clinical Dentistry - Center for Clinical Dental Research; University of Bergen; Bergen Norway
| | - Kamal Mustafa
- Faculty of Medicine and Dentistry; Department of Clinical Dentistry - Center for Clinical Dental Research; University of Bergen; Bergen Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology; KTH Royal Institute of Technology; Stockholm Sweden
| | - Ulf M. E. Wikesjö
- Laboratory for Applied Periodontal & Craniofacial Regeneration; Georgia Regents University; Augusta GA, USA
| | - Knut N. Leknes
- Faculty of Medicine and Dentistry; Department of Clinical Dentistry - Periodontics; University of Bergen; Bergen Norway
| |
Collapse
|
18
|
Docheva D, Popov C, Alberton P, Aszodi A. Integrin signaling in skeletal development and function. ACTA ACUST UNITED AC 2014; 102:13-36. [DOI: 10.1002/bdrc.21059] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/14/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Denitsa Docheva
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Cvetan Popov
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| |
Collapse
|
19
|
A Comparative Evaluation between New Ternary Zirconium Alloys as Alternative Metals for Orthopedic and Dental Prosthetic Devices. Int J Artif Organs 2014; 37:149-64. [DOI: 10.5301/ijao.5000287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 11/20/2022]
Abstract
Purpose We assessed in vitro the corrosion behavior and biocompatibility of four Zr-based alloys (Zr97.5 Nb1.5 VM1.0 ; VM, valve metal: Ti, Mo, W, Ta; at%) to be used as implant materials, comparing the results with grade-2 titanium, a biocompatible metal standard. Methods Corrosion resistance was investigated by open circuit potential and electrochemical impedance spectroscopy measurements as a function of exposure time to an artificial physiological environment (Ringer's solution). Human bone marrow stromal cells were used to evaluate biocompatibility of the alloys and their influence on growth kinetics and cell osteogenic differentiation through histochemical and gene expression analyses. Results Open circuit potential values indicated that Zr-based alloys and grade-2 Ti undergo spontaneous passivation in the simulated aggressive environment. High impedance values for all samples demonstrated improved corrosion resistance of the oxide film, with the best protection characteristics displayed by Zr97.5 Nb1.5 Ta1.0. Cells seeded on all surfaces showed the same growth kinetics, although matrix mineralization and alkaline phosphatase activity were maximal on Zr97.5 Nb1.5 Mo1.0 and Zr97.5 Nb1.5 Ta1.0. Markers of ongoing proliferation, however, such as podocalyxin and CD49f, were still overexpressed on Zr97.5 Nb1.5 Mo1.0 even upon osteoinduction. No relevant effects were noted for the CD146-expressing population of bone progenitors. Nonetheless, the presence of a more differentiated cell population on Zr97.5 Nb1.5 Ta1.0 samples was inferable by comparing mineralization data and transcript levels of osteogenic markers (osteocalcin, osteopontin, bone sialoprotein, and RUNX2). Conclusions The combination of passivation, corrosion resistance and satisfactory biotolerance to bone progenitors make the Zr-based alloys promising implant materials. Among those we tested, Zr97.5 Nb1.5 Ta1.0 seems to be the most appealing.
Collapse
|
20
|
Chatakun P, Núñez-Toldrà R, Díaz López EJ, Gil-Recio C, Martínez-Sarrà E, Hernández-Alfaro F, Ferrés-Padró E, Giner-Tarrida L, Atari M. The effect of five proteins on stem cells used for osteoblast differentiation and proliferation: a current review of the literature. Cell Mol Life Sci 2014; 71:113-42. [PMID: 23568025 PMCID: PMC11113514 DOI: 10.1007/s00018-013-1326-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/13/2013] [Accepted: 03/14/2013] [Indexed: 01/04/2023]
Abstract
Bone-tissue engineering is a therapeutic target in the field of dental implant and orthopedic surgery. It is therefore essential to find a microenvironment that enhances the growth and differentiation of osteoblasts both from mesenchymal stem cells (MSCs) and those derived from dental pulp. The aim of this review is to determine the relationship among the proteins fibronectin (FN), osteopontin (OPN), tenascin (TN), bone sialoprotein (BSP), and bone morphogenetic protein (BMP2) and their ability to coat different types of biomaterials and surfaces to enhance osteoblast differentiation. Pre-treatment of biomaterials with FN during the initial phase of osteogenic differentiation on all types of surfaces, including slotted titanium and polymers, provides an ideal microenvironment that enhances adhesion, morphology, and proliferation of pluripotent and multipotent cells. Likewise, in the second stage of differentiation, surface coating with BMP2 decreases the diameter and the pore size of the scaffold, causing better adhesion and reduced proliferation of BMP-MSCs. Coating oligomerization surfaces with OPN and BSP promotes cell adhesion, but it is clear that the polymeric coating material BSP alone is insufficient to induce priming of MSCs and functional osteoblastic differentiation in vivo. Finally, TN is involved in mineralization and can accelerate new bone formation in a multicellular environment but has no effect on the initial stage of osteogenesis.
Collapse
Affiliation(s)
- P. Chatakun
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Police General Hospital, Bangkok, Thailand
| | - R. Núñez-Toldrà
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Chair of Regenerative Implantology MIS-UIC, Universitat Internacional de Catalunya, Barcelona, Spain
| | - E. J. Díaz López
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - C. Gil-Recio
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Chair of Regenerative Implantology MIS-UIC, Universitat Internacional de Catalunya, Barcelona, Spain
| | - E. Martínez-Sarrà
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Chair of Regenerative Implantology MIS-UIC, Universitat Internacional de Catalunya, Barcelona, Spain
| | - F. Hernández-Alfaro
- Surgery and Oral Implantology Department, College of Dentistry, Universitat Internacional de Catalunya, Barcelona, Spain
| | - E. Ferrés-Padró
- Surgery and Oral Implantology Department, College of Dentistry, Universitat Internacional de Catalunya, Barcelona, Spain
- Oral and Maxillofacial Surgery Department, Fundacio Hospital de Nens de Barcelona, Barcelona, Spain
| | - L. Giner-Tarrida
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Chair of Regenerative Implantology MIS-UIC, Universitat Internacional de Catalunya, Barcelona, Spain
| | - M. Atari
- Laboratory for Regenerative Medicine, College of Dentistry, Universitat Internacional de Catalunya, C/Josep Trueta s/n, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Chair of Regenerative Implantology MIS-UIC, Universitat Internacional de Catalunya, Barcelona, Spain
- Surgery and Oral Implantology Department, College of Dentistry, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
21
|
A systems-pharmacology analysis of herbal medicines used in health improvement treatment: predicting potential new drugs and targets. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:938764. [PMID: 24369484 PMCID: PMC3863530 DOI: 10.1155/2013/938764] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 09/23/2013] [Accepted: 10/04/2013] [Indexed: 11/24/2022]
Abstract
For thousands of years, tonic herbs have been successfully used all around the world to improve health, energy, and vitality. However, their underlying mechanisms of action in molecular/systems levels are still a mystery. In this work, two sets of tonic herbs, so called Qi-enriching herbs (QEH) and Blood-tonifying herbs (BTH) in TCM, were selected to elucidate why they can restore proper balance and harmony inside body, organ and energy system. Firstly, a pattern recognition model based on artificial neural network and discriminant analysis for assessing the molecular difference between QEH and BTH was developed. It is indicated that QEH compounds have high lipophilicity while BTH compounds possess high chemical reactivity. Secondly, a systematic investigation integrating ADME (absorption, distribution, metabolism, and excretion) prediction, target fishing and network analysis was performed and validated on these herbs to obtain the compound-target associations for reconstructing the biologically-meaningful networks. The results suggest QEH enhance physical strength, immune system and normal well-being, acting as adjuvant therapy for chronic disorders while BTH stimulate hematopoiesis function in body. As an emerging approach, the systems pharmacology model might facilitate to understand the mechanisms of action of the tonic herbs, which brings about new development for complementary and alternative medicine.
Collapse
|
22
|
Konttinen YT, Kaivosoja E, Stegaev V, Wagner HD, Levón J, Tiainen VM, Mackiewicz Z. Extracellular Matrix and Tissue Regeneration. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
23
|
Müller WEG, Wang X, Grebenjuk V, Diehl-Seifert B, Steffen R, Schloßmacher U, Trautwein A, Neumann S, Schröder HC. Silica as a morphogenetically active inorganic polymer. Biomater Sci 2013; 1:669-678. [DOI: 10.1039/c3bm00001j] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Biogenic Inorganic Polysilicates (Biosilica): Formation and Biomedical Applications. BIOMEDICAL INORGANIC POLYMERS 2013; 54:197-234. [DOI: 10.1007/978-3-642-41004-8_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Mosig RA, Martignetti JA. Loss of MMP-2 in murine osteoblasts upregulates osteopontin and bone sialoprotein expression in a circuit regulating bone homeostasis. Dis Model Mech 2012; 6:397-403. [PMID: 22917927 PMCID: PMC3597021 DOI: 10.1242/dmm.007914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multicentric osteolysis with arthropathy (MOA; MIM 605156) is an inherited osteolyses and arthritis syndrome resulting from loss of matrix metalloproteinase 2 (MMP-2). We recently demonstrated that Mmp2–/– mice represent a unique model for the study of the human disease, sharing many features of the human syndrome including skeletal dysplasia and defects in osteoblast behavior. We therefore sought to explore the secondary molecular effects of MMP-2 loss, which coexist with the underlying skeletal and osteoblast phenotypes. We used quantitative real-time RT-PCR (qRT-PCR) to measure osteoblast-related gene expression through ex vivo osteoblast differentiation of bone marrow stromal cells (BMSC) from Mmp2−/− and Mmp2+/+ mice. We used western blot to measure osteopontin (OPN) serum levels and immunohistochemical staining to examine bone expression. MMP-2 expression was inhibited in SaOS2 cells using siRNA, and decreased MMP-2 expression at both RNA and protein levels was confirmed by qRT-PCR and western blot, respectively. Mmp2−/− BMSC induced to differentiate into osteoblasts were shown to significantly upregulate OPN and bone sialoprotein (BSP) expression levels compared with controls. Transcriptional upregulation was maintained in vivo, as demonstrated by increased levels of OPN in serum and bone in Mmp2−/− mice. These effects are generalizable because siRNA-mediated inhibition in cultured cells also upregulated OPN and BSP. OPN and BSP are known to affect MMP-2 expression and activity but have not previously been shown to be regulated by MMP-2. Identification of this newly defined circuitry provides insight into the potential molecular landscape underlying the MOA phenotype and highlights a pathway that might play a role in normal bone homeostasis.
Collapse
Affiliation(s)
- Rebecca A Mosig
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
26
|
Suzuki S, Haruyama N, Nishimura F, Kulkarni AB. Dentin sialophosphoprotein and dentin matrix protein-1: Two highly phosphorylated proteins in mineralized tissues. Arch Oral Biol 2012; 57:1165-75. [PMID: 22534175 DOI: 10.1016/j.archoralbio.2012.03.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/23/2012] [Accepted: 03/20/2012] [Indexed: 12/15/2022]
Abstract
Dentin sialophosphoprotein (DSPP) and dentin matrix protein-1 (DMP-1) are highly phosphorylated proteins that belong to the family of small integrin-binding ligand N-linked glycoproteins (SIBLINGs), and are essential for proper development of hard tissues such as teeth and bones. In order to understand how they contribute to tissue organization, DSPP and DMP-1 have been analyzed for over a decade using both in vivo and in vitro techniques. Among the five SIBLINGs, the DSPP and DMP-1 genes are located next to each other and their gene and protein structures are most similar. In this review we examine the phenotypes of the genetically engineered mouse models of DSPP and DMP-1 and also introduce complementary in vitro studies into the molecular mechanisms underlying these phenotypes. DSPP affects the mineralization of dentin more profoundly than DMP-1. In contrast, DMP-1 significantly affects bone mineralization and importantly controls serum phosphate levels by regulating serum FGF-23 levels, whereas DSPP does not show any systemic effects. DMP-1 activates integrin signalling and is endocytosed into the cytoplasm whereupon it is translocated to the nucleus. In contrast, DSPP only activates integrin-dependent signalling. Thus it is now clear that both DSPP and DMP-1 contribute to hard tissue mineralization and the tissues affected by each are different presumably as a result of their different expression levels. In fact, in comparison with DMP-1, the functional analysis of cell signalling by DSPP remains relatively unexplored.
Collapse
Affiliation(s)
- Shigeki Suzuki
- Department of Dental Science for Health Promotion, Division of Cervico-Gnathostomatology, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan.
| | | | | | | |
Collapse
|
27
|
Yamamoto N, Maeda H, Tomokiyo A, Fujii S, Wada N, Monnouchi S, Kono K, Koori K, Teramatsu Y, Akamine A. Expression and effects of glial cell line-derived neurotrophic factor on periodontal ligament cells. J Clin Periodontol 2012; 39:556-64. [PMID: 22512503 DOI: 10.1111/j.1600-051x.2012.01881.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2012] [Indexed: 12/15/2022]
Abstract
AIM To investigate Glial cell line-derived neurotrophic factor (GDNF) expression in normal and wounded rat periodontal ligament (PDL) and the effects of GDNF on human PDL cells (HPDLCs) migration and extracellular matrix expression in HPDLCs. MATERIAL AND METHODS The expression of GDNF and GDNF receptors was examined by immunocyto/histochemical analyses. Gene expression in HPDLCs treated with GDNF, interleukin-1 beta (IL-1β), or tumour necrosis factor-alpha (TNF-α) was quantified by quantitative RT-PCR (qRT-PCR). In addition, we examined the migratory effect of GDNF on HPDLCs. RESULTS GDNF was expressed in normal rat PDL and cultured HPDLCs. HPDLCs also expressed GDNF receptors. In wounded rat PDL, GDNF expression was up-regulated. QRT-PCR analysis revealed that IL-1β and TNF-α significantly increased the expression of GDNF in HPDLCs. Furthermore, GDNF induced migration of HPDLCs, which was blocked by pre-treatment with the peptide including Arg-Gly-Asp (RGD) sequence, or neutralizing antibodies against integrin αVβ3 or GDNF. Also, GDNF up-regulated expression of bone sialoprotein (BSP) and fibronectin in HPDLCs. CONCLUSIONS GDNF expression is increased in rat wounded PDL tissue and HPDLCs treated with pro-inflammatory cytokines. GDNF enhances the expression of BSP and fibronectin, and migration in an RGD-dependent manner via the integrin αVβ3. These findings suggest that GDNF may contribute to wound healing in PDL tissue.
Collapse
Affiliation(s)
- Naohide Yamamoto
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Higashi-ku, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Barthelemi S, Robinet J, Garnotel R, Antonicelli F, Schittly E, Hornebeck W, Lorimier S. Mechanical forces-induced human osteoblasts differentiation involves MMP-2/MMP-13/MT1-MMP proteolytic cascade. J Cell Biochem 2012; 113:760-72. [DOI: 10.1002/jcb.23401] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
29
|
|
30
|
Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival and engraftment of bone marrow stromal cells in an ectopic model of bone regeneration. Tissue Eng Part A 2010; 16:489-99. [PMID: 19712045 DOI: 10.1089/ten.tea.2009.0041] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In tissue-engineered applications bone marrow stromal cells (BMSCs) are combined with scaffolds to target bone regeneration; animal models have been devised and the cells' long-term engraftment has been widely studied. However, in regenerated bone, the cell number is severely reduced with respect to the initially seeded BMSCs. This reflects the natural low cellularity of bone but underlines the selectivity of the differentiation processes. In this respect, we evaluated the short-term survival of BMSCs, transduced with the luciferase gene, after implantation of cell-seeded scaffolds in a nude mouse model. Cell proliferation/survival was assessed by bioluminescence imaging: light production was decreased by 30% on the first day, reaching a 50% loss within 48 h. Less than 5% of the initial signal remained after 2 months in vivo. Apoptotic BMSCs were detected within the first 2 days of implantation. Interestingly, the initial frequency of clonogenic progenitors matched the percentage of in vivo surviving cells. Cytokines and inflammation may contribute to the apoptotic onset at the implant milieu. However, preculturing cells with tumor necrosis factor alpha enhanced survival, allowing detection of 8.1% of the seeded BMSCs 2 months after implantation. Thus culturing conditions may reduce the apoptotic overload of seeded osteoprogenitors, strengthening the constructs' osteogenic potential.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Advanced Biotechnology Center, Genova, Italy.
| | | | | | | | | | | |
Collapse
|
31
|
Tsagaraki I, Tsilibary EC, Tzinia AK. TIMP-1 interaction with αvβ3 integrin confers resistance to human osteosarcoma cell line MG-63 against TNF-α-induced apoptosis. Cell Tissue Res 2010; 342:87-96. [PMID: 20798956 DOI: 10.1007/s00441-010-1025-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 07/21/2010] [Indexed: 10/19/2022]
Abstract
Tumor necrosis factor-α (TNF-α) is a pleiotropic cytokine affecting diverse cellular responses. TNF-α is cytotoxic in many systems, but it can also act as an anti-apoptotic signal to promote cell survival pathways activated through integrins and extracellular matrix components. This is particularly evident in cancer cells. To unravel the basis of resistance to TNF-α-induced apoptosis, human osteosarcoma MG-63 cell line was used. Our data showed that resistance to apoptosis was accompanied by high levels of TIMP-1 expression in part mediated by NF-κB activation, whereas under apoptotic conditions, in the presence of cycloheximide (CHX), TIMP-1 and αvβ3 integrin protein levels were significantly reduced. Silencing TIMP-1 using siRNA led to increased apoptosis following treatment with TNF-α, whereas exogenously-added recombinant TIMP-1 reduced the extent of apoptosis. Immunoprecipitation and confocal microscopy experiments demonstrated that TIMP-1 interacted with αvβ3 integrins. The biological role of this interaction was revealed by the use of echistatin, an antagonist of αvβ3 integrin. In the presence of echistatin, decreased protection against apoptosis by recombinant TIMP-1 was observed.
Collapse
Affiliation(s)
- Ioanna Tsagaraki
- Institute of Biology, National Center for Scientific Research Demokritos, 15310 Agia Paraskevi, Athens, Greece
| | | | | |
Collapse
|
32
|
Dissection of genetic factors modulating fetal growth in cattle indicates a substantial role of the non-SMC condensin I complex, subunit G (NCAPG) gene. Genetics 2009; 183:951-64. [PMID: 19720859 DOI: 10.1534/genetics.109.106476] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The increasing evidence of fetal developmental effects on postnatal life, the still unknown fetal growth mechanisms impairing offspring generated by somatic nuclear transfer techniques, and the impact on stillbirth and dystocia in conventional reproduction have generated increasing attention toward mammalian fetal growth. We identified a highly significant quantitative trait locus (QTL) affecting fetal growth on bovine chromosome 6 in a specific resource population, which was set up by consistent use of embryo transfer and foster mothers and, thus, enabled dissection of fetal-specific genetic components of fetal growth. Merging our data with results from other cattle populations differing in historical and geographical origin and with comparative data from human whole-genome association mapping suggests that a nonsynonymous polymorphism in the non-SMC condensin I complex, subunit G (NCAPG) gene, NCAPG c.1326T>G, is the potential cause of the identified QTL resulting in divergent bovine fetal growth. NCAPG gene expression data in fetal placentomes with different NCAPG c.1326T>G genotypes, which are in line with recent results about differential NCAPG expression in placentomes from studies on assisted reproduction techniques, indicate that the NCAPG locus may give valuable information on the specific mechanisms regulating fetal growth in mammals.
Collapse
|
33
|
Manduca P, Castagnino A, Lombardini D, Marchisio S, Soldano S, Ulivi V, Zanotti S, Garbi C, Ferrari N, Palmieri D. Role of MT1-MMP in the osteogenic differentiation. Bone 2009; 44:251-65. [PMID: 19027888 DOI: 10.1016/j.bone.2008.10.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 09/27/2008] [Accepted: 10/16/2008] [Indexed: 11/16/2022]
Abstract
Metalloproteinase MT1-MMP is induced and Pro-MMP-2 up modulated early in rat preosteoblasts (ROB) set to differentiate. We here show that the induction of MMPs, accompanied by activation of Pro-MMP-2, occurs by 6 h of adhesion on endogenous extracellular matrix (ECM), Fibronectin (FN) and Collagen type I (CI). These events do not occur after adhesion on Collagen III (CIII), Vitronectin (VN) or BSA. Within the first hour on inducing substrata or plastic, FAK is unchanged and ERK(1,2), is activated, but this activation is not sufficient for MT1-MMP induction. The function of p38 MAPK and PTKs is not required for the induction by substrata of MMPs. Six hours after plating preosteoblasts on MMP-inducing substrata, complexes of beta1 integrin with MT1-MMP are formed, that contain integrin dimers specifically engaged by the substratum, alpha4 and alpha5 chains for cells plated on FN, and alpha2 chain for cells plated on CI and ECM. Induction of MT1-MMP and its expression during osteogenesis pleiotropically regulate alkaline phosphatase (AP) expression. During differentiation, variant clones derived from preosteoblasts and MMPs-over-expressing osteoblasts show high MT1-MMP level associated with high AP level both persisting in time, while inhibition of MMPs is accompanied by inhibition of AP. Up or down modulation of AP, transcriptionally or by inhibition of the enzyme activity, has no effect on level or timing of expression of MT1-MMP and Pro-MMP-2. The persistence in expression of MT1-MMP during differentiation, and the associated persistence in expression of AP, as well as their inhibition, both impair the formation of nodules and mineral deposition. A transient pattern of expression of MT1-MMP is required for the establishment of nodules, and MT1-MMP decrease is permissive for nodule mineralization. The expression of AP is required for nodule formation and its level modulates the mineralization. MT1-MMP has multiple functions and is implicated in multiple steps of the differentiation process, acting to regulate homeostasis of the osteogenic differentiation.
Collapse
Affiliation(s)
- Paola Manduca
- Genetics, DiBio, University of Genoa, 26, C. Europa, Genoa 16132, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Nakamura M, Nagai A, Tanaka Y, Sekijima Y, Yamashita K. Polarized hydroxyapatite promotes spread and motility of osteoblastic cells. J Biomed Mater Res A 2009; 92:783-90. [DOI: 10.1002/jbm.a.32404] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
35
|
Dentin phosphophoryn promotes cellular migration of human dental pulp cells. J Endod 2008; 34:575-8. [PMID: 18436037 DOI: 10.1016/j.joen.2008.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 02/06/2008] [Accepted: 02/12/2008] [Indexed: 11/22/2022]
Abstract
Dentin phosphophoryn (DPP) is a dentin sialophosphoprotein gene product that has an RGD motif and repeat sequences of aspartic acid and phosphoserine. To date, the function of DPP in the early stage of reparative dentin formation still remains unclear. The objective of this study was to evaluate the effects of DPP on pulp cell migration and proliferation. DPP promoted cell migration in a concentration-dependent manner, thus increasing it by about 3-fold at 1000 ng/mL compared with the control, but it had no effect on cell proliferation. Dephosphorylated DPP also promoted cell migration, similarly to DPP. However, cell migration was significantly suppressed by the addition of alphavbeta3 integrin antibody to the medium. Furthermore, porcine DPP-derived RGD peptide, but not its mutant RAD peptide, significantly promoted cell migration. These results indicated that the RGD motif of DPP plays an important role in the migration of human dental pulp cells.
Collapse
|
36
|
Globus RK. Extracellular Matrix and Integrin Interactions in the Skeletal Responses to Mechanical Loading and Unloading. Clin Rev Bone Miner Metab 2007. [DOI: 10.1007/s12018-008-9013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
37
|
Morrissey C, Vessella RL. The role of tumor microenvironment in prostate cancer bone metastasis. J Cell Biochem 2007; 101:873-86. [PMID: 17387734 DOI: 10.1002/jcb.21214] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Prostate cancer (PCa) epithelial cells require a number of factors to facilitate their establishment and growth at a distant site of metastasis. Their ability to adapt to their microenvironment, proliferate and recruit an underlying stroma is integral to the survival and growth of the metastasis. PCa predominantly metastasizes to the bone, and bone metastases are the main cause of morbidity. The bone marrow provides a permissive environment for the formation of a metastasis. In some cases, the cells may remain dormant for some time, eventually proliferating in response to an unknown "trigger." The marrow is rich in progenitor cells that differentiate into numerous cell types, producing new blood vessels, supporting fibroblasts, and an underlying extracellular matrix (ECM) that form the reactive stroma. By secreting a number of cytokines, growth factors and proteases they recruit auxiliary cells required to produce a functional stroma. These components are involved in a reciprocal interaction between the stroma and the PCa cells, allowing for the growth and survival of the tumor. Left unchecked, once a PCa tumor has established itself in the bone marrow it will eventually replace the marrow, interrupting bone homeostasis and typically promoting an osteoblastic response in the bone including osteoclastic events. The abundant deposition of new woven bone results in nerve compression, bone pain and an increase in fractures in patients with PCa bone metastases. This review will examine the tumor microenvironment, its role in facilitating tumor dissemination, growth and the resultant pathologies associated with PCa bone metastasis.
Collapse
Affiliation(s)
- Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
38
|
Gordon JAR, Tye CE, Sampaio AV, Underhill TM, Hunter GK, Goldberg HA. Bone sialoprotein expression enhances osteoblast differentiation and matrix mineralization in vitro. Bone 2007; 41:462-73. [PMID: 17572166 DOI: 10.1016/j.bone.2007.04.191] [Citation(s) in RCA: 242] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 04/16/2007] [Accepted: 04/23/2007] [Indexed: 11/20/2022]
Abstract
Bone sialoprotein (BSP) is an acidic, noncollagenous glycoprotein abundantly expressed in mineralized tissues. Although BSP is frequently used as a marker of osteoblast differentiation, the role of the protein in osteoblast function is unclear. BSP belongs to the SIBLING (Small Integrin-binding LIgand N-linked Glycoprotein) family of RGD-containing matrix proteins, several members of which have been shown to affect cell differentiation. The normal levels of BSP expression in osteoblasts were specifically altered by CMV-mediated adenoviral overexpression in primary osteoblasts or inhibition by an RNA interference-based strategy in the MC3T3E1 cell line. Alternatively, osteoblast cultures were supplemented with recombinant BSP protein. Quantitative real-time PCR was used to monitor the mRNA levels of the osteoblast-related transcription factors Osterix and Runx2 as well as the osteoblast-specific gene osteocalcin. As markers of osteoblast differentiation, alkaline phosphatase enzyme activity, Runx2-luciferase reporter activity and calcein incorporation into mineralized cultures were also measured. The overexpression of BSP increased osteoblast-related gene expression as well as calcium incorporation and nodule formation by osteoblast cultures. Similarly, supplementation of osteoblast cultures with recombinant BSP increased several markers of osteoblast differentiation. Conversely, suppression of BSP expression by small-hairpin RNA-encoding plasmids inhibited expression of osteoblast markers and nodule formation. Overexpression of several functional-domain mutants of BSP demonstrated that increases in osteoblast-related gene expression and matrix mineralization observed in BSP overexpression models are mediated by the integrin-binding RGD motif found near the C-terminus of the protein. These results demonstrate that BSP may serve as a matrix-associated signal directly promoting osteoblast differentiation resulting in the increased production of a mineralized matrix.
Collapse
Affiliation(s)
- Jonathan A R Gordon
- Department of Biochemistry and Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
39
|
Rehn AP, Birch MA, Karlström E, Wendel M, Lind T. ADAMTS-1 increases the three-dimensional growth of osteoblasts through type I collagen processing. Bone 2007; 41:231-8. [PMID: 17560840 DOI: 10.1016/j.bone.2007.04.187] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 04/02/2007] [Accepted: 04/15/2007] [Indexed: 11/28/2022]
Abstract
The multi-domain neutral endopeptidase, ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin repeats) is induced by parathyroid hormone (PTH) in rat osteoblasts and has therefore been suggested to be involved in initiation of bone remodeling. However, its function(s) in bone cells have not been studied. Here, we first establish that ADAMTS-1 protein is rapidly and transiently produced by human primary osteoblasts in response to PTH (1-34). We also show that ADAMTS-1 is specifically in close proximity to collagen fibrils in bone tissue using ultrastructural immunolabeling. To study the consequence(s) of ADAMTS-1 metalloprotease production in osteoblastic cells, human osteosarcoma cells (SaOS-2), were forced to express either wild-type (wtATS) or a point-mutated (pmATS) metalloprotease dead ADAMTS-1. SaOS-2 cells expressing wtATS had a growth advantage and increased collagenolytic activity when seeded inside a collagen type I gel but exhibited a reduced migration in a scratch wound assay. Immunolabeling of moving cells shows ADAMTS-1 to be located towards the direction of cellular migration. Finally, Western analysis demonstrated excess accumulation of mature collagen type I alpha1 species in the extracellular matrix together with increased release of distinct small collagen fragments into the conditioned media, by cultures of wtATS cells compared to pmATS cells. These results show that ADAMTS-1 has both the opportunity in bone and capability in vitro to induce collagen type I processing, together with a positive influence on osteoblastic three-dimensional growth. Although it is not clear at present if ADAMTS-1 promotes collagen degradation directly or indirectly, it shows that ADAMTS-1 activity can have a profound influence on the osteoblast phenotype, inhibiting migration on a planar substrate but enhancing growth in a collagen scaffold. These findings further establish ADAMTS-1 as a potentially important protein in PTH induced bone remodeling.
Collapse
Affiliation(s)
- Anders P Rehn
- Center for Oral Biology, Karolinska Institute, PO Box 4064, SE-141 04 Huddinge, Sweden.
| | | | | | | | | |
Collapse
|
40
|
Docheva D, Popov C, Mutschler W, Schieker M. Human mesenchymal stem cells in contact with their environment: surface characteristics and the integrin system. J Cell Mol Med 2007; 11:21-38. [PMID: 17367499 PMCID: PMC4401218 DOI: 10.1111/j.1582-4934.2007.00001.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The identification of mesenchymal stem cells (MSCs) in adult human tissues and the disclosure of their self-renew-al and multi-lineage differentiation capabilities have provided exciting prospects for cell-based regeneration and tis-sue engineering. Although a considerable amount of data is available describing MSCs, there is still lack of information regarding the molecular mechanisms that govern their adhesion and migration. In this work, we will review the current state of knowledge on integrins and other adhesion molecules found to be expressed on MSCs. The discussed topics include the characteristics of MSCs and their clinical applications, integrins and their central role in cell-matrix attachment and migration, and comments on mobilization, differentiation and contribution to tumour development. Finally, by understanding the complex and fundamental pathways by which MSCs attach and migrate, it might be possible to fine-tune the strategies for effective and safe use of MSCs in regenerative therapies.
Collapse
Affiliation(s)
- Denitsa Docheva
- Experimental Surgery and Regenerative Medicine, Department of Surgery,Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | |
Collapse
|