1
|
Zhang Y, Remy M, Leste-Lasserre T, Durrieu MC. Manipulating Stem Cell Fate with Disordered Bioactive Cues on Surfaces: The Role of Bioactive Ligand Selection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18474-18489. [PMID: 38581548 DOI: 10.1021/acsami.4c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2024]
Abstract
The development of 2D or 3D bioactive platforms for rapidly isolating pure populations of cells from adult stem cells holds promise for advancing the understanding of cellular mechanisms, drug testing, and tissue engineering. Over the years, methods have emerged to synthesize bioactive micro- and nanostructured 2D materials capable of directing stem cell fate. We introduce a novel method for randomly micro- or nanopatterning any protein/peptide onto both 2D and 3D scaffolds via spray technology. Our goal is to investigate the impact of arranging bioactive micropatterns (ordered vs disordered) on surfaces to guide human mesenchymal stem cell (hMSC) differentiation. The spray technology efficiently coats materials with controlled, cost-effective bioactive micropatterns in various sizes and shapes. BMP-2 mimetic peptides were covalently grafted, individually or in combination with RGD peptides, onto activated polyethylene terephthalate (PET) surfaces through a spraying process, incorporating nano/microscale parameters like size, shape, and composition. The study explores different peptide distributions on surfaces and various peptide combinations. Four surfaces were homogeneously functionalized with these peptides (M1 to M4 with various densities of peptides), and six surfaces with disordered micro- and nanopatterns of peptides (S0 to S5 with different sizes of peptide patterns) were synthesized. Fluorescence microscopy assessed peptide distribution, followed by hMSC culture for 2 weeks, and evaluated osteogenic differentiation via immunocytochemistry and RT-qPCR for osteoblast and osteocyte markers. Cells on uniformly peptide-functionalized surfaces exhibited cuboidal forms, while those on surfaces with disordered patterns tended toward columnar or cuboidal shapes. Surfaces S4 and S5 showed dendrite-like formations resembling an osteocyte morphology. S5 showed significant overexpression of osteoblast (OPN) and osteocyte markers (E11, DMP1, and SOST) compared to control surfaces and other micropatterned surfaces. Notably, despite sharing an equivalent quantity of peptides with a homogeneous functionalized surface, S5 displayed a distinct distribution of peptides, resulting in enhanced osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Yujie Zhang
- CNRS, Bordeaux INP, CBMN, Univ. Bordeaux, UMR 5248, Pessac33600,France
| | - Murielle Remy
- CNRS, Bordeaux INP, CBMN, Univ. Bordeaux, UMR 5248, Pessac33600,France
| | | | | |
Collapse
|
2
|
Takeshita N, Takano-Yamamoto T. Analysis of Chemotactic Property of CCN2/CTGF in Intramembranous Osteogenesis. Methods Mol Biol 2023; 2582:237-253. [PMID: 36370354 DOI: 10.1007/978-1-0716-2744-0_16] [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] [Indexed: 06/16/2023]
Abstract
Chemotaxis is a directed migration of cells in response to a gradient of extracellular molecules called chemoattractants. Development, growth, remodeling, and fracture healing of bones are advanced through intramembranous osteogenesis. Chemotaxis of preosteoblasts toward future bone formation sites observed in the early stage of intramembranous osteogenesis is a critical cellular process for normal bone formation. However, molecular biological mechanisms of the chemotaxis of preosteoblasts are not fully understood. We have recently clarified, for the first time, the critical role of the cellular communication network factor 2 (CCN2)/connective tissue growth factor (CTGF)-integrin α5-Ras axis for chemotaxis of preosteoblasts during new bone formation through intramembranous osteogenesis. In this chapter, we describe in detail the procedures of the in vivo and in vitro assays to investigate the chemotactic property of CCN2/CTGF and its underlying molecular biological mechanisms during intramembranous osteogenesis.
Collapse
Affiliation(s)
- Nobuo Takeshita
- Section of Orthodontics and Dentofacial Orthopedics, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan.
- Department of Biomaterials and Bioengineering, Faculty of Dental Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.
| |
Collapse
|
3
|
Shi C, Sun B, Wu H, Zhang R, Wu L, Guo L, Li C, Xi Y, Yuan W, Zhang Y, Xu G. Dysfunction of Caveolae-Mediated Endocytic TβRI Degradation Results in Hypersensitivity of TGF-β/Smad Signaling in Osteogenesis Imperfecta. J Bone Miner Res 2023; 38:103-118. [PMID: 36321807 DOI: 10.1002/jbmr.4734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/22/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022]
Abstract
Osteogenesis imperfecta (OI) is a genetic disorder caused by mutations of type I collagen-related genes, and excessive transforming growth factor-beta (TGF-β) signaling is a common mechanism. TGF-β/Smad signaling has inhibitory effects on osteoblast differentiation and maturation and is mainly transduced and regulated by the internalization of a tetrameric receptor complex comprising types I and II TGF-β receptors (TβRI and TβRII). During internalization, clathrin-mediated endocytosis enhances TGF-β/Smad signaling via Smad2/3 phosphorylation and receptors recycling, while caveolae-mediated endocytosis turns off TGF-β/Smad signaling by promoting receptor ubiquitination and degradation. In this study, using an animal model of OI (Colla2oim , osteogenesis imperfecta murine [oim]/oim mouse), we found that osteoblastic cells of oim/oim mice were more sensitive to the inhibitory effects of TGF-β on osteoblast differentiation and maturation and had much higher cell membrane protein levels of TGF-β receptors than those of wild-type (wt)/wt mice. Further results showed that clathrin-mediated endocytosis of TβRI was enhanced, whereas caveolae-mediated TβRI endocytic degradation was reduced in oim/oim mice, combined with reduced caveolin-1 (Cav-1) phosphorylation. In addition, type I collagen downregulated TβRI via focal adhesion kinase (FAK) and Src activation-dependent Cav-1 phosphorylation. To further examine this mechanism, 4-week-old oim/oim and wt/wt mice were treated with either TβRI kinase inhibitor (SD-208) or vehicle for 8 weeks. SD-208 treatment significantly reduced the fracture incidence in oim/oim mice. Micro-computed tomography and biomechanical testing showed that femoral bone mass and strength were significantly improved with SD-208 treatment in both genotypes. Additionally, SD-208 significantly promoted osteoblast differentiation and bone formation and inhibited bone resorption. In conclusion, dysfunction of caveolae-mediated endocytic TβRI degradation is a possible mechanism for the enhanced TGF-β/Smad signaling in OI. Targeting this mechanism using a TβRI kinase inhibitor effectively reduced fractures and improved bone mass and strength in OI model and, thus, may offer a new strategy for the treatment of OI. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Changgui Shi
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bin Sun
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huiqiao Wu
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Rongcheng Zhang
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lecheng Wu
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lei Guo
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changwei Li
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopedics and Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanhai Xi
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wen Yuan
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ying Zhang
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Guohua Xu
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Liu Y, Li S, Liu H, Li B. Osteogenic peptides in collagen hydrolysates: Stimulate differentiation of MC3T3-E1 cells via β1 integrin-FAK-ERK1/2 signaling pathway and Smad1 protein. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
5
|
Targeted activation of androgen receptor signaling in the periosteum improves bone fracture repair. Cell Death Dis 2022; 13:123. [PMID: 35136023 PMCID: PMC8826926 DOI: 10.1038/s41419-022-04595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/30/2021] [Accepted: 01/27/2022] [Indexed: 12/03/2022]
Abstract
Low testosterone level is an independent predictor of osteoporotic fracture in elderly men as well as increased fracture risk in men undergoing androgen deprivation. Androgens and androgen receptor (AR) actions are essential for bone development and homeostasis but their linkage to fracture repair remains unclear. Here we found that AR is highly expressed in the periosteum cells and is co-localized with a mesenchymal progenitor cell marker, paired-related homeobox protein 1 (Prrx1), during bone fracture repair. Mice lacking the AR gene in the periosteum expressing Prrx1-cre (AR-/Y;Prrx1::Cre) but not in the chondrocytes (AR-/Y;Col-2::Cre) exhibits reduced callus size and new bone volume. Gene expression data analysis revealed that the expression of several collagens, integrins and cell adhesion molecules were downregulated in periosteum-derived progenitor cells (PDCs) from AR-/Y;Prrx1::Cre mice. Mechanistically, androgens-AR signaling activates the AR/ARA55/FAK complex and induces the collagen-integrin α2β1 gene expression that is required for promoting the AR-mediated PDCs migration. Using mouse cortical-defect and femoral graft transplantation models, we proved that elimination of AR in periosteum of host mice impairs fracture healing, regardless of AR existence of transplanted donor graft. While testosterone implanted scaffolds failed to complete callus bridging across the fracture gap in AR-/Y;Prrx1::Cre mice, cell-based transplantation using DPCs re-expressing AR could lead to rescue bone repair. In conclusion, targeting androgen/AR axis in the periosteum may provide a novel therapy approach to improve fracture healing.
Collapse
|
6
|
Jiang W, Takeshita N, Maeda T, Sogi C, Oyanagi T, Kimura S, Yoshida M, Sasaki K, Ito A, Takano-Yamamoto T. Connective tissue growth factor promotes chemotaxis of preosteoblasts through integrin α5 and Ras during tensile force-induced intramembranous osteogenesis. Sci Rep 2021; 11:2368. [PMID: 33504916 PMCID: PMC7841149 DOI: 10.1038/s41598-021-82246-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
In vertebrates, new bone formation via intramembranous osteogenesis is a critical biological event for development, remodeling, and fracture healing of bones. Chemotaxis of osteoblast lineage cells is an essential cellular process in new bone formation. Connective tissue growth factor (CTGF) is known to exert chemotactic properties on various cells; however, details of CTGF function in the chemotaxis of osteoblast lineage cells and underlying molecular biological mechanisms have not been clarified. The aim of the present study was to evaluate the chemotactic properties of CTGF and its underlying mechanisms during active bone formation through intramembranous osteogenesis. In our mouse tensile force-induced bone formation model, preosteoblasts were aggregated at the osteogenic front of calvarial bones. CTGF was expressed at the osteogenic front, and functional inhibition of CTGF using a neutralizing antibody suppressed the aggregation of preosteoblasts. In vitro experiments using μ-slide chemotaxis chambers showed that a gradient of CTGF induced chemotaxis of preosteoblastic MC3T3-E1 cells, while a neutralizing integrin α5 antibody and a Ras inhibitor inhibited the CTGF-induced chemotaxis of MC3T3-E1 cells. These findings suggest that the CTGF-integrin α5-Ras axis is an essential molecular mechanism to promote chemotaxis of preosteoblasts during new bone formation through intramembranous osteogenesis.
Collapse
Affiliation(s)
- Wei Jiang
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Nobuo Takeshita
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Toshihiro Maeda
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Chisumi Sogi
- Department of Pediatrics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, 980-8574, Japan
| | - Toshihito Oyanagi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Seiji Kimura
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Michiko Yoshida
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Kiyo Sasaki
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Arata Ito
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, 980-8575, Japan. .,Department of Biomaterials and Bioengineering, Faculty of Dental Medicine, Hokkaido University, Sapporo, Hokkaido, 060-8586, Japan.
| |
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
|
Buarque de Gusmão CV, Batista NA, Vidotto Lemes VT, Maia Neto WL, de Faria LD, Alves JM, Belangero WD. Effect of Low-Intensity Pulsed Ultrasound Stimulation, Extracorporeal Shockwaves and Radial Pressure Waves on Akt, BMP-2, ERK-2, FAK and TGF-β1 During Bone Healing in Rat Tibial Defects. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2140-2161. [PMID: 31101448 DOI: 10.1016/j.ultrasmedbio.2019.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/24/2019] [Accepted: 04/07/2019] [Indexed: 06/09/2023]
Abstract
An experimental study was conducted to determine whether low-intensity pulsed ultrasound stimulation (LIPUS), extracorporeal shockwave treatment (ESWT) and radial pressure wave treatment (RPWT) modulate Akt, bone morphogenetic protein-2 (BMP-2), extracellular signal-regulated kinase-2 (ERK-2), focal adhesion kinase (FAK) and transforming growth factor-β1 (TGF-β1) during bone healing in rat tibial defects. Rat tibial defects were exposed to 500 shots of ESWT delivered at 0.12 mJ/mm2, 500 impulses of RPWT operated at 2.0 bar or to daily 20-min 30 mW/cm2 LIPUS. Following 1, 3 and 6 wk, bones were harvested to determine the expression and activity of Akt, BMP-2, ERK-2, FAK and TGF-β1. Animals exposed to ultrasound were followed up to 3 wk. Protein expression and activity were unchanged following LIPUS treatment. ESWT increased Akt activity 2.11-fold (p = 0.043) and TGF-β1 expression 9.11-fold (p = 0.016) at 1 wk and increased FAK activity 2.16-fold (p = 0.047) at 3 wk. RPWT increased FAK activity 2.6-fold (p = 0.028) at 3 wk and decreased Akt expression 0.52-fold (p = 0.05) at 6 wk. In conclusion, the protocols employed for ESWT and RPWT modulated distinct signaling pathways during fracture healing, while LIPUS standard protocol did not change the usual signaling pathways of the proteins investigated. Future studies are required to monitor osteogenesis so that the biologic meaning of our results can be clarified.
Collapse
Affiliation(s)
- Carlos Vinícius Buarque de Gusmão
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Nilza Alzira Batista
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Valeria Trombini Vidotto Lemes
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Wilson Leite Maia Neto
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Lidia Dornelas de Faria
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José Marcos Alves
- Electrical Engineering Department, College of Engineering of São Carlos, University of São Paulo (USP), São Carlos, São Paulo, Brazil
| | - William Dias Belangero
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
9
|
Nokin MJ, Bellier J, Durieux F, Peulen O, Rademaker G, Gabriel M, Monseur C, Charloteaux B, Verbeke L, van Laere S, Roncarati P, Herfs M, Lambert C, Scheijen J, Schalkwijk C, Colige A, Caers J, Delvenne P, Turtoi A, Castronovo V, Bellahcène A. Methylglyoxal, a glycolysis metabolite, triggers metastasis through MEK/ERK/SMAD1 pathway activation in breast cancer. Breast Cancer Res 2019; 21:11. [PMID: 30674353 PMCID: PMC6343302 DOI: 10.1186/s13058-018-1095-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/27/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Elevated aerobic glycolysis rate is a biochemical alteration associated with malignant transformation and cancer progression. This metabolic shift unavoidably generates methylglyoxal (MG), a potent inducer of dicarbonyl stress through the formation of advanced glycation end products (AGEs). We have previously shown that the silencing of glyoxalase 1 (GLO1), the main MG detoxifying enzyme, generates endogenous dicarbonyl stress resulting in enhanced growth and metastasis in vivo. However, the molecular mechanisms through which MG stress promotes metastasis development remain to be unveiled. METHODS In this study, we used RNA sequencing analysis to investigate gene-expression profiling of GLO1-depleted breast cancer cells and we validated the regulated expression of selected genes of interest by RT-qPCR. Using in vitro and in vivo assays, we demonstrated the acquisition of a pro-metastatic phenotype related to dicarbonyl stress in MDA-MB-231, MDA-MB-468 and MCF7 breast cancer cellular models. Hyperactivation of MEK/ERK/SMAD1 pathway was evidenced using western blotting upon endogenous MG stress and exogenous MG treatment conditions. MEK and SMAD1 regulation of MG pro-metastatic signature genes in breast cancer cells was demonstrated by RT-qPCR. RESULTS High-throughput transcriptome profiling of GLO1-depleted breast cancer cells highlighted a pro-metastatic signature that establishes novel connections between MG dicarbonyl stress, extracellular matrix (ECM) remodeling by neoplastic cells and enhanced cell migration. Mechanistically, we showed that these metastasis-related processes are functionally linked to MEK/ERK/SMAD1 cascade activation in breast cancer cells. We showed that sustained MEK/ERK activation in GLO1-depleted cells notably occurred through the down-regulation of the expression of dual specificity phosphatases in MG-stressed breast cancer cells. The use of carnosine and aminoguanidine, two potent MG scavengers, reversed MG stress effects in in vitro and in vivo experimental settings. CONCLUSIONS These results uncover for the first time the key role of MG dicarbonyl stress in the induction of ECM remodeling and the activation of migratory signaling pathways, both in favor of enhanced metastatic dissemination of breast cancer cells. Importantly, the efficient inhibition of mitogen-activated protein kinase (MAPK) signaling using MG scavengers further emphasizes the need to investigate their therapeutic potential across different malignancies.
Collapse
Affiliation(s)
- Marie-Julie Nokin
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Justine Bellier
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Florence Durieux
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Gilles Rademaker
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Maude Gabriel
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Christine Monseur
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | | | - Lieven Verbeke
- Department of Information Technology, Ghent University, Ghent, Belgium
| | - Steven van Laere
- Translational Cancer Research Unit, University of Antwerp, Antwerp, Belgium
| | - Patrick Roncarati
- Laboratory of Experimental Pathology, GIGA-Cancer, ULiège, Liège, Belgium
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, ULiège, Liège, Belgium
| | - Charles Lambert
- Laboratory of Connective Tissues Biology, GIGA-Cancer, ULiège, Liège, Belgium
| | - Jean Scheijen
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Casper Schalkwijk
- Laboratory for Metabolism and Vascular Medicine, Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-Cancer, ULiège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Inflammation, Infection and Immunity, ULiège, Liège, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology, GIGA-Cancer, ULiège, Liège, Belgium
| | - Andrei Turtoi
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier, France
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège (ULiège), Pathology Tour, +4 level, Building 23, Avenue Hippocrate 13, 4000, Liège, Belgium.
| |
Collapse
|
10
|
Zhang L, Zhang S, Song H, Li B. Effect of Collagen Hydrolysates from Silver Carp Skin ( Hypophthalmichthys molitrix) on Osteoporosis in Chronologically Aged Mice: Increasing Bone Remodeling. Nutrients 2018; 10:nu10101434. [PMID: 30287779 PMCID: PMC6212965 DOI: 10.3390/nu10101434] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/27/2018] [Accepted: 09/30/2018] [Indexed: 12/20/2022] Open
Abstract
Osteoporosis is a common skeletal disorder in humans and gelatin hydrolysates from mammals have been reported to improve osteoporosis. In this study, 13-month-old mice were used to evaluate the effects of collagen hydrolysates (CHs) from silver carp skin on osteoporosis. No significant differences were observed in mice body weight, spleen or thymus indices after daily intake of antioxidant collagen hydrolysates (ACH; 200 mg/kg body weight (bw) (LACH), 400 mg/kg bw (MACH), 800 mg/kg bw (HACH)), collagenase hydrolyzed collagen hydrolysates (CCH) or proline (400 mg/kg body weight) for eight weeks, respectively. ACH tended to improve bone mineral density, increase bone hydroxyproline content, enhance alkaline phosphatase (ALP) level and reduce tartrate-resistant acid phosphatase 5b (TRAP-5b) activity in serum, with significant differences observed between the MACH and model groups (p < 0.05). ACH exerted a better effect on osteoporosis than CCH at the identical dose, whereas proline had no significant effect on repairing osteoporosis compared to the model group. Western blotting results demonstrated that CHs mainly increased bone remodeling by stimulating the transforming growth factor β1 (TGF-β1)/Smad signaling pathway and improving the interaction between collagen and α2β1 integrin. The results indicated that CHs from fish could be applied to alleviate osteoporosis or treat bone loss.
Collapse
Affiliation(s)
- Ling Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Siqi Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Hongdong Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Bo Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
- Beijing Higher Institution Engineering Research Center of Animal Product, Beijing 100083, China.
| |
Collapse
|
11
|
Rajshankar D, Wang Y, McCulloch CA. Osteogenesis requires FAK-dependent collagen synthesis by fibroblasts and osteoblasts. FASEB J 2016; 31:937-953. [PMID: 27881487 DOI: 10.1096/fj.201600645r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 11/14/2016] [Indexed: 11/11/2022]
Abstract
Focal adhesion kinase (FAK) is critical in adhesion-dependent signaling, but its role in osteogenesis in vivo is ill defined. We deleted Fak in fibroblasts and osteoblasts in Floxed-Fak mice bred with those expressing Cre-recombinase driven by 3.6-kb α1(I)-collagen promoter. Compared with wild-type (WT), conditional FAK-knockout (CFKO) mice were shorter (2-fold; P < 0.0001) and had crooked, shorter tails (50%; P < 0.0001). Microcomputed tomography analysis showed reduced bone volume (4-fold in tails; P < 0.0001; 2-fold in mandibles; P < 0.0001), whereas bone surface area/bone volume increased (3-fold in tails; P < 0.0001; 2.5-fold in mandibles; P < 0.001). Collagen density and fiber alignment in periodontal ligament were reduced by 4-fold (P < 0.0001) and 30% (P < 0.05), respectively, in CFKO mice. In cultured CFKO osteoblasts, mineralization at d 7 and mineralizing colony-forming units at d 21 were 30% (P < 0.0001) and >3-fold less than WT, respectively. Disruptions of FAK function in osteoblasts by conditional knockout, siRNA-knockdown, or FAK inhibitor reduced mRNA and protein expression of Runx2 (>30%), Osterix (>25%), and collagen-1 (2-fold). Collagen synthesis was abrogated in WT osteoblasts with Runx2 knockdown and in Fak-null fibroblasts transfected with an FAK kinase domain mutant or a kinase-impaired mutant (Y397F). These data indicate that FAK regulates osteogenesis through transcription factors that regulate collagen synthesis.-Rajshankar, D., Wang, Y., McCulloch, C. A. Osteogenesis requires FAK-dependent collagen synthesis by fibroblasts and osteoblasts.
Collapse
Affiliation(s)
- Dhaarmini Rajshankar
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Yongqiang Wang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Christopher A McCulloch
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Modulation of BMP signalling by integrins. Biochem Soc Trans 2016; 44:1465-1473. [DOI: 10.1042/bst20160111] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/11/2016] [Accepted: 07/15/2016] [Indexed: 12/17/2022]
Abstract
The bone morphogenetic protein (BMP) pathway is a major conserved signalling pathway with diverse roles in development and homeostasis. Given that cells exist in three-dimensional environments, one important area is to understand how the BMP pathway operates within such complex cellular environments. The extracellular matrix contains information regarding tissue architecture and its mechanical properties that is transmitted to the cell via integrin receptors. In this review, I describe various examples of modulation of the BMP pathway by integrins. In the case of the Drosophila embryo and some cell line-based studies, integrins have been found to enhance BMP responses through different mechanisms, such as enhancement of BMP ligand–receptor binding and effects on Smad phosphorylation or stability. In these contexts, BMP-dependent activation of integrins is a common theme. However, I also discuss examples where integrins inhibit the BMP pathway, highlighting the context-dependent nature of integrin–BMP cross-talk.
Collapse
|
13
|
RGD and BMP-2 mimetic peptide crosstalk enhances osteogenic commitment of human bone marrow stem cells. Acta Biomater 2016; 36:132-42. [PMID: 27000551 DOI: 10.1016/j.actbio.2016.03.032] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/19/2016] [Accepted: 03/17/2016] [Indexed: 12/16/2022]
Abstract
UNLABELLED Human bone marrow mesenchymal stem cells (hBMSCs) commitment and differentiation are dictated by bioactive molecules sequestered within their Extra Cellular Matrix (ECM). One common approach to mimic the physiological environment is to functionalize biomaterial surfaces with ECM-derived peptides able to recruit stem cells and trigger their linage-specific differentiation. The objective of this work was to investigate the effect of RGD and BMP-2 ligands crosstalk and density on the extent of hBMSCs osteogenic commitment, without recourse to differentiation medium. RGD peptide promotes cell adhesion via cell transmembrane integrin receptors, while BMP-2 peptide, corresponding to residues 73-92 of Bone Morphogenetic Protein-2, was shown to induce hBMSCs osteoblast differentiation. The immobilization of peptides on aminated glass was ascertained by X-ray Photoelectron Spectroscopy (XPS), the density of grafted peptides was quantified by fluorescence microscopy and the surface roughness was evaluated using Atomic Force Microscopy (AFM). The osteogenic commitment of hBMSCs cultured on RGD and/or BMP-2 surfaces was characterized by immunohistochemistry using STRO-1 as specific stem cells marker and Runx-2 as an earlier osteogenic marker. Biological results showed that the osteogenic commitment of hBMSCs was enhanced on bifunctionalized surfaces as compared to surfaces containing BMP-2, while on RGD surfaces cells mainly preserved their stemness character. These results demonstrated that RGD and BMP-2 mimetic peptides act synergistically to enhance hBMSCs osteogenesis without supplementing the media with osteogenic factors. These findings contribute to the development of biomimetic materials, allowing a deeper understanding of signaling pathways that govern the transition of stem cells towards the osteoblastic lineage. STATEMENT OF SIGNIFICANCE For a long time, scientists thought that the differentiation of Mesenchymal Stem Cells (MSCs) into bone cells was dictated by growth factors. This manuscript shed light on other ligands that play a crucial role in regulating MSCs fate. In concrete terms, it was demonstrated that the osteoinductive effect of BMP-2 peptide is 2 folds improved in the presence of adhesive RGD peptide. Compared to previous works highlighting this synergistic cooperation between RGD and BMP-2 peptides, the main strength of this work lies to the use of primitive human cells (hMSCs) and well-defined biomimetic material surfaces (controlled surface roughness and peptide densities). This work provides valuable insights to develop custom-designed in vitro cell culture models, capable of targeting the desired cell response.
Collapse
|
14
|
Rasi Ghaemi S, Delalat B, Cetó X, Harding FJ, Tuke J, Voelcker NH. Synergistic influence of collagen I and BMP 2 drives osteogenic differentiation of mesenchymal stem cells: A cell microarray analysis. Acta Biomater 2016. [PMID: 26196081 DOI: 10.1016/j.actbio.2015.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell microarrays are a novel platform for the high throughput discovery of new biomaterials. By re-creating a multitude of cell microenvironments on a single slide, this approach can identify the optimal surface composition to drive a desired cell response. To systematically study the effects of molecular microenvironments on stem cell fate, we designed a cell microarray based on parallel exposure of mesenchymal stem cells (MSCs) to surface-immobilised collagen I (Coll I) and bone morphogenetic protein 2 (BMP 2). This was achieved by means of a reactive coating on a slide surface, enabling the covalent anchoring of Coll I and BMP 2 as microscale spots printed by a robotic contact printer. The surface between the printed protein spots was passivated using poly (ethylene glycol) bisamine 10,000Da (A-PEG). MSCs were then captured and cultured on array spots composed of binary mixtures of Coll I and BMP 2, followed by automated image acquisition and quantitative, multi-parameter analysis of cellular responses. Surface compositions that gave the highest osteogenic differentiation were determined using Runx2 expression and calcium deposition. Quantitative single cell analysis revealed subtle concentration-dependent effects of surface-immobilised proteins on the extent of osteogenic differentiation obscured using conventional analysis. In particular, the synergistic interaction of Coll I and BMP 2 in supporting osteogenic differentiation was confirmed. Our studies demonstrate the value of cell microarray platforms to decipher the combinatorial interactions at play in stem cell niche microenvironments.
Collapse
|
15
|
Sika Deer Antler Collagen Type I-Accelerated Osteogenesis in Bone Marrow Mesenchymal Stem Cells via the Smad Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2109204. [PMID: 27066099 PMCID: PMC4809101 DOI: 10.1155/2016/2109204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/06/2015] [Indexed: 11/17/2022]
Abstract
Deer antler preparations have been used to strengthen bones for centuries. It is particularly rich in collagen type I. This study aimed to unravel part of the purported bioremedial effect of Sika deer antler collagen type I (SDA-Col I) on bone marrow mesenchymal stem cells. The results suggest that SDA-Col I might be used to promote and regulate osteoblast proliferation and differentiation. SDA-Col I might potentially provide the basis for novel therapeutic strategies in the treatment of bone injury and/or in scaffolds for bone replacement strategies. Finally, isolation of SDA-Col I from deer antler represents a renewable, green, and uncomplicated way to obtain a biomedically valuable therapeutic.
Collapse
|
16
|
Moon JS, Kim SH, Oh SH, Jeong YW, Kang JH, Park JC, Son HJ, Bae S, Park BI, Kim MS, Koh JT, Ko HM. Relaxin augments BMP-2-induced osteoblast differentiation and bone formation. J Bone Miner Res 2014; 29:1586-96. [PMID: 24643989 DOI: 10.1002/jbmr.2197] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 01/26/2014] [Accepted: 02/06/2014] [Indexed: 12/19/2022]
Abstract
Relaxin (Rln), a polypeptide hormone of the insulin superfamily, is an ovarian peptide hormone that is involved in a diverse range of physiological and pathological reactions. In this study, we investigated the effect of Rln on bone morphogenetic protein 2 (BMP-2)-induced osteoblast differentiation and bone formation. Expression of Rln receptors was examined in the primary mouse bone marrow stem cells (BMSCs) and mouse embryonic fibroblast cell line C3H/10T1/2 cells by RT-PCR and Western blot during BMP-2-induced osteoblast differentiation. The effect of Rln on osteoblast differentiation and mineralization was evaluated by measuring the alkaline phosphatase activity, osteocalcin production, and Alizarin red S staining. For the in vivo evaluation, BMP-2 and/or Rln were administered with type I collagen into the back of mice, and after 3 weeks, bone formation was analyzed by micro-computed tomography (µCT). Western blot was performed to determine the effect of Rln on osteoblast differentiation-related signaling pathway. Expression of Rxfp 1 in BMSCs and C3H/10T1/2 cells was significantly increased by BMP-2. In vitro, Rln augmented BMP-2-induced alkaline phosphatase expression, osteocalcin production, and matrix mineralization in BMSCs and C3H/10T1/2 cells. In addition, in vivo administration of Rln enhanced BMP-2-induced bone formation in a dose-dependent manner. Interestingly, Rln synergistically increased and sustained BMP-2-induced Smad, p38, and transforming growth factor-β activated kinase (TAK) 1 phosphorylation. BMP-2-induced Runx 2 expression and activity were also significantly augmented by Rln. These results show that Rln enhanced synergistically BMP-2-induced osteoblast differentiation and bone formation through its receptor, Rxfp 1, by augmenting and sustaining BMP-2-induced Smad and p38 phosphorylation, which upregulate Runx 2 expression and activity. These results suggest that Rln might be useful for therapeutic application in destructive bone diseases.
Collapse
Affiliation(s)
- Jung-Sun Moon
- Dental Science Research Institute, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bone regeneration using an alpha 2 beta 1 integrin-specific hydrogel as a BMP-2 delivery vehicle. Biomaterials 2014; 35:5453-61. [PMID: 24726536 DOI: 10.1016/j.biomaterials.2014.03.055] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/21/2014] [Indexed: 01/01/2023]
Abstract
Non-healing bone defects present tremendous socioeconomic costs. Although successful in some clinical settings, bone morphogenetic protein (BMP) therapies require supraphysiological dose delivery for bone repair, raising treatment costs and risks of complications. We engineered a protease-degradable poly(ethylene glycol) (PEG) synthetic hydrogel functionalized with a triple helical, α2β1 integrin-specific peptide (GFOGER) as a BMP-2 delivery vehicle. GFOGER-functionalized hydrogels lacking BMP-2 directed human stem cell differentiation and produced significant enhancements in bone repair within a critical-sized bone defect compared to RGD hydrogels or empty defects. GFOGER functionalization was crucial to the BMP-2-dependent healing response. Importantly, these engineered hydrogels outperformed the current clinical carrier in repairing non-healing bone defects at low BMP-2 doses. GFOGER hydrogels provided sustained in vivo release of encapsulated BMP-2, increased osteoprogenitor localization in the defect site, enhanced bone formation and induced defect bridging and mechanically robust healing at low BMP-2 doses which stimulated almost no bone regeneration when delivered from collagen sponges. These findings demonstrate that GFOGER hydrogels promote bone regeneration in challenging defects with low delivered BMP-2 doses and represent an effective delivery vehicle for protein therapeutics with translational potential.
Collapse
|
18
|
Abstract
Understanding the processes by which stem cells give rise to de novo tissues is an active focus of stem cell biology and bioengineering disciplines. Instructive morphogenic cues surrounding the stem cell during morphogenesis create what is referred to as the stem cell microenvironment. An emerging paradigm in stem cell bioengineering involves "biologically driven assembly," in which stem cells are encouraged to largely define their own morphogenesis processes. However, even in the case of biologically driven assembly, stem cells do not act alone. The properties of the surrounding microenvironment can be critical regulators of cell fate. Stem cell-material interactions are among the most well-characterized microenvironmental effectors of stem cell fate and establish a signaling "context" that can define the mode of influence for morphogenic cues. Here we describe illustrative examples of cell-material interactions that occur during in vitro stem cell studies, with an emphasis on how cell-material interactions create instructive contexts for stem cell differentiation and morphogenesis.
Collapse
Affiliation(s)
- Andrew S. Khalil
- Department of Biomedical Engineering, Orthopedics University of Wisconsin, Madison, Wisconsin 53705, USA
| | - Angela W. Xie
- Department of Biomedical Engineering, Orthopedics University of Wisconsin, Madison, Wisconsin 53705, USA
| | - William L. Murphy
- Department of Biomedical Engineering, Orthopedics University of Wisconsin, Madison, Wisconsin 53705, USA
- Department of Biomedical Rehabilitation, and Material Science University of Wisconsin, Madison, Wisconsin 53705, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin 53705, USA
| |
Collapse
|
19
|
Sista S, Wen C, Hodgson PD, Pande G. Expression of cell adhesion and differentiation related genes in MC3T3 osteoblasts plated on titanium alloys: role of surface properties. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:1573-82. [DOI: 10.1016/j.msec.2012.12.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/31/2012] [Accepted: 12/17/2012] [Indexed: 01/23/2023]
|
20
|
Schofer MD, Tünnermann L, Kaiser H, Roessler PP, Theisen C, Heverhagen JT, Hering J, Voelker M, Agarwal S, Efe T, Fuchs-Winkelmann S, Paletta JRJ. Functionalisation of PLLA nanofiber scaffolds using a possible cooperative effect between collagen type I and BMP-2: impact on colonization and bone formation in vivo. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:2227-33. [PMID: 22718044 PMCID: PMC3431465 DOI: 10.1007/s10856-012-4697-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 05/27/2012] [Indexed: 06/01/2023]
Abstract
The reconstruction of large bone defects after injury or tumor resection often requires the use of bone substitution. Artificial scaffolds based on synthetic biomaterials can overcome disadvantages of autologous bone grafts, like limited availability and donor side morbidity. Among them, scaffolds based on nanofibers offer great advantages. They mimic the extracellular matrix, can be used as a carrier for growth factors and allow the differentiation of human mesenchymal stem cells. Differentiation is triggered by a series of signaling processes, including integrin and bone morphogenetic protein (BMP), which act in a cooperative manner. The aim of this study was to analyze whether these processes can be remodeled in artificial poly-(l)-lactide acid (PLLA) based nanofiber scaffolds in vivo. Electrospun matrices composed of PLLA-collagen type I or BMP-2 incorporated PLLA-collagen type I were implanted in calvarial critical size defects in rats. Cranial CT-scans were taken 4, 8 and 12 weeks after implantation. Specimens obtained after euthanasia were processed for histology and immunostainings on osteocalcin, BMP-2 and Smad5. After implantation the scaffolds were inhomogeneously colonized and cells were only present in wrinkle- or channel-like structures. Ossification was detected only in focal areas of the scaffold. This was independent of whether BMP-2 was incorporated in the scaffold. However, cells that migrated into the scaffold showed an increased ratio of osteocalcin and Smad5 positive cells compared to empty defects. Furthermore, in case of BMP-2 incorporated PLLA-collagen type I scaffolds, 4 weeks after implantation approximately 40 % of the cells stained positive for BMP-2 indicating an autocrine process of the ingrown cells. These findings indicate that a cooperative effect between BMP-2 and collagen type I can be transferred to PLLA nanofibers and furthermore, that this effect is active in vivo. However, this had no effect on bone formation. The reason for this seems to be an unbalanced colonization of the scaffolds with cells, due to insufficient pore size.
Collapse
Affiliation(s)
- Markus D. Schofer
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Lisa Tünnermann
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Hendric Kaiser
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Philip P. Roessler
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Christina Theisen
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Johannes T. Heverhagen
- Department of Radiology, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Jacqueline Hering
- Department of Radiology, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Maximilian Voelker
- Department of Radiology, University Hospital Marburg, Baldingerstraße, 35033 Marburg, Germany
| | - Seema Agarwal
- Department of Macromolecular Chemistry, Philipps-University Marburg, Hans-Meerwein-Straße, 35032 Marburg, Germany
| | - Turgay Efe
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Susanne Fuchs-Winkelmann
- Department of Orthopedics and Rheumatology, University Hospital Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Department of Orthopedics and Rheumatology, Philipps-University, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
21
|
Ge C, Yang Q, Zhao G, Yu H, Kirkwood KL, Franceschi RT. Interactions between extracellular signal-regulated kinase 1/2 and p38 MAP kinase pathways in the control of RUNX2 phosphorylation and transcriptional activity. J Bone Miner Res 2012; 27:538-51. [PMID: 22072425 PMCID: PMC4285380 DOI: 10.1002/jbmr.561] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
RUNX2, a key transcription factor for osteoblast differentiation, is regulated by ERK1/2 and p38 MAP kinase-mediated phosphorylation. However, the specific contribution of each kinase to RUNX2-dependent transcription is not known. Here we investigate ERK and p38 regulation of RUNX2 using a unique P-RUNX2-specific antibody. Both MAP kinases stimulated RUNX2 Ser319 phosphorylation and transcriptional activity. However, a clear preference for ERK1 versus p38α/β was found when the ability of these MAPKs to phosphorylate and activate RUNX2 was compared. Similarly, ERK1 preferentially bound to a consensus MAPK binding site on RUNX2 that was essential for the activity of either kinase. To assess the relative contribution of ERK1/2 and p38 to osteoblast gene expression, MC3T3-E1 preosteoblast cells were grown in control or ascorbic acid (AA)-containing medium ± BMP2/7. AA-induced gene expression, which requires collagen matrix synthesis, was associated with parallel increases in P-ERK and RUNX2-S319-P in the absence of any changes in P-p38. This response was blocked by ERK, but not p38, inhibition. Significantly, in the presence of AA, BMP2/7 synergistically stimulated RUNX2 S319 phosphorylation and transcriptional activity without affecting total RUNX2 and this response was totally dependent on ERK/MAPK activity. In contrast, although p38 inhibition partially blocked BMP-dependent transcription, it did not affect RUNX2 S319 phosphorylation, suggesting the involvement of other phosphorylation sites and/or transcription factors in this response. Based on this work, we conclude that extracellular matrix and BMP regulation of RUNX2 phosphorylation and transcriptional activity in osteoblasts is predominantly mediated by ERK rather than p38 MAPKs.
Collapse
Affiliation(s)
- Chunxi Ge
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | - Qian Yang
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | - Guisheng Zhao
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | - Hong Yu
- Department of Craniofacial Biology and the Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Keith L. Kirkwood
- Department of Craniofacial Biology and the Center for Oral Health Research, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Renny T. Franceschi
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
- Department of Biological Chemistry, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| |
Collapse
|
22
|
Skarżyńska J, Damulewicz M, Filipowska J, Madej W, Leboy PS, Osyczka AM. Modification of Smad1 linker modulates BMP-mediated osteogenesis of adult human MSC. Connect Tissue Res 2011; 52:408-14. [PMID: 21405981 DOI: 10.3109/03008207.2010.551568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We examined whether bone morphogenetic protein (BMP)-mediated osteogenesis of adult human mesenchymal stem cells (MSCs) is regulated by extracellular signal-regulated kinase phosphorylation of Smad1. Adenoviral constructs carrying either unmodified human Smad1 or Smad1 mutated in the linker region to preclude extracellular signal-regulated kinase phosphorylation were expressed in human and rodent cells. Unlike unmodified Smad1, expression of mutated Smad1 facilitated BMP-stimulated expression of osteoblast markers in human MSC but had no effect on either rat MSC or mouse pre-osteoblastic MC3T3-E1 cells. Expression of mutated Smad1 in adult human MSC cultures also resulted in increased nuclear accumulation of BMP-activated Smads and elevated gene transcripts characteristic of differentiating osteoblasts. These results may partly explain the poor efficacy of BMP in some human bone therapies and indicate an important mechanism regulating BMP-mediated bone formation in adults.
Collapse
Affiliation(s)
- Joanna Skarżyńska
- Department of Cytology and Histology, Jagiellonian University , Kraków , Poland
| | | | | | | | | | | |
Collapse
|
23
|
Schneider GB, Zaharias R, Seabold D, Stanford C. Integrin-associated tyrosine kinase FAK affects Cbfa1 expression. J Orthop Res 2011; 29:1443-7. [PMID: 21412826 DOI: 10.1002/jor.21382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 01/18/2011] [Indexed: 02/04/2023]
Abstract
Following cell adhesion, focal adhesion kinase (FAK) autophosphorylates on tyrosine and regulates intracellular signaling cascades that regulate cell growth and differentiation. The hypothesis of this study was FAK mediates osteoblast differentiation dependent Cbfa1 expression. Slowly mineralizing UI and rapidly mineralizing UMR-106-01 BSP osteoblasts formed focal adhesions; however, the level of FAK in UI focal adhesions was less than that seen in BSP cells. UI cultures had less FAK expression (p < 0.05) along with elevated levels of FAK phosphotyrosine in comparison to rapidly mineralizing BSP cultures. Mineralization decreased in a dose-dependent manner in response to Herbimycin A, a tyrosine kinase inhibitor. Overexpression of FAK in UI cells led to a fourfold increase in Cbfa1 gene expression (p < 0.02), and an increase in Cbfa1 protein expression. These results suggest that the integrin-associated tyrosine kinase FAK contributes to the regulation of the osteoblast differentiation in part through the regulation of Cbfa1 expression.
Collapse
Affiliation(s)
- Galen B Schneider
- Department of Prosthodontics, University of Iowa College of Dentistry, The University of Iowa, Iowa City, Iowa 52242-1001, USA.
| | | | | | | |
Collapse
|
24
|
Schofer MD, Veltum A, Theisen C, Chen F, Agarwal S, Fuchs-Winkelmann S, Paletta JRJ. Functionalisation of PLLA nanofiber scaffolds using a possible cooperative effect between collagen type I and BMP-2: impact on growth and osteogenic differentiation of human mesenchymal stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2011; 22:1753-62. [PMID: 21604139 PMCID: PMC3127010 DOI: 10.1007/s10856-011-4341-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 05/07/2011] [Indexed: 05/21/2023]
Abstract
Mesenchymal stem cell differentiation of osteoblasts is triggered by a series of signaling processes including integrin and bone morphogenetic protein (BMP), which therefore act in a cooperative manner. The aim of this study was to analyze whether these processes can be remodeled in an artificial poly-(L)-lactide acid (PLLA) based nanofiber scaffold. Matrices composed of PLLA-collagen type I or BMP-2 incorporated PLLA-collagen type I were seeded with human mesenchymal stem cells (hMSC) and cultivated over a period of 22 days, either under growth or osteoinductive conditions. During the course of culture, gene expression of alkaline phosphatase (ALP), osteocalcin (OC) and collagen I (COL-I) as well as Smad5 and focal adhesion kinase (FAK), two signal transduction molecules involved in BMP-2 or integrin signaling were analyzed. Furthermore, calcium and collagen I deposition, as well as cell densities and proliferation, were determined using fluorescence microscopy. The incorporation of BMP-2 into PLLA-collagen type I nanofibers resulted in a decrease in diameter as well as pore sizes of the scaffold. Mesenchymal stem cells showed better adherence and a reduced proliferation on BMP-containing scaffolds. This was accompanied by an increase in gene expression of ALP, OC and COL-I. Furthermore the presence of BMP-2 resulted in an upregulation of FAK, while collagen had an impact on the gene expression of Smad5. Therefore these different strategies can be combined in order to enhance the osteoblast differentiation of hMSC on PLLA based nanofiber scaffold. By doing this, different signal transduction pathways seem to be up regulated.
Collapse
Affiliation(s)
- Markus D. Schofer
- Department of Orthopedics, University Hospital of Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Anne Veltum
- Department of Orthopedics, University Hospital of Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Christina Theisen
- Department of Orthopedics, University Hospital of Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Fei Chen
- Department of Chemistry, University of Marburg, Hans-Meerwein-Straße, 35032 Marburg, Germany
| | - Seema Agarwal
- Department of Chemistry, University of Marburg, Hans-Meerwein-Straße, 35032 Marburg, Germany
| | - Susanne Fuchs-Winkelmann
- Department of Orthopedics, University Hospital of Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Department of Orthopedics, University Hospital of Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
25
|
Yang L, Wang NL, Cai GP. Maohuoside A promotes osteogenesis of rat mesenchymal stem cells via BMP and MAPK signaling pathways. Mol Cell Biochem 2011; 358:37-44. [PMID: 21698346 DOI: 10.1007/s11010-011-0918-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 06/07/2011] [Indexed: 11/29/2022]
Abstract
Osteoporosis is becoming a more prevalent health problem with the aging of the population around the world. Epimedium koreanum Nakai is one of the most used herbs in East Asia for curing osteoporosis, with its major ingredient, icariin, mostly explored by researchers. In this article, maohuoside A (MHA), a single isolated compound from the herb, was identified to be more potent than icariin in promoting osteogenesis of rat bone marrow-derived mesenchymal stem cells (rMSCs) (increasing by 16.6, 33.3, and 15.8% on D3, D7, and D11, respectively). Alkaline phosphatase (ALP) assay and calcium content measurement were assigned to quantify the promoted osteogenesis and alizarin red S (ARS) staining was conducted to visualize it. Quantitative real-time PCR (Q-PCR) was assayed to evaluate the mRNA expression of marker genes in osteogenesis and master regulators in BMP pathway. Moreover, PD98059 (PD) and SB203580 (SB), inhibitor of ERK1/2 and p38 MAPK pathway, were administered to assess the involvement of MAPK pathway in the promotion process. In conclusion, MHA pronouncedly enhanced the osteogenesis of rMSC, plausibly via the BMP and MAPK signaling pathways.
Collapse
Affiliation(s)
- Lei Yang
- School of Life Science, Tsinghua University, Beijing 100084, People's Republic of China
| | | | | |
Collapse
|
26
|
Alcantara EH, Lomeda RAR, Feldmann J, Nixon GF, Beattie JH, Kwun IS. Zinc deprivation inhibits extracellular matrix calcification through decreased synthesis of matrix proteins in osteoblasts. Mol Nutr Food Res 2011; 55:1552-60. [PMID: 21656670 DOI: 10.1002/mnfr.201000659] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2010] [Revised: 03/15/2011] [Accepted: 03/21/2011] [Indexed: 01/25/2023]
Abstract
SCOPE Zinc is implicated as an activator for bone formation, however, its influence on bone calcification has not been reported. This study examined how zinc regulates the bone matrix calcification in osteoblasts. METHODS AND RESULTS Two osteoblastic MC3T3-E1 cell subclones (SC 4 and SC 24 as high and low osteogenic differentiation, respectively) were cultured in normal osteogenic (OSM), Zinc deficient (Zn-, 1 μM), or adequate (Zn+, 15 μM) media up to 20 days. Cells (SC 4) were also supplemented with (50 μg/mL) or no ascorbic acid (AA) in combination with Zinc treatment. Zn- decreased collagen synthesis and matrix accumulation. Although AA is essential for collagen formation, its supplementation could not compensate for Zinc deficiency-induced detrimental effects on extracellular matrix mineralization. Zn- also decreased the medium and cell layer alkaline phosphatase ALP activity. This decreased ALP activity might cause the decrease of Pi accumulation in response to Zn-, as measured by von Kossa staining. Ca deposition in cell layers, measured by Alizarin red S staining, was also decreased by Zn(-) . CONCLUSION Our findings suggest that zinc deprivation inhibits extracellular matrix calcification in osteoblasts by decreasing the synthesis and activity of matrix proteins, type I collagen and ALP, and decreasing Ca and Pi accumulation. Therefore zinc deficiency can be considered as risk factor for poor extracellular matrix calcification.
Collapse
Affiliation(s)
- Ethel H Alcantara
- Department of Food Science and Nutrition, Andong National University, 388 Songchundong,Andong, Kyungbook, South Korea
| | | | | | | | | | | |
Collapse
|
27
|
Anderson JM, Vines JB, Patterson JL, Chen H, Javed A, Jun HW. Osteogenic differentiation of human mesenchymal stem cells synergistically enhanced by biomimetic peptide amphiphiles combined with conditioned medium. Acta Biomater 2011; 7:675-82. [PMID: 20728586 PMCID: PMC2999640 DOI: 10.1016/j.actbio.2010.08.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 08/12/2010] [Accepted: 08/17/2010] [Indexed: 12/18/2022]
Abstract
An attractive strategy for bone tissue engineering is the use of extracellular matrix (ECM) analogous biomaterials capable of governing biological response based on synthetic cell-ECM interactions. In this study, peptide amphiphiles (PAs) were investigated as an ECM-mimicking biomaterial to provide an instructive microenvironment for human mesenchymal stem cells (hMSCs) in an effort to guide osteogenic differentiation. PAs were biologically functionalized with ECM isolated ligand sequences (i.e. RGDS, DGEA), and the osteoinductive potential was studied with or without conditioned medium, containing the supplemental factors of dexamethasone, β-glycerol phosphate and ascorbic acid. It was hypothesized that the ligand-functionalized PAs would synergistically enhance osteogenic differentiation in combination with conditioned medium. Concurrently, comparative evaluations independent of osteogenic supplements investigated the differentiating potential of the functionalized PA scaffolds as promoted exclusively by the inscribed ligand signals, thus offering the potential for therapeutic effectiveness under physiological conditions. Osteoinductivity was assessed by histochemical staining for alkaline phosphatase (ALP) and quantitative real-time polymerase chain reaction analysis of key osteogenic markers. Both of the ligand-functionalized PAs were found to synergistically enhance the level of visualized ALP activity and osteogenic gene expression compared to the control surfaces lacking biofunctionality. Guided osteoinduction was also observed without supplemental aid on the PA scaffolds, but at a delayed response and not to the same phenotypic levels. Thus, the biomimetic PAs foster a symbiotic enhancement of osteogenic differentiation, demonstrating the potential of ligand-functionalized biomaterials for future bone tissue repair.
Collapse
Affiliation(s)
- Joel M. Anderson
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy B. Vines
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessica L. Patterson
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Haiyan Chen
- Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amjad Javed
- Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
28
|
Shekaran A, García AJ. Extracellular matrix-mimetic adhesive biomaterials for bone repair. J Biomed Mater Res A 2010; 96:261-72. [PMID: 21105174 DOI: 10.1002/jbm.a.32979] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/07/2010] [Accepted: 09/08/2010] [Indexed: 01/12/2023]
Abstract
Limited osseointegration of current orthopedic biomaterials contributes to the failure of implants such as arthroplasties, bone screws, and bone grafts, which present a large socioeconomic cost within the United States. These implant failures underscore the need for biomimetic approaches that modulate host cell-implant material responses to enhance implant osseointegration and bone formation. Bioinspired strategies have included functionalizing implants with extracellular matrix (ECM) proteins or ECM-derived peptides or protein fragments, which engage integrins and direct osteoblast adhesion and differentiation. This review discusses (1) bone ECM composition and key integrins implicated in osteogenic differentiation, (2) the use of implants functionalized with ECM-mimetic peptides/protein fragments, and (3) growth factor-derived peptides to promote the mechanical fixation of implants to bone and to enhance bone healing within large defects.
Collapse
Affiliation(s)
- Asha Shekaran
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | |
Collapse
|
29
|
Lu Z, Zreiqat H. The Osteoconductivity of Biomaterials Is Regulated by Bone Morphogenetic Protein 2 Autocrine Loop Involving α2β1 Integrin and Mitogen-Activated Protein Kinase/Extracellular Related Kinase Signaling Pathways. Tissue Eng Part A 2010; 16:3075-84. [DOI: 10.1089/ten.tea.2010.0204] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- ZuFu Lu
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney, Sydney, Australia
| |
Collapse
|
30
|
Fernandes H, Dechering K, Van Someren E, Steeghs I, Apotheker M, Leusink A, Bank R, Janeczek K, Van Blitterswijk C, de Boer J. The role of collagen crosslinking in differentiation of human mesenchymal stem cells and MC3T3-E1 cells. Tissue Eng Part A 2010; 15:3857-67. [PMID: 19694522 DOI: 10.1089/ten.tea.2009.0011] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Collagen is the main protein component of the extracellular matrix of bone, and it has structural and instructive properties. Collagen undergoes many post-translational modifications, including extensive crosslinking. Although defective crosslinking has been implicated in human syndromes (e.g., osteogenesis imperfecta or Ehlers-Danlos syndrome), it is not clear to what extent crosslinking is necessary for collagen's instructive properties during bone formation. Here we report that inhibition of collagen crosslinking in the mouse pre-osteoblast cell line MC3T3-E1 impairs the osteogenic program. Genome-wide expression profiling of beta-aminopropionitrile-treated and control cells revealed that matrix deposition by MC3T3-E1 cells provides a feed back signal, driving cells through the differentiation process, that is strongly impaired when crosslinking is inhibited. Inhibition of crosslinking did not affect osteogenic differentiation of human mesenchymal stem cells (hMSCs), shown by the expression of alkaline phosphatase and genome-wide gene expression analysis, although it enhances matrix mineralization. In conclusion, collagen crosslinking harbors instructive properties in MC3T3-E1 differentiation but plays a more-passive role in differentiation of bone marrow-derived hMSCs.
Collapse
Affiliation(s)
- Hugo Fernandes
- Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Johnson MD, O'Connell MJ, Vito F, Pilcher W. Bone morphogenetic protein 4 and its receptors are expressed in the leptomeninges and meningiomas and signal via the Smad pathway. J Neuropathol Exp Neurol 2009; 68:1177-83. [PMID: 19816200 DOI: 10.1097/nen.0b013e3181bc6642] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The roles of bone morphogenetic proteins (BMPs) and their receptors (BMPRs) in meningioma biology are not known. In this study, frozen tissues from 26 World Health Organization Grades I to III meningiomas were analyzed by Western blot for BMP-2/4, BMPR IA, and BMPR II, and activation of downstream p-Smad1, p38 mitogen-activated protein kinase (MAPK), and p44/42 MAPK signaling molecules. Sections from 20 normal leptomeninges, 2 arachnoid cysts, and 51 meningiomas were analyzed for BMP-4 and p44/42 MAPK by immunohistochemistry. Primary meningioma cultures from 11 meningiomas were treated with BMP-4 and evaluated for cell proliferation and signaling pathway activation. Conditioned media from 7 cultures were analyzed for BMP-4 by ELISA. Bone morphogenetic protein 4 was variably detected in adult leptomeninges but was detected in 89% or 84% of Grade I meningiomas and in 60% of Grade II meningiomas by Western blot and immunohistochemistry, respectively. Bone morphogenetic protein receptors IA and II were detected in leptomeninges and in all meningiomas studied, and activated Smad1 was detected in all meningiomas studied. Bone morphogenetic protein 4 stimulated meningioma cell proliferation and phosphorylation/activation of Smad1 but not p38 MAPK or p44/42 MAPK in vitro, and it was detected in conditioned media from 4 of 7 cultures. These findings suggest that BMP-4 and BMPRs may play autocrine/paracrine roles and interact with other transforming growth factor-beta superfamily members in regulating meningioma growth and differentiation.
Collapse
Affiliation(s)
- Mahlon D Johnson
- Department of Pathology, Division of Neuropathology and Neurosurgery, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | | | |
Collapse
|
32
|
Watanabe-Takano H, Takano K, Keduka E, Endo T. M-Ras is activated by bone morphogenetic protein-2 and participates in osteoblastic determination, differentiation, and transdifferentiation. Exp Cell Res 2009; 316:477-90. [PMID: 19800879 DOI: 10.1016/j.yexcr.2009.09.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 09/26/2009] [Accepted: 09/28/2009] [Indexed: 11/19/2022]
Abstract
The small GTPase M-Ras is highly expressed in the central nervous system and plays essential roles in neuronal differentiation. However, its other cellular and physiological functions remain to be elucidated. Here, we clarify the novel functions of M-Ras in osteogenesis. M-Ras was prominently expressed in developing mouse bones particularly in osteoblasts and hypertrophic chondrocytes. Its expression was elevated in C3H/10T1/2 (10T1/2) mesenchymal cells and in MC3T3-E1 preosteoblasts during differentiation into osteoblasts. Treatment of C2C12 skeletal muscle myoblasts with bone morphogenetic protein-2 (BMP-2) to bring about transdifferentiation into osteoblasts also induced M-Ras mRNA and protein expression. Moreover, the BMP-2 treatment activated the M-Ras protein. Stable expression of the constitutively active M-Ras(G22V) in 10T1/2 cells facilitated osteoblast differentiation. M-Ras(G22V) also induced transdifferentiation of C2C12 cells into osteoblasts. In contrast, knockdown of endogenous M-Ras by RNAi interfered with osteoblast differentiation in 10T1/2 and MC3T3-E1 cells. Osteoblast differentiation in M-Ras(G22V)-expressing C2C12 cells was inhibited by treatment with inhibitors of p38 MAP kinase (MAPK) and c-Jun N-terminal kinase (JNK) but not by inhibitors of MAPK and ERK kinase (MEK) or phosphatidylinositol 3-kinase. These results imply that M-Ras, induced and activated by BMP-2 signaling, participates in the osteoblastic determination, differentiation, and transdifferentiation under p38 MAPK and JNK regulation.
Collapse
Affiliation(s)
- Haruko Watanabe-Takano
- Department of Biology, Graduate School of Science, Chiba University, Inageku, Chiba 263-8522, Japan
| | | | | | | |
Collapse
|
33
|
Dore JJ, DeWitt JC, Setty N, Donald MD, Joo E, Chesarone MA, Birren SJ. Multiple signaling pathways converge to regulate bone-morphogenetic-protein-dependent glial gene expression. Dev Neurosci 2009; 31:473-86. [PMID: 19325229 DOI: 10.1159/000210187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Accepted: 01/12/2009] [Indexed: 12/22/2022] Open
Abstract
A fundamental problem in developmental neuroscience is understanding how extracellular cues link to complex intracellular signaling pathways to drive stage-specific developmental decisions. During the formation of the mammalian peripheral nervous system, bone morphogenetic proteins (BMPs) promote neuronal differentiation. BMPs also maintain the expression of early glial genes such as GFAP, while blocking the acquisition of a mature, myelinating Schwann cell phenotype. We investigated the BMP-activated signaling pathways that contribute to early glial gene expression to address the question of how specific signaling interactions contribute to cell fate decisions in neural crest lineages. Using a neural-crest-derived cell line that exhibits the characteristics of immature Schwann cells, we found that BMP2 promotes GFAP expression using Smad signaling as well as the phosphoinositide-3 kinase (PI3K) and mitogen-activated protein kinase1/2extracellular signal-regulated kinase- (MEK1/2/ERK) pathways. The GFAP promoter does not contain known Smad consensus sites, suggesting that Smads may act indirectly to promote GFAP expression. We provide evidence that this indirect effect may be mediated via induction of immediate early genes and the transcription factor Sp1 by demonstrating that these transcriptional regulators are induced by BMP2 and contribute to GFAP promoter activity. These findings demonstrate new roles for intracellular kinase pathways in mediating the effects of BMPs during the early stages of glial differentiation and suggest that differential contributions by signaling and transcriptional networks may contribute to the range of effects of BMPs on neuronal and glial development during the formation of the peripheral nervous system.
Collapse
Affiliation(s)
- Justin J Dore
- Department of Biology and Volen National Center for Complex Systems, Brandeis University, Waltham, Mass. 02454, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Vohra S, Hennessy KM, Sawyer AA, Zhuo Y, Bellis SL. Comparison of mesenchymal stem cell and osteosarcoma cell adhesion to hydroxyapatite. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2008; 19:3567-74. [PMID: 18626747 PMCID: PMC3677517 DOI: 10.1007/s10856-008-3525-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2007] [Accepted: 06/25/2008] [Indexed: 05/24/2023]
Abstract
Immortalized cells are often used to model the behavior of osteogenic cells on orthopaedic and dental biomaterials. In the current study we compared the adhesive behavior of two osteosarcoma cell lines, MG-63 and Saos-2, with that of mesenchymal stem cells (MSCs) on hydroxyapatite (HA). It was found that osteosarcoma cells demonstrated maximal binding to fibronectin-coated HA, while MSCs alternately preferred HA coated with collagen-I. Interesting, the binding of MG-63 and Saos-2 cells to fibronectin was mediated by both alpha5 and alphav-containing integrin heterodimers, whereas only alphav integrins were used by MSCs. Cell spreading was also markedly different for the three cell types. Osteosarcoma cells exhibited optimal spreading on fibronectin, but poor spreading on HA disks coated with fetal bovine serum. In contrast, MSCs spread very well on serum-coated surfaces, but less extensively on fibronectin. Finally, we evaluated integrin expression and found that MSCs have higher levels of alpha2 integrin subunits relative to MG-63 or Saos-2 cells, which may explain the enhanced adhesion of MSCs on collagen-coated HA. Collectively our results suggest that osteosarcoma cells utilize different mechanisms than MSCs during initial attachment to protein-coated HA, thereby calling into question the suitability of these cell lines as in vitro models for cell/biomaterial interactions.
Collapse
Affiliation(s)
- Shikhar Vohra
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | |
Collapse
|
35
|
Insulin potentiates the proliferation and bone morphogenetic protein-2-induced osteogenic differentiation of rat spinal ligament cells via extracellular signal-regulated kinase and phosphatidylinositol 3-kinase. Spine (Phila Pa 1976) 2008; 33:2394-402. [PMID: 18923314 DOI: 10.1097/brs.0b013e3181838fe5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN This study was designed to confirm the correlation of hyperinsulinemia with ossification of posterior longitudinal ligament (OPLL) of the cervical spine in vitro. OBJECTIVE To investigate the effects of insulin on the proliferation, collagen synthesis, and osteogenic differentiation of isolated rat spinal ligament cell in the presence or absence of bone morphogenetic protein-2 (BMP-2). SUMMARY OF BACKGROUND DATA Noninsulin-dependent diabetes mellitus is an independent risk factor for the onset of OPLL, but the mechanism is still unknown. We have hypothesized that insulin may exert direct effects on the proliferation and osteogenic differentiation of spinal ligament cells. METHODS Cells isolated from rat spinal ligaments were stimulated by different concentrations of insulin in the presence or absence of BMP-2. The proliferation of the cell was measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromid (MTT) and direct cell counting and the procollagen type I amino-terminal peptide was measured by enzyme-linked immunosorbent assay. The gene expressions of insulin receptor, alkaline phosphatase, osteocalcin and Runx2 were examined by quantitative reverse transcriptase-polymerase chain reaction. PI3-K/Akt and extracellular signal-regulated kinase (ERK) were analyzed by western blotting. RESULTS Insulin positively regulated the expression of its receptor in the cells and stimulated the proliferation of the cells in a time- and dose-dependent manner. Insulin alone did not affect the synthesis of procollagen type I amino-terminal peptide synthesis or osteogenesis differentiation in the cells. However, high concentration of insulin (1000 nmol/L) significantly promoted BMP-2-induced alkaline phosphatase expression and activation, which was associated with the up-regulation of PI3-K/Akt and down-regulation of ERK in the cells. CONCLUSION Hyperinsulinemia may contribute to the onset and progression of OPLL in subjects with noninsu-lin-dependent diabetes mellitus by stimulating the proliferation and BMP-2-induced osteogenic differentiation of ligament cells via the activation of insulin receptor and PI3-K/Akt pathway and the suppression of ERK.
Collapse
|
36
|
Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from the laboratory to the clinic, part I (basic concepts). J Tissue Eng Regen Med 2008; 2:1-13. [PMID: 18293427 DOI: 10.1002/term.63] [Citation(s) in RCA: 230] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Discovered in 1965, bone morphogenetic proteins (BMPs) are a group of cytokines from the transforming growth factor-beta (TGFbeta) superfamily with significant roles in bone and cartilage formation. BMPs are used as powerful osteoinductive components of diverse tissue-engineering products for the healing of bone. Several BMPs with different physiological roles have been identified in humans. The purpose of this review is to cover the biological function of the main members of BMP family, the latest research on BMPs signalling pathways and advances in the production of recombinant BMPs for tissue engineering purposes.
Collapse
Affiliation(s)
- P C Bessa
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | | | | |
Collapse
|
37
|
Boehrs J, Zaharias RS, Laffoon J, Ko YJ, Schneider GB. Three-Dimensional Culture Environments Enhance Osteoblast Differentiation. J Prosthodont 2008; 17:517-21. [DOI: 10.1111/j.1532-849x.2008.00330.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
38
|
Reyes CD, Petrie TA, García AJ. Mixed extracellular matrix ligands synergistically modulate integrin adhesion and signaling. J Cell Physiol 2008; 217:450-8. [PMID: 18613064 DOI: 10.1002/jcp.21512] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cell adhesion to extracellular matrix (ECM) components through cell-surface integrin receptors is essential to the formation, maintenance and repair of numerous tissues, and therefore represents a central theme in the design of bioactive materials that successfully interface with the body. While the adhesive responses associated with a single ligand have been extensively analyzed, the effects of multiple integrin subtypes binding to multivalent ECM signals remain poorly understood. In the present study, we generated a high throughput platform of non-adhesive surfaces presenting well-defined, independent densities of two integrin-specific engineered ligands for the type I collagen (COL-I) receptor alpha(2)beta(1) and the fibronectin (FN) receptor alpha(5)beta(1) to evaluate the effects of integrin cross-talk on adhesive responses. Engineered surfaces displayed ligand density-dependent adhesive effects, and mixed ligand surfaces significantly enhanced cell adhesion strength and focal adhesion assembly compared to single FN and COL-I ligand surfaces. Moreover, surfaces presenting mixed COL-I/FN ligands synergistically enhanced FAK activation compared to the single ligand substrates. The enhanced adhesive activities of the mixed ligand surfaces also promoted elevated proliferation rates. Our results demonstrate interplay between multivalent ECM ligands in adhesive responses and downstream cellular signaling.
Collapse
Affiliation(s)
- Catherine D Reyes
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | | | |
Collapse
|
39
|
Ko YJ, Zaharias RS, Seabold DA, Lafoon J, Schneider GB. Osteoblast differentiation is enhanced in rotary cell culture simulated microgravity environments. J Prosthodont 2007; 16:431-8. [PMID: 17559538 DOI: 10.1111/j.1532-849x.2007.00204.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
PURPOSE As the aging population increases, more people will become reliant on regenerative dental medicine for implant therapy. The objective of this study was to test the hypothesis that 3D rotary cell culture (RCC) environments created by simulated microgravity would enhance osteogenic gene expression using integrin mediated pathways. MATERIALS AND METHODS Human embryonic palatal mesenchymal (HEPM, ATCC 1486) pre-osteoblasts were cultured in either RCC to create 3D environments or in 2D monolayers for 72 hours. Gross phenotypic analysis was performed using Alizarin Red S staining for calcium and microscopy. Real-time PCR analysis was used to detect differences in osteoblast gene expression. Aggregates developed in 3D RCC environments were treated with or without antibody to the collagen-I integrin receptor alpha2beta1 to determine whether this molecular pathway might contribute to the development of a mineralized matrix. RESULTS Microscopic analysis demonstrated that RCC environments promoted 3D aggregate formation by 72 hours without any scaffold. The mass appeared osseous-like with a white, shiny, translucent surface. The center was amorphous with areas of vacuolization, tubule-like structures, and fibrous-like extensions. Real-time PCR data showed that 3D environments enhanced osteogenic gene expression as compared with 2D monolayer culturing conditions. At 72 hours, changes in levels of osteogenic gene expression were noted. Cbfa1, a necessary transcription factor for osteoblast differentiation, was expressed 33% higher (p= 0.26); Collagen 1, 69% higher (p= 0.05); Osterix, 49% higher (p= 0.001); and BSPII, 54% higher (p= 0.001) than osteoblasts cultured for 72 hours in standard 2D monolayer conditions. When cultured in the presence of collagen alpha2beta1 integrin receptor antibody, 3D aggregates had decreased levels of mineralization as compared with non-treated aggregates. CONCLUSION RCC enhances osteoblast differentiation using integrin mediated pathways.
Collapse
Affiliation(s)
- Y Joon Ko
- Department of Prosthodontics, College of Dentistry, University of Iowa, Iowa City, IA, USA
| | | | | | | | | |
Collapse
|
40
|
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: 227] [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
|
41
|
Ge C, Xiao G, Jiang D, Franceschi RT. Critical role of the extracellular signal-regulated kinase-MAPK pathway in osteoblast differentiation and skeletal development. ACTA ACUST UNITED AC 2007; 176:709-18. [PMID: 17325210 PMCID: PMC2064027 DOI: 10.1083/jcb.200610046] [Citation(s) in RCA: 398] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) pathway provides a major link between the cell surface and nucleus to control proliferation and differentiation. However, its in vivo role in skeletal development is unknown. A transgenic approach was used to establish a role for this pathway in bone. MAPK stimulation achieved by selective expression of constitutively active MAPK/ERK1 (MEK-SP) in osteoblasts accelerated in vitro differentiation of calvarial cells, as well as in vivo bone development, whereas dominant-negative MEK1 was inhibitory. The involvement of the RUNX2 transcription factor in this response was established in two ways: (a) RUNX2 phosphorylation and transcriptional activity were elevated in calvarial osteoblasts from TgMek-sp mice and reduced in cells from TgMek-dn mice, and (b) crossing TgMek-sp mice with Runx2+/- animals partially rescued the hypomorphic clavicles and undemineralized calvaria associated with Runx2 haploinsufficiency, whereas TgMek-dn; Runx2+/- mice had a more severe skeletal phenotype. This work establishes an important in vivo function for the ERK-MAPK pathway in bone that involves stimulation of RUNX2 phosphorylation and transcriptional activity.
Collapse
Affiliation(s)
- Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
42
|
Reyes CD, Petrie TA, Burns KL, Schwartz Z, García AJ. Biomolecular surface coating to enhance orthopaedic tissue healing and integration. Biomaterials 2007; 28:3228-35. [PMID: 17448533 PMCID: PMC2034748 DOI: 10.1016/j.biomaterials.2007.04.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2007] [Accepted: 04/02/2007] [Indexed: 11/22/2022]
Abstract
Implant osseointegration is a prerequisite for clinical success in orthopaedic and dental applications, many of which are restricted by loosening. Biomaterial surface modification approaches, including calcium-phosphate ceramic coatings and macro/microporosity, have had limited success in promoting integration. To improve osseointegration, titanium surfaces were coated with the glycine-phenylalanine-hydroxyproline-glycine-glutamate-arginine (GFOGER) collagen-mimetic peptide, selectively promoting alpha2beta1 integrin binding, a crucial event for osteoblastic differentiation. Titanium surfaces presenting GFOGER triggered osteoblastic differentiation and mineral deposition in bone marrow stromal cells, leading to enhanced osteoblastic function compared to unmodified titanium. Furthermore, this integrin-targeted coating significantly improved in vivo peri-implant bone regeneration and osseointegration, as characterized by bone-implant contact and mechanical fixation, compared to untreated titanium in a rat cortical bone-implant model. GFOGER-modified implants also significantly enhanced osseointegration compared to surfaces modified with full-length type I collagen, highlighting the importance of presenting specific biofunctional domains within the native ligand. In addition, this biomimetic implant coating is generated using a simple, single-step procedure that readily translates to a clinical environment with minimal processing and cytotoxicity concerns. Therefore, this study establishes a biologically active and clinically relevant implant-coating strategy that enhances bone repair and orthopaedic implant integration.
Collapse
Affiliation(s)
- Catherine D Reyes
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
43
|
Hayami T, Zhang Q, Kapila Y, Kapila S. Dexamethasone's enhancement of osteoblastic markers in human periodontal ligament cells is associated with inhibition of collagenase expression. Bone 2007; 40:93-104. [PMID: 16934542 DOI: 10.1016/j.bone.2006.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 07/05/2006] [Accepted: 07/09/2006] [Indexed: 11/26/2022]
Abstract
Although dexamethasone (Dex) substantially enhances the osteoblastic phenotype in osteogenic cells, including human periodontal ligament (PDL) cells, the basis for this response remains poorly understood. Since the accretion of a collagenous matrix is important for an osteoblastic response and dexamethasone is known to decrease collagenase expression, we examined whether osteoblastic differentiation mediated by Dex is linked to a decrease in collagenase expression in PDL cells. Early passage human PDL cells were exposed to Dex, or ascorbic acid (AA) or beta-glycerophosphate (betaGP) alone, or in various combinations in serum-free media for 3 or 5 days. Cells exposed to Dex alone or any combinations of treatments that included Dex demonstrated increased core binding factor alpha 1 (Cbfa1), alkaline phosphatase (AP), osteonectin (ON), osteopontin (OP), bone sialoprotein (BSP) and collagen I (alpha1) expression when compared to control cells or those exposed to AA or betaGP. The induction of these osteoblastic markers was accompanied by a decrease in collagenase-1 expression. Collagenase activity showed a statistically significant strong negative relationship to Cbfa1 (Pearson's r=-0.97), AP (r=-0.87), OP (r=-0.95) and BSP (r=-0.82) in 5-day cultures, and moderately strong relationship to ON (r=-0.74) from 3 days culture. Dex also produced a dose-dependent increase in AP that was paralleled by a decrease in collagenase activity (r=-0.98). Addition of collagenase inhibitors increased AP expression while concomitantly suppressing collagenase activity. Conversely, addition of exogenous collagenase decreased the AP phenotype of the cells, which was more marked in the absence then in the presence of Dex. The findings indicate that Dex enhances specific markers of osteoblastic differentiation in PDL cells by decreasing collagenase expression, and suggest that endogenous collagenase may regulate osteoblastic differentiation of these cells.
Collapse
Affiliation(s)
- Takayuki Hayami
- University of Michigan, 1011 North University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Aaron Schindeler
- Department of Orthopaedic Research and Biotechnology, The Children's Hospital at Westmeade, Sydney, Australia.
| | | |
Collapse
|
45
|
Abstract
Many serious adverse physiological changes occur during spaceflight. In the search for underlying mechanisms and possible new countermeasures, many experimental tools and methods have been developed to study microgravity caused physiological changes, ranging from in vitro bioreactor studies to spaceflight investigations. Recently, genomic and proteomic approaches have gained a lot of attention; after major scientific breakthroughs in the fields of genomics and proteomics, they are now widely accepted and used to understand biological processes. Understanding gene and/or protein expression is the key to unfolding the mechanisms behind microgravity-induced problems and, ultimately, finding effective countermeasures to spaceflight-induced alterations. Significant progress has been made in identifying the genes/proteins responsible for these changes. Although many of these genes and/or proteins were observed to be either upregulated or downregulated, however, no large-scale genomics and proteomics studies have been published so far. This review aims at summarizing the current status of microgravity-related genomics and proteomics studies and stimulating large-scale proteomics and genomics research activities.
Collapse
Affiliation(s)
- Heather L Nichols
- Clemson-Medical University of South Carolina Bioengineering Program, Department of Bioengineering, Clemson University, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
46
|
Hartung A, Sieber C, Knaus P. Yin and Yang in BMP signaling: Impact on the pathology of diseases and potential for tissue regeneration. ACTA ACUST UNITED AC 2006. [DOI: 10.1002/sita.200600098] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
47
|
Woo KM, Jun JH, Chen VJ, Seo J, Baek JH, Ryoo HM, Kim GS, Somerman MJ, Ma PX. Nano-fibrous scaffolding promotes osteoblast differentiation and biomineralization. Biomaterials 2006; 28:335-43. [PMID: 16854461 DOI: 10.1016/j.biomaterials.2006.06.013] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2006] [Accepted: 06/13/2006] [Indexed: 01/05/2023]
Abstract
Nano-fibrous poly(L-lactic acid) (PLLA) scaffolds with interconnected pores were developed under the hypothesis that nano-fibrous scaffolding would mimic a morphological function of collagen fibrils to create a more favorable microenvironment for cells versus solid-walled scaffolds. In this study, an in vitro system was used to examine biological properties of the nano-fibrous scaffolds compared with those of solid-walled scaffolds for their potential use in bone tissue engineering. Biomineralization was enhanced substantially on the nano-fibrous scaffolds compared to solid-walled scaffolds, and this was confirmed by von Kossa staining, measurement of calcium contents, and transmission electron microscopy. In support of this finding, osteoblasts cultured on the nano-fibrous scaffolds exhibited higher alkaline phosphatase activity and an earlier and enhanced expression of the osteoblast phenotype versus solid-walled scaffolds. Most notable were the increases in runx2 protein and in bone sialoprotein mRNA in cells cultured on nano-fibrous scaffolds versus solid-walled scaffolds. At the day 1 of culture, alpha2 and beta1 integrins as well as alphav and beta3 integrins were highly expressed on the surface of cells seeded on nano-fibrous scaffolds, and linked to this were higher levels of phospho-Paxillin and phospho-FAK in cell lysates. In contrast, cells seeded on solid-walled scaffolds expressed significantly lower levels of these integrins, phospho-Paxillin, and phospho-FAK. To further examine the role of nano-fibrous architecture, we inhibited the formation of collagen fibrils by adding 3,4-dehydroproline to cultures and then assayed cells for expression of alpha2 integrin. Cells seeded on nano-fibrous scaffolds sustained expression of alpha2 integrin in the presence of dehydroproline, while suppression of alpha2 integrin was evident in cells seeded on solid-walled scaffolds. These results provide initial evidence that synthetic nano fibers may exhibit certain properties that are comparable to natural collagen fibers, and thus, the nano-fibrous architecture may serve as a superior scaffolding versus solid-walled architecture for promoting osteoblast differentiation and biomineralization.
Collapse
Affiliation(s)
- Kyung Mi Woo
- School of Dentistry, Department of Cell and Developmental Biology, Seoul National University, Seoul 110-740, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Moradian-Oldak J, Wen HB, Schneider GB, Stanford CM. Tissue engineering strategies for the future generation of dental implants. Periodontol 2000 2006; 41:157-76. [PMID: 16686932 DOI: 10.1111/j.1600-0757.2006.00153.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Janet Moradian-Oldak
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, California, USA
| | | | | | | |
Collapse
|
49
|
Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev 2006; 16:251-63. [PMID: 15871923 DOI: 10.1016/j.cytogfr.2005.01.009] [Citation(s) in RCA: 648] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2004] [Accepted: 01/20/2005] [Indexed: 12/25/2022]
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-beta (TGF-beta) superfamily, bind to two different serine/threonine kinase receptors, and mediate their signals through Smad-dependent and Smad-independent pathways. Receptor regulated-Smad (R-Smad) proteins specific for the BMP pathways interact with various proteins, including transcription factor Runx, and transmit specific signals in target cells. The recent development of DNA microarray techniques has allowed us to identify many BMP target genes. BMP signaling is modulated by various molecules, including inhibitory Smads (I-Smads). Moreover, recent findings have revealed that BMP pathways interact with other signaling pathways, and such signaling cross-talk plays pivotal roles in growth and differentiation of target cells.
Collapse
Affiliation(s)
- Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | | | | |
Collapse
|
50
|
Langenfeld EM, Kong Y, Langenfeld J. Bone morphogenetic protein 2 stimulation of tumor growth involves the activation of Smad-1/5. Oncogene 2006; 25:685-92. [PMID: 16247476 DOI: 10.1038/sj.onc.1209110] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Morphogenetic protein 2 (BMP-2) is normally expressed in the embryo promoting the development of several organs. Aberrant expression of BMP-2 occurs in approximately 98% of lung carcinomas, however, its role in regulating tumor growth is poorly understood. We show that BMP-2 induces Id-1 expression in lung cancer cell lines through its activation of Smad-1/5, which is dependent on cell culture conditions. A549 cells in DMEM/5% FCS BMP-2 activated Smad-1/5 and caused a transient increase in proliferation. In serum-free medium, BMP-2 induced significantly less Smad-1/5 activation and Id-1 expression, and produced significant growth inhibition. The affect of BMP-2 on tumor growth in vivo was substantially more significant. Recombinant BMP-2 coinjected with A549 cells, into nude mice increased proliferation and produced an increase in Id-1 expression. Forced expression of BMP-2 in A549 cells significantly enhanced tumor growth in the lungs following intravenous injection but not of subcutaneous tumors. Tumors in the lung were found to have an activated Smad-1/5 and expressed Id-1. Subcutaneous tumors expressed less activated Smad-1/5 and Id-1 than that of controls. Human lung carcinomas were also found to express an activated Smad-1/5 and Id-1. We provide evidence that BMP-2 promotes tumor growth. This paper highlights that cell culture experiments may not reveal the full biological affects of BMP-2, and its activity varies depending of the local environment.
Collapse
Affiliation(s)
- E M Langenfeld
- Division of Thoracic Surgery, Department of Surgery, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ 08903-0019, USA
| | | | | |
Collapse
|