1
|
Li W, Huang Y, Gu D, Peng S, Zhang B, Peng F, Zhang D, Li M, Xiao J, Jia Z, Qiu L. Ascorbate-loaded MgFe layered double hydroxide for osteomyelitis treatment. J Control Release 2025; 378:1045-1060. [PMID: 39740696 DOI: 10.1016/j.jconrel.2024.12.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Bacterial infections evoke considerable apprehension in orthopedics. Traditional antibiotic treatments exhibit cytotoxic effects and foster bacterial resistance, thereby presenting an ongoing and formidable obstacle in the realm of therapeutic interventions. Achieving bacterial eradication and osteogenesis are critical requirements for bone infection treatment. Herein, we design and fabricate a nanoenzyme-mimicking drug through the co-precipitation process, integrating MgFe layered double hydroxide with ascorbic acid (AA@LDH), to facilitate the simultaneous presence of these two unique functionalities. Within a bacterial acidic milieu, the degradation of the AA@LDH nanosystem prompts ascorbic acid to undergo a pro-oxidative transformation, generating an abundance of reactive oxygen species (ROS). These ROS overwhelm bacterial cellular processes, including nucleic acid replication, cell wall construction, virulence factor production, biosynthetic pathways, and energy generation. This disruption culminates in substantial bacterial mortality, as substantiated by RNA sequencing data. Hence, the AA@LDH nano system exhibits an in vitro antibacterial rate of approximately 100 % and 99 %, against S.aureus and E. coli, respectivaly. Additionally, the AA@LDH could directly accelerate osteogenic differentiation in vitro, evidenced by a 50 % increase in alkaline phosphatase activity and a 270 % improvement in extracellular matrix mineralization capability. Moreover, it enhances osteointegration process in vivo by favorably reshaping the osteogenic immune microenvironment. This innovative nanosystem for delivery offers new strategies that concurrently combat bacterial infections, mitigate inflammation, and induce tissue regeneration, marking a significant advancement in the realm of advanced materials and its applications.
Collapse
Affiliation(s)
- Wei Li
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yuliang Huang
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Dongqiang Gu
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Sijun Peng
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Bao Zhang
- Department of Orthopedics, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Feng Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Dongdong Zhang
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mingjun Li
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Jin Xiao
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Zhiwei Jia
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, China.
| | - Longhai Qiu
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China.
| |
Collapse
|
2
|
Hudacova E, Abaffy P, Kaplan MM, Krausova M, Kubista M, Machon O. Single-cell transcriptomic resolution of osteogenesis during craniofacial morphogenesis. Bone 2025; 190:117297. [PMID: 39461490 DOI: 10.1016/j.bone.2024.117297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Craniofacial morphogenesis depends on complex cell fate decisions during the differentiation of post-migratory cranial neural crest cells. Molecular mechanisms of cell differentiation of mesenchymal cells to developing bones, cartilage, teeth, tongue, and other craniofacial tissues are still poorly understood. We performed single-cell transcriptomic analysis of craniofacial mesenchymal cells derived from cranial NCCs in mouse embryo. Using FACS sorting of Wnt1-Cre2 progeny, we carefully mapped the cell heterogeneity in the craniofacial region during the initial stages of cartilage and bone formation. Transcriptomic data and in vivo validations identified molecular determinants of major cell populations involved in the development of lower and upper jaw, teeth, tongue, dermis, or periocular mesenchyme. Single-cell transcriptomic analysis of Meis2-deficient mice revealed critical gene expression differences, including increased osteogenic and cell adhesion markers. This leads to affected mesenchymal cell differentiation and increased ossification, resulting in impaired bone, cartilage, and tongue formation.
Collapse
Affiliation(s)
- Erika Hudacova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 12000 Prague, Czech Republic.
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Mehmet Mahsum Kaplan
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Michaela Krausova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Ondrej Machon
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| |
Collapse
|
3
|
Tricou LP, Mouton W, Cara A, Trouillet-Assant S, Bouvard D, Laurent F, Diot A, Josse J. Staphylococcus aureus can use an alternative pathway to be internalized by osteoblasts in absence of β1 integrins. Sci Rep 2024; 14:28643. [PMID: 39562631 PMCID: PMC11576967 DOI: 10.1038/s41598-024-78754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Staphylococcus aureus main internalization mechanism in osteoblasts relies on a tripartite interaction between bacterial fibronectin-binding proteins, extracellular matrix soluble fibronectin, and osteoblasts' β1 integrins. Caveolins, and particularly caveolin-1, have been shown to limit the plasma membrane microdomain mobility, and consequently reduce the uptake of S. aureus in keratinocytes. In this study, we aimed to deepen our understanding of the molecular mechanisms underlying S. aureus internalization in osteoblasts. Mechanistically, S. aureus internalization requires endosomal recycling of β1 integrins as well as downstream effectors such as Src, Rac1, and PAK1. Surprisingly, in β1 integrin deficient osteoblasts, S. aureus internalization is restored when Caveolin-1 is absent and requires αvβ3/5 integrins as backup fibronectin receptors. Altogether, our data support that β1 integrins regulate the level of detergent-resistant membrane at the plasma membrane in a an endosomal and Caveolin-1 dependent manner.
Collapse
Affiliation(s)
- Léo-Paul Tricou
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
- Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - William Mouton
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Andréa Cara
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Sophie Trouillet-Assant
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Daniel Bouvard
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, Université de Montpellier, Montpellier, France
| | - Frédéric Laurent
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Centre Interrégional de Référence pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Alan Diot
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Josse
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France.
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France.
- Centre Interrégional de Référence pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
4
|
Janssen JN, Kalev-Altman R, Shalit T, Sela-Donenfeld D, Monsonego-Ornan E. Differential gene expression in the calvarial and cortical bone of juvenile female mice. Front Endocrinol (Lausanne) 2023; 14:1127536. [PMID: 37378024 PMCID: PMC10291685 DOI: 10.3389/fendo.2023.1127536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Both the calvarial and the cortical bones develop through intramembranous ossification, yet they have very different structures and functions. The calvaria enables the rapid while protected growth of the brain, whereas the cortical bone takes part in locomotion. Both types of bones undergo extensive modeling during embryonic and post-natal growth, while bone remodeling is the most dominant process in adults. Their shared formation mechanism and their highly distinct functions raise the fundamental question of how similar or diverse the molecular pathways that act in each bone type are. Methods To answer this question, we aimed to compare the transcriptomes of calvaria and cortices from 21-day old mice by bulk RNA-Seq analysis. Results The results revealed clear differences in expression levels of genes related to bone pathologies, craniosynostosis, mechanical loading and bone-relevant signaling pathways like WNT and IHH, emphasizing the functional differences between these bones. We further discussed the less expected candidate genes and gene sets in the context of bone. Finally, we compared differences between juvenile and mature bone, highlighting commonalities and dissimilarities of gene expression between calvaria and cortices during post-natal bone growth and adult bone remodeling. Discussion Altogether, this study revealed significant differences between the transcriptome of calvaria and cortical bones in juvenile female mice, highlighting the most important pathway mediators for the development and function of two different bone types that originate both through intramembranous ossification.
Collapse
Affiliation(s)
- Jerome Nicolas Janssen
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Rotem Kalev-Altman
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
- The Koret School of Veterinary Medicine, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tali Shalit
- The Ilana and Pascal Mantoux Institute for Bioinformatics, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- The Koret School of Veterinary Medicine, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry, Food Science and Nutrition, The Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
5
|
Franceschi RT, Hallett SA, Ge C. Discoidin domain receptors; an ancient family of collagen receptors has major roles in bone development, regeneration and metabolism. FRONTIERS IN DENTAL MEDICINE 2023; 4:1181817. [PMID: 38222874 PMCID: PMC10785288 DOI: 10.3389/fdmed.2023.1181817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
The extracellular matrix (ECM) niche plays a critical role in determining cellular behavior during bone development including the differentiation and lineage allocation of skeletal progenitor cells to chondrocytes, osteoblasts, or marrow adipocytes. As the major ECM component in mineralized tissues, collagen has instructive as well as structural roles during bone development and is required for bone cell differentiation. Cells sense their extracellular environment using specific cell surface receptors. For many years, specific β1 integrins were considered the main collagen receptors in bone, but, more recently, the important role of a second, more primordial collagen receptor family, the discoidin domain receptors, has become apparent. This review will specifically focus on the roles of discoidin domain receptors in mineralized tissue development as well as related functions in abnormal bone formation, regeneration and metabolism.
Collapse
Affiliation(s)
- Renny T. Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Shawn A. Hallett
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Chen S, He T, Zhong Y, Chen M, Yao Q, Chen D, Shao Z, Xiao G. Roles of focal adhesion proteins in skeleton and diseases. Acta Pharm Sin B 2023; 13:998-1013. [PMID: 36970189 PMCID: PMC10031257 DOI: 10.1016/j.apsb.2022.09.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The skeletal system, which contains bones, joints, tendons, ligaments and other elements, plays a wide variety of roles in body shaping, support and movement, protection of internal organs, production of blood cells and regulation of calcium and phosphate metabolism. The prevalence of skeletal diseases and disorders, such as osteoporosis and bone fracture, osteoarthritis, rheumatoid arthritis, and intervertebral disc degeneration, increases with age, causing pain and loss of mobility and creating a huge social and economic burden globally. Focal adhesions (FAs) are macromolecular assemblies that are composed of the extracellular matrix (ECM), integrins, intracellular cytoskeleton and other proteins, including kindlin, talin, vinculin, paxillin, pinch, Src, focal adhesion kinase (FAK) and integrin-linked protein kinase (ILK) and other proteins. FA acts as a mechanical linkage connecting the ECM and cytoskeleton and plays a key role in mediating cell-environment communications and modulates important processes, such as cell attachment, spreading, migration, differentiation and mechanotransduction, in different cells in skeletal system by impacting distinct outside-in and inside-out signaling pathways. This review aims to integrate the up-to-date knowledge of the roles of FA proteins in the health and disease of skeletal system and focuses on the specific molecular mechanisms and underlying therapeutic targets for skeletal diseases.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
7
|
Binrayes A, Ge C, Mohamed F, Franceschi R. Role of Discoidin Domain Receptor 2 in Craniofacial Bone Regeneration. J Dent Res 2021; 100:1359-1366. [PMID: 33899571 PMCID: PMC8532241 DOI: 10.1177/00220345211007447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bone loss caused by trauma, neoplasia, congenital defects, or periodontal disease is a major cause of disability and human suffering. Skeletal progenitor cell-extracellular matrix interactions are critical for bone regeneration. Discoidin domain receptor 2 (DDR2), an understudied collagen receptor, plays an important role in skeletal development. Ddr2 loss-of-function mutations in humans and mice cause severe craniofacial and skeletal defects, including altered cranial shape, dwarfing, reduced trabecular and cortical bone, alveolar bone/periodontal defects, and altered dentition. However, the role of this collagen receptor in craniofacial regeneration has not been examined. To address this, calvarial subcritical-size defects were generated in wild-type (WT) and Ddr2-deficient mice. The complete bridging seen in WT controls at 4 wk postsurgery was not observed in Ddr2-deficient mice even after 12 wk. Quantitation of defect bone area by micro-computed tomography also revealed a 50% reduction in new bone volume in Ddr2-deficient mice. Ddr2 expression during calvarial bone regeneration was measured using Ddr2-LacZ knock-in mice. Expression was restricted to periosteal surfaces of uninjured calvarial bone and, after injury, was detected in select regions of the defect site by 3 d postsurgery and expanded during the healing process. The impaired bone healing associated with Ddr2 deficiency may be related to reduced osteoprogenitor or osteoblast cell proliferation and differentiation since knockdown/knockout of Ddr2 in a mesenchymal cell line and primary calvarial osteoblast cultures reduced osteoblast differentiation while Ddr2 overexpression was stimulatory. In conclusion, Ddr2 is required for cranial bone regeneration and may be a novel target for therapy.
Collapse
Affiliation(s)
- A. Binrayes
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Prosthetic Dental Sciences, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - C. Ge
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - F.F. Mohamed
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - R.T. Franceschi
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Biological Chemistry, School of Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Shim NY, Heo JS. Performance of the Polydopamine-Graphene Oxide Composite Substrate in the Osteogenic Differentiation of Mouse Embryonic Stem Cells. Int J Mol Sci 2021; 22:ijms22147323. [PMID: 34298943 PMCID: PMC8303500 DOI: 10.3390/ijms22147323] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Graphene oxide (GO) is a biocompatible material considered a favorable stem cell culture substrate. In this study, GO was modified with polydopamine (PDA) to facilitate depositing GO onto a tissue culture polystyrene (PT) surface, and the osteogenic performance of the PDA/GO composite in pluripotent embryonic stem cells (ESCs) was investigated. The surface chemistry of the PDA/GO-coated PT surface was analyzed by scanning electron microscopy (SEM) and X-ray photoelectron spectroscopy (XPS). A high cell viability of ESCs cultured on the PDA/GO composite-coated surface was initially ensured. Then, the osteogenic differentiation of the ESCs in response to the PDA/GO substrate was assessed by alkaline phosphatase (ALP) activity, intracellular calcium levels, matrix mineralization assay, and evaluation of the mRNA and protein levels of osteogenic factors. The culture of ESCs on the PDA/GO substrate presented higher osteogenic potency than that on the uncoated control surface. ESCs cultured on the PDA/GO substrate expressed significantly higher levels of integrin α5 and β1, as well as bone morphogenetic protein receptor (BMPR) types I and II, compared with the control groups. The phosphorylation of extracellular signal-regulated kinase (ERK)1/2, p38, and c-Jun-N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) was observed in ESCs culture on the PDA/GO substrate. Moreover, BMP signal transduction by SMAD1/5/8 phosphorylation was increased more in cells on PDA/GO than in the control. The nuclear translocation of SMAD1/5/8 in cells was also processed in response to the PDA/GO substrate. Blocking activation of the integrin α5/β1, MAPK, or SMAD signaling pathways downregulated the PDA/GO-induced osteogenic differentiation of ESCs. These results suggest that the PDA/GO composite stimulates the osteogenic differentiation of ESCs via the integrin α5/β1, MAPK, and BMPR/SMAD signaling pathways.
Collapse
|
9
|
Nedeva IR, Vitale M, Elson A, Hoyland JA, Bella J. Role of OSCAR Signaling in Osteoclastogenesis and Bone Disease. Front Cell Dev Biol 2021; 9:641162. [PMID: 33912557 PMCID: PMC8072347 DOI: 10.3389/fcell.2021.641162] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Formation of mature bone-resorbing cells through osteoclastogenesis is required for the continuous remodeling and repair of bone tissue. In aging and disease this process may become aberrant, resulting in excessive bone degradation and fragility fractures. Interaction of receptor-activator of nuclear factor-κB (RANK) with its ligand RANKL activates the main signaling pathway for osteoclastogenesis. However, compelling evidence indicates that this pathway may not be sufficient for the production of mature osteoclast cells and that co-stimulatory signals may be required for both the expression of osteoclast-specific genes and the activation of osteoclasts. Osteoclast-associated receptor (OSCAR), a regulator of osteoclast differentiation, provides one such co-stimulatory pathway. This review summarizes our present knowledge of osteoclastogenesis signaling and the role of OSCAR in the normal production of bone-resorbing cells and in bone disease. Understanding the signaling mechanism through this receptor and how it contributes to the production of mature osteoclasts may offer a more specific and targeted approach for pharmacological intervention against pathological bone resorption.
Collapse
Affiliation(s)
- Iva R Nedeva
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Mattia Vitale
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Jordi Bella
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Haag SL, Schiele NR, Bernards MT. Enhancement and mechanisms of MC3T3-E1 osteoblast-like cell adhesion to albumin through calcium exposure. Biotechnol Appl Biochem 2021; 69:492-502. [PMID: 33586804 DOI: 10.1002/bab.2126] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/09/2021] [Indexed: 01/06/2023]
Abstract
Serum albumin is the most prominent protein in blood, and it aids in bone fracture healing, though the manner through which enhanced healing occurs is not well understood. This study investigates the influence of calcium on the bioactivity of albumin due to the prevalence of calcium at bone injury sites. Bovine serum albumin (BSA) was exposed to varying concentrations of calcium, adsorbed to tissue culture polystyrene, and the subsequent BSA-coated surfaces were evaluated with calcium titration, and cell adhesion, viability, and binding inhibition studies. Calcium-modified BSA improved overall MC3T3-E1 osteoblast-like cell adhesion, although high calcium concentrations induced cell death. Inhibiting specific integrins revealed that without calcium exposure, cell binding to BSA was primarily mediated by integrins that typically bind to the GFOGER sequence of collagen. As calcium exposure increases, the primary binding interaction transitioned to integrins known to bind RGD. However, cell binding to calcium-modified BSA was not completely eliminated during the inhibition studies indicating additional unidentified binding interactions occur. Overall, these results suggest that the exposure to calcium induces conformational changes that affect the cell-binding bioactivity of BSA, which may explain the beneficial impact of albumin in bone tissue.
Collapse
Affiliation(s)
- Stephanie L Haag
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, ID, USA
| | - Nathan R Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, ID, USA
| | - Matthew T Bernards
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, ID, USA
| |
Collapse
|
11
|
Hu P, Zhu X, Zhao C, Hu J, Luo E, Ye B. Fak silencing impairs osteogenic differentiation of bone mesenchymal stem cells induced by uniaxial mechanical stretch. J Dent Sci 2019; 14:225-233. [PMID: 31528249 PMCID: PMC6739265 DOI: 10.1016/j.jds.2019.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/06/2019] [Indexed: 02/05/2023] Open
Abstract
Background/purpose Mechanical stretch plays a key role in promoting proliferation and differentiation of bone marrow mesenchymal stem cells (BMSCs) in distraction osteogenesis (DO). A better understanding of how the extracellular biomechanical stimulation is transferred to intracellular signal expression will benefit DO. Focal adhesion kinase (FAK) is a key factor in integrin signaling pathway. However, little is known about the effect of integrin-FAK signaling during the process of stretch induced osteogenic differentiation of BMSCs. Materials and methods A specific short hairpin RNAs (shRNAs) lentiviral expression vector was used to silence Fak gene and a well-established in vitro uniaxial dynamic stretching device was applied to stimulate DO. Fak silencing was confirmed by fluorescence microscopy and the detection of Fak mRNA and FAK, p-FAK protein expression. Alkaline phosphatase (ALP) activity, expression of osteogenic differentiation markers - runt-related transcription factor 2 (RUNX2/Runx2) and alkaline phosphatase (Alp) together with integrin upstream signal transduction molecules integrin beta-1 (ITGB1/Itgb1) and downstream signal transduction molecules integrin-linked kinase (ILK) were detected after the stretch. Results The results showed that mechanical stretch in control groups significantly induced the osteogenic differentiation of BMSCs with increased ALP activity, expression of RUNX2/Runx2 and Alp, together with upregulated ITGB1/Itgb1 and ILK, which all vanished in Fak silencing group. Conclusion Silencing of the Fak gene inhibited the osteogenic differentiation of rat BMSCs induced by in vitro mechanical stretch through integrin signaling pathway.
Collapse
Affiliation(s)
- Pei Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaowen Zhu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chuang Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jing Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,West China School of Stomatology, Sichuan University, Chengdu, China
| | - En Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,West China School of Stomatology, Sichuan University, Chengdu, China
| | - Bin Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Brunner M, Mandier N, Gautier T, Chevalier G, Ribba AS, Guardiola P, Block MR, Bouvard D. β1 integrins mediate the BMP2 dependent transcriptional control of osteoblast differentiation and osteogenesis. PLoS One 2018; 13:e0196021. [PMID: 29677202 PMCID: PMC5909894 DOI: 10.1371/journal.pone.0196021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/04/2018] [Indexed: 12/05/2022] Open
Abstract
Osteoblast differentiation is a highly regulated process that requires coordinated information from both soluble factors and the extracellular matrix. Among these extracellular stimuli, chemical and physical properties of the matrix are sensed through cell surface receptors such as integrins and transmitted into the nucleus to drive specific gene expression. Here, we showed that the conditional deletion of β1 integrins in the osteo-precursor population severely impacts bone formation and homeostasis both in vivo and in vitro. Mutant mice displayed a severe bone deficit characterized by bone fragility and reduced bone mass. We showed that β1 integrins are required for proper BMP2 dependent signaling at the pre-osteoblastic stage, by positively modulating Smad1/5-dependent transcriptional activity at the nuclear level. The lack of β1 integrins results in a transcription modulation that relies on a cooperative defect with other transcription factors rather than a plain blunted BMP2 response. Our results point to a nuclear modulation of Smad1/5 transcriptional activity by β1 integrins, allowing a tight control of osteoblast differentiation.
Collapse
Affiliation(s)
- Molly Brunner
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Noémie Mandier
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Thierry Gautier
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Genevieve Chevalier
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Anne-Sophie Ribba
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Philippe Guardiola
- Centre Hospitalier Universitaire and University of Angers, SNP Plateform, Institute for Biological Health, Transcriptome and Epigenomic, Angers, France
| | - Marc R. Block
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
| | - Daniel Bouvard
- Centre de Recherche INSERM 1209, CNRS 5309, Institute for Advanced Bioscience; Université Grenoble Alpes, Grenoble, France
- * E-mail:
| |
Collapse
|
13
|
Assis-Ribas T, Forni MF, Winnischofer SMB, Sogayar MC, Trombetta-Lima M. Extracellular matrix dynamics during mesenchymal stem cells differentiation. Dev Biol 2018; 437:63-74. [PMID: 29544769 DOI: 10.1016/j.ydbio.2018.03.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/05/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are stromal cells that display self-renewal and multipotent differentiation capacity. The repertoire of mature cells generated ranges but is not restricted to: fat, bone and cartilage. Their potential importance for both cell therapy and maintenance of in vivo homeostasis is indisputable. Nonetheless, both their in vivo identity and use in cell therapy remain elusive. A drawback generated by this fact is that little is known about the MSC niche and how it impacts differentiation and homeostasis maintenance. Hence, the roles played by the extracellular matrix (ECM) and its main regulators namely: the Matrix Metalloproteinases (MMPs) and their counteracting inhibitors (TIMPs and RECK) upon stem cells differentiation are only now beginning to be unveiled. Here, we will focus on mesenchymal stem cells and review the main mechanisms involved in adipo, chondro and osteogenesis, discussing how the extracellular matrix can impact not only lineage commitment, but, also, their survival and potentiality. This review critically analyzes recent work in the field in an effort towards a better understanding of the roles of Matrix Metalloproteinases and their inhibitors in the above-cited events.
Collapse
Affiliation(s)
- Thais Assis-Ribas
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil
| | - Maria Fernanda Forni
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | | | - Mari Cleide Sogayar
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil; Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Marina Trombetta-Lima
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil.
| |
Collapse
|
14
|
Dejaeger M, Böhm AM, Dirckx N, Devriese J, Nefyodova E, Cardoen R, St-Arnaud R, Tournoy J, Luyten FP, Maes C. Integrin-Linked Kinase Regulates Bone Formation by Controlling Cytoskeletal Organization and Modulating BMP and Wnt Signaling in Osteoprogenitors. J Bone Miner Res 2017; 32:2087-2102. [PMID: 28574598 DOI: 10.1002/jbmr.3190] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 12/19/2022]
Abstract
Cell-matrix interactions constitute a fundamental aspect of skeletal cell biology and play essential roles in bone homeostasis. These interactions are primarily mediated by transmembrane integrin receptors, which mediate cell adhesion and transduce signals from the extracellular matrix to intracellular responses via various downstream effectors, including integrin-linked kinase (ILK). ILK functions as adaptor protein at focal adhesion sites, linking integrins to the actin cytoskeleton, and has been reported to act as a kinase phosphorylating signaling molecules such as GSK-3β and Akt. Thereby, ILK plays important roles in cellular attachment, motility, proliferation and survival. To assess the in vivo role of ILK signaling in osteoprogenitors and the osteoblast lineage cells descending thereof, we generated conditional knockout mice using the Osx-Cre:GFP driver strain. Mice lacking functional ILK in osterix-expressing cells and their derivatives showed no apparent developmental or growth phenotype, but by 5 weeks of age they displayed a significantly reduced trabecular bone mass, which persisted into adulthood in male mice. Histomorphometry and serum analysis indicated no alterations in osteoclast formation and activity, but provided evidence that osteoblast function was impaired, resulting in reduced bone mineralization and increased accumulation of unmineralized osteoid. In vitro analyses further substantiated that absence of ILK in osteogenic cells was associated with compromised collagen matrix production and mineralization. Mechanistically, we found evidence for both impaired cytoskeletal functioning and reduced signal transduction in osteoblasts lacking ILK. Indeed, loss of ILK in primary osteogenic cells impaired F-actin organization, cellular adhesion, spreading, and migration, indicative of defective coupling of cell-matrix interactions to the cytoskeleton. In addition, BMP/Smad and Wnt/β-catenin signaling was reduced in the absence of ILK. Taken together, these data demonstrate the importance of integrin-mediated cell-matrix interactions and ILK signaling in osteoprogenitors in the control of osteoblast functioning during juvenile bone mass acquisition and adult bone remodeling and homeostasis. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marian Dejaeger
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anna-Marei Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Naomi Dirckx
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Joke Devriese
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Elena Nefyodova
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ruben Cardoen
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - René St-Arnaud
- Shriners Hospital for Children, McGill University, Montreal, Canada
| | - Jos Tournoy
- Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium.,Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Franceschi RT, Ge C. Control of the Osteoblast Lineage by Mitogen-Activated Protein Kinase Signaling. ACTA ACUST UNITED AC 2017; 3:122-132. [PMID: 29057206 DOI: 10.1007/s40610-017-0059-5] [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: 12/28/2022]
Abstract
PURPOSE OF THE REVIEW This review will provide a timely assessment of MAP kinase actions in bone development and homeostasis with particular emphasis on transcriptional control of the osteoblast lineage. RECENT FINDINGS ERK and p38 MAP kinases function as transducers of signals initiated by the extracellular matrix, mechanical loading, TGF-β, BMPs and FGF2. MAPK signals may also affect and/or interact with other important pathways such as WNT and HIPPO. ERK and p38 MAP kinase pathways phosphorylate specific osteogenic transcription factors including RUNX2, Osterix, ATF4 and DLX5. For RUNX2, phosphorylation at specific serine residues initiates epigenetic changes in chromatin necessary for decondensation and increased transcription. MAPK also suppresses marrow adipogenesis by phosphorylating and inhibiting PPARγ, which may explain the well-known relationship between reduced skeletal loading and marrow fat accumulation. SUMMARY MAPKs transduce signals from the extracellular environment to the nucleus allowing bone cells to respond to changes in hormonal/growth factor signaling and mechanical loading thereby optimizing bone structure to meet physiological and mechanical needs of the body.
Collapse
Affiliation(s)
- Renny T Franceschi
- Departments of Periodontics and Oral Medicine, University of Michigan School of Dentistry and Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-1078
| | - Chunxi Ge
- Departments of Periodontics and Oral Medicine, University of Michigan School of Dentistry and Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-1078
| |
Collapse
|
16
|
Groen N, Yuan H, Hebels DGAJ, Koçer G, Mbuyi F, LaPointe V, Truckenmüller R, van Blitterswijk CA, Habibović P, de Boer J. Linking the Transcriptional Landscape of Bone Induction to Biomaterial Design Parameters. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1603259. [PMID: 27991696 DOI: 10.1002/adma.201603259] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/07/2016] [Indexed: 06/06/2023]
Abstract
New engineering possibilities allow biomaterials to serve as active orchestrators of the molecular and cellular events of tissue regeneration. Here, the molecular control of tissue regeneration for calcium phosphate (CaP)-based materials is established by defining the parameters critical for tissue induction and those are linked to the molecular circuitry controlling cell physiology. The material properties (microporosity, ion composition, protein adsorption) of a set of synthesized osteoinductive and noninductive CaP ceramics are parameterized and these properties are correlated to a transcriptomics profile of osteogenic cells grown on the materials in vitro. Using these data, a genetic network controlling biomaterial-induced bone formation is built. By isolating the complex material properties into single-parameter test conditions, it is verified that a subset of these genes is indeed controlled by surface topography and ions released from the ceramics, respectively. The gene network points to a decisive role for extracellular matrix deposition in osteoinduction by genes such as tenascin C and hyaluronic acid synthase 2, which are controlled by calcium and phosphate ions as well as surface topography. This work provides insight into the biomaterial composition and material engineering aspects of bone void filling and can be used as a strategy to explore the interface between biomaterials and tissue regeneration.
Collapse
Affiliation(s)
- Nathalie Groen
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Huipin Yuan
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
- Xpand Biotechnology B.V, Professor Bronkhorstlaan 10, 3723, MB, Bilthoven, The Netherlands
| | - Dennie G A J Hebels
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Gülistan Koçer
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Faustin Mbuyi
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
| | - Vanessa LaPointe
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Clemens A van Blitterswijk
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| | - Jan de Boer
- Department of Tissue Regeneration, University of Twente, Drienerlolaan 5, 7522, NB, Enschede, The Netherlands
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229, ER, Maastricht, The Netherlands
| |
Collapse
|
17
|
Zhang F, Zhang Z, Sun D, Dong S, Xu J, Dai F. Periostin: A Downstream Mediator of EphB4-Induced Osteogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:7241829. [PMID: 26788070 PMCID: PMC4695675 DOI: 10.1155/2016/7241829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/05/2015] [Accepted: 08/24/2015] [Indexed: 01/06/2023] Open
Abstract
Erythropoietin-producing hepatocyte B4 (EphB4) has been reported to be a key molecular switch in the regulation of bone homeostasis, but the underlying mechanism remains poorly understood. In this study, we investigated the role of EphB4 in regulating the expression of periostin (POSTN) within bone marrow-derived mesenchymal stem cells (MSCs) and assessed its effect and molecular mechanism of osteogenic induction in vitro. Treatment with ephrinB2-FC significantly increased the expression of POSTN in MSCs, and the inhibition of EphB4 could abrogate this effect. In addition, osteogenic markers were upregulated especially in MSCs overexpressing EphB4. To elucidate the underlying mechanism of cross talk between EphB4 and the Wnt pathway, we detected the change in protein expression of phosphorylated-glycogen synthase kinase 3β-serine 9 (p-GSK-3β-Ser9) and β-catenin, as well as the osteogenic markers Runx2 and COL1. The results showed that GSK-3β activation and osteogenic marker expression levels were downregulated by ephrinB2-FC treatment, but these effects were inhibited by blocking integrin αvβ3 in MSCs. Our findings demonstrate that EphB4 can promote osteogenic differentiation of MSCs via upregulation of POSTN expression. It not only helps to reveal the interaction mechanism between EphB4 and Wnt pathway but also brings a better understanding of EphB4/ephrinB2 signaling in bone homeostasis.
Collapse
Affiliation(s)
- Fei Zhang
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zehua Zhang
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Dong Sun
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shiwu Dong
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Jianzhong Xu
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
- *Jianzhong Xu: and
| | - Fei Dai
- National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University, Chongqing 400038, China
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
- *Fei Dai:
| |
Collapse
|
18
|
Glycosylation of Dentin Matrix Protein 1 is critical for osteogenesis. Sci Rep 2015; 5:17518. [PMID: 26634432 PMCID: PMC4669440 DOI: 10.1038/srep17518] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/05/2015] [Indexed: 01/23/2023] Open
Abstract
Proteoglycans play important roles in regulating osteogenesis. Dentin matrix protein 1 (DMP1) is a highly expressed bone extracellular matrix protein that regulates both bone development and phosphate metabolism. After glycosylation, an N-terminal fragment of DMP1 protein was identified as a new proteoglycan (DMP1-PG) in bone matrix. In vitro investigations showed that Ser89 is the key glycosylation site in mouse DMP1. However, the specific role of DMP1 glycosylation is still not understood. In this study, a mutant DMP1 mouse model was developed in which the glycosylation site S89 was substituted with G89 (S89G-DMP1). The glycosylation level of DMP1 was down-regulated in the bone matrix of S89G-DMP1 mice. Compared with wild type mice, the long bones of S89G-DMP1 mice showed developmental changes, including the speed of bone remodeling and mineralization, the morphology and activities of osteocytes, and activities of both osteoblasts and osteoclasts. These findings indicate that glycosylation of DMP1 is a key posttranslational modification process during development and that DMP1-PG functions as an indispensable proteoglycan in osteogenesis.
Collapse
|
19
|
Ren H, Ren H, Li X, Yu D, Mu S, Chen Z, Fu Q. Effects of intermedin on proliferation, apoptosis and the expression of OPG/RANKL/M-CSF in the MC3T3-E1 osteoblast cell line. Mol Med Rep 2015; 12:6711-7. [PMID: 26398911 PMCID: PMC4626169 DOI: 10.3892/mmr.2015.4328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/28/2015] [Indexed: 01/21/2023] Open
Abstract
Bone remodeling is a vital physiological process of healthy bone tissue in humans. It is characterized by the formation of bone by osteoblasts and its resorption by osteoclasts, and the bone resorbed by osteoclasts is replaced through the differentiation and activity of osteoblasts. Imbalances in this vital process lead to pathological conditions, including osteoporosis. Intermedin (IMD) as a newly discovered peptide in the calcitonin (CT) family of peptides, which shares similar functions with CT, calcitonin gene‑related peptide and amylin in bone resorption. However, the mechanism underlying its effect remains to be elucidated. This was investigated in the present study using the osteoblastic MC3T3‑E1 cell line, which was treated with different doses of IMD (0, 1, 10 and 100 nM). Cell proliferation, apoptosis and the expression of receptor activator of NF‑κB ligand (RANKL), osteoprotegerin (OPG) and macrophage colony‑stimulating factor (M‑CSF) were measured following treatment using multiple detection techniques, including an MTT assay, flow cytometry, reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The resulting data demonstrated that IMD significantly inhibited the apoptosis of MC3T3‑E1 cells induced by serum‑free culture and dexamethasone, however, no significant effects on MC3T3‑E1 cell proliferation were observed. IMD had additional functions on the MC3T3‑E1 cells, including inhibition of the expression of RANKL and M‑CSF, and promotion of the expression of OPG. Previous studies have also demonstrated that RANKL and M‑CSF are two vital factor produced by osteoblasts to promote the maturation and differentiation of osteoclasts, and it has been reported that IMD can inhibit the osteoclast formation stimulated by RANKL and M‑CSF. Together with these findings, the present study concluded that IMD reduces bone resorption by inhibiting osteoblast apoptosis, decreasing the RANKL/OPG ratio and the expression of M-CSF, and inhibiting osteoclast maturation and differentiation.
Collapse
Affiliation(s)
- Hongfei Ren
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongyu Ren
- Department of Orthopedic Surgery, Gaizhou Central Hospital, Yingkou, Liaoning 115200, P.R. China
| | - Xue Li
- Department of Orthopedic Surgery, First Affiliated Hospital of Liaoning Medical University, Jingzhou, Liaoning 121001, P.R. China
| | - Dongdong Yu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Shuai Mu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhiguang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Qin Fu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
20
|
Lu Y, Zheng Q, Lu W, Yue J, Zhou W, Zhou X, Zhang L, Huang D. Compressive mechanical stress may activate IKK-NF-κB through proinflammatory cytokines in MC3T3-E1 cells. Biotechnol Lett 2015; 37:1729-35. [DOI: 10.1007/s10529-015-1849-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/01/2015] [Indexed: 01/13/2023]
|
21
|
Saidak Z, Le Henaff C, Azzi S, Marty C, Da Nascimento S, Sonnet P, Marie PJ. Wnt/β-catenin signaling mediates osteoblast differentiation triggered by peptide-induced α5β1 integrin priming in mesenchymal skeletal cells. J Biol Chem 2015; 290:6903-12. [PMID: 25631051 DOI: 10.1074/jbc.m114.621219] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The α5β1 integrin is a key fibronectin (FN) receptor that binds to RGD-containing peptides to mediate cell adhesion. We previously reported that α5β1 integrin promotes osteogenic differentiation in mesenchymal skeletal cells (MSCs), but the underlying mechanisms are not fully understood. In this study, we determined the signaling mechanisms induced by α5β1 integrin interaction with its high-affinity ligand CRRETAWAC in murine and human MSCs and in vivo. We show that cyclized CRRETAWAC fully displaced MSC adhesion to FN, whereas related peptides lacking the full RRET sequence produced a partial displacement, indicating that RRET acts as an RGD-like sequence that is required to antagonize FN-mediated cell adhesion. However, all peptides increased focal adhesion kinase phosphorylation, OSE2 transcriptional activity, osteoblast gene expression, and matrix mineralization in MSCs, indicating that peptide-induced α5β1 integrin priming can promote osteogenic differentiation independently of the RRET sequence. Biochemical analyses showed that peptide-induced α5β1 integrin priming transiently increased PI3K/Akt phosphorylation and promoted Wnt/β-catenin transcriptional activity independently of RRET. Consistently, pharmacological inhibition of PI3K activity reduced osteoblast differentiation and abolished Wnt regulatory gene expression induced by α5β1 integrin priming. In vivo, systemic delivery of cyclized GACRETAWACGA linked to (DSS)6 to allow delivery to bone-forming sites for 6 weeks increased serum osteocalcin levels and improved long bone mass and microarchitecture in SAMP-6 senescent osteopenic mice. The results support a mechanism whereby α5β1 integrin priming by high-affinity ligands integrates Wnt/β-catenin signaling to promote osteoblast differentiation independently of cell adhesion, which could be used to improve bone mass and microarchitecture in the aging skeleton.
Collapse
Affiliation(s)
- Zuzana Saidak
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Carole Le Henaff
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Sofia Azzi
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Caroline Marty
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Sophie Da Nascimento
- the Equipe Théra, Laboratoire de Glycochimie, des Antimicrobiens, et des Agroressources (LG2A)-FRE-CNRS 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 80037 Amiens Cedex 1, France
| | - Pascal Sonnet
- the Equipe Théra, Laboratoire de Glycochimie, des Antimicrobiens, et des Agroressources (LG2A)-FRE-CNRS 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 80037 Amiens Cedex 1, France
| | - Pierre J Marie
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| |
Collapse
|
22
|
Xu WX, Liu Y, Liu SZ, Zhang Y, Qiao GF, Yan J. Arsenic trioxide exerts a double effect on osteoblast growth in vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:412-9. [PMID: 25128771 DOI: 10.1016/j.etap.2014.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 05/25/2023]
Abstract
Arsenic trioxide (ATO) is a promising antitumor agent used to treat acute promyelocytic leukemia (APL) and, recently solid tumor. The present study was designed to evaluate the effect of ATO proliferation of osteoblast that plays very important roles in maintaining the structure integrity and function of bone. Cell survives, apoptosis, collagen, and molecular targets were identified by multiple detecting techniques, including MTT assay, electron microscopy, collagen detecting kit, TUNEL kit, and western blot in hFOB1.19 human osteoblasts cell line. The results showed that low dose of ATO (0.25, 0.5, and 1μM) remarkably enhanced the viability of cultured osteoblasts in a concentration- and time-dependent manner. Intriguingly, a dual effect of high dose of ATO (5, 10, and 20μM) was also observed showing significant reduction in viability of culture osteoblasts at concentration- and time-dependent fashion. Moreover, low dose of ATO promoted secretion and synthesis of collagen, whereas high dose of ATO induced typical morphological characteristics of apoptosis in osteoblasts. Mechanically, western blot results demonstrated that low dose of ATO dramatically up-regulated TGF-β1 protein and activated p-AKT proliferative signaling. And, high dose of ATO increased Bax/Bcl-2 ratio in a time-dependent fashion and activated caspase-3 apoptotic signaling. These results demonstrate at the first time that ATO exerts a double effect on osteoblast function depending upon the concentration and provide a clue to rationally use ATO for clinicians to pay more attention to protect bone from the adverse effects of therapeutic dose of ATO during tumor therapy.
Collapse
Affiliation(s)
- Wen-Xiao Xu
- Department of Orthopedics, The Second Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Sheng-Zhi Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Yu Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Guo-Fen Qiao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
23
|
The reversal phase of the bone-remodeling cycle: cellular prerequisites for coupling resorption and formation. BONEKEY REPORTS 2014; 3:561. [PMID: 25120911 PMCID: PMC4130129 DOI: 10.1038/bonekey.2014.56] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/19/2014] [Indexed: 12/31/2022]
Abstract
The reversal phase couples bone resorption to bone formation by generating an osteogenic environment at remodeling sites. The coupling mechanism remains poorly understood, despite the identification of a number of ‘coupling' osteogenic molecules. A possible reason is the poor attention for the cells leading to osteogenesis during the reversal phase. This review aims at creating awareness of these cells and their activities in adult cancellous bone. It relates cell events (i) on the bone surface, (ii) in the mesenchymal envelope surrounding the bone marrow and appearing as a canopy above remodeling surfaces and (iii) in the bone marrow itself within a 50-μm distance of this canopy. When bone remodeling is initiated, osteoprogenitors at these three different levels are activated, likely as a result of a rearrangement of cell–cell and cell–matrix interactions. Notably, canopies are brought under the osteogenic influence of capillaries and osteoclasts, whereas bone surface cells become exposed to the eroded matrix and other osteoclast products. In several diverse pathophysiological situations, including osteoporosis, a decreased availability of osteoprogenitors from these local reservoirs coincides with decreased osteoblast recruitment and impaired initiation of bone formation, that is, uncoupling. Overall, this review stresses that coupling does not only depend on molecules able to activate osteogenesis, but that it also demands the presence of osteoprogenitors and ordered cell rearrangements at the remodeling site. It points to protection of local osteoprogenitors as a critical strategy to prevent bone loss.
Collapse
|