1
|
Hsuuw YD, Su YT, Chan WH, Wu CC, Tsai YC, Chen HC, Huang FJ, Lin CF. Co-culture with adipose mesenchymal stem cells promotes Blastocyst formation and gene expression in embryos from aged mice. Regen Ther 2025; 29:319-327. [PMID: 40242085 PMCID: PMC12002604 DOI: 10.1016/j.reth.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/06/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Recent studies have highlighted the positive effects of co-culturing embryos with stem cells on embryo development in various mammalian systems. Stem cells secrete numerous factors, including cytokines, growth factors, and microRNAs, which promote embryo development. However, the impact of stem cells on the development of embryos derived from aged mice's oocytes remains poorly understood. This study evaluated the co-culture effects of adipose tissue-derived mesenchymal stem cells (ADMSCs) on zygotes, focusing on the developmental potential of fertilized embryos. Embryo quality was assessed through staining techniques to measure trophectoderm (TE), inner cell mass (ICM), and total blastocyst cell numbers during in vitro culture. Results demonstrated that ADMSC co-culture significantly improved zygote cleavage and blastocyst development rates, particularly in embryos derived from aged mice. Enhanced implantation and post-implantation potential were observed in embryos from both young and aged mice. Notably, co-culture increased TE, ICM, and total blastocyst cell numbers in aged mice-derived embryos without inducing apoptosis in blastocysts. Gene expression analysis revealed upregulation of OCT4 and G6PDH, associated with pluripotency and glucose metabolism, particularly in embryos from aged mice, while the heat stress marker HSP70 showed no significant changes. These findings demonstrate the potential of ADMSC co-culture as a beneficial protocol for improving embryo development. These findings from this study could offer an important basis for future mechanistic studies in this area.
Collapse
Affiliation(s)
- Yan-Der Hsuuw
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Yu-Ting Su
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung City 83301, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City 32023, Taiwan
| | - Cheng-Chun Wu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Yu-Chieh Tsai
- Department of Obstetrics and Gynecology, An- An Women and Children Clinic & ART Center, Kaohsiung City 80752, Taiwan
| | - Hou-Chun Chen
- Laboratory Animal Center, Office of Advanced Science and Technology, Thammasat University, Krung Thep Maha Nakhon 10200, Thailand
| | - Fu-Jen Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung City 83301, Taiwan
- Department of Obstetrics and Gynecology, An- An Women and Children Clinic & ART Center, Kaohsiung City 80752, Taiwan
| | - Chuen-Fu Lin
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Animal Disease Diagnostic Center, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| |
Collapse
|
2
|
Gao Y, Chen C, Liu R, Zhang Z, Zhao X, Ma H. Research progress of connexin 43 mediated gap junction communication regulating bone metabolism in glucocorticoid-induced osteonecrosis of the femoral head. Exp Cell Res 2025; 449:114598. [PMID: 40339781 DOI: 10.1016/j.yexcr.2025.114598] [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: 11/13/2024] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic disease that commonly affects young and middle-aged individuals. Long-term and high-dose use of glucocorticoids (GCs) is one of the main causes. Currently, the pathological mechanism of GCs-induced ONFH remains unclear, which poses difficulties for clinical prevention and treatment. This article focuses on reviewing the roles of gap junctions (GJs) and connexin 43 (Cx43) in GCs-induced ONFH. Under normal circumstances, cells in bone tissue form a network structure through GJs to maintain bone metabolic balance. However, GCs can obstruct the normal connections and signal transmission between bone tissue cells, leading to bone metabolic imbalance and triggering ONFH. As a key component of GJs in bone tissue, Cx43 is of great significance in bone metabolism. It not only participates in the construction of the osteocyte network but also regulates osteocyte activity, osteoblast differentiation, and osteogenic activities. Meanwhile, in vascular endothelial cells, Cx43 plays an important role in angiogenesis and maintaining vascular homeostasis, and is closely related to the vascularization of bone tissue. In addition, Cx43 is associated with the release of prostaglandin E2 (PGE2). GCs can inhibit the activity of Cx43, reduce the release of PGE2, and disrupt the balance of bone metabolism. Studies have shown that measuring changes in the expression level of Cx43 is expected to become an early diagnostic biomarker for GCs-induced ONFH. Enhancing its expression through small - molecule drugs, biological agents, and gene therapy may be potential treatment approaches for ONFH. This article proposes the PI3K/Akt/GSK3β/β-catenin pathway and conducts research on the regulatory mechanism of Cx43-mediated GJ-based intercellular communication, aiming to provide new ideas for the treatment of ONFH and bone metabolism-related diseases.
Collapse
Affiliation(s)
- Yang Gao
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Changjun Chen
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Rongxing Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Zhongkai Zhang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Xin Zhao
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
| | - Huanzhi Ma
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
| |
Collapse
|
3
|
Guerrero J, Maevskaia E, Pfister P, Dominguez AP, Ghayor C, Bhattacharya I, Scherberich A, Weber FE. Mineralized Osteoblast-Derived Exosomes and 3D-printed Ceramic-based Scaffolds for Enhanced Bone Healing: A Preclinical Exploration. Acta Biomater 2025:S1742-7061(25)00376-9. [PMID: 40409510 DOI: 10.1016/j.actbio.2025.05.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/25/2025] [Accepted: 05/21/2025] [Indexed: 05/25/2025]
Abstract
In regenerative medicine, addressing the complex challenge of bone tissue regeneration demands innovative strategies. Exosomes, nanoscale vesicles rich in bioactive molecules, have shown great promise in tissue repair. This study focuses on exosomes derived from mineralized osteoblasts (MOBs), which play a pivotal role in bone formation. We investigated the therapeutic potential of exosomes isolated from osteoblasts cultured in osteogenic medium for 21 days, delivered via 3D-printed gyroid scaffolds composed of hydroxyapatite (HA) and tricalcium phosphate (TCP). The exosomes were characterized through nanoparticle tracking analysis to determine size, morphology, and concentration, while proteomics revealed their cargo contents. In vitro, rabbit bone marrow mesenchymal stromal cells (rBMSCs) were cultured as monolayers and within ceramic scaffolds, where MOB-derived exosomes were shown to promote osteogenic differentiation. In vivo, their osteoconductive and bone augmentation capabilities were evaluated in two rabbit calvarial models, while the osteoinductive potential was further tested in a heterotopic mouse model. Neo-bone formation was assessed using µCT and histological analysis. Our findings demonstrated that MOB-derived exosomes upregulated bone-related gene expression and promoted mineralization in rBMSCs, even in the absence of osteogenic medium. Proteomics confirmed the presence of bone-associated proteins in these exosomes. In rabbit models, however, exosomes did not significantly enhance bone formation. In contrast, in the heterotopic mouse model, exosomes functionalized onto ceramic scaffolds exhibited strong osteoinductive activity. This study highlights the potential of MOB-derived exosomes to enhance 3D-printed ceramic scaffolds for bone regeneration, offering a promising avenue for bone healing without the need for additional growth factors or stem cells. STATEMENT OF SIGNIFICANCE: The here presented report of our project not only advances our understanding of the role of exosome-functionalized scaffolds in bone regeneration but also proposes a promising alternative to traditional growth factor- or cell-based approaches. We are confident that this study represents a novel and impactful contribution to the field.
Collapse
Affiliation(s)
- Julien Guerrero
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Ekaterina Maevskaia
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Pablo Pfister
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Ana Pérez Dominguez
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Chafik Ghayor
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Indranil Bhattacharya
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Arnaud Scherberich
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Franz E Weber
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland; CABMM, Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Wu J, Wang Y, Wang L, Xie W, Wan Q, Wang J, Chen J, Pei X, Zhu Z. A 3D Co-Culture System Inspired by Fracture Healing Cell Interactions for Bone Tissue Engineering. Adv Healthc Mater 2025:e2500534. [PMID: 40394923 DOI: 10.1002/adhm.202500534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Peri-bone fibroblasts play a crucial role in regulating bone regeneration during early fracture healing. Inspired by the synergy between osteoblasts and fibroblasts at fracture sites, a biomimetic three-dimensional (3D) indirect co-culture system is developed, comprising a 3D scaffold and co-cultured cells. To mimic cellular interactions in the fracture healing zone, the scaffold features an inner-outer ring structure with communication channels that support indirect cell co-culture. This setup provides fibroblasts and osteoblasts with a 3D culture environment resembling the in vivo extracellular matrix, enhancing intercellular signaling while minimizing risks of direct contact. Mechanically tunable bioinks are formulated by incorporating hyaluronic acid methacrylate (HAMA) hydrogel into gelatin methacryloyl (GelMA) hydrogel to construct the scaffold. The optimal co-culture ratio is established in vitro, where fibroblasts are found to regulate the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) via zinc ion transport mechanisms. In vivo validations are conducted, including ectopic bone formation in nude mice and bone regeneration in rat cranial defect and tooth extraction socket models. This 3D indirect co-culture system enhances osteogenesis by promoting functional fibroblast-osteoblast interactions, offering a novel platform for co-culture studies and a promising strategy for clinical bone regeneration applications.
Collapse
Affiliation(s)
- Jicenyuan Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuxuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liang Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjia Xie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
5
|
Cai X, Zhang S, Xiao C, Dang Z, Huang W, Xu W, Wu G. Orchestrating Macrophage and Bone Mesenchymal Stem Cells to Promote Bone Regeneration via Modulation of the Internal Surface Morphology inside 3D Printed Scaffolds. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:8853-8865. [PMID: 40146177 DOI: 10.1021/acs.langmuir.5c00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Surface morphology has been widely used to orchestrate multicellular function. However, most studies are mainly based on two-dimensional (2D) surface morphology. Therefore, a new scaffold that could be used to design and obtain controllable internal surface morphology was fabricated to explore the effect of a micropatterned scaffold on bone repair. In this study, through the combination of three-dimensional (2D) printing and soft lithography, a controllable micropatterned poly(ε-caprolactone) scaffold was obtained, which realized the transformation from 2D micropattern research to 3D research. Pit micropatterns with morphology sizes of 0, 25, and 45 μm (Flat, P25, and P45) were constructed. In vitro, the results showed that the P25 micropattern had a better effect on the promotion of M2 polarization, inhibition of the M1 polarization of RAW264.7 cells, and promotion of the osteogenic differentiation of bone marrow stromal stem cells (BMSCs). Direct and indirect coculture models of macrophages and BMSCs were constructed to study the bone immunomodulation of the pit micropatterns. Compared with the Flat and P45 groups, the P25 group could promote the secretion of M2 markers, inhibit the secretion of M1 markers, and immunomodulate the promotion of osteogenic differentiation of BMSCs. In vivo, the results also showed that the P25 group had a lower proinflammatory effect and better performance than scaffolds without micropatterned surfaces and a bigger morphology size (the P45 group), which could regulate the immune function of macrophages, reduce the inflammatory response, and accelerate bone regeneration and repair. This work provides a new strategy for the preparation of scaffolds for bone defect regeneration.
Collapse
Affiliation(s)
- Xiayu Cai
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Shaohui Zhang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Chujie Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- School of Biology and Biological Engineering, South China University of Technology, No. 381 Wushan Road, Tianhe District, Guangzhou, Guangdong 510641, China
| | - Zhaohui Dang
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
| | - Weihua Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
- The Second Affiliated Hospital of Guangzhou Medical University, The Second Clinical Medicine School of Guangzhou Medical University, No. 250, Changgang East Road, Haizhu District, Guangzhou, Guangdong 510260, China
- Department of Orthopedic Surgery, Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, Guangdong 511518, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangdong Institute of Medical Instruments, No. 1307, Guangzhou Avenue Central, Tianhe District, Guangzhou, Guangdong 510500, China
- Guangdong Chinese Medicine Intelligent Diagnosis and Treatment Engineering Technology Research Center, Jianghai Avenue Central, Haizhu District, Guangzhou, Guangdong 510316, China
| | - Gang Wu
- National Engineering Research Center for Human Tissue Restoration and Function Reconstruction, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong 510006, China
| |
Collapse
|
6
|
Xu M, Liu H, Zhang J, Xu M, Zhao X, Wang J. Functionalized zeolite regulates bone metabolic microenvironment. Mater Today Bio 2025; 31:101558. [PMID: 40034985 PMCID: PMC11874869 DOI: 10.1016/j.mtbio.2025.101558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
The regulation of bone metabolic microenvironment imbalances in diseases such as osteoporosis, bone defects, infections, and tumors remains a significant challenge in orthopedics. Therefore, it has become urgent to develop biomaterials with effective bone metabolic microenvironmental regulatory functions. Zeolites, as advanced biomedical materials, possess distinctive physicochemical properties such as multi-level pore structures, adjustable frameworks, easily modifiable surfaces, and excellent adsorption capabilities. These advantageous characteristics give zeolites broad application prospects in regulating the bone metabolic microenvironment. Therefore, this paper first classifies zeolites used to regulate the bone metabolic microenvironment based on their topological structures and compositional frameworks. Subsequently, it provides a detailed description of modification strategies for zeolite materials aimed at regulating this microenvironment. Next, a comprehensive summary was provided on the preparation strategies for zeolite materials aimed at regulating the bone metabolic microenvironment. Additionally, the paper focuses on the specific applications of zeolite materials in conditions of bone metabolic imbalance, such as osteoporosis, bone defects, orthopedic infections, and bone tumors, highlighting their potential in enhancing osteogenic microenvironments, controlling infections, and treating bone tumors. Finally, it outlines the prospects and challenges associated with the application of zeolites in regulating the bone metabolic microenvironment. This review comprehensively summarizes zeolites used for bone metabolic regulation, aiming to provide guidance for future research and application development.
Collapse
Affiliation(s)
| | | | - Jiaxin Zhang
- Orthopedic Institute of Jilin Province, Orthopedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Meng Xu
- Orthopedic Institute of Jilin Province, Orthopedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Xin Zhao
- Orthopedic Institute of Jilin Province, Orthopedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Jincheng Wang
- Orthopedic Institute of Jilin Province, Orthopedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, China
| |
Collapse
|
7
|
Bastos AR, Maia FR, Oliveira JM, Reis RL, Correlo VM. In vitro Bone Tissue Engineering Strategies: The Relevance of Cells and Culturing Methods in Bone Formation and Remodeling. Macromol Biosci 2025; 25:e2400453. [PMID: 39932135 DOI: 10.1002/mabi.202400453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/06/2024] [Indexed: 04/15/2025]
Abstract
The most recent advances in bone tissue engineering (BTE) approaches step forward in the field of three-dimensional (3D) tissue models, enabling the development of more realistic tools to study bone disorders, such as osteoporosis. BTE field aims to mimic native bone tissue more truthfully, providing an appropriate environment for tissue regeneration and repair through the combination of 3D porous scaffolds, specific growth factors, and cells. Currently, the scientific community is focused on developing and improving new biomaterials that in combination with growth factors and specific cell types, that can accurately emulate the native bone microenvironment. However, most of the reported studies in the BTE field are focused on bone formation, disregarding the entire bone remodeling steps, which also involve bone resorption. In this review, the currently available mono and co-culturing methods, types of biomaterials used in several strategies that combine scaffolds and relevant cells (e.g., osteoblasts (OBs), osteoclasts (OCs), and osteocytes (OCys)), envisioning a healthy bone formation and remodeling process, the gold-standard drug delivery systems, and bioengineered-based systems to tackle bone diseases are described.
Collapse
Affiliation(s)
- Ana Raquel Bastos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia Rua Ave 1, Edifício 1 (Sede), Barco, 4805-694, GMR - Portugal
- ICVS/3B's - PT Government Associated Laboratory, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Fátima Raquel Maia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia Rua Ave 1, Edifício 1 (Sede), Barco, 4805-694, GMR - Portugal
- ICVS/3B's - PT Government Associated Laboratory, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia Rua Ave 1, Edifício 1 (Sede), Barco, 4805-694, GMR - Portugal
- ICVS/3B's - PT Government Associated Laboratory, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Rui Luís Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia Rua Ave 1, Edifício 1 (Sede), Barco, 4805-694, GMR - Portugal
- ICVS/3B's - PT Government Associated Laboratory, Campus de Gualtar, Braga, 4710-057, Portugal
| | - Vítor Manuel Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Parque de Ciência e Tecnologia Rua Ave 1, Edifício 1 (Sede), Barco, 4805-694, GMR - Portugal
- ICVS/3B's - PT Government Associated Laboratory, Campus de Gualtar, Braga, 4710-057, Portugal
| |
Collapse
|
8
|
Agnes CJ, Li L, Bertrand D, Murshed M, Willie BM, Tabrizian M. Assessment of bone regeneration potential for a 6-bromoindirubin-3'-oxime (BIO) encapsulated chitosan based scaffold in a mouse critical sized bone defect model. Int J Biol Macromol 2025; 304:140995. [PMID: 39952511 DOI: 10.1016/j.ijbiomac.2025.140995] [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: 08/26/2024] [Revised: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Development of an effective treatment to guide bone repair in critical size bone defect applications remains a major unmet challenge. Current clinical gold standards display significant disadvantages, thereby necessitating that research focus on designing and producing a suitable alternative for healing. In this study, we comprehensively assessed the bone regenerative potential of a newly formulated 6-Bromoindirubin-3'-Oxime (BIO) incorporated chitosan-based scaffold using a mouse femoral defect model. Live 3D in vivo micro-CT imaging enabled us to monitor the progression of bone formation over 56 days, without needing additional replicates. Results demonstrated smaller distances between bone ends (1.033 ± 0.512 mm) compared to controls (1.474 ± 0.465 mm) at later timepoints (p = 0.0430), suggesting improved bone formation. This observed effect was supported with serum procollagen type I N-propeptide levels, where BIO scaffolds showed marked increases in collagen synthesis. As vascularization is often-overlooked, blood vessel density at 56 days was also assessed, showing an additional benefit of BIO incorporated scaffolds (9.264 ± 0.578) over controls (6.667 ± 1.300) on angiogenesis. Although BIO's incorporation did not lead to bony bridging or a significant difference in bone volume compared to controls at day 56, our findings suggest the BIO incorporated scaffold's ability to improve healing outcomes through enhancement of Wnt signaling. Further studies aimed at optimizing the dose to target this pathway are warranted, as a means to more completely regenerate bone in challenging healing scenarios.
Collapse
Affiliation(s)
- Celine J Agnes
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children, Montreal, QC, Canada.
| | - Ling Li
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
| | - David Bertrand
- Shriners Hospital for Children, Montreal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
| | - Monzur Murshed
- Shriners Hospital for Children, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| | - Bettina M Willie
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
De Pace R, Iaquinta MR, Benkhalqui A, D'Agostino A, Trevisiol L, Nocini R, Mazziotta C, Rotondo JC, Bononi I, Tognon M, Martini F, Mazzoni E. Revolutionizing bone healing: the role of 3D models. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:7. [PMID: 40113735 PMCID: PMC11926310 DOI: 10.1186/s13619-025-00225-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
The increasing incidence of bone diseases has driven research towards Bone Tissue Engineering (BTE), an innovative discipline that uses biomaterials to develop three-dimensional (3D) scaffolds capable of mimicking the natural environment of bone tissue. Traditional approaches relying on two-dimensional (2D) models have exhibited significant limitations in simulating cellular interactions and the complexity of the bone microenvironment. In response to these challenges, 3D models such as organoids and cellular spheroids have emerged as effective tools for studying bone regeneration. Adult mesenchymal stem cells have proven crucial in this context, as they can differentiate into osteoblasts and contribute to bone tissue repair. Furthermore, the integration of composite biomaterials has shown substantial potential in enhancing bone healing. Advanced technologies like microfluidics offer additional opportunities to create controlled environments for cell culture, facilitating more detailed studies on bone regeneration. These advancements represent a fundamental step forward in the treatment of bone pathologies and the promotion of skeletal health. In this review, we report on the evolution of in vitro culture models applied to the study of bone healing/regrowth, starting from 2 to 3D cultures and microfluids. The different methodologies of in vitro model generation, cells and biomaterials are presented and discussed.
Collapse
Affiliation(s)
- Raffaella De Pace
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, 44121, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Assia Benkhalqui
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Department of Surgery, University of Verona, Verona, Italy
| | | | - Lorenzo Trevisiol
- Centre for Medical Sciences (CISMed), University of Trento, Trento, Italy
- Unit of Maxillofacial Surgery, Santa Chiara Regional Hospital, APSS, Trento, Italy
| | | | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Bononi
- Centralized Laboratory of Pre-Clinical Research, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, 44121, Italy.
| |
Collapse
|
10
|
Kozalak G, Koşar A. Bone-on-a-Chip Systems for Hematological Cancers. BIOSENSORS 2025; 15:176. [PMID: 40136973 PMCID: PMC11940066 DOI: 10.3390/bios15030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Hematological malignancies originating from blood, bone marrow, and lymph nodes include leukemia, lymphoma, and myeloma, which necessitate the use of a distinct chemotherapeutic approach. Drug resistance frequently complicates their treatment, highlighting the need for predictive tools to guide therapeutic decisions. Conventional 2D/3D cell cultures do not fully encompass in vivo criteria, and translating disease models from mice to humans proves challenging. Organ-on-a-chip technology presents an avenue to surmount genetic disparities between species, offering precise design, concurrent manipulation of various cell types, and extrapolation of data to human physiology. The development of bone-on-a-chip (BoC) systems is crucial for accurately representing the in vivo bone microenvironment, predicting drug responses for hematological cancers, mitigating drug resistance, and facilitating personalized therapeutic interventions. BoC systems for modeling hematological cancers and drug research can encompass intricate designs and integrated platforms for analyzing drug response data to simulate disease scenarios. This review provides a comprehensive examination of BoC systems applicable to modeling hematological cancers and visualizing drug responses within the intricate context of bone. It thoroughly discusses the materials pertinent to BoC systems, suitable in vitro techniques, the predictive capabilities of BoC systems in clinical settings, and their potential for commercialization.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
11
|
Guillaumin S, Rossoni A, Zeugolis D. State-of the-art and future perspective in co-culture systems for tendon engineering. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100110. [PMID: 40130022 PMCID: PMC11932666 DOI: 10.1016/j.bbiosy.2025.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Tendon is a connective tissue that links bone to muscle, allowing for maintenance of skeleton posture, joint movement, energy storage and transmission of muscle force to bone. Tendon is a hypocellular and hypovascular tissue of poor self-regeneration capacity. Current surgical treatments are of limited success, frequently resulting in reinjury. Upcoming cell therapies are primarily based on tenocytes, a cell population of limited self-renewal capacity in vitro or mesenchymal stromal cells, a cell population prone to ectopic bone formation in vivo. Over the years mono- or multi- factorial cell culture technologies have failed to effectively maintain tenocyte phenotype in culture during expansion or to prime mesenchymal stromal cells towards tenogenic lineage prior to implantation. Upon these limitations the concept of co-culture was conceived. Here, we comprehensively review and discuss tenogenic differentiation of mesenchymal stromal cells through direct or indirect culture with tenocytes in an attempt to generate a tenocyte or a tendon-like cell population for regenerative medicine purposes.
Collapse
Affiliation(s)
- Salomé Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
12
|
Kontogianni GI, Loukelis K, Bonatti AF, Batoni E, De Maria C, Vozzi G, Naseem R, Dalgarno K, Shin H, Vitale-Brovarone C, Chatzinikolaidou M. A Mechanically Stimulated Co-culture in 3-Dimensional Composite Scaffolds Promotes Osteogenic and Anti-osteoclastogenic Activity and M2 Macrophage Polarization. Biomater Res 2025; 29:0135. [PMID: 39911306 PMCID: PMC11794764 DOI: 10.34133/bmr.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 02/07/2025] Open
Abstract
Bone is subjected to a plethora of mechanical stresses, which have been found to directly influence the equilibrium between bone resorption and formation. Taking this into account, we present herein a novel biomimicking 3-dimensional model that applies cyclic uniaxial compression onto cells co-cultured on 3-dimensionally printed scaffolds consisting of poly L-lactic acid/poly(ε-caprolactone)/poly(3-hydroxybutyrate-co-3-hydroxyvalerate)/Sr-nanohydroxyapatite. The aim is to investigate how compression can modulate the balance between osteogenesis and osteoclastogenesis in co-culture, as well as the polarization of macrophages. One of the key aspects of the current study is the unprecedented development of a growth-factor-free co-culture, sustainable solely by the cross talk between human bone marrow mesenchymal stem cells and human peripheral blood mononuclear cells for their survival and osteogenic/osteoclastogenic differentiation capacity, respectively. Real-time polymerase chain reaction gene expression analysis of the mechanically stimulated constructs revealed up-regulation of the osteogenesis-related markers osteocalcin, osteoprotegerin, and runt-related transcription factor 2, with concurrent down-regulation of the osteoclastogenic markers dendritic-cell-specific transmembrane protein, nuclear factor of activated T cells 1, and tartrate acid phosphatase. The secretion of the receptor activator of nuclear factor kappa-Β ligand and macrophage colony-stimulating factor, as determined from enzyme-linked immunosorbent assay, was also found to depict lower levels compared to static conditions. Finally, macrophage polarization was examined via confocal imaging of tumor necrosis factor-α and interleukin-10 secretion levels, as well as through nitric oxide synthase and arginase 1 markers' gene expression, with the results indicating stronger commitment toward the M2 phenotype after mechanical stimulation.
Collapse
Affiliation(s)
| | - Konstantinos Loukelis
- Department of Materials Science and Engineering,
University of Crete, 70013 Heraklion, Greece
| | - Amedeo Franco Bonatti
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Elisa Batoni
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio, Department of Information Engineering,
University of Pisa, 56126 Pisa, Italy
| | - Raasti Naseem
- School of Engineering,
Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Kenneth Dalgarno
- School of Engineering,
Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Heungsoo Shin
- Department of Bioengineering,
Hanyang University, Seoul 04763, Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader,
Hanyang University, Seoul 04763, Korea
| | | | - Maria Chatzinikolaidou
- Department of Materials Science and Engineering,
University of Crete, 70013 Heraklion, Greece
- Foundation for Research and Technology Hellas (FORTH)-IESL, 70013 Heraklion, Greece
| |
Collapse
|
13
|
Han D, Wang W, Gong J, Ma Y, Li Y. Controlled delivery of mesenchymal stem cells via biodegradable scaffolds for fracture healing. Nanomedicine (Lond) 2025; 20:207-224. [PMID: 39686770 PMCID: PMC11731254 DOI: 10.1080/17435889.2024.2439242] [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: 09/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Biodegradable controlled delivery systems for mesenchymal stem cells (MSCs) have emerged as novel advancements in the field of regenerative medicine, particularly for accelerating bone fracture healing. This detailed study emphasizes the importance of quick and adequate fracture treatment and the limitations of existing methods. New approaches employing biodegradable scaffolds can be placed within a fracture to serve as a mechanical support and allow controlled release of in situ MSCs and bioactive agents. They are made up of polymers and composites which degrade over time, aiding in natural tissue regrowth. The fabrication methods, including 3D printing, electrospinning, and solvent casting, with particulate leaching that enable precise control over scaffold architecture and properties, are discussed. Progress in controlled drug delivery systems including encapsulation techniques and release kinetics is described, highlighting the potential of such strategies to maintain therapeutic benefits over a prolonged time as well as improving outcomes for fracture repair. MSCs play a role in bone regeneration through differentiation using biodegradable scaffolds, paracrine effects, and regulation of inflammation focusing on fracture healing. Current trends and future directions in scaffold technology and MSC delivery, including smart scaffolds with growth factor incorporation and innovative delivery approaches for fracture healing are also discussed.
Collapse
Affiliation(s)
- Dong Han
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Weijiao Wang
- Otolaryngology Department, Yantaishan Hospital, Yantai, China
| | - Jinpeng Gong
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yupeng Ma
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yu Li
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| |
Collapse
|
14
|
Mathis K, Meckes B. Precision Cell-Cell Assembly Through Light-Mediated DNA Interactions. Methods Mol Biol 2025; 2902:173-182. [PMID: 40029603 DOI: 10.1007/978-1-0716-4402-7_11] [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: 03/05/2025]
Abstract
Complex interactions between diverse cell populations influence processes ranging from cell division to programmed cell death. Replicating this complexity with reproducibility remains challenging. We introduce an innovative approach that combines DNA-mediated interactions with photolithography to achieve controlled cell-cell interactions. By coating cells with DNA sequences responsive to light, we enable meticulous spatial organization through light activation, facilitating rapid and programmable construction of intricate cell structures. This method not only addresses the challenges of orchestrating cell arrangements in vitro, particularly in three-dimensional settings but also offers a new avenue for the on-demand creation of complex cellular architectures.
Collapse
Affiliation(s)
- Katelyn Mathis
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
| |
Collapse
|
15
|
Shariati K, Bedar M, Huang KX, Moghadam S, Mirzaie S, LaGuardia JS, Chen W, Kang Y, Ren X, Lee JC. Biomaterial Cues for Regulation of Osteoclast Differentiation and Function in Bone Regeneration. ADVANCED THERAPEUTICS 2025; 8:2400296. [PMID: 39867107 PMCID: PMC11756815 DOI: 10.1002/adtp.202400296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 01/28/2025]
Abstract
Tissue regeneration involves dynamic dialogue between and among different cells and their surrounding matrices. Bone regeneration is specifically governed by reciprocity between osteoblasts and osteoclasts within the bone microenvironment. Osteoclast-directed resorption and osteoblast-directed formation of bone are essential to bone remodeling, and the crosstalk between these cells is vital to curating a sequence of events that culminate in the creation of bone tissue. Among bone biomaterial strategies, many have investigated the use of different material cues to direct the development and activity of osteoblasts. However, less attention has been given to exploring features that similarly target osteoclast formation and activity, with even fewer strategies demonstrating or integrating biomaterial-directed modulation of osteoblast-osteoclast coupling. This review aims to describe various biomaterial cues demonstrated to influence osteoclastogenesis and osteoclast function, emphasizing those that enhance a material construct's ability to achieve bone healing and regeneration. Additionally discussed are approaches that influence the communication between osteoclasts and osteoblasts, particularly in a manner that takes advantage of their coupling. Deepening our understanding of how biomaterial cues may dictate osteoclast differentiation, function, and influence on the microenvironment may enable the realization of bone-replacement interventions with enhanced integrative and regenerative capacities.
Collapse
Affiliation(s)
- Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Sarah Mirzaie
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
16
|
Piao X, Wu X, Yan Y, Li Y, Li N, Xue L, He F. Targeting EZH2 attenuates the ferroptosis-mediated osteoblast-osteoclast imbalance in rheumatoid arthritis. Int Immunopharmacol 2024; 143:113201. [PMID: 39353382 DOI: 10.1016/j.intimp.2024.113201] [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: 06/23/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE The enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) can regulate osteogenesis and osteoclastogenesis. This study aimed to further explore the effects of EZH2 modification on ferroptosis and the osteoblast-osteoclast balance in rheumatoid arthritis (RA) in vitro and in vivo. METHODS Bone marrow mesenchymal stromal cells were transfected with EZH2 overexpression (oeEZH2) and EZH2 shRNA (shEZH2) plasmids with or without ferrostatin-1 (Fer-1) treatment and subjected to an osteoblast differentiation assay. The cells were then cocultured with bone marrow-derived macrophages and subjected to an osteoclast differentiation assay. Collagen-induced arthritis (CIA) mice were generated and injected with shEZH2 adeno-associated virus (AAV). RESULTS OeEZH2 repressed osteoblast differentiation, as reflected by decreased ALP and Alizarin Red S staining and increased OPN, RUNX2, OPG and RANKL; however, shEZH2 had the opposite effects. Besides, oeEZH2 promoted osteoblast ferroptosis, as suggested by increased MDA, Fe2+, ROS, and PTGS2 but decreased GPX4 and SLC7A11; these effects could be attenuated by Fer-1 treatment. In contrast, shEZH2 ameliorated osteoblast ferroptosis. OeEZH2 subsequently increased osteoclast differentiation, as indicated by increased TRAP+ multinucleated cells, NFATC1, CTSK, and c-FOS; however, shEZH2 had the opposite effect, except that it did not regulate CTSK. In CIA mice, shEZH2 AAV decreased the clinical symptom score, histological score of cartilage, and systemic inflammation (TNF-α and IL-6) and repressed bone ferroptosis and restored the osteoblast-osteoclast balance to some extent, as reflected by immunohistochemical staining of related markers. CONCLUSION Targeting EZH2 attenuates the ferroptosis-mediated osteoblast-osteoclast imbalance in RA, revealing its potential as a treatment target.
Collapse
Affiliation(s)
- Xuemei Piao
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiangxiang Wu
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yixin Yan
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Yongming Li
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Na Li
- Department of Internal Medicine, The Third People's Hospital of Chongming District, Shanghai 202153, China
| | - Luan Xue
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Feng He
- The Center for Cancer Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
17
|
De Vos B, Kasonga AE, Joubert AM, Nyakudya TT. Exploring the In Vitro Effects of Zingerone on Differentiation and Signalling Pathways in Bone Cell Lines. Metabolites 2024; 14:693. [PMID: 39728474 DOI: 10.3390/metabo14120693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVE Ensuring adequate bone health is crucial for preventing conditions such as osteoporosis and fractures. Zingerone, a phytonutrient isolated from cooked ginger, has gained attention for its potential benefits in bone health. This study evaluated the osteoprotective potential of zingerone and its effects on differentiation and signalling pathways in vitro using SAOS-2 osteosarcoma and RAW264.7 macrophage cell lines, aiming to elucidate its mechanism of action in bone remodelling. METHODS SAOS-2 osteosarcoma and RAW264.7 macrophage cells were treated with zingerone at concentrations of 200 µM. Osteoblast differentiation was assessed by alkaline phosphatase (ALP) activity, bone mineralisation via Alizarin Red S stain, and gene expression markers (ALP, runt-related transcription factor 2 (Runx2), and osteocalcin) via quantitative polymerase chain reaction (q-PCR). Osteoclast differentiation was evaluated by tartrate-resistant acid phosphatase (TRAP) staining, TRAP activity, and mitogen-activated protein kinase (MAPK) pathways. RESULTS Treatment with zingerone was non-toxic at 200 µM. Zingerone (200 µM) significantly stimulated the gene expression of ALP and Runx2 in SAOS-2 cells (p < 0.05) without statistically significantly enhancing SAOS-2 mineralisation via calcium deposits. Moreover, zingerone significantly inhibited osteoclast differentiation in RAW264.7 cells as evidenced by reduced TRAP staining and activity (p < 0.05). CONCLUSIONS Zingerone shows promise in reducing osteoclast activity and supporting early osteoblast differentiation, suggesting its potential as a dietary supplement for bone health. Further in vivo and clinical studies are needed to confirm its role in managing osteoporosis.
Collapse
Affiliation(s)
- Brunhildé De Vos
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| | - Abe E Kasonga
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| | - Anna M Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| | - Trevor T Nyakudya
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| |
Collapse
|
18
|
Zheng J, Li X, Zhang F, Li C, Zhang X, Wang F, Qi J, Cui W, Deng L. Targeting Osteoblast-Osteoclast Cross-Talk Bone Homeostasis Repair Microcarriers Promotes Intervertebral Fusion in Osteoporotic Rats. Adv Healthc Mater 2024; 13:e2402117. [PMID: 39155412 DOI: 10.1002/adhm.202402117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Balancing osteoblast-osteoclast (OB-OC) cross-talk is crucial for restoring bone tissue structure and function. Current clinical drugs targeting either osteogenesis or osteoclastogenesis fail to effectively regulate cross-talk, impeding efficient bone repair in osteoporosis patients. Ubiquitin-specific protease 26 (USP26) is shown to coordinate OB-OC cross-talk by independently regulating β-catenin and Iκb-α. However, effective drugs for activating USP26 are still lacking. Here, they constructed bone homeostasis repair microcarriers (BHRC) that encapsulate Usp26 mRNA-loaded lipid nanoparticles (mRNA@LNP) within MMPs-responsive GelMA hydrogel microspheres. These microcarriers target the osteoporotic microenvironment and regulate OB-OC cross-talk, thereby facilitating intervertebral fusion in osteoporotic rats. Results demonstrate that mRNA@LNP exhibits uniform particle size and high transfection efficiency, while GelMA hydrogel microspheres possess excellent biocompatibility and MMP responsiveness, providing favorable cell survival space and controllable release of mRNA@LNP. The released LNP upregulates USP26 protein expression, effectively promoting osteogenesis while suppressing osteoclast formation. In vivo experiments show that injecting BHRC into the defect site of intervertebral discs in osteoporotic rats significantly promotes tail vertebrae fusion by responding to the microenvironment and regulating cell-to-cell cross-talk. Thus, the BHRC holds great potential in regulating osteoporotic homeostasis, particularly in challenging bone defects such as intervertebral fusion in osteoporotic environments.
Collapse
Affiliation(s)
- Jiancheng Zheng
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiaoyan Li
- Department of Orthopedic, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, 272029, P. R. China
| | - Fangke Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Changwei Li
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xingkai Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Fei Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Jin Qi
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
19
|
Zhong C, Tang Z, Yu X, Wang L, Ren C, Qin L, Zhou P. Advances in the Construction and Application of Bone-on-a-Chip Based on Microfluidic Technologies. J Biomed Mater Res B Appl Biomater 2024; 112:e35502. [PMID: 39555794 DOI: 10.1002/jbm.b.35502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bone-on-a-chip (BOC) models that based on microfluidic technology have widely applied to understand bone physiology and the pathogenesis of related diseases. In this review, we provide an overview of bone biology and related diseases, explain the advantages and applications of microfluidic technology in the construction of BOC models, and summarize their progress in physiology, pathology, and drug development. Finally, we discussed the problems to be solved and the future directions of microfluidic technology and BOC platforms, so as to provide a reference for researchers to design better BOC models.
Collapse
Affiliation(s)
- Chang Zhong
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Zihui Tang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Xin Yu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Lu Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Chenyuan Ren
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Gansu Health Vocational College, Lanzhou, China
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
20
|
Sun YS, Huang HH, Tsai YH, Kuo YL, Lee JW, Lee YJ, Linn TY, Chen P. Creating an extracellular matrix-like three-dimension structure to enhance the corrosion resistance and biological responses of titanium implants. J Dent Sci 2024; 19:S70-S80. [PMID: 39807433 PMCID: PMC11725070 DOI: 10.1016/j.jds.2024.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Indexed: 01/16/2025] Open
Abstract
Background/purpose Titanium (Ti) is extensively used in dental and orthopedic implants due to its excellent mechanical properties. However, its smooth and biologically inert surface does not support the ingrowth of new bone, and Ti ions may have adverse biological effects. The purpose is to improve the corrosion resistance of titanium and create a 3D structured coating to enhance osseointegration through a very simple and fast surface treatment. Materials and methods This study investigated the use of sandblasting, acid etching, and NaOH leaching to produce porous Ti implants with enhanced biological activity and corrosion resistance. Results These surface modifications generated a mixed oxide layer resembling the extracellular matrix (ECM), consisting of a dense amorphous TiO2 inner layer (50-100 nm thick) and a TiO2 outer layer with interconnected pores (pore size 50-500 nm; 150-200 nm thick). The inner layer significantly improved corrosion resistance, while the hydrophilic outer layer, with its porous structure, facilitated protein albumin adsorption and promoted the attachment, proliferation, and mineralization of human bone marrow mesenchymal stem cells. Conclusion The combined surface treatment approach of sandblasting, acid etching, and NaOH leaching offers a comprehensive solution to the challenges associated with titanium implants' biological inertness and corrosion susceptibility. By enhancing both the biological activity and corrosion resistance of Ti surfaces, this protocol holds significant promise for improving dental and orthopedic implants' success rates and longevity. Future studies should focus on in vivo assessments and long-term clinical trials to further validate these findings and explore the potential for widespread clinical adoption.
Collapse
Affiliation(s)
- Ying-Sui Sun
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Her-Hsiung Huang
- Department of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Tsai
- Institute of Oral Biology, School of Dentistry Graduate, National Taiwan University, Taipei, Taiwan
| | - Yu-Lin Kuo
- Department of Mechanical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Jyh-Wei Lee
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei, Taiwan
| | - Yun-Jung Lee
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Thu Ya Linn
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Peng Chen
- Liaison Center for Innovative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| |
Collapse
|
21
|
Bousch JF, Beyersdorf C, Schultz K, Windolf J, Suschek CV, Maus U. Proinflammatory Cytokines Enhance the Mineralization, Proliferation, and Metabolic Activity of Primary Human Osteoblast-like Cells. Int J Mol Sci 2024; 25:12358. [PMID: 39596421 PMCID: PMC11594863 DOI: 10.3390/ijms252212358] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoporosis is a progressive metabolic bone disease characterized by decreased bone density and microarchitectural deterioration, leading to an increased risk of fracture, particularly in postmenopausal women and the elderly. Increasing evidence suggests that inflammatory processes play a key role in the pathogenesis of osteoporosis and are strongly associated with the activation of osteoclasts, the cells responsible for bone resorption. In the present study, we investigated, for the first time, the influence of proinflammatory cytokines on the osteogenic differentiation, proliferation, and metabolic activity of primary human osteoblast-like cells (OBs) derived from the femoral heads of elderly patients. We found that all the proinflammatory cytokines, IL-1β, TNF-α, IL-6, and IL-8, enhanced the extracellular matrix mineralization of OBs under differentiation-induced cell culture conditions. In the cases of IL-1β and TNF-α, increased mineralization was correlated with increased osteoblast proliferation. Additionally, IL-1β- and TNF-α-increased osteogenesis was accompanied by a rise in energy metabolism due to improved glycolysis or mitochondrial respiration. In conclusion, we show here, for the first time, that, in contrast to findings obtained with cell lines, mesenchymal stem cells, or animal models, human OBs obtained from patients exhibited significantly enhanced osteogenesis upon exposure to proinflammatory cytokines, probably in part via a mechanism involving enhanced cellular energy metabolism. This study significantly contributes to the field of osteoimmunology by examining a clinically relevant cell model that can help to develop treatments for inflammation-related metabolic bone diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Uwe Maus
- Department for Orthopedics and Trauma Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany; (J.F.B.); (C.B.); (K.S.); (J.W.); (C.V.S.)
| |
Collapse
|
22
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024; 13:e2401307. [PMID: 39175382 PMCID: PMC11582516 DOI: 10.1002/adhm.202401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | - Janis Locs
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and BioengineeringFaculty of Natural Sciences and TechnologyRiga Technical UniversityPulka Street 3RigaLV‐1007Latvia
- Baltic Biomaterials Centre of ExcellenceHeadquarters at Riga Technical UniversityRigaLV‐1007Latvia
| |
Collapse
|
23
|
Martinez DC, Borkam-Schuster A, Helmholz H, Dobkowska A, Luthringer-Feyerabend B, Płociński T, Willumeit-Römer R, Święszkowski W. Bone cells influence the degradation interface of pure Mg and WE43 materials: Insights from multimodal in vitro analysis. Acta Biomater 2024; 187:471-490. [PMID: 39168423 DOI: 10.1016/j.actbio.2024.08.015] [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: 04/09/2024] [Revised: 06/28/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
In this study, the interaction of pure Mg and WE43 alloy under the presence of osteoblast (OB) and osteoclast (OC) cells and their influence on the degradation of materials have been deeply analyzed. Since OB and OC interaction has an important role in bone remodeling, we examined the surface morphology and dynamic changes in the chemical composition and thickness of the corrosion layers formed on pure Mg and WE43 alloy by direct monoculture and coculture of pre-differentiated OB and OC cells in vitro. Electrochemical techniques examined the corrosion performance. The corrosion products were characterized using a combination of the focused ion beam (FIB), scanning electron microscopy (SEM), and energy-dispersive X-ray spectroscopy (EDX). Cell viability and morphology were assessed by fluorescent microscopy and SEM. Our findings demonstrate cell spread and attachment variations, which differ depending on the Mg substrates. It was clearly shown that cell culture groups delayed degradation processes with the lowest corrosion rate observed in the presence of OBOC coculture for the WE43 substrate. Ca-P enrichment was observed in the outer-middle region of the corrosion layer but only after 7 days of OBOC coculture on WE43 and after 14 days on the pure Mg specimens. STATEMENT OF SIGNIFICANCE: Magnesium metallic materials that can degrade over time provide distinct opportunities for orthopedic application. However, there is still a lack, especially in elucidating cell-material interface characterization. This study investigated the influence of osteoblast-osteoclast coculture in direct Mg-material contact. Our findings demonstrated that pre-differentiated osteoblasts and osteoclasts cocultured on Mg substrates influenced the chemistry of the corrosion layers. The cell spread and attachment were Mg substrate-dependent. The findings of coculturing bone cells directly on Mg materials within an in vitro model provide an effective approach for studying the dynamic degradation processes of Mg alloys while also elucidating cell behavior and their potential contribution to the degradation of these alloys.
Collapse
Affiliation(s)
- Diana C Martinez
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland
| | - Anke Borkam-Schuster
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon GmbH, 21502 Geesthacht, Germany
| | - Heike Helmholz
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon GmbH, 21502 Geesthacht, Germany
| | - Anna Dobkowska
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland
| | | | - Tomasz Płociński
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland
| | - Regine Willumeit-Römer
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon GmbH, 21502 Geesthacht, Germany
| | - Wojciech Święszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska 141, 02-507 Warsaw, Poland.
| |
Collapse
|
24
|
Shen S, Lin Y, Sun J, Liu Y, Chen Y, Lu J. A New Tissue Engineering Strategy to Promote Tendon-bone Healing: Regulation of Osteogenic and Chondrogenic Differentiation of Tendon-derived Stem Cells. Orthop Surg 2024; 16:2311-2325. [PMID: 39043618 PMCID: PMC11456719 DOI: 10.1111/os.14152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
In the field of sports medicine, repair surgery for anterior cruciate ligament (ACL) and rotator cuff (RC) injuries are remarkably common. Despite the availability of relatively effective treatment modalities, outcomes often fall short of expectations. This comprehensive review aims to thoroughly examine current strategies employed to promote tendon-bone healing and analyze pertinent preclinical and clinical research. Amidst ongoing investigations, tendon-derived stem cells (TDSCs), which have comparatively limited prior exploration, have garnered increasing attention in the context of tendon-bone healing, emerging as a promising cell type for regenerative therapies. This review article delves into the potential of combining TDSCs with tissue engineering methods, with ACL reconstruction as the main focus. It comprehensively reviews relevant research on ACL and RC healing to address the issues of graft healing and bone tunnel integration. To optimize tendon-bone healing outcomes, our emphasis lies in not only reconstructing the original microstructure of the tendon-bone interface but also achieving proper bone tunnel integration, encompassing both cartilage and bone formation. In this endeavor, we thoroughly analyze the transcriptional and molecular regulatory variables governing TDSCs differentiation, incorporating a retrospective analysis utilizing single-cell sequencing, with the aim of unearthing relevant signaling pathways and processes. By presenting a novel strategy rooted in TDSCs-driven osteogenic and chondrogenic differentiation for tendon-bone healing, this study paves the way for potential future research avenues and promising therapeutic applications. It is anticipated that the findings herein will contribute to advancing the field of tendon-bone healing and foster the exploration of TDSCs as a viable option for regenerative therapies in the future.
Collapse
Affiliation(s)
- Sinuo Shen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yucheng Lin
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jiachen Sun
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuanhao Liu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Yuzhi Chen
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Jun Lu
- School of MedicineSoutheast UniversityNanjingChina
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
25
|
Borciani G, Montalbano G, Perut F, Ciapetti G, Baldini N, Vitale-Brovarone C. Osteoblast and osteoclast activity on collagen-based 3D printed scaffolds enriched with strontium-doped bioactive glasses and hydroxyapatite nanorods for bone tissue engineering. Biomed Mater 2024; 19:065007. [PMID: 39173660 DOI: 10.1088/1748-605x/ad72c3] [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: 03/15/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Bone tissue engineering (BTE) aims to promote bone regeneration by means of the synergistic effect of biomaterials, cells, and other factors, as potential alternative to conventional treatments for bone fractures. To this aim, a composite material was developed, based on collagen type I, strontium-enriched mesoporous bioactive glasses, and hydroxyapatite nanorods as bioactive and biomimetic components. Nanostructured scaffolds were 3D printed and subsequently chemically crosslinked with genipin to improve mechanical properties and stability. The developed nanostructured system was maintained in culture until 3 weeks with a co-culture of human bone cells to provide anex vivomodel of bone microenvironment and examine the cellular crosstalk and signaling pathways through paracrine cell activities. Human osteoblasts (OBs), derived from trabecular bone, and human osteoclast precursors (OCs), isolated from buffy coat samples were involved, with OBs seeded on the scaffold and OC precursors seeded in a transwell device. When compared to the material without inorganic components, the bioactive and biomimetic scaffold positively influenced cell proliferation and cell metabolic activity, boosting alkaline phosphatase activity of OBs, and reducing OC differentiation. Thus, the bioactive and biomimetic system promoted an enhanced cellular response, highlighting its potential application in BTE.
Collapse
Affiliation(s)
- Giorgia Borciani
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Giorgia Montalbano
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Francesca Perut
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Gabriela Ciapetti
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Nicola Baldini
- Biomedical Science and Technologies and Nanobiotechnology Lab, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - Chiara Vitale-Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
26
|
Waidi YO, Debnath S, Datta S, Chatterjee K. 3D-Printed Silk Proteins for Bone Tissue Regeneration and Associated Immunomodulation. Biomacromolecules 2024; 25:5512-5540. [PMID: 39133748 DOI: 10.1021/acs.biomac.4c00540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Current bone repair methods have limitations, prompting the exploration of innovative approaches. Tissue engineering emerges as a promising solution, leveraging biomaterials to craft scaffolds replicating the natural bone environment, facilitating cell growth and differentiation. Among fabrication techniques, three-dimensional (3D) printing stands out for its ability to tailor intricate scaffolds. Silk proteins (SPs), known for their mechanical strength and biocompatibility, are an excellent choice for engineering 3D-printed bone tissue engineering (BTE) scaffolds. This article comprehensively reviews bone biology, 3D printing, and the unique attributes of SPs, specifically detailing criteria for scaffold fabrication such as composition, structure, mechanics, and cellular responses. It examines the structural, mechanical, and biological attributes of SPs, emphasizing their suitability for BTE. Recent studies on diverse 3D printing approaches using SPs-based for BTE are highlighted, alongside advancements in their 3D and four-dimensional (4D) printing and their role in osteo-immunomodulation. Future directions in the use of SPs for 3D printing in BTE are outlined.
Collapse
Affiliation(s)
- Yusuf Olatunji Waidi
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Souvik Debnath
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Sudipto Datta
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Bioengineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
- Department of Materials Engineering, Indian Institute of Science, C. V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
27
|
Tian K, He X, Lin X, Chen X, Su Y, Lu Z, Chen Z, Zhang L, Li P, Ma L, Lan Z, Zhao X, Fen G, Hai Q, Xue D, Jin Q. Unveiling the Role of Sik1 in Osteoblast Differentiation: Implications for Osteoarthritis. Mol Cell Biol 2024; 44:411-428. [PMID: 39169784 PMCID: PMC11485870 DOI: 10.1080/10985549.2024.2385633] [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: 03/25/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by subchondral osteosclerosis, mainly due to osteoblast activity. This research investigates the function of Sik1, a member of the AMP-activated protein kinase family, in OA. Proteomic analysis was conducted on clinical samples from 30 OA patients, revealing a negative correlation between Sik1 expression and OA. In vitro experiments utilized BMSCs to examine the effect of Sik1 on osteogenic differentiation. BMSCs were cultured and induced toward osteogenesis with specific media. Sik1 overexpression was achieved through lentiviral transfection, followed by analysis of osteogenesis-associated proteins using Western blotting, RT-qPCR, and alkaline phosphate staining. In vivo experiments involved destabilizing the medial meniscus in mice to establish an OA model, assessing the therapeutic potential of Sik1. The CT scans and histological staining were used to analyze subchondral bone alterations and cartilage damage. The findings show that Sik1 downregulation correlates with advanced OA and heightened osteogenic differentiation in BMSCs. Sik1 overexpression inhibits osteogenesis-related markers in vitro and reduces cartilage damage and subchondral osteosclerosis in vivo. Mechanistically, Sik1 modulates osteogenesis and subchondral bone changes through Runx2 activity regulation. The research emphasizes Sik1 as a promising target for treating OA, suggesting its involvement in controlling bone formation and changes in the subchondral osteosclerosis.
Collapse
Affiliation(s)
- Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxin He
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yajing Su
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhidong Lu
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhirong Chen
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Liang Zhang
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Peng Li
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xin Zhao
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Fen
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qinqin Hai
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qunhua Jin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| |
Collapse
|
28
|
Aslam Khan MU, Aslam MA, Bin Abdullah MF, Stojanović GM. Current Perspectives of Protein in Bone Tissue Engineering: Bone Structure, Ideal Scaffolds, Fabrication Techniques, Applications, Scopes, and Future Advances. ACS APPLIED BIO MATERIALS 2024; 7:5082-5106. [PMID: 39007509 DOI: 10.1021/acsabm.4c00362] [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: 07/16/2024]
Abstract
In view of their exceptional approach, excellent inherent biocompatibility and biodegradability properties, and interaction with the local extracellular matrix, protein-based polymers have received attention in bone tissue engineering, which is a multidisciplinary field that repairs and regenerates fractured bones. Bone is a multihierarchical complex structure, and it performs several essential biofunctions, including maintaining mineral balance and structural support and protecting soft organs. Protein-based polymers have gained interest in developing ideal scaffolds as emerging biomaterials for bone fractured healing and regeneration, and it is challenging to design ideal bone substitutes as perfect biomaterials. Several protein-based polymers, including collagen, keratin, gelatin, serum albumin, etc., are potential materials due to their inherent cytocompatibility, controlled biodegradability, high biofunctionalization, and tunable mechanical characteristics. While numerous studies have indicated the encouraging possibilities of proteins in BTE, there are still major challenges concerning their biodegradability, stability in physiological conditions, and continuous release of growth factors and bioactive molecules. Robust scaffolds derived from proteins can be used to replace broken or diseased bone with a biocompatible substitute; proteins, being biopolymers, provide excellent scaffolds for bone tissue engineering. Herein, recent developments in protein polymers for cutting-edge bone tissue engineering are addressed in this review within 3-5 years, with a focus on the significant challenges and future perspectives. The first section discusses the structural fundamentals of bone anatomy and ideal scaffolds, and the second section describes the fabrication techniques of scaffolds. The third section highlights the importance of proteins and their applications in BTE. Hence, the recent development of protein polymers for state-of-the-art bone tissue engineering has been discussed, highlighting the significant challenges and future perspectives.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Muhammad Azhar Aslam
- Department of Physics, University of Engineering and Technology, Lahore 39161, Pakistan
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus Kubang Kerian 16150, Kota Bharu, Kelantan, Malaysia
- Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus Kubang Kerian 16150, Kota Bharu, Kelantan, Malaysia
| | - Goran M Stojanović
- Faculty of Technical Sciences, University of Novi Sad, T. D. Obradovica 6, 21000 Novi Sad, Serbia
| |
Collapse
|
29
|
Yuan X, Zhu W, Yang Z, He N, Chen F, Han X, Zhou K. Recent Advances in 3D Printing of Smart Scaffolds for Bone Tissue Engineering and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403641. [PMID: 38861754 DOI: 10.1002/adma.202403641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Indexed: 06/13/2024]
Abstract
The repair and functional reconstruction of bone defects resulting from severe trauma, surgical resection, degenerative disease, and congenital malformation pose significant clinical challenges. Bone tissue engineering (BTE) holds immense potential in treating these severe bone defects, without incurring prevalent complications associated with conventional autologous or allogeneic bone grafts. 3D printing technology enables control over architectural structures at multiple length scales and has been extensively employed to process biomimetic scaffolds for BTE. In contrast to inert and functional bone grafts, next-generation smart scaffolds possess a remarkable ability to mimic the dynamic nature of native extracellular matrix (ECM), thereby facilitating bone repair and regeneration. Additionally, they can generate tailored and controllable therapeutic effects, such as antibacterial or antitumor properties, in response to exogenous and/or endogenous stimuli. This review provides a comprehensive assessment of the progress of 3D-printed smart scaffolds for BTE applications. It begins with an introduction to bone physiology, followed by an overview of 3D printing technologies utilized for smart scaffolds. Notable advances in various stimuli-responsive strategies, therapeutic efficacy, and applications of 3D-printed smart scaffolds are discussed. Finally, the review highlights the existing challenges in the development and clinical implementation of smart scaffolds, as well as emerging technologies in this field.
Collapse
Affiliation(s)
- Xun Yuan
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Wei Zhu
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Zhongyuan Yang
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Ning He
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Feng Chen
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Xiaoxiao Han
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Kun Zhou
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
30
|
Zainal Ariffin SH, Megat Abdul Wahab R, Abdul Razak M, Yazid MD, Shahidan MA, Miskon A, Zainol Abidin IZ. Evaluation of in vitro osteoblast and osteoclast differentiation from stem cell: a systematic review of morphological assays and staining techniques. PeerJ 2024; 12:e17790. [PMID: 39071131 PMCID: PMC11283775 DOI: 10.7717/peerj.17790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Background Understanding human stem cell differentiation into osteoblasts and osteoclasts is crucial for bone regeneration and disease modeling. Numerous morphological techniques have been employed to assess this differentiation, but a comprehensive review of their application and effectiveness is lacking. Methods Guided by the PRISMA framework, we conducted a rigorous search through the PubMed, Web of Science and Scopus databases, analyzing 254 articles. Each article was scrutinized against pre-defined inclusion criteria, yielding a refined selection of 14 studies worthy of in-depth analysis. Results The trends in using morphological approaches were identified for analyzing osteoblast and osteoclast differentiation. The three most used techniques for osteoblasts were Alizarin Red S (mineralization; six articles), von Kossa (mineralization; three articles) and alkaline phosphatase (ALP; two articles) followed by one article on Giemsa staining (cell morphology) and finally immunochemistry (three articles involved Vinculin, F-actin and Col1 biomarkers). For osteoclasts, tartrate-resistant acid phosphatase (TRAP staining) has the highest number of articles (six articles), followed by two articles on DAPI staining (cell morphology), and immunochemistry (two articles with VNR, Cathepsin K and TROP2. The study involved four stem cell types: peripheral blood monocyte, mesenchymal, dental pulp, and periodontal ligament. Conclusion This review offers a valuable resource for researchers, with Alizarin Red S and TRAP staining being the most utilized morphological procedures for osteoblasts and osteoclasts, respectively. This understanding provides a foundation for future research in this rapidly changing field.
Collapse
Affiliation(s)
- Shahrul Hisham Zainal Ariffin
- Department of Science Biology and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Rohaya Megat Abdul Wahab
- Centre of Family Dental Health, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Muhammad Abdul Razak
- Board of Director Office, 6th Floor, Chancellery Building, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Universiti Kebangsaan Malaysia, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Muhammad Ashraf Shahidan
- Department of Science Biology and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Azizi Miskon
- Department of Electrical and Electronics Engineering, Faculty of Engineering, National Defence University of Malaysia, Sungai Besi, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Intan Zarina Zainol Abidin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Cyberjaya, Cyberjaya, Selangor, Malaysia
| |
Collapse
|
31
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [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: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
32
|
Li C, He W, Song Y, Zhang X, Sun J, Zhou Z. Advances of 3D Cell Co-Culture Technology Based on Microfluidic Chips. BIOSENSORS 2024; 14:336. [PMID: 39056612 PMCID: PMC11274478 DOI: 10.3390/bios14070336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Cell co-culture technology aims to study the communication mechanism between cells and to better reveal the interactions and regulatory mechanisms involved in processes such as cell growth, differentiation, apoptosis, and other cellular activities. This is achieved by simulating the complex organismic environment. Such studies are of great significance for understanding the physiological and pathological processes of multicellular organisms. As an emerging cell cultivation technology, 3D cell co-culture technology, based on microfluidic chips, can efficiently, rapidly, and accurately achieve cell co-culture. This is accomplished by leveraging the unique microchannel structures and flow characteristics of microfluidic chips. The technology can simulate the native microenvironment of cell growth, providing a new technical platform for studying intercellular communication. It has been widely used in the research of oncology, immunology, neuroscience, and other fields. In this review, we summarize and provide insights into the design of cell co-culture systems on microfluidic chips, the detection methods employed in co-culture systems, and the applications of these models.
Collapse
Affiliation(s)
- Can Li
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.L.); (Y.S.); (X.Z.)
| | - Wei He
- Department of Clinical Medical Engineering, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| | - Yihua Song
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.L.); (Y.S.); (X.Z.)
| | - Xia Zhang
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.L.); (Y.S.); (X.Z.)
| | - Jianfei Sun
- State Key Laboratory of Bioelectronics and Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, China
| | - Zuojian Zhou
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China; (C.L.); (Y.S.); (X.Z.)
| |
Collapse
|
33
|
Cazzolla AP, Brescia V, Lovero R, Fontana A, Giustino A, Dioguardi M, Di Comite MS, Di Serio F, Ciavarella D, Crincoli V. Evaluation of Biomarkers of Bone Metabolism on Salivary Matrix in the Remodeling of Periodontal Tissue during Orthodontic Treatment. Dent J (Basel) 2024; 12:209. [PMID: 39056996 PMCID: PMC11276302 DOI: 10.3390/dj12070209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/23/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The aim of this study was to evaluate changes in the concentration of N-terminal type I collagen extension pro-peptide (PINP), tartrate-resistant acid phosphatase (TRAcP), and parathyroid hormone-related protein (PTHrP) in saliva during orthodontic treatment in order to evaluate whether changes in bone turnover marker (BTM) concentration can help highlight the effects of orthodontic mechanical loading in the absence of clinical evidence of tooth movement in terms of tooth movement. Saliva samples from 25 apparently healthy young subjects (10 females and 15 males) were collected using Salivette® (Sarstedt) with cotton swabs and the concentrations of PTHrP, TRAcP 5b, and PINP were analyzed at time 0 (T1), 25 days (T2), and at 45 days (T3). Differences in the median value of biomarker levels between baseline T1 and follow-up of the different groups (T2 and T3) were assessed using the non-parametric Mann-Whitney U test. Trough concentrations of P1NP, PTHrP, and TRAcP were 0.80 µg/L, 0.21 ng/mL, and 0.90 U/L above the method LOD. The non-parametric Mann-Whitney U test confirmed a statistically significant difference in T1 versus concentrations of T2 and T3. All subjects evaluated had a statistically significant difference between T1 vs. T3. when compared with the specific critical difference (RCV) for the analyte The results obtained demonstrate that the evaluation of BTM changes in saliva can help the evaluation of orthodontic procedures and the monitoring of biomechanical therapy.
Collapse
Affiliation(s)
- Angela Pia Cazzolla
- Department of Clinical and Experimental Medicine, Università degli Studi di Foggia, 71100 Foggia, Italy; (M.D.); (D.C.)
| | - Vincenzo Brescia
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari—Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (R.L.); (A.F.); (F.D.S.)
| | - Roberto Lovero
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari—Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (R.L.); (A.F.); (F.D.S.)
| | - Antonietta Fontana
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari—Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (R.L.); (A.F.); (F.D.S.)
| | - Arcangela Giustino
- Department of Biomedical Sciences and Human Oncology, Aldo Moro, University of Bari, 70121 Bari, Italy;
| | - Mario Dioguardi
- Department of Clinical and Experimental Medicine, Università degli Studi di Foggia, 71100 Foggia, Italy; (M.D.); (D.C.)
| | - Maria Severa Di Comite
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Human Anatomy Section, Aldo Moro, University of Bari, 70121 Bari, Italy;
| | - Francesca Di Serio
- Clinical Pathology Unit, AOU Policlinico Consorziale di Bari—Ospedale Giovanni XXIII, 70124 Bari, Italy; (V.B.); (R.L.); (A.F.); (F.D.S.)
| | - Domenico Ciavarella
- Department of Clinical and Experimental Medicine, Università degli Studi di Foggia, 71100 Foggia, Italy; (M.D.); (D.C.)
| | - Vito Crincoli
- Interdisciplinary Department of Medicine, Aldo Moro, University of Bari, 70121 Bari, Italy
| |
Collapse
|
34
|
Peng Z, Ahsan N, Yang Z. Proteomics Analysis of Interactions between Drug-Resistant and Drug-Sensitive Cancer Cells: Comparative Studies of Monoculture and Coculture Cell Systems. J Proteome Res 2024; 23:2608-2618. [PMID: 38907724 PMCID: PMC11425778 DOI: 10.1021/acs.jproteome.4c00338] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Cell-cell interactions, which allow cells to communicate with each other through molecules in their microenvironment, are critical for the growth, health, and functions of cells. Previous studies show that drug-resistant cells can interact with drug-sensitive cells to elevate their drug resistance level, which is partially responsible for cancer recurrence. Studying protein targets and pathways involved in cell-cell communication provides essential information for fundamental cell biology studies and therapeutics of human diseases. In the current studies, we performed direct coculture and indirect coculture of drug-resistant and drug-sensitive cell lines, aiming to investigate intracellular proteins responsible for cell communication. Comparative studies were carried out using monoculture cells. Shotgun bottom-up proteomics results indicate that the P53 signaling pathway has a strong association with drug resistance mechanisms, and multiple TP53-related proteins were upregulated in both direct and indirect coculture systems. In addition, cell-cell communication pathways, including the phagosome and the HIF-signaling pathway, contribute to both direct and indirect coculture systems. Consequently, AK3 and H3-3A proteins were identified as potential targets for cell-cell interactions that are relevant to drug resistance mechanisms. We propose that the P53 signaling pathway, in which mitochondrial proteins play an important role, is responsible for inducing drug resistance through communication between drug-resistant and drug-sensitive cancer cells.
Collapse
Affiliation(s)
- Zongkai Peng
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Nagib Ahsan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
35
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
36
|
Shen J, Zhang S, Zhang J, Wei X, Wang Z, Han B. Osteogenic mechanism of chlorogenic acid and its application in clinical practice. Front Pharmacol 2024; 15:1396354. [PMID: 38873428 PMCID: PMC11169668 DOI: 10.3389/fphar.2024.1396354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Natural polyphenols may have a role in counteracting oxidative stress, which is associated with aging and several bone-related diseases. Chlorogenic acid (CGA) is a naturally occurring polyphenolic compound formed by the esterification of caffeic and quininic acids with osteogenic, antioxidant, and anti-inflammatory properties. This review discusses the potential of CGA to enhance osteogenesis by increasing the osteogenic capacity of mesenchymal stem cells (MSCs), osteoblast survival, proliferation, differentiation, and mineralization, as well as its ability to attenuate osteoclastogenesis by enhancing osteoclast apoptosis and impeding osteoclast regeneration. CGA can be involved in bone remodeling by acting directly on pro-osteoclasts/osteoblasts or indirectly on osteoclasts by activating the nuclear factor kB (RANK)/RANK ligand (RANKL)/acting osteoprotegerin (OPG) system. Finally, we provide perspectives for using CGA to treat bone diseases.
Collapse
Affiliation(s)
- Jiayu Shen
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| | - Shichen Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| | - Jiayu Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| | - Xin Wei
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| | - Zilin Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| | - Bing Han
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Jilin University, Changchun, China
| |
Collapse
|
37
|
Marozin S, Simon-Nobbe B, Huth A, Beyerer E, Weber L, Nüssler A, Lepperdinger G. Aggregation of human osteoblasts unlocks self-reliant differentiation and constitutes a microenvironment for 3D-co-cultivation with other bone marrow cells. Sci Rep 2024; 14:10345. [PMID: 38710795 PMCID: PMC11074281 DOI: 10.1038/s41598-024-60986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
Skeletal bone function relies on both cells and cellular niches, which, when combined, provide guiding cues for the control of differentiation and remodeling processes. Here, we propose an in vitro 3D model based on human fetal osteoblasts, which eases the study of osteocyte commitment in vitro and thus provides a means to examine the influences of biomaterials, substances or cells on the regulation of these processes. Aggregates were formed from human fetal osteoblasts (hFOB1.19) and cultivated under proliferative, adipo- and osteoinductive conditions. When cultivated under osteoinductive conditions, the vitality of the aggregates was compromised, the expression levels of the mineralization-related gene DMP1 and the amount of calcification and matrix deposition were lower, and the growth of the spheroids stalled. However, within spheres under growth conditions without specific supplements, self-organization processes occur, which promote extracellular calcium deposition, and osteocyte-like cells develop. Long-term cultivated hFOB aggregates were free of necrotic areas. Moreover, hFOB aggregates cultivated under standard proliferative conditions supported the co-cultivation of human monocytes, microvascular endothelial cells and stromal cells. Overall, the model presented here comprises a self-organizing and easily accessible 3D osteoblast model for studying bone marrow formation and in vitro remodeling and thus provides a means to test druggable molecular pathways with the potential to promote life-long bone formation and remodeling.
Collapse
Affiliation(s)
- Sabrina Marozin
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria.
| | - Birgit Simon-Nobbe
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria
| | - Astrid Huth
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria
| | - Evelyn Beyerer
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria
| | - Laurenz Weber
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria
| | - Andreas Nüssler
- Siegfried Weller Institut (SWI) | BG Klinik Tübingen, Tübingen, Germany
| | - Günter Lepperdinger
- Department of Biosciences and Medical Biology, University Salzburg, 5020, Salzburg, Austria
| |
Collapse
|
38
|
Guo DG, Zhu J, Wang HJ, Pan BW. Investigating the Effects and Mechanisms of Cyclomorusin on Osteoclasts in a High Glucose Environment. Chem Biodivers 2024; 21:e202301741. [PMID: 38477870 DOI: 10.1002/cbdv.202301741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
Diabetes mellitus is an endocrine disease characterized by prolonged hyperglycemia. Prolonged high blood sugar levels interfere with the differentiation and maturation process of OBs and OCs, leading to the onset of osteoporosis. However, OCs differentiation and maturation is a complex regulatory process. In this study, we used a co-culture system of RAW264.7 and MC3T3-E1 cells under HG concentration to explore the effect of CYM on OCs in a HG environment. The effects of CYM on the formation and function of OCs were observed using TRAP-positive cell counts and bone resorption pits. Then, mRNA and protein expression levels of OCs-related genes were detected by real-time qPCR and western blotting. The results showed that CYM had an inhibitory effect on OCs differentiation and bone resorption, reduced mRNAs expression of OCs-associated genes, and downregulated RANKL/RANK/TRAF6 pathway that mediates OCs differentiation. CYM could be a promising natural compound against diabetic osteoporosis.
Collapse
Affiliation(s)
- Dong-Gui Guo
- College of Food and Pharmaceutical Engineering, Guizhou Institute of Technology, Guiyang, 550025, China
| | - Jun Zhu
- National & Local Joint Engineering Research Center for the Exploition of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Hui-Juan Wang
- National & Local Joint Engineering Research Center for the Exploition of Homology Resources of Southwest Medicine and Food, Guizhou University, Guiyang, 550025, China
| | - Bo-Wen Pan
- College of pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| |
Collapse
|
39
|
Gupta N, Kanwar N, Arora A, Khatri K, Kanwal A. The interplay of rheumatoid arthritis and osteoporosis: exploring the pathogenesis and pharmacological approaches. Clin Rheumatol 2024; 43:1421-1433. [PMID: 38499817 DOI: 10.1007/s10067-024-06932-5] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
Rheumatoid arthritis (RA) and osteoporosis are two chronic disorders that are often seen together. RA is an autoimmune disorder that causes pain and inflammation in the joints, while osteoporosis is a disorder in which the bones become weak and fragile. Risk factors for bone loss in RA include disease activity, longer disease duration, erosive disease, autoantibody positivity, and joint damage leading to impaired physical activity. Recent research has shown that there is a complex interplay between immune cells, cytokines, and bone remodeling processes in both RA and osteoporosis. The bone remodeling process is regulated by cytokines and immune system signaling pathways, with osteoclasts activated through the RANK/RANKL/OPG pathway and the Wnt/DKK1/sclerostin pathway. Understanding these mechanisms can aid in developing targeted therapies for treatment of osteoporosis in RA patients. Current pharmacological approaches include anti-osteoporotic drugs such as bisphosphonates, denosumab, teriparatide, abaloparatide, raloxifene, and romosozumab. Conventional disease-modifying antirheumatic drugs such as methotrexate and biologicals including TNF inhibitors, IL-6 inhibitors, rituximab, and abatacept lower disease activity in RA and can improve bone metabolism by reducing inflammation but have limited impact on bone mineral density. This review will shed light on the relationship between osteoporosis and rheumatoid arthritis as well as the various factors that influence the onset of osteoporosis in RA patients. We also explore several treatment approaches to effectively managing osteoporosis in RA patients.
Collapse
Affiliation(s)
- Nikhil Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
| | - Anchal Arora
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Kavin Khatri
- Department of Orthopedics, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India.
| |
Collapse
|
40
|
Faqeer A, Liu J, Zhang L, Wang C, Zhou G, Zhang Y. Establishment and validation of an efficient method for the 3D culture of osteoclasts in vitro. J Dent 2024; 144:104957. [PMID: 38527517 DOI: 10.1016/j.jdent.2024.104957] [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: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION Osteoclasts (OCs) play a crucial role in maintaining bone health. Changes in OC activity are linked to different bone diseases, making them an intriguing focus for research. However, most studies on OCs have relied on 2D cultures, limiting our understanding of their behavior. Yet, there's a lack of knowledge regarding platforms that effectively support osteoclast formation in 3D cultures. METHODS In our investigation, we explored the capacity of collagen and GelMA hydrogels to facilitate osteoclast development in 3D culture settings. We assessed the osteoclast development by using different hydrogels and cell seeding strategies and optimizing cell seeding density and cytokine concentration. The osteoclast development in 3D cultures was further validated by biochemical assays and immunochemical staining. RESULTS Our findings revealed that 0.3 % (w/v) collagen was conducive to osteoclast formation in both 2D and 3D cultures, demonstrated by increased multinucleation and higher TRAP activity compared to 0.6 % collagen and 5 % to 10 % (w/v) GelMA hydrogels. Additionally, we devised a "sandwich" technique using collagen substrates and augmented the initial macrophage seeding density and doubling cytokine concentrations, significantly enhancing the efficiency of OC culture in 3D conditions. Notably, we validated osteoclasts derived from macrophages in our 3D cultures express key osteoclast markers like cathepsin K and TRAP. CONCLUSIONS To conclude, our study contributes to establishing an effective method for cultivating osteoclasts in 3D environments in vitro. This innovative approach not only promises a more physiologically relevant platform to study osteoclast behavior during bone remodeling but also holds potential for applications in bone tissue engineering. CLINICAL SIGNIFICANCE This study introduces an efficient method for cultivating osteoclasts in 3D environments in vitro. It offers a more physiologically relevant platform to investigate osteoclast behavior and holds promise to advance research in bone biology and regenerative dentistry.
Collapse
Affiliation(s)
- Abdullah Faqeer
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China; School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
| | - Jie Liu
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Geriatric Orthopeadics, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518118, China
| | - Li Zhang
- Department of Stomatology, Shenzhen Children's Hospital, Shenzhen 518026, China
| | - Changde Wang
- Department of Geriatric Orthopeadics, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518118, China
| | - Guangqian Zhou
- School of Basic Medicine, Shenzhen University Medical School, Shenzhen 518015, China.
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China; School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China.
| |
Collapse
|
41
|
Kontogianni GI, Bonatti AF, De Maria C, Naseem R, Coelho C, Alpantaki K, Batsali A, Pontikoglou C, Quadros P, Dalgarno K, Vozzi G, Vitale-Brovarone C, Chatzinikolaidou M. Cell Instructive Behavior of Composite Scaffolds in a Co-Culture of Human Mesenchymal Stem Cells and Peripheral Blood Mononuclear Cells. J Funct Biomater 2024; 15:116. [PMID: 38786628 PMCID: PMC11122527 DOI: 10.3390/jfb15050116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
The in vitro evaluation of 3D scaffolds for bone tissue engineering in mono-cultures is a common practice; however, it does not represent the native complex nature of bone tissue. Co-cultures of osteoblasts and osteoclasts, without the addition of stimulating agents for monitoring cellular cross-talk, remains a challenge. In this study, a growth factor-free co-culture of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and human peripheral blood mononuclear cells (hPBMCs) has been established and used for the evaluation of 3D-printed scaffolds for bone tissue engineering. The scaffolds were produced from PLLA/PCL/PHBV polymeric blends, with two composite materials produced through the addition of 2.5% w/v nanohydroxyapatite (nHA) or strontium-substituted nanohydroxyapatite (Sr-nHA). Cell morphology data showed that hPBMCs remained undifferentiated in co-culture, while no obvious differences were observed in the mono- and co-cultures of hBM-MSCs. A significantly increased alkaline phosphatase (ALP) activity and osteogenic gene expression was observed in co-culture on Sr-nHA-containing scaffolds. Tartrate-resistant acid phosphatase (TRAP) activity and osteoclastogenic gene expression displayed significantly suppressed levels in co-culture on Sr-nHA-containing scaffolds. Interestingly, mono-cultures of hPBMCs on Sr-nHA-containing scaffolds indicated a delay in osteoclasts formation, as evidenced from TRAP activity and gene expression, demonstrating that strontium acts as an osteoclastogenesis inhibitor. This co-culture study presents an effective 3D model to evaluate the regenerative capacity of scaffolds for bone tissue engineering, thus minimizing time-consuming and costly in vivo experiments.
Collapse
Affiliation(s)
| | - Amedeo Franco Bonatti
- Research Center E. Piaggio, Department of Information Engineering, University of Pisa, 56126 Pisa, Italy; (A.F.B.); (C.D.M.); (G.V.)
| | - Carmelo De Maria
- Research Center E. Piaggio, Department of Information Engineering, University of Pisa, 56126 Pisa, Italy; (A.F.B.); (C.D.M.); (G.V.)
| | - Raasti Naseem
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (R.N.); (K.D.)
| | | | - Kalliopi Alpantaki
- Department of Orthopaedics and Trauma, Venizeleion General Hospital of Heraklion, 70013 Heraklion, Greece;
| | - Aristea Batsali
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 70013 Heraklion, Greece; (A.B.); (C.P.)
| | - Charalampos Pontikoglou
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, 70013 Heraklion, Greece; (A.B.); (C.P.)
| | - Paulo Quadros
- FLUIDINOVA, S.A., 4475-188 Maia, Portugal; (C.C.); (P.Q.)
| | - Kenneth Dalgarno
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (R.N.); (K.D.)
| | - Giovanni Vozzi
- Research Center E. Piaggio, Department of Information Engineering, University of Pisa, 56126 Pisa, Italy; (A.F.B.); (C.D.M.); (G.V.)
| | | | - Maria Chatzinikolaidou
- Department of Materials Science and Engineering, University of Crete, 70013 Heraklion, Greece;
- Foundation for Research and Technology Hellas (FO.R.T.H)-IESL, 70013 Heraklion, Greece
| |
Collapse
|
42
|
Li X, Chen W, Liu D, Chen P, Wang S, Li F, Chen Q, Lv S, Li F, Chen C, Guo S, Yuan W, Li P, Hu Z. Pathological progression of osteoarthritis: a perspective on subchondral bone. Front Med 2024; 18:237-257. [PMID: 38619691 DOI: 10.1007/s11684-024-1061-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 04/16/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.
Collapse
Affiliation(s)
- Xuefei Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenhua Chen
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pinghua Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiyun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangfang Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qian Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shunyi Lv
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangyu Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Suxia Guo
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weina Yuan
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pan Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhijun Hu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
43
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
44
|
Pinho AR, Gomes MC, Costa DCS, Mano JF. Bioactive Self-Regulated Liquified Microcompartments to Bioengineer Bone-Like Microtissues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305029. [PMID: 37847901 DOI: 10.1002/smll.202305029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Designing a microenvironment that drives autonomous stromal cell differentiation toward osteogenesis while recapitulating the complexity of bone tissue remains challenging. In the current study, bone-like microtissues are created using electrohydrodynamic atomization to form two distinct liquefied microcapsules (mCAPs): i) hydroxypyridinone (HOPO)-modified gelatin (GH mCAPs, 7.5% w/v), and ii) HOPO-modified gelatin and dopamine-modified gelatin (GH+GD mCAPs, 7.5%+1.5% w/v). The ability of HOPO to coordinate with iron ions at physiological pH allows the formation of a semipermeable micro-hydrogel shell. In turn, the dopamine affinity for calcium ions sets a bioactive milieu for bone-like microtissues. After 21 days post encapsulation, GH and GH+GD mCAPs potentiate autonomous osteogenic differentiation of mesenchymal stem cells accompanied by collagen type-I gene upregulation, increased alkaline phosphatase (ALP) expression, and formation of mineralized extracellular matrix. However, the GH+GD mCAPs show higher levels of osteogenic markers starting on day 14, translating into a more advanced and organized mineralized matrix. The GH+GD system also shows upregulation of the receptor activator of nuclear factor kappa-B ligand (RANK-L) gene, enabling the autonomous osteoclastic differentiation of monocytes. These catechol-based mCAPs offer a promising approach to designing multifunctional and autonomous bone-like microtissues to study in vitro bone-related processes at the cell-tissue interface, angiogenesis, and osteoclastogenesis.
Collapse
Affiliation(s)
- Ana R Pinho
- CICECO, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Maria C Gomes
- CICECO, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Dora C S Costa
- CICECO, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- CICECO, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
45
|
Tan Y, Zhang M, Kong Y, Zhang F, Wang Y, Huang Y, Song W, Li Z, Hou L, Liang L, Guo X, Liu Q, Feng Y, Zhang C, Fu X, Huang S. Fibroblasts and endothelial cells interplay drives hypertrophic scar formation: Insights from in vitro and in vivo models. Bioeng Transl Med 2024; 9:e10630. [PMID: 38435816 PMCID: PMC10905555 DOI: 10.1002/btm2.10630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/23/2023] [Accepted: 11/24/2023] [Indexed: 03/05/2024] Open
Abstract
Hypertrophic scar formation is influenced by the intricate interplay between fibroblasts and endothelial cells. In this study, we investigated this relationship using in vitro and in vivo models. Clinical observations revealed distinct morphological changes and increased vascularity at pathological scar sites. Further analysis using OCTA, immunohistochemistry, and immunofluorescence confirmed the involvement of angiogenesis in scar formation. Our indirect co-culture systems demonstrated that endothelial cells enhance the proliferation and migration of fibroblasts through the secretion of cytokines including VEGF, PDGF, bFGF, and TGF-β. Additionally, a suspended co-culture multicellular spheroid model revealed molecular-level changes associated with extracellular matrix remodeling, cellular behaviors, inflammatory response, and pro-angiogenic activity. Furthermore, KEGG pathway analysis identified the involvement of TGF-β, IL-17, Wnt, Notch, PI3K-Akt, and MAPK pathways in regulating fibroblasts activity. These findings underscore the critical role of fibroblasts-endothelial cells crosstalk in scar formation and provide potential targets for therapeutic intervention. Understanding the molecular mechanisms underlying this interplay holds promise for the development of innovative approaches to treat tissue injuries and diseases.
Collapse
Affiliation(s)
- Yaxin Tan
- College of GraduateTianjin Medical UniversityTianjinPR China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Mengde Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Yi Kong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Fanliang Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Yuzhen Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Yuyan Huang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Linhao Hou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Liting Liang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Xu Guo
- College of GraduateTianjin Medical UniversityTianjinPR China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Qinghua Liu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Yu Feng
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Chao Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Xiaobing Fu
- College of GraduateTianjin Medical UniversityTianjinPR China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingPR China
| |
Collapse
|
46
|
Jasiński T, Turek B, Kaczorowski M, Brehm W, Skierbiszewska K, Bonecka J, Domino M. Equine Models of Temporomandibular Joint Osteoarthritis: A Review of Feasibility, Biomarkers, and Molecular Signaling. Biomedicines 2024; 12:542. [PMID: 38540155 PMCID: PMC10968442 DOI: 10.3390/biomedicines12030542] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 11/11/2024] Open
Abstract
Osteoarthritis (OA) of the temporomandibular joint (TMJ) occurs spontaneously in humans and various animal species, including horses. In humans, obtaining tissue samples is challenging and clinical symptoms appear late in the disease progression. Therefore, genetically modified, induced, and naturally occurring animal models play a crucial role in understanding the pathogenesis and evaluating potential therapeutic interventions for TMJ OA. Among the naturally occurring models, the equine TMJ OA model is characterized by slow, age-related progression, a wide range of clinical examinations, and imaging modalities that can be performed on horses, as well as easy tissue and synovial fluid collection. The morphological and functional similarities of TMJ structures in both species make the equine model of TMJ OA an excellent opportunity to track disease progression and response to treatment. However, much work remains to be carried out to determine the utility of human TMJ OA biomarkers in horses. Among the main TMJ OA biomarkers, IL-1, IL-6, TGF-β, TNF-α, and PGE2 have been recently investigated in the equine model. However, the majority of biomarkers for cartilage degradation, chondrocyte hypertrophy, angiogenesis, and TMJ overload-as well as any of the main signaling pathways-have not been studied so far. Therefore, it would be advisable to focus further research on equine specimens, considering both mediators and signaling.
Collapse
Affiliation(s)
- Tomasz Jasiński
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland; (T.J.); (K.S.)
| | - Bernard Turek
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland; (T.J.); (K.S.)
| | | | - Walter Brehm
- Department for Horses, Veterinary Teaching Hospital, University of Leipzig, 04103 Leipzig, Germany;
| | - Katarzyna Skierbiszewska
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland; (T.J.); (K.S.)
| | - Joanna Bonecka
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland;
| | - Małgorzata Domino
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland; (T.J.); (K.S.)
| |
Collapse
|
47
|
Capobianco CA, Hankenson KD, Knights AJ. Temporal dynamics of immune-stromal cell interactions in fracture healing. Front Immunol 2024; 15:1352819. [PMID: 38455063 PMCID: PMC10917940 DOI: 10.3389/fimmu.2024.1352819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Bone fracture repair is a complex, multi-step process that involves communication between immune and stromal cells to coordinate the repair and regeneration of damaged tissue. In the US, 10% of all bone fractures do not heal properly without intervention, resulting in non-union. Complications from non-union fractures are physically and financially debilitating. We now appreciate the important role that immune cells play in tissue repair, and the necessity of the inflammatory response in initiating healing after skeletal trauma. The temporal dynamics of immune and stromal cell populations have been well characterized across the stages of fracture healing. Recent studies have begun to untangle the intricate mechanisms driving the immune response during normal or atypical, delayed healing. Various in vivo models of fracture healing, including genetic knockouts, as well as in vitro models of the fracture callus, have been implemented to enable experimental manipulation of the heterogeneous cellular environment. The goals of this review are to (1): summarize our current understanding of immune cell involvement in fracture healing (2); describe state-of-the art approaches to study inflammatory cells in fracture healing, including computational and in vitro models; and (3) identify gaps in our knowledge concerning immune-stromal crosstalk during bone healing.
Collapse
Affiliation(s)
- Christina A. Capobianco
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Alexander J. Knights
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
48
|
Şeker Ş, Aral D, Elçin AE, Yaşar Murat E. Biomimetic mineralization of platelet lysate/oxidized dextran cryogel as a macroporous 3D composite scaffold for bone repair. Biomed Mater 2024; 19:025006. [PMID: 38194711 DOI: 10.1088/1748-605x/ad1c9a] [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: 08/18/2023] [Accepted: 01/09/2024] [Indexed: 01/11/2024]
Abstract
Scaffold development approaches using autologous sources for tissue repair are of great importance in obtaining bio-active/-compatible constructs. Platelet-rich plasma (PRP) containing various growth factors and platelet lysate (PL) derived from PRP are autologous products that have the potential to accelerate the tissue repair response by inducing a transient inflammatory event. Considering the regenerative capacity of PRP and PL, PRP/PL-based scaffolds are thought to hold great promise for tissue engineering as a natural source of autologous growth factors and a provider of mechanical support for cells. Here, a bio-mineralized PRP-based scaffold was developed using oxidized dextran (OD) and evaluated for future application in bone tissue engineering. Prepared PL/OD scaffolds were incubated in simulated body fluid (SBF) for 7, 14 and 21 d periods. Mineralized PL/OD scaffolds were characterized using Fourier transform infrared spectroscopy, x-ray diffraction spectroscopy, scanning electron microscopy (SEM), thermogravimetric analysis, porosity and compression tests. SEM and energy-dispersive x-ray spectroscopy analyses revealed mineral accumulation on the PL/OD scaffold as a result of SBF incubation.In vitrocytotoxicity andin vitrohemolysis tests revealed that the scaffolds were non-toxic and hemocompatible. Additionally, human osteoblasts (hOBs) exhibited good attachment and spreading behavior on the scaffolds and maintained their viability throughout the culture period. The alkaline phosphatase activity assay and calcium release results revealed that PL/OD scaffolds preserved the osteogenic properties of hOBs. Overall, findings suggest that mineralized PL/OD scaffold may be a promising scaffold for bone tissue engineering.
Collapse
Affiliation(s)
- Şükran Şeker
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Dilara Aral
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Elçin Yaşar Murat
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
- Biovalda Health Technologies, Inc., Ankara, Turkey
| |
Collapse
|
49
|
Zhao J, Zhou C, Xiao Y, Zhang K, Zhang Q, Xia L, Jiang B, Jiang C, Ming W, Zhang H, Long H, Liang W. Oxygen generating biomaterials at the forefront of regenerative medicine: advances in bone regeneration. Front Bioeng Biotechnol 2024; 12:1292171. [PMID: 38282892 PMCID: PMC10811251 DOI: 10.3389/fbioe.2024.1292171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Globally, an annual count of more than two million bone transplants is conducted, with conventional treatments, including metallic implants and bone grafts, exhibiting certain limitations. In recent years, there have been significant advancements in the field of bone regeneration. Oxygen tension regulates cellular behavior, which in turn affects tissue regeneration through metabolic programming. Biomaterials with oxygen release capabilities enhance therapeutic effectiveness and reduce tissue damage from hypoxia. However, precise control over oxygen release is a significant technical challenge, despite its potential to support cellular viability and differentiation. The matrices often used to repair large-size bone defects do not supply enough oxygen to the stem cells being used in the regeneration process. Hypoxia-induced necrosis primarily occurs in the central regions of large matrices due to inadequate provision of oxygen and nutrients by the surrounding vasculature of the host tissues. Oxygen generating biomaterials (OGBs) are becoming increasingly significant in enhancing our capacity to facilitate the bone regeneration, thereby addressing the challenges posed by hypoxia or inadequate vascularization. Herein, we discussed the key role of oxygen in bone regeneration, various oxygen source materials and their mechanism of oxygen release, the fabrication techniques employed for oxygen-releasing matrices, and novel emerging approaches for oxygen delivery that hold promise for their potential application in the field of bone regeneration.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Yang Xiao
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Kunyan Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Qiang Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Bo Jiang
- Rehabilitation Department, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chanyi Jiang
- Department of Pharmacy, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenyi Ming
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengjian Zhang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
50
|
Park Y, Sato T, Lee J. Functional and analytical recapitulation of osteoclast biology on demineralized bone paper. Nat Commun 2023; 14:8092. [PMID: 38062034 PMCID: PMC10703810 DOI: 10.1038/s41467-023-44000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Osteoclasts are the primary target for osteoporosis drug development. Recent animal studies revealed the crucial roles of osteoblasts in regulating osteoclastogenesis and the longer lifespans of osteoclasts than previously thought with fission and recycling. However, existing culture platforms are limited to replicating these newly identified cellular processes. We report a demineralized bone paper (DBP)-based osteoblast culture and osteoclast assay platform that replicates osteoclast fusion, fission, resorption, and apoptosis with high fidelity and analytical power. An osteoid-inspired DBP supports rapid and structural mineral deposition by osteoblasts. Coculture osteoblasts and bone marrow monocytes under biochemical stimulation recapitulate osteoclast differentiation and function. The DBP-based bone model allows longitudinal quantitative fluorescent monitoring of osteoclast responses to bisphosphonate drug, substantiating significantly reducing their number and lifespan. Finally, we demonstrate the feasibility of humanizing the bone model. The DBP-based osteo assay platforms are expected to advance bone remodeling-targeting drug development with improved prediction of clinical outcomes.
Collapse
Affiliation(s)
- Yongkuk Park
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Tadatoshi Sato
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|