1
|
Kabir A, B M, A N, Selvaraj V, Sudhakar S. Protein Nano Coop Complexes Promote Fracture Healing and Bone Regeneration in a Zebrafish Osteoporosis Model. Biomacromolecules 2024. [PMID: 39449233 DOI: 10.1021/acs.biomac.4c00931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Nanotherapeutic techniques are becoming increasingly important in the treatment of bone disorders owing to their targeted drug delivery. This study formulates zein nano coop composites containing chimeric antioxidants (ascorbic acid, luteolin, resveratrol, and coenzyme Q) (AZN) and evaluates its application in bone regeneration using osteoblasts and a Zebrafish osteoporosis model. In vitro experiments with human osteoblast-like MG63 cells showed enhancement of bone mineralization and regeneration. It further exhibited high biocompatibility in Zebrafish larvae, with increased calcium/phosphorus deposition and upregulation of osteogenic genes. The study has unequivocally demonstrated the potential of AZN in bone regeneration and fracture healing in both normal and osteoporosis models, underscoring the significance of this research. Further investigations using higher animal models are warranted to expand on these findings. The impact of this research seems far-reaching, with the possible development of new, effective, and safe treatment options for osteoporosis, addressing the limitations of the currently available treatments.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Mukilarasi B
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Natarajan A
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| |
Collapse
|
2
|
Orji C, Ojo C, Onobun DE, Igbokwe K, Khaliq F, Ononye R. Fracture Non-Union in Osteoporotic Bones: Current Practice and Future Directions. Cureus 2024; 16:e69778. [PMID: 39429299 PMCID: PMC11491133 DOI: 10.7759/cureus.69778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
Given the compromised bone quality and altered healing environment, fracture non-union in osteoporotic bones presents a complex challenge in orthopedics. As global populations age, the incidence of osteoporotic fractures rises, leading to increased delayed healing and non-union cases. The pathophysiology underlying non-union in osteoporotic patients involves impaired bone regeneration, reduced osteoblast function, and poor vascularity. Traditional management strategies - ranging from pharmacological interventions like bisphosphonates and teriparatides to surgical approaches such as bone grafting and mechanical fixation - often yield limited success due to the weakened bone structure. Recent advances, however, have introduced novel therapies such as growth factors, stem cell applications, gene therapy, and bioactive scaffolds that offer more targeted and biologically driven solutions. Emerging technologies like three-dimensional printing and nanotechnology further contribute to customized treatment strategies that hold promise for improved outcomes. Diagnostic approaches have also evolved, integrating radiological assessments and biomarkers to identify patients at risk for non-union better. Despite these advancements, challenges remain, including the high costs, technical complexities, and the need for more robust clinical evidence. Future directions involve optimizing these innovative treatments, validating their effectiveness in more extensive clinical trials, and integrating personalized medicine approaches to cater to the individual needs of osteoporotic patients. Overall, integrating these emerging therapeutic strategies alongside traditional practices represents a significant shift towards more effective and personalized management of fracture non-union in osteoporotic bones.
Collapse
Affiliation(s)
- Chijioke Orji
- Department of Trauma and Orthopedics, Liverpool University Hospitals NHS Foundation Trust, Liverpool, GBR
| | - Charles Ojo
- Department of Emergency Medicine, United Lincolnshire Hospitals NHS Trust, Boston, GBR
| | - Daniel E Onobun
- Department of Neurological Surgery, Wellington Neurosurgery Centre, Abuja, NGA
| | | | - Farihah Khaliq
- Department of Trauma and Orthopedics, Aintree University Hospital, Liverpool, GBR
| | - Reginald Ononye
- Department of Neurological Surgery, Wellington Neurosurgery Centre, Abuja, NGA
| |
Collapse
|
3
|
Chen C, Wang B, Zhao X, Luo Y, Fu L, Qi X, Ying Z, Chen L, Wang Q, Sun S, Chen D, Kang P. Lithium Promotes Osteogenesis via Rab11a-Facilitated Exosomal Wnt10a Secretion and β-Catenin Signaling Activation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30793-30809. [PMID: 38833412 DOI: 10.1021/acsami.4c04199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Both bone mesenchymal stem cells (BMSCs) and their exosomes suggest promising therapeutic tools for bone regeneration. Lithium has been reported to regulate BMSC function and engineer exosomes to improve bone regeneration in patients with glucocorticoid-induced osteonecrosis of the femoral head. However, the mechanisms by which lithium promotes osteogenesis have not been elucidated. Here, we demonstrated that lithium promotes the osteogenesis of BMSCs via lithium-induced increases in the secretion of exosomal Wnt10a to activate Wnt/β-catenin signaling, whose secretion is correlated with enhanced MARK2 activation to increase the trafficking of the Rab11a and Rab11FIP1 complexes together with exosomal Wnt10a to the plasma membrane. Then, we compared the proosteogenic effects of exosomes derived from lithium-treated or untreated BMSCs (Li-Exo or Con-Exo) both in vitro and in vivo. We found that, compared with Con-Exo, Li-Exo had superior abilities to promote the uptake and osteogenic differentiation of BMSCs. To optimize the in vivo application of these hydrogels, we fabricated Li-Exo-functionalized gelatin methacrylate (GelMA) hydrogels, which are more effective at promoting osteogenesis and bone repair than Con-Exo. Collectively, these findings demonstrate the mechanism by which lithium promotes osteogenesis and the great promise of lithium for engineering BMSCs and their exosomes for bone regeneration, warranting further exploration in clinical practice.
Collapse
Affiliation(s)
- Changjun Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Baoning Wang
- Department of Microbiology, West China of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xin Zhao
- Department of Orthopedic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, China
| | - Yue Luo
- Department of Orthopedic Surgery, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Li Fu
- Research Core Facility, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Qi
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhendong Ying
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Liyile Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuru Wang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuo Sun
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dailing Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Pengde Kang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Gao J, Ren J, Ye H, Chu W, Ding X, Ding L, Fu Y. Thymosin beta 10 loaded ZIF-8/sericin hydrogel promoting angiogenesis and osteogenesis for bone regeneration. Int J Biol Macromol 2024; 267:131562. [PMID: 38626832 DOI: 10.1016/j.ijbiomac.2024.131562] [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: 02/08/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Angiogenesis is pivotal for osteogenesis during bone regeneration. A hydrogel that promotes both angiogenesis and osteogenesis is essential in bone tissue engineering. However, creating scaffolds with the ideal balance of biodegradability, osteogenic, and angiogenic properties poses a challenge. Thymosin beta 10 (TMSB10), known for its dual role in angiogenesis and osteogenesis differentiation, faces limitations due to protein activity preservation. To tackle this issue, ZIF-8 was engineered as a carrier for TMSB10 (TMSB10@ZIF-8), and subsequently integrated into the self-assembled sericin hydrogel. The efficacy of the composite hydrogel in bone repair was assessed using a rat cranial defect model. Characterization of the nanocomposites confirmed the successful synthesis of TMSB10@ZIF-8, with a TMSB10 encapsulation efficiency of 88.21 %. The sustained release of TMSB10 from TMSB10@ZIF-8 has significantly enhanced tube formation in human umbilical vein endothelial cells (HUVECs) in vitro and promoted angiogenesis in the chicken chorioallantoic membrane (CAM) model in vivo. It has markedly improved the osteogenic differentiation ability of MC 3 T3-E1 cells in vitro. 8 weeks post-implantation, the TMSB10@ZIF-8/ Sericin hydrogel group exhibited significant bone healing (86.77 ± 8.91 %), outperforming controls. Thus, the TMSB10@ZIF-8/Sericin hydrogel, leveraging ZIF-8 for TMSB10 delivery, emerges as a promising bone regeneration scaffold with substantial clinical application potential.
Collapse
Affiliation(s)
- Jia Gao
- Taizhou Key Laboratory of Biomass Functional Materials Development and Application, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, PR China; College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin 130000, PR China
| | - Hanjie Ye
- Taizhou Key Laboratory of Biomass Functional Materials Development and Application, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, PR China
| | - Wenhui Chu
- Taizhou Key Laboratory of Biomass Functional Materials Development and Application, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, PR China.
| | - Xuankai Ding
- Taizhou Key Laboratory of Biomass Functional Materials Development and Application, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, PR China; College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, PR China
| | - Lingzhi Ding
- Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang 318000, PR China
| | - Yongqian Fu
- Taizhou Key Laboratory of Biomass Functional Materials Development and Application, School of Life Science, Taizhou University, Taizhou, Zhejiang 318000, PR China.
| |
Collapse
|
5
|
Chen J, Wu W, Xian C, Wang T, Hao X, Chai N, Liu T, Shang L, Wang B, Gao J, Bi L. Analysis of risk factors and development of a nomogram-based prediction model for defective bony non-union. Heliyon 2024; 10:e28502. [PMID: 38586399 PMCID: PMC10998093 DOI: 10.1016/j.heliyon.2024.e28502] [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: 12/19/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Objective To explore risk factors for defective non-union of bone and develop a nomogram-based prediction model for such an outcome. Methods This retrospective study analysed the case data of patients with defective bony non-unions who were treated at the authors' hospital between January 2010 and December 2020. Patients were divided into the union and non-union groups according to their Radiographic Union Score for Tibia scores 1 year after surgery. Univariate analysis was performed to assess factors related to demographic characteristics, laboratory investigations, surgery, and trauma in both groups. Subsequently, statistically significant factors were included in the multivariate logistic regression analysis to identify independent risk factors. A nomogram-based prediction model was established using statistically significant variables in the multivariate analysis. The accuracy and stability of the model were evaluated using receiver operating characteristic (ROC) and calibration curves. The clinical applicability of the nomogram model was evaluated using decision curve analysis. Results In total, 204 patients (171 male, 33 female; mean [±SD] age, 39.75 ± 13.00 years) were included. The mean body mass index was 22.95 ± 3.64 kg/m2. Among the included patients, 29 were smokers, 18 were alcohol drinkers, and 21 had a previous comorbid systemic disease (PCSD). Univariate analysis revealed that age, occupation, PCSD, smoking, drinking, interleukin-6, C-reactive protein (CRP), procalcitonin, alkaline phosphatase, glucose, and uric acid levels; blood calcium ion concentration; and bone defect size (BDS) were correlated with defective bone union (all P < 0.05). Multivariate logistic regression analysis revealed that PCSD, smoking, interleukin-6, CRP, and glucose levels; and BDS were associated with defective bone union (all P < 0.05), and the variables in the multivariate analysis were included in the nomogram-based prediction model. The value of the area under the ROC curve for the predictive model for bone defects was 0.95. Conclusion PCSD, smoking, interleukin-6, CRP, and glucose levels; and BDS were independent risk factors for defective bony non-union, and the incidence of such non-union was predicted using the nomogram. These findings are important for clinical interventions and decision-making.
Collapse
Affiliation(s)
- Jingdi Chen
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
- Department of Orthopedics, 95829 Military Hospital in PLA, Wuhan, 430000, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Chunxing Xian
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Taoran Wang
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Xiaotian Hao
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Na Chai
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Tao Liu
- Department of Orthopedics, 95829 Military Hospital in PLA, Wuhan, 430000, China
| | - Lei Shang
- Department of Health Statistics, Air Force Medical University, Xi'an, 710032, China
| | - Bo Wang
- Department of Epidemiology, School of Public Health, Air Force Medical University, Xi'an, 710032, China
| | - Jiakai Gao
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Long Bi
- Department of Orthopedics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| |
Collapse
|
6
|
Souza ATP, Freitas GP, Lopes HB, Weffort D, Adolpho LF, Gomes MPO, Oliveira FS, Almeida ALG, Beloti MM, Rosa AL. Mesenchymal stem cell-based therapy for osteoporotic bones: Effects of the interaction between cells from healthy and osteoporotic rats on osteoblast differentiation and bone repair. Life Sci 2024; 340:122463. [PMID: 38286209 DOI: 10.1016/j.lfs.2024.122463] [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: 11/24/2023] [Revised: 01/09/2024] [Accepted: 01/23/2024] [Indexed: 01/31/2024]
Abstract
AIMS Cell therapy utilizing mesenchymal stem cells (MSCs) from healthy donors (HE-MSCs) is a promising strategy for treating osteoporotic bone defects. This study investigated the effects of interaction between HE-MSCs and MSCs from osteoporotic donors (ORX-MSCs) on osteoblast differentiation of MSCs and of HE-MSCs on bone formation in calvarial defects of osteoporotic rats. MATERIALS AND METHODS Osteoporosis was induced by orchiectomy (ORX) and its effects on the bone were evaluated by femur microtomography (μCT) and osteoblast differentiation of bone marrow MSCs. HE- and ORX-MSCs were cocultured, and osteoblast differentiation was evaluated using genotypic and phenotypic parameters. HE-MSCs were injected into the calvarial defects of osteoporotic rats, and bone formation was evaluated by μCT, histology, and gene expression of osteoblast markers. KEY FINDINGS ORX-induced osteoporosis was revealed by reduced bone morphometric parameters and osteoblast differentiation in ORX-MSCs. HE-MSCs partially recovered the osteogenic potential of ORX-MSCs, whereas HE-MSCs were mildly affected by ORX-MSCs. Additionally, the bone morphogenetic protein and wingless-related integration site signaling pathway components were similarly modulated in cocultures involving ORX-MSCs. HE-MSCs induced meaningful bone formation, highlighting the effectiveness of cell therapy even in osteoporotic bones. SIGNIFICANCE These results provide new perspectives on the development of cell-based therapies to regenerate bone defects in patients with disorders that affect bone tissue.
Collapse
Affiliation(s)
- Alann Thaffarell Portilho Souza
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil; Centro Universitário Metropolitano da Amazônia, Belém 66053-000, PA, Brazil
| | - Gileade Pereira Freitas
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Helena Bacha Lopes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Denise Weffort
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Leticia Faustino Adolpho
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Maria Paula Oliveira Gomes
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Fabiola Singaretti Oliveira
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | | | - Marcio Mateus Beloti
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Adalberto Luiz Rosa
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil.
| |
Collapse
|
7
|
Wen J, Cai D, Gao W, He R, Li Y, Zhou Y, Klein T, Xiao L, Xiao Y. Osteoimmunomodulatory Nanoparticles for Bone Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13040692. [PMID: 36839060 PMCID: PMC9962115 DOI: 10.3390/nano13040692] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 05/27/2023]
Abstract
Treatment of large bone fractures remains a challenge for orthopedists. Bone regeneration is a complex process that includes skeletal cells such as osteoblasts, osteoclasts, and immune cells to regulate bone formation and resorption. Osteoimmunology, studying this complicated process, has recently been used to develop biomaterials for advanced bone regeneration. Ideally, a biomaterial shall enable a timely switch from early stage inflammatory (to recruit osteogenic progenitor cells) to later-stage anti-inflammatory (to promote differentiation and terminal osteogenic mineralization and model the microstructure of bone tissue) in immune cells, especially the M1-to-M2 phenotype switch in macrophage populations, for bone regeneration. Nanoparticle (NP)-based advanced drug delivery systems can enable the controlled release of therapeutic reagents and the delivery of therapeutics into specific cell types, thereby benefiting bone regeneration through osteoimmunomodulation. In this review, we briefly describe the significance of osteoimmunology in bone regeneration, the advancement of NP-based approaches for bone regeneration, and the application of NPs in macrophage-targeting drug delivery for advanced osteoimmunomodulation.
Collapse
Affiliation(s)
- Jingyi Wen
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Donglin Cai
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222, Australia
| | - Wendong Gao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Ruiying He
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430061, China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200231, China
| | - Yinghong Zhou
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Travis Klein
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Lan Xiao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Yin Xiao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
8
|
Lončar SR, Halcrow SE, Swales D. Osteoimmunology: The effect of autoimmunity on fracture healing and skeletal analysis. Forensic Sci Int Synerg 2023; 6:100326. [PMID: 37091290 PMCID: PMC10120377 DOI: 10.1016/j.fsisyn.2023.100326] [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: 01/24/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 04/25/2023]
Abstract
Understanding factors that affect bone response to trauma is integral to forensic skeletal analysis. It is essential in forensic anthropology to identify if impaired fracture healing impacts assessment of post-traumatic time intervals and whether a correction factor is required. This paper presents a synthetic review of the intersection of the literature on the immune system, bone biology, and osteoimmunological research to present a novel model of interactions that may affect fracture healing under autoimmune conditions. Results suggest that autoimmunity likely impacts fracture healing, the pathogenesis however, is under researched, but likely multifactorial. With autoimmune diseases being relatively common, significant clinical history should be incorporated when assessing skeletal remains. Future research includes the true natural healing rate of bone; effect of autoimmunity on this rate; variation of healing with different autoimmune diseases; and if necessary, development of a correction factor on the natural healing rate to account for impairment in autoimmunity.
Collapse
Affiliation(s)
- Stephie R. Lončar
- Centre for Anatomy and Human Identification, School of Science and Engineering, University of Dundee, Scotland, United Kingdom
- Department of Anatomy, University of Otago, New Zealand
- Corresponding author. Centre for Anatomy and Human Identification School of Science and Engineering, MSI/WTB Complex, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, United Kingdom.
| | - Siân E. Halcrow
- Department of Anatomy, University of Otago, New Zealand
- Corresponding author. Biological Anthropology Research Group, Department of Anatomy, 270 Great King Street, University of Otago, Dunedin, 9016, New Zealand.
| | - Diana Swales
- Centre for Anatomy and Human Identification, School of Science and Engineering, University of Dundee, Scotland, United Kingdom
| |
Collapse
|
9
|
Ge YW, Chu M, Zhu ZY, Ke QF, Guo YP, Zhang CQ, Jia WT. Nacre-inspired magnetically oriented micro-cellulose fibres/nano-hydroxyapatite/chitosan layered scaffold enhances pro-osteogenesis and angiogenesis. Mater Today Bio 2022; 16:100439. [PMID: 36245833 PMCID: PMC9557728 DOI: 10.1016/j.mtbio.2022.100439] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 12/01/2022] Open
Affiliation(s)
- Yu-Wei Ge
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Min Chu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| | - Zi-Yang Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qin-Fei Ke
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| | - Ya-Ping Guo
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
- Corresponding author.
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
- Corresponding author.
| | - Wei-Tao Jia
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
- Corresponding author.
| |
Collapse
|
10
|
Qiu W, Liu B, Li X, Tian Y. Novel application of 3D printed microporous prosthesis to repair humeral nonunion with segmental bone defects: a case report. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:840. [PMID: 36034998 PMCID: PMC9403914 DOI: 10.21037/atm-22-447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023]
Abstract
Background Nonunion of the humeral shaft can turn into bone defects. There is no consensus on the optimal treatment of humeral shaft nonunion with bone defects. Herein, we presented a single case of a patient with a 9.5 cm humerus shaft bone defect treated with a 3D printed Ti6Al4V microporous prosthesis after internal fixation failure of a middle-inferior humerus fracture. Case Description A 53-year-old female who injured her left upper limb by falling was diagnosed with a fracture of the left humeral shaft. The fracture was treated with open reduction and internal fixation. Nine months postoperatively, radiography examination indicated humeral nonunion with a 9.5 cm segmental bone defect. A 3D printing technology was then used to design and fabricate a customized microporous prosthesis with an intramedullary nail and lateral plates. A two-stage surgical strategy was performed, including radical debridement, temporary fixation for the induced membrane formation, and the implantation of the prosthesis. At 18 months of follow-up, encouraging clinical outcomes were observed. The prosthesis remained stable in the original implantation area and callus formation was found at the contact end of the prosthesis and bone stump. The upper limb functions returned to normal with a satisfactory functional score. Also, no complications were found. Conclusions Reconstruction with a 3D printed microporous prosthesis might be used as an alternative for the repair of large segmental bone defects of limbs.
Collapse
Affiliation(s)
- Weipeng Qiu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China;,Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Bingchuan Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China;,Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Xingcai Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China;,Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Yun Tian
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China;,Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
11
|
Mukherjee P, Roy S, Ghosh D, Nandi SK. Role of animal models in biomedical research: a review. Lab Anim Res 2022; 38:18. [PMID: 35778730 PMCID: PMC9247923 DOI: 10.1186/s42826-022-00128-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
The animal model deals with the species other than the human, as it can imitate the disease progression, its’ diagnosis as well as a treatment similar to human. Discovery of a drug and/or component, equipment, their toxicological studies, dose, side effects are in vivo studied for future use in humans considering its’ ethical issues. Here lies the importance of the animal model for its enormous use in biomedical research. Animal models have many facets that mimic various disease conditions in humans like systemic autoimmune diseases, rheumatoid arthritis, epilepsy, Alzheimer’s disease, cardiovascular diseases, Atherosclerosis, diabetes, etc., and many more. Besides, the model has tremendous importance in drug development, development of medical devices, tissue engineering, wound healing, and bone and cartilage regeneration studies, as a model in vascular surgeries as well as the model for vertebral disc regeneration surgery. Though, all the models have some advantages as well as challenges, but, present review has emphasized the importance of various small and large animal models in pharmaceutical drug development, transgenic animal models, models for medical device developments, studies for various human diseases, bone and cartilage regeneration model, diabetic and burn wound model as well as surgical models like vascular surgeries and surgeries for intervertebral disc degeneration considering all the ethical issues of that specific animal model. Despite, the process of using the animal model has facilitated researchers to carry out the researches that would have been impossible to accomplish in human considering the ethical prohibitions.
Collapse
Affiliation(s)
- P Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - S Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - D Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - S K Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India.
| |
Collapse
|
12
|
Effects of Systemic or Local Administration of Mesenchymal Stem Cells from Patients with Osteoporosis or Osteoarthritis on Femoral Fracture Healing in a Mouse Model. Biomolecules 2022; 12:biom12050722. [PMID: 35625649 PMCID: PMC9138345 DOI: 10.3390/biom12050722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
The purpose of this study was to analyze the regenerative capacity of mesenchymal stem cells (MSCs) in the treatment of fractures. MSCs extracted from patients with osteoporotic hip fractures or hip osteoarthritis undergoing hip replacement surgeries were cultured and injected into mice with femoral fracture. Two experimental models were established, one for the systemic administration of MSCs (n = 29) and another one for local administration (n = 30). Fracture consolidation was assessed by micro-CT and histology. The degree of radiological consolidation and corticalization was better with MSCs from osteoporosis than from osteoarthritis, being significant after systemic administration (p = 0.0302 consolidation; p = 0.0243 corticalization). The histological degree of consolidation was also better with MSCs from osteoporosis than from osteoarthritis. Differences in histological scores after systemic infusion were as follows: Allen, p = 0.0278; Huo, p = 0.3471; and Bone Bridge, p = 0.0935. After local administration at the fracture site, differences in histological scores were as follows: Allen, p = 0.0764; Huo, p = 0.0256; and Bone Bridge, p = 0.0012. As osteoporosis and control groups were similar, those differences depended on an inhibitory influence by MSCs from patients with osteoarthritis. In conclusion, we found an unexpected impairment of consolidation induced by MSCs from patients with osteoarthritis. However, MSCs from patients with osteoporosis compared favorably with cells from patients with osteoarthritis. In other words, based on this study and previous studies, MSCs from patients with osteoporosis do not appear to have worse bone-regenerating capabilities than MSCs from non-osteoporotic individuals of similar age.
Collapse
|
13
|
Strontium based Astragalus polysaccharides promote osteoblasts differentiation and mineralization. Int J Biol Macromol 2022; 205:761-771. [PMID: 35318083 DOI: 10.1016/j.ijbiomac.2022.03.088] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/23/2022]
Abstract
Bone formation and repair represent a clinical challenge. In this work, we designed and synthesized strontium Astragalus polysaccharide (APS-Sr), a novel polysaccharide compound that should have therapeutic effects on both anti-inflammation and promoting bone formation. Using material characterization techniques, including SEM, FITR, XRD, etc., we verified the successful synthesis of this compound. Moreover, we examined the potential of this compound for promoting bone repair and inhibiting inflammatory response by cell proliferation assay, ALP and Alizarin Red staining experiments and RT-qPCR. The biological experiment results showed that APS-Sr can effectively inhibit inflammatory factors, promote osteogenic differentiation and up-regulate the bone growth factors. It is therefore believed that APS-Sr should be a promising polysaccharide compound in bone-related biomedical applications.
Collapse
|
14
|
Sun X, Jiao X, Yang X, Ma J, Wang T, Jin W, Li W, Yang H, Mao Y, Gan Y, Zhou X, Li T, Li S, Chen X, Wang J. 3D bioprinting of osteon-mimetic scaffolds with hierarchical microchannels for vascularized bone tissue regeneration. Biofabrication 2022; 14. [PMID: 35417902 DOI: 10.1088/1758-5090/ac6700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/13/2022] [Indexed: 11/12/2022]
Abstract
The integration of three-dimensional (3D) bioprinted scaffold's structure and function for critical-size bone defect repair is of immense significance. Inspired by the basic component of innate cortical bone tissue--osteons, many studies focus on biomimetic strategy. However, the complexity of hierarchical microchannels in the osteon, the requirement of mechanical strength of bone, and the biological function of angiogenesis and osteogenesis remain challenges in the fabrication of osteon-mimetic scaffolds. Therefore, we successfully built mimetic scaffolds with vertically central medullary canals, peripheral Haversian canals, and transverse Volkmann canals structures simultaneously by 3D bioprinting technology using polycaprolactone and bioink loading with bone marrow mesenchymal stem cells (BMSCs) and bone morphogenetic protein-4 (BMP-4). Subsequently, endothelial progenitor cells (EPCs) were seeded into the canals to enhance angiogenesis. The porosity and compressive properties of bioprinted scaffolds could be well controlled by altering the structure and canal numbers of the scaffolds. The osteon-mimetic scaffolds showed satisfactory biocompatibility and promotion of angiogenesis and osteogenesis in vitro and prompted the new blood vessels and new bone formation in vivo. In summary, this study proposes a biomimetic strategy for fabricating structured and functionalized 3D bioprinted scaffolds for vascularized bone tissue regeneration.
Collapse
Affiliation(s)
- Xin Sun
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Xin Jiao
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Xue Yang
- College of Medicine, Southwest JiaoTong University, No. 111 2nd Ring Rd, Chengdu, 610031, CHINA
| | - Jie Ma
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Tianchang Wang
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Wenjie Jin
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Wentao Li
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Han Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai, 200030, CHINA
| | - Yuanqing Mao
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Yaokai Gan
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Xiaojun Zhou
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, No. 2999 North Renmin Road, Shanghai, 201620, CHINA
| | - Tao Li
- Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, CHINA
| | - Shuai Li
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| | - Xiaodong Chen
- Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, CHINA
| | - Jinwu Wang
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200001, CHINA
| |
Collapse
|
15
|
Zhang H, Fu X, Zhang S, Li Q, Chen Y, Liu H, Zhou W, Wei S. Strategy of Stem Cell Transplantation for Bone Regeneration with Functionalized Biomaterials and Vascularized Tissues in Immunocompetent Mice. ACS Biomater Sci Eng 2022; 8:1656-1666. [PMID: 35341241 DOI: 10.1021/acsbiomaterials.1c01512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The use of human bone marrow mesenchymal stem cells (hBMSCs) to regenerate and repair bone tissue defects is a complex research field of bone tissue engineering; nevertheless, it is a hot topic. One of the biggest problems is the limited survival and osteogenic capacity of the transplanted cells within the host tissue. Even for hBMSCs with their low immunogenicity, the body will still cause a local immune-inflammatory response directed against the allogeneic cells and thereby reduce the activity of the transplanted cells. Even in the case of successful transplantation, the lack of vascularization at the transplantation site makes it difficult for the transplanted cells to exchange nutrients and metabolic wastes that ultimately affects bone regeneration. In this study, we covalently modified alginate with RGD and QK peptides that were injected subcutaneously into immunocompetent mice. Histological analysis, as well as ELISA techniques, proved that this method is able to provide bioactive stem cell transplant beds containing functionalized biomaterials and vascularized surrounding tissues. Inflammation-related factors, such as IL-2, IL-6, TNF-α, and IFN-γ, around the cell graft beds decreased with time and were lowest at the second week. Then, the hBMSCs were injected into the cell transplantation beds intended to form vascularized bonelike tissues that were evaluated by micro-computed tomography (Micro CT), histological, and immunohistochemical analyses. The results showed that the expression of osteogenesis-related proteins RUNX2, COL1A1, and OPN, as well as the expression of angiogenic factor vWF and cartilage-related protein COL2A1 were significantly upregulated in the hBMSC-derived osteogenic tissue. These results suggest that the stem cell transplantation strategy by constructing bioactive cell transplant beds is effective to enhance the bone regeneration capacity of hBMSCs and holds great potential in bone tissue engineering.
Collapse
Affiliation(s)
- He Zhang
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P. R. China
| | - Xiaoming Fu
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P. R. China
| | - Siqi Zhang
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P. R. China
| | - Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P. R. China
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P. R. China
| | - Hao Liu
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P. R. China
| | - Wen Zhou
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P. R. China
| | - Shicheng Wei
- Central Laboratory, and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P. R. China.,Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P. R. China
| |
Collapse
|
16
|
Wei M, Hsu YI, Asoh TA, Sung MH, Uyama H. Design of Injectable Poly(γ-glutamic acid)/Chondroitin Sulfate Hydrogels with Mineralization Ability. ACS APPLIED BIO MATERIALS 2022; 5:1508-1518. [PMID: 35286062 DOI: 10.1021/acsabm.1c01269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Biocompatible hydrogels are considered promising agents for application in bone tissue engineering. However, the design of reliable hydrogels with satisfactory injectability, mechanical strength, and a rapid biomineralization rate for bone regeneration remains challenging. Herein, injectable hydrogels are fabricated using hydrazide-modified poly(γ-glutamic acid) and oxidized chondroitin sulfate by combining acylhydrazone bonds and ionic bonding of carboxylic acid groups or sulfate groups with calcium ions (Ca2+). The resulting hydrogels display a fast gelation rate and good self-healing ability due to the acylhydrazone bonds. The introduction of Ca2+ at a moderate concentration enhances the mechanical strength of the hydrogels. The self-healing capacity of hydrogels is improved, with a healing efficiency of 87.5%, because the addition of Ca2+ accelerates the healing process of hydrogels. Moreover, the hydrogels can serve as a robust template for biomineralization. The mineralized hydrogels with increasing Ca2+ concentration exhibit rapid formation and high crystallization of apatite after immersion in simulated body fluid. The hydrogels containing the aldehyde groups possess good bioadhesion to the bone and cartilage tissues. With these superior properties, the developed hydrogels demonstrate potential applicability in bone tissue engineering.
Collapse
Affiliation(s)
- Meng Wei
- Department of Applied Chemistry, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yu-I Hsu
- Department of Applied Chemistry, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taka-Aki Asoh
- Department of Applied Chemistry, Osaka University, Suita, Osaka 565-0871, Japan
| | - Moon-Hee Sung
- Department of Advanced Fermentation Fusion Science and Technology, Kookmin University, Seongbuk-gu, Seoul 136-702, Korea
| | - Hiroshi Uyama
- Department of Applied Chemistry, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Yan MD, Ou YJ, Lin YJ, Liu RM, Fang Y, Wu WL, Zhou L, Yao X, Chen J. Does the incorporation of strontium into calcium phosphate improve bone repair? A meta-analysis. BMC Oral Health 2022; 22:62. [PMID: 35260122 PMCID: PMC8905839 DOI: 10.1186/s12903-022-02092-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 02/21/2022] [Indexed: 12/09/2022] Open
Abstract
Background The application of calcium phosphate (CaP)-based bone substitutes plays an important role in periodontal regeneration, implant dentistry and alveolar bone reconstruction. The incorporation of strontium (Sr) into CaP-based bone substitutes appears to improve their biological properties, but the reported in vivo bone repair performance is inconsistent among studies. Herein, we conducted a systematic review and meta-analysis to investigate the in vivo performance of Sr-doped materials. Methods We searched PubMed, EMBASE (via OVIDSP), and reference lists to identify relevant animal studies. The search, study selection, and data extraction were performed independently by two investigators. Meta-analyses and sub-group analyses were conducted using Revman version 5.4.1. The heterogeneity between studies were assessed by I2. Publication bias was investigated through a funnel plot. Results Thirty-five studies were finally enrolled, of which 16 articles that reported on new bone formation (NBF) were included in the meta-analysis, covering 31 comparisons and 445 defects. The overall effect for NBF was 2.25 (95% CI 1.61–2.90, p < 0.00001, I2 = 80%). Eight comparisons from 6 studies reported the outcomes of bone volume/tissue volume (BV/TV), with an overall effect of 1.42 (95% CI 0.65–2.18, p = 0.0003, I2 = 75%). Fourteen comparisons reported on the material remaining (RM), with the overall effect being -2.26 (95% CI − 4.02 to − 0.50, p = 0.0009, I2 = 86%). Conclusions Our study revealed that Sr-doped calcium phosphate bone substitutes improved in vivo performance of bone repair. However, more studies are also recommended to further verify this conclusion. Supplementary Information The online version contains supplementary material available at 10.1186/s12903-022-02092-7.
Collapse
Affiliation(s)
- Ming-Dong Yan
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China
| | - Yan-Jing Ou
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China.,Department of Oral Implantology, Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, 350002, China
| | - Yan-Jun Lin
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China
| | - Rui-Min Liu
- ORAL Center, Fujian Provincial Governmental Hospital (Affiliated Hospital of Fujian Health College), Fuzhou, 350003, China
| | - Yan Fang
- Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, 350007, China
| | - Wei-Liang Wu
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China
| | - Lin Zhou
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China
| | - Xiu Yao
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China.,Institute of Stomatology and Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jiang Chen
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China.
| |
Collapse
|
18
|
Chakraborty N, Zamarioli A, Gautam A, Campbell R, Mendenhall SK, Childress PJ, Dimitrov G, Sowe B, Tucker A, Zhao L, Hammamieh R, Kacena MA. Gene-metabolite networks associated with impediment of bone fracture repair in spaceflight. Comput Struct Biotechnol J 2021; 19:3507-3520. [PMID: 34194674 PMCID: PMC8220416 DOI: 10.1016/j.csbj.2021.05.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/26/2021] [Accepted: 05/30/2021] [Indexed: 01/05/2023] Open
Abstract
Adverse effects of spaceflight on musculoskeletal health increase the risk of bone injury and impairment of fracture healing. Its yet elusive molecular comprehension warrants immediate attention, since space travel is becoming more frequent. Here we examined the effects of spaceflight on bone fracture healing using a 2 mm femoral segmental bone defect (SBD) model. Forty, 9-week-old, male C57BL/6J mice were randomized into 4 groups: 1) Sham surgery on Ground (G-Sham); 2) Sham surgery housed in Spaceflight (FLT-Sham); 3) SBD surgery on Ground (G-Surgery); and 4) SBD surgery housed in Spaceflight (FLT-Surgery). Surgery procedures occurred 4 days prior to launch; post-launch, the spaceflight mice were house in the rodent habitats on the International Space Station (ISS) for approximately 4 weeks before euthanasia. Mice remaining on the Earth were subjected to identical housing and experimental conditions. The right femur from half of the spaceflight and ground groups was investigated by micro-computed tomography (µCT). In the remaining mice, the callus regions from surgery groups and corresponding femoral segments in sham mice were probed by global transcriptomic and metabolomic assays. µCT confirmed escalated bone loss in FLT-Sham compared to G-Sham mice. Comparing to their respective on-ground counterparts, the morbidity gene-network signal was inhibited in sham spaceflight mice but activated in the spaceflight callus. µCT analyses of spaceflight callus revealed increased trabecular spacing and decreased trabecular connectivity. Activated apoptotic signals in spaceflight callus were synchronized with inhibited cell migration signals that potentially hindered the wound site to recruit growth factors. A major pro-apoptotic and anti-migration gene network, namely the RANK-NFκB axis, emerged as the central node in spaceflight callus. Concluding, spaceflight suppressed a unique biomolecular mechanism in callus tissue to facilitate a failed regeneration, which merits a customized intervention strategy.
Collapse
Affiliation(s)
| | - Ariane Zamarioli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Orthopaedics and Anaesthesiology, Ribeirão Preto Medical School, SP, Brazil
| | - Aarti Gautam
- Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
| | - Ross Campbell
- Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
- Geneva Foundation, Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
| | - Stephen K Mendenhall
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul J. Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George Dimitrov
- Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
- Geneva Foundation, Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
| | - Bintu Sowe
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- ORISE, Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
| | - Aamir Tucker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liming Zhao
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, CMPN, WRAIR, Silver Spring, MD, USA
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
19
|
Musculoskeletal tissue engineering: Regional gene therapy for bone repair. Biomaterials 2021; 275:120901. [PMID: 34091300 DOI: 10.1016/j.biomaterials.2021.120901] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/24/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023]
Abstract
Bone loss associated with fracture nonunion, revision total joint arthroplasty (TJA), and pseudoarthrosis of the spine presents a challenging clinical scenario for the orthopaedic surgeon. Current treatment options including autograft, allograft, bone graft substitutes, and bone transport techniques are associated with significant morbidity, high costs, and prolonged treatment regimens. Unfortunately, these treatment strategies have proven insufficient to safely and consistently heal bone defects in the stringent biological environments often encountered in clinical cases of bone loss. The application of tissue engineering (TE) to musculoskeletal pathology has uncovered exciting potential treatment strategies for challenging bone loss scenarios in orthopaedic surgery. Regional gene therapy involves the local implantation of nucleic acids or genetically modified cells to direct specific protein expression, and has shown promise as a potential TE technique for the regeneration of bone. Preclinical studies in animal models have demonstrated the ability of regional gene therapy to safely and effectively heal critical sized bone defects which otherwise do not heal. The purpose of the present review is to provide a comprehensive overview of the current status of gene therapy applications for TE in challenging bone loss scenarios, with an emphasis on gene delivery methods and models, scaffold biomaterials, preclinical results, and future directions.
Collapse
|
20
|
Chu M, Sun Z, Fan Z, Yu D, Mao Y, Guo Y. Bi-directional regulation functions of lanthanum-substituted layered double hydroxide nanohybrid scaffolds via activating osteogenesis and inhibiting osteoclastogenesis for osteoporotic bone regeneration. Am J Cancer Res 2021; 11:6717-6734. [PMID: 34093849 PMCID: PMC8171081 DOI: 10.7150/thno.56607] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: Osteoporotic patients suffer symptoms of excessive osteoclastogenesis and impaired osteogenesis, resulting in a great challenge to treat osteoporosis-related bone defects. Based on the positive effect of rare earth elements on bone metabolism and bone regeneration, we try to prove the hypothesis that the La3+ dopants in lanthanum-substituted MgAl layered double hydroxide (La-LDH) nanohybrid scaffolds simultaneously activate osteogenesis and inhibit osteoclastogenesis. Methods: A freeze-drying technology was employed to construct La-LDH nanohybrid scaffolds. The in vitro osteogenic and anti-osteoclastogenic activities of La-LDH nanohybrid scaffolds were evaluated by using ovariectomized rat bone marrow stromal cells (rBMSCs-OVX) and bone marrow-derived macrophages (BMMs) as cell models. The in vivo bone regeneration ability of the scaffolds was investigated by using critical-size calvarial bone defect model of OVX rats. Results: La-LDH nanohybrid scaffolds exhibited three-dimensional macroporous structure, and La-LDH nanoplates arranged perpendicularly on chitosan organic matrix. The La3+ dopants in the scaffolds promote proliferation and osteogenic differentiation of rBMSCs-OVX by activating Wnt/β-catenin pathway, leading to high expression of ALP, Runx-2, COL-1 and OCN genes. Moreover, La-LDH scaffolds significantly suppressed RANKL-induced osteoclastogenesis by inhibiting NF-κB signaling pathway. As compared with the scaffolds without La3+ dopants, La-LDH scaffolds provided more favourable microenvironment to induce new bone in-growth along macroporous channels. Conclusion: La-LDH nanohybrid scaffolds possessed the bi-directional regulation functions on osteogenesis and osteoclastogenesis for osteoporotic bone regeneration. The modification of La3+ dopants in bone scaffolds provides a novel strategy for osteoporosis-related bone defect healing.
Collapse
|
21
|
Parathyroid Hormone Secretion and Receptor Expression Determine the Age-Related Degree of Osteogenic Differentiation in Dental Pulp Stem Cells. J Pers Med 2021; 11:jpm11050349. [PMID: 33925324 PMCID: PMC8144966 DOI: 10.3390/jpm11050349] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022] Open
Abstract
Objective: To demonstrate the levels of parathyroid hormone secretion and genetic expressions of parathyroid hormone (PTH) and PTH1 receptor (PTH1R) genes in the dental pulp stem cells (DPSCs) from different age groups before and after induction of osteogenic differentiation. In addition, we also wanted to check their correlation with the degree of osteogenic differentiation. Methods: Human primary DPSCs from three age groups (milk tooth (SHEDs), 7–12 years old; young DPSCs (yDPSCs), 20–40 years old; old DPSCs (oDPSCs), 60+ years old) were characterized for mesenchymal stem cell (MSC) markers. DPSCs were subjected to osteogenic differentiation and functional staining. Gene expression levels were analyzed by qRT-PCR. Surface receptor analysis was done by flow cytometry. Comparative protein levels were evaluated by ELISA. Results: All SHEDs, yDPSCs, and oDPSCs were found to be expressing mesenchymal stem cell markers. SHEDs showed more mineralization than yDPSCs and oDPSCs after osteogenic induction. SHEDs exhibited higher expression of PTH and PTH1R before and after osteogenic induction, and after osteogenic induction, SHEDs showed more expression for RUNX2, ALPL, and OCN. Higher levels of PTH were observed in SHEDs and yDPSCs, and the number of PTH1R positive cells was relatively lower in yDPSCs and oDPSCs than in SHEDs. After osteogenic induction, SHEDs were superior in the secretion of OPG, and the secretions of ALPL and PTH and the number of PTH1R positive cells were relatively low in the oDPSCs. Conclusions: The therapeutic quality of dental pulp stem cells is largely based on their ability to retain their stemness characteristics. This study emphasizes the criterion of aging, which affects the secretion of PTH by these cells, which in turn attenuates their osteogenic potential.
Collapse
|
22
|
Zhang R, Yang M, Li Y, Liu H, Ren M, Tao ZS. Effect of alendronate on the femoral metaphyseal defect under carbamazepine in ovariectomized rats. J Orthop Surg Res 2021; 16:14. [PMID: 33407695 PMCID: PMC7788868 DOI: 10.1186/s13018-020-02151-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The use of antiepileptic drugs and estrogen deficiency put forward higher requirements for bone defect regeneration. The present study investigated the effects of alendronate (ALN) on femoral bone defect in ovariectomized (OVX) rats under the influence of carbamazepine (CBZ). METHODS One hundred female SD rats at 3 months of age were either sham-operated or OVX and divided into four groups: sham control (CON); OVX control (OVX); ovariectomized rats treated with CBZ via gavage (75 mg/kg/day; CBZ); ovariectomized rats treated with CBZ plus ALN (2 mg/kg/day; CBZ-ALN). A critical-sized femoral metaphyseal bone defect was established in all female SD rats. Animals from the CBZ and CBZ-ALN groups received drugs by gavage the day after bone defect surgery was performed. After the rats were sacrificed, the defected area located in the distal femur was harvested for evaluation by microcomputed tomography (micro-CT), hematoxylin and eosin (HE) staining, and Masson's trichrome staining. The samples were also analyzed by biomechanics and immunohistochemical evaluation (IHC). Besides, biochemical analysis evaluates all serum samples. RESULTS The present study showed that ovariectomy changed the microstructural parameters of bone. The use of CBZ further decreased femur bone mass while treatment with ALN prevented bone loss. Compared to OVX and CBZ groups, CBZ-ALN group promoted bone neoformation and enhanced the ultimate load of the femur bone. However, the group of CBZ-ALN did not return to normal levels compared with the CON group. Besides, we noticed that CBZ-ALN group reduced tartrate-resistant acid phosphatase-5b (Tracp-5b) expression and had no significant effect on the expression of osteocalcin (OCN) and type I collagen (Col-I) in IHC compared with CBZ group. Biochemical analysis results presented that systemic delivery of CBZ showed pernicious effects on bone formation and resorption in ovariectomized rats, with the worse effects on C-terminal crosslinked telopeptide of type I collagen (CTX-1). Besides, a significant decrease in CTX-1 levels was observed in CBZ-ALN group as compared to the group of CBZ. CONCLUSION These results demonstrated that ALN can effectively reverse the effects of CBZ on the microarchitectural properties of bone, and thus can have a positive effect on local bone neoformation in rats with osteoporosis. CLINICAL RELEVANCE The dose of 2 mg/kg ALN improves the negative effect of prescription of CBZ at 75 mg/kg and promotes bone neoformation of femoral bony deficits.
Collapse
Affiliation(s)
- Ruotian Zhang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China
| | - Min Yang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China.
| | - Yang Li
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China
| | - Hedong Liu
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China
| | - Maoxian Ren
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China
| | - Zhou-Shan Tao
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital, No. 2, Zhe shan Xi Road, Anhui, 241001, Wuhu, People's Republic of China
| |
Collapse
|
23
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
24
|
Quercetin as an Agent for Protecting the Bone: A Review of the Current Evidence. Int J Mol Sci 2020; 21:ijms21176448. [PMID: 32899435 PMCID: PMC7503351 DOI: 10.3390/ijms21176448] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 01/02/2023] Open
Abstract
Quercetin is a flavonoid abundantly found in fruits and vegetables. It possesses a wide spectrum of biological activities, thus suggesting a role in disease prevention and health promotion. The present review aimed to uncover the bone-sparing effects of quercetin and its mechanism of action. Animal studies have found that the action of quercetin on bone is largely protective, with a small number of studies reporting negative outcomes. Quercetin was shown to inhibit RANKL-mediated osteoclastogenesis, osteoblast apoptosis, oxidative stress and inflammatory response while promoting osteogenesis, angiogenesis, antioxidant expression, adipocyte apoptosis and osteoclast apoptosis. The possible underlying mechanisms involved are regulation of Wnt, NF-κB, Nrf2, SMAD-dependent, and intrinsic and extrinsic apoptotic pathways. On the other hand, quercetin was shown to exert complex and competing actions on the MAPK signalling pathway to orchestrate bone metabolism, resulting in both stimulatory and inhibitory effects on bone in parallel. The overall interaction is believed to result in a positive effect on bone. Considering the important contributions of quercetin in regulating bone homeostasis, it may be considered an economical and promising agent for improving bone health. The documented preclinical findings await further validation from human clinical trials.
Collapse
|
25
|
Asgari M, Gazor R, Abdollahifar MA, Fadaei Fathabady F, Zare F, Norouzian M, Amini A, Khosravipour A, Kiani P, Atashgah RB, Rezaei F, Ghoreishi SK, Chien S, Hamblin MR, Bayat M. Combined therapy of adipose-derived stem cells and photobiomodulation on accelerated bone healing of a critical size defect in an osteoporotic rat model. Biochem Biophys Res Commun 2020; 530:173-180. [PMID: 32828282 DOI: 10.1016/j.bbrc.2020.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/03/2020] [Indexed: 12/29/2022]
Abstract
We investigated the impact of human demineralized bone matrix (hDBM) plus adipose-derived stem cells (hADS) plus photobiomodulation (PBM) on a critical-sized femoral defect (CSFD) in ovariectomy induced osteoporosis in rats. There were 6 groups as follows. In group 1 (control, C), only CSFDs were created. Groups 2-6 were implanted with DBM into the CSFD (DBM-CSFD). In group 2 (S), only DBM was transplanted into the CSFD. In group 3 (S + PBM), the DBM-CSFDs were treated with PBM. In group 4, the DBM-CSFDs were treated with alendronate (S + ALN). In group 5, ADSs were seeded into DBM-CSFD (S + ADS). In group 6, ADSs were seeded into DBM-CSFD and the CSFDs were treated with PBM (S + PBM + ADS). At week eight (catabolic phase of bone repair), the S + ALN, S + PBM + ADS, S + PBM, and S + ADS groups all had significantly increased bone strength than the S group (ANOVA, p = 0.000). The S + PBM, S + PBM + ADS, and S + ADS groups had significantly increased Hounsfield unit than the S group (ANOVA, p = 0.000). ALN, ADS, and PBM significantly increased healed bone strength in an experimental model of DBM-treated CSFD in the catabolic phase of bone healing in osteoporotic rats. However, ALN alone and PBM plus ADS were superior to the other protocols.
Collapse
Affiliation(s)
- Mehrdad Asgari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rouhallah Gazor
- Department of Anatomy, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Zare
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Armin Khosravipour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pejman Kiani
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Science, Tehran, Iran.
| | - Rahimeh B Atashgah
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 13169-43551, Iran.
| | - Fatemehsadat Rezaei
- University of Kentucky College of Pharmacy, 789 South Limestone, Lexington, KY, 40536, USA.
| | | | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, Noveratech LLC, Louisville, KY, USA.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Price Institute of Surgical Research, University of Louisville, Noveratech LLC, Louisville, KY, USA.
| |
Collapse
|
26
|
Goodman SB, Lin T. Modifying MSC Phenotype to Facilitate Bone Healing: Biological Approaches. Front Bioeng Biotechnol 2020; 8:641. [PMID: 32671040 PMCID: PMC7328340 DOI: 10.3389/fbioe.2020.00641] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Healing of fractures and bone defects normally follows an orderly series of events including formation of a hematoma and an initial stage of inflammation, development of soft callus, formation of hard callus, and finally the stage of bone remodeling. In cases of severe musculoskeletal injury due to trauma, infection, irradiation and other adverse stimuli, deficient healing may lead to delayed or non-union; this results in a residual bone defect with instability, pain and loss of function. Modern methods of mechanical stabilization and autologous bone grafting are often successful in achieving fracture union and healing of bone defects; however, in some cases, this treatment is unsuccessful because of inadequate biological factors. Specifically, the systemic and local microenvironment may not be conducive to bone healing because of a loss of the progenitor cell population for bone and vascular lineage cells. Autologous bone grafting can provide the necessary scaffold, progenitor and differentiated lineage cells, and biological cues for bone reconstruction, however, autologous bone graft may be limited in quantity or quality. These unfavorable circumstances are magnified in systemic conditions with chronic inflammation, including obesity, diabetes, chronic renal disease, aging and others. Recently, strategies have been devised to both mitigate the necessity for, and complications from, open procedures for harvesting of autologous bone by using minimally invasive aspiration techniques and concentration of iliac crest bone cells, followed by local injection into the defect site. More elaborate strategies (not yet approved by the U.S. Food and Drug Administration-FDA) include isolation and expansion of subpopulations of the harvested cells, preconditioning of these cells or inserting specific genes to modulate or facilitate bone healing. We review the literature pertinent to the subject of modifying autologous harvested cells including MSCs to facilitate bone healing. Although many of these techniques and technologies are still in the preclinical stage and not yet approved for use in humans by the FDA, novel approaches to accelerate bone healing by modifying cells has great potential to mitigate the physical, economic and social burden of non-healing fractures and bone defects.
Collapse
Affiliation(s)
- Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| |
Collapse
|
27
|
Kronemberger GS, Dalmônico GML, Rossi AL, Leite PEC, Saraiva AM, Beatrici A, Silva KR, Granjeiro JM, Baptista LS. Scaffold- and serum-free hypertrophic cartilage tissue engineering as an alternative approach for bone repair. Artif Organs 2020; 44:E288-E299. [PMID: 31950507 DOI: 10.1111/aor.13637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/25/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | | | | | - Paulo Emílio Correa Leite
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Antonio M Saraiva
- Laboratory of Macromolecules, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Anderson Beatrici
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Scientific and Technological Metrology Division (Dimci), National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói, Brazil
| | - Leandra Santos Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| |
Collapse
|
28
|
Zuchuat J, Manzano A, Decco O. Image-based analysis of trabecular bone in osteoporotic rabbits: correlation of morphological features and bone mineral density profile. Biomed Phys Eng Express 2019; 6:015005. [PMID: 33438593 DOI: 10.1088/2057-1976/ab575f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To analyse a set of morphological features from digital radiographs (RX) and bone mineral density (BMD) values estimated from quantitative computerized tomography scans (QCT scans) of the knee joint of an osteoporotic rabbits, and to determine the relationship and correlation of these variables to be considered as an alternative diagnosis method. METHODS The knee joint of rabbits (N = 9 ovariectomized and injected with of methylprednisolone sodium succinate (OVX + MPSS) and 3 sham operated healthy controls) were subjected to radiographic examination before the beginning of the study and after 6 weeks; after sacrifice, they were immediately scanned with a 64-channel CT. A set of morphological features was extracted from RX images and then subjected to Principal Component Analysis (PCA); BMD-values were calculated at different depths from the articular surface of the femur and the tibia. The selected morphological variables and the BMD values were correlated in order to determine useful information in medical diagnostics. RESULTS Ten morphological variables explained 80.39% of the total variability, but only some of them demonstrated significant differences between baseline and OVX + SSMP states and between OVX + SSMP and control. Spearman analysis showed higher positive and negative correlation of these parameters among them (r = 0.67, 0.81, 0.83 and 1); and a moderate correlation with the BMD values (r = 0.50, 0.52, 0.52 and 0.55) during the progression of osteoporosis (OP). CONCLUSION The Spearman's rank test supported the overall significant correlation between both, the morphological features and BMD values, making them as a reliable alternative option to the diagnosis of osteoporosis.
Collapse
Affiliation(s)
- Jésica Zuchuat
- Bioimplants Laboratory, Faculty of Engineering-National University of Entre Rios, Oro Verde-Entre Rios, Argentina
| | | | | |
Collapse
|
29
|
Ball AN, Phillips JN, McIlwraith CW, Kawcak CE, Samulski RJ, Goodrich LR. Genetic modification of scAAV-equine-BMP-2 transduced bone-marrow-derived mesenchymal stem cells before and after cryopreservation: An "off-the-shelf" option for fracture repair. J Orthop Res 2019; 37:1310-1317. [PMID: 30578639 PMCID: PMC8366205 DOI: 10.1002/jor.24209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/12/2018] [Indexed: 02/04/2023]
Abstract
Optimizing the environment of complex bone healing and improving treatment of catastrophic bone fractures and segmental bone defects remains an unmet clinical need both human and equine veterinary medical orthopaedics. The objective of this study was to determine whether scAAV-equine-BMP-2 transduced cells would induce osteogenesis in equine bone marrow derived mesenchymal stem cells (BMDMSCs) in vitro, and if these cells could be cryopreserved in an effort to osteogenically prime them as an "off-the-shelf" gene therapeutic approach for fracture repair. Our study found that transgene expression is altered by cell expansion, as would be expected by a transduction resulting in episomal transgene expression, and that osteoinductive levels could still be achieved 5 days after recovery, and protein expression would continue up to 14 days after transduction. This is the first evidence that cryopreservation of genetically modified BMDMSCs would not alter the osteoinductive potential or clinical use of allogeneic donor cells in cases of equine fracture repair. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1310-1317, 2019.
Collapse
Affiliation(s)
- Alyssa N. Ball
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jennifer N. Phillips
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Christopher E. Kawcak
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Richard J. Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Laurie R. Goodrich
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
30
|
Cai L, Huo Z, Yang H, He F, Cao Z, Wu F, Liu L, Sun B. Gene co-expression network analysis identifies BRCC3 as a key regulator in osteogenic differentiation of osteoblasts through a β-catenin signaling dependent pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:173-178. [PMID: 30834083 PMCID: PMC6396986 DOI: 10.22038/ijbms.2018.29498.7123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 09/25/2018] [Indexed: 11/17/2022]
Abstract
OBJECTIVES The prognosis of osteoporosis is very poor, and it is very important to identify a biomarker for prevention of osteoporosis. In this study, we aimed to identify candidate markers in osteoporosis and to investigate the role of candidate markers in osteogenic differentiation. MATERIALS AND METHODS Using Weighted Gene Co-Expression Network analysis, we identified three hub genes might associate with osteoporosis. The mRNA expression of hub genes in osteoblasts from osteoporosis patients or healthy donor was detected by qRT-PCR. Using siRNA and overexpression, we investigated the role of hub gene BRCC3 in osteogenic differentiation by alkaline phosphatase staining and Alizarin red staining. Moreover, the role of β-catenin signaling in the osteogenic differentiation was detected by using β-catenin signaling inhibitor XAV939. RESULTS We identified three hub genes that might associate with osteoporosis including BRCC3, UBE2N, and UBE2K. UBE2N mRNA and UBE2K mRNA were not changed in osteoblasts isolated from osteoporosis patients, compared with healthy donors, whereas BRCC3 mRNA was significantly increased. Depletion of BRCC3 promoted the activation of alkaline phosphatase and formation of calcified nodules in osteoblasts isolated from osteoporosis patients and up-regulated β-catenin expression. XAV939 reversed the BRCC3 siRNA-induced osteogenic differentiation. Additionally, inhibited osteogenic differentiation was also observed after BACC3 overexpression, and this was accompanied by decreased β-catenin expression. CONCLUSION BRCC3 is an important regulator for osteogenic differentiation of osteoblasts through β-catenin signaling, and it might be a promising target for osteoporosis treatment.
Collapse
Affiliation(s)
- Lixiong Cai
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Zhiqian Huo
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Haiyun Yang
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Fengchun He
- Department of Spine Osteopathy, Nanhai Hospital of Southen Medical University, Foshan 528000, China
| | - Zhenglin Cao
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Feng Wu
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Lianjun Liu
- Department of Ophthalmology Myopia Treatment, Foshan Aier Eye Hospital, Foshan 528000, China
| | - Bingyin Sun
- Department of Orthopaedics, Foshan Jiangxiang Hospital, Foshan 528200, China
| |
Collapse
|
31
|
Gong M, Huang C, Huang Y, Li G, Chi C, Ye J, Xie W, Shi R, Zhang L. Core-sheath micro/nano fiber membrane with antibacterial and osteogenic dual functions as biomimetic artificial periosteum for bone regeneration applications. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:124-136. [PMID: 30668985 DOI: 10.1016/j.nano.2019.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 02/07/2023]
Abstract
The traditional Chinese medicine icariin (ICA) and broad-spectrum antibacterial drug moxifloxacin hydrochloride (MOX) were introduced into a polycaprolactone core and gelatin shell, respectively, to develop osteogenic and antibacterial biomimetic periosteum by coaxial electrospinning. The physical properties, drug release, degradation, antibacterial property, in vitro and in vivo osteogenesis performances were investigated. Results demonstrated that stepwise and controlled drug release profiles were achieved based on the core-shell configuration and disparate degradation rate of PCL and gelatin. Only 20% ICA was released from this dual drug-loaded membrane after 1 month while the release of MOX was almost completed. Moreover, clear in vitro antibacterial effect and enhancement in osteogenic marker expressions including osteocalcin, type-I collagen expression, and calcium deposition were observed. Notably, the dual drug-loaded membrane displayed fascinating properties contributing to in vivo bone formation in terms of quality and quantity in a rabbit radius defect model.
Collapse
Affiliation(s)
- Min Gong
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China
| | - Chenlin Huang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China
| | - Yuelong Huang
- Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, PR China
| | - Guangping Li
- Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, PR China
| | - Cheng Chi
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China
| | - Jingjing Ye
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China
| | - Wenqi Xie
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China
| | - Rui Shi
- Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, PR China.
| | - Liqun Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, PR China; State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, PR China.
| |
Collapse
|
32
|
Sun X, Guo Q, Wei W, Robertson S, Yuan Y, Luo X. Current Progress on MicroRNA-Based Gene Delivery in the Treatment of Osteoporosis and Osteoporotic Fracture. Int J Endocrinol 2019; 2019:6782653. [PMID: 30962808 PMCID: PMC6431398 DOI: 10.1155/2019/6782653] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/28/2018] [Accepted: 12/31/2018] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence demonstrates that microRNAs, as important endogenous posttranscriptional regulators, are essential for bone remodeling and regeneration. Undoubtedly, microRNA-based gene therapies show great potential to become novel approaches against bone-related diseases, including osteoporosis and associated fractures. The major obstacles for continued advancement of microRNA-based therapies in clinical application include their poor in vivo stability, nonspecific biodistribution, and unwanted side effects. Appropriate chemical modifications and delivery vectors, which improve the biological performance and potency of microRNA-based drugs, hold the key to translating miRNA technologies into clinical practice. Thus, this review summarizes the current attempts and existing deficiencies of chemical modifications and delivery systems applied in microRNA-based therapies for osteoporosis and osteoporotic fractures to inform further explorations.
Collapse
Affiliation(s)
- Xi Sun
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, 138# Tongzipo Road, Changsha, Hunan 410007, China
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Wenhua Wei
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Stephen Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Ying Yuan
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
33
|
Wu D, Wang Z, Zheng Z, Geng Y, Zhang Z, Li Q, Zhou Q, Cao Y, Zhang ZY. Effects of physiological aging factor on bone tissue engineering repair based on fetal BMSCs. J Transl Med 2018; 16:324. [PMID: 30470235 PMCID: PMC6251213 DOI: 10.1186/s12967-018-1686-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/09/2018] [Indexed: 12/30/2022] Open
Abstract
Background At present, many laboratories and hospitals all over the world are attempting and exploring the clinical transformation of this tissue engineered bone graft (TEBG) strategy. Many successful cases of bone tissue engineering (BTE) repair were based on young individuals. But there are little studies about the effectiveness of TEBG strategy in physiological aged individuals. Methods In this research, we studied whether aging factor has influence on the skull repair effect of Fetal-TEBG, at the level of the large animal models. We used the fetal bone marrow stromal cells (Fetal-BMSCs) as the seed cells, combining the decalcified bone matrix (DBM) scaffolds, to repair the skull defects of the aged goats and the young goats. The repair effects on both aged goat and young goat were compared by Micro-CT and histology examination. Results The skull defects of the young goats could be repaired better than that of the aged goats after 6 months by Fetal-TEBG; In the aged goats, although not completely repaired, the defects repaired by Fetal-TEBG was better than that repaired by the Control DBM scaffold. Conclusions Aging factor has impact on the bone repair effect of Fetal-TEBG; and the BTE strategy is still efficacious even in the aged individuals. The improvement of the aged state may promote the repair effect of the BTE in the aged individuals. ![]()
Collapse
Affiliation(s)
- Dingyu Wu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwei Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Yingnan Geng
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhanzhao Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Qiannan Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Quan Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China.,Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Key Laboratory of Tissue Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China. .,Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou City, 510150, Guangdong Province, China.
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou City, 510150, Guangdong Province, China. .,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China.
| |
Collapse
|
34
|
Whitely M, Cereceres S, Dhavalikar P, Salhadar K, Wilems T, Smith B, Mikos A, Cosgriff-Hernandez E. Improved in situ seeding of 3D printed scaffolds using cell-releasing hydrogels. Biomaterials 2018; 185:194-204. [PMID: 30245387 DOI: 10.1016/j.biomaterials.2018.09.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/11/2018] [Accepted: 09/16/2018] [Indexed: 12/31/2022]
Abstract
The design of tissue engineered scaffolds based on polymerized high internal phase emulsions (polyHIPEs) has emerged as a promising bone grafting strategy. We previously reported the ability to 3D print emulsion inks to better mimic the structure and mechanical properties of native bone while precisely matching defect geometry. In the current study, redox-initiated hydrogel carriers were investigated for in situ delivery of human mesenchymal stem cells (hMSCs) utilizing the biodegradable macromer, poly(ethylene glycol)-dithiothreitol. Hydrogel carrier properties including network formation time, sol-gel fraction, and swelling ratio were modulated to achieve rapid cure without external stimuli and a target cell-release period of 5-7 days. These in situ carriers enabled improved distribution of hMSCs in 3D printed polyHIPE grafts over standard suspension seeding. Additionally, carrier-loaded polyHIPEs supported sustained cell viability and osteogenic differentiation of hMSCs post-release. In summary, these findings demonstrate the potential of this in situ curing hydrogel carrier to enhance the cell distribution and retention of hMSCs in bone grafts. Although initially focused on improving bone regeneration, the ability to encapsulate cells in a hydrogel carrier without relying on external stimuli that can be attenuated in large grafts or tissues is expected to have a wide range of applications in tissue engineering.
Collapse
Affiliation(s)
- Michael Whitely
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA.
| | - Stacy Cereceres
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA.
| | - Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Thomas Wilems
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Brandon Smith
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Antonios Mikos
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | | |
Collapse
|