1
|
Wang H, Zhang J, Li Z, Liu J, Chang H, Jia S, Di Z, Liu H, Wang J, Gao D, Wang C, Li G, Zhao X. NIR-programmable 3D-printed shape-memory scaffold with dual-thermal responsiveness for precision bone regeneration and bone tumor management. J Nanobiotechnology 2025; 23:300. [PMID: 40247322 PMCID: PMC12007331 DOI: 10.1186/s12951-025-03375-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Clinically, intraoperative treatment of bone tumors presents several challenges, including the effective inactivation of tumors and filling of irregular bone defects after tumor removal. In this study, intelligent thermosensitive composite materials with shape-memory properties were constructed using polylactic acid (PLA) and polycaprolactone (PCL), which have excellent biocompatibility and degradability. Additionally, beta-tricalcium phosphate (β-TCP), with its osteogenic properties, and magnesium (Mg) powder, with its photothermal and bone-promoting abilities, were incorporated to improve the osteogenic potential of the composite and enable the material to respond intelligently to near-infrared (NIR) light. Utilizing 3D printing technology, the composite material was prepared into an NIR-responsive shape-memory bone-filling implant that deforms when the scaffold temperature increases to 48 ℃ under NIR laser irradiation. Moreover, at a lower temperature of 42 ℃, mild photothermal therapy promotes macrophage polarization toward the M2 phenotype. This process regulates the secretion of interleukin (IL)-4, IL-10, tumor necrosis factor-α, IL-6, and bone morphogenetic protein (BMP)-2, reducing local inflammation, enhancing the release of pro-healing factors, and improving osteogenesis. Overall, this innovative scaffold is a promising and efficient treatment for filling irregular bone defects after bone tumor surgery.
Collapse
Affiliation(s)
- Hui Wang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Jiaxin Zhang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zuhao Li
- Department of Orthopaedics, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jiaqi Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Haoran Chang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Shipu Jia
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zexin Di
- Department of Orthopaedics, School of Economics and Management, Beihua University, Jilin, 132013, China
| | - He Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Jincheng Wang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Delong Gao
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, 130022, China.
| | - Chenyu Wang
- Department of Plastic & Reconstruct Surgery, First Hospital of Jilin University, Changchun, 130061, China.
| | - Guiwei Li
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun, 130025, China.
| | - Xin Zhao
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
2
|
Gao X, Yang J, Liu L, Hu Z, Lin R, Tang L, Yu M, Chen Z, Gao C, Zhang M, Li L, Ruan C, Liu Y. An electrostatic encapsulation strategy to motivate 3D-printed polyelectrolyte scaffolds for repair of osteoporotic bone defects. Bioact Mater 2025; 46:1-20. [PMID: 39719966 PMCID: PMC11665476 DOI: 10.1016/j.bioactmat.2024.12.007] [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: 08/03/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Repair of osteoporotic bone defects (OBD) remains a clinical challenge due to dysregulated bone homeostasis, characterized by impaired osteogenesis and excessive osteoclast activity. While drug-loaded 3D-printed scaffolds hold great potential in the restoration of bone homeostasis for enhanced OBD repair, achieving the controlled release and targeted delivery of drugs in a 3D-printed scaffold is still unmet. Herein, we developed an electrostatic encapsulation strategy to motivate 3D-printed polyelectrolyte scaffolds (APS@P) with bone-targeting liposome formulation of salvianolic acid B (SAB-BTL). Benefiting from this strategy, SAB, an unstable and untargetable plant-derived osteogenic compound, was effectively encapsulated in APS@P, demonstrating stable and precise delivery with improved therapeutic efficacy. Owing to SAB-mediated bone homeostasis, APS@P significantly promoted angiogenesis and new bone formation while suppressing bone resorption, resulting in a significant 146 % increase in bone mass and improved microstructure compared to the OBD group. It was confirmed that the encapsulation of SAB into APS@P could promote the osteogenic differentiation of MSCs by stimulating Tph2/Wnt/β-catenin signaling axis, coupled with the stimulation of type H angiogenesis and the suppression of RANKL-mediate bone resorption, thereby enhance OBD repair. This study provides a universal platform for enhancing the bioactivity of tissue-engineered scaffolds, offering an effective solution for the efficient regeneration of osteoporotic bone.
Collapse
Affiliation(s)
- Xiang Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Jirong Yang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingna Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Zilong Hu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Rui Lin
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Lan Tang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mei Yu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Zhiping Chen
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Chongjian Gao
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Li Li
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| | - Changshun Ruan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, China
| |
Collapse
|
3
|
Sun X, Zhong R, Wu C, Ye S, Yuan H, Fang Z, Chen J, Cheng D, Hao L, Chu L, Wang L. 3D Printed Titanium Scaffolds with Bi-Directional Gradient QK-Functionalized Surface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2406421. [PMID: 39811993 DOI: 10.1002/adma.202406421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 12/09/2024] [Indexed: 01/16/2025]
Abstract
3D printed titanium scaffold has promising applications in orthopedics. However, the bioinert titanium presents challenges for promoting vascularization and tissue growth within the porous scaffold for stable osteointegration. In this study, a modular porous titanium scaffold is created using 3D printing and a gradient-surface strategy to immobilize QK peptide on the surface with a bi-directional gradient distribution. This design featured high peptide density in the interior and low peptide density on both ends, aiming to induce cell migration from ends to interior and subsequently enhance vascularization and osteointegration within the scaffold. In vitro results showed that besides the inherent bioactivity, the gradient distribution of QK positively correlated with endothelial cell migration and promoted angiogenesis. In vivo assay was performed by a segmental bone defect model in rabbit and a spine repair model in sheep. Various staining and Micro-CT results demonstrated that compared to that with uniformly QK-functionalized surface, the scaffold with bi-directional gradient QK-functionalized surface (Ti-G) significantly encouraged new tissue growth toward the interior of the scaffold, subsequently facilitated angiogenesis and osteointegration. This study provides an effective strategy for enhancing the bioactivity of peptide-functionalized scaffolds through the concept of bi-directional gradients, and holds potential for various 3D printed scaffolds.
Collapse
Affiliation(s)
- Xiaoyun Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Ru Zhong
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Congcong Wu
- Jinan Center for Disease Control and Prevention, Jinan, 250001, China
| | - Silin Ye
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Haipeng Yuan
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Zhou Fang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| | - Junjian Chen
- Guangzhou Huare Medical Equipment Co., Ltd., Guangzhou, 511447, China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510000, China
| | - Delin Cheng
- Guangzhou Huare Medical Equipment Co., Ltd., Guangzhou, 511447, China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510000, China
| | - Lijing Hao
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Guangdong Institute of Advanced Biomaterials and Medical Devices, Guangzhou, 510000, China
| | - Lei Chu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lin Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- School of Material Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
4
|
Zhu S, Sun H, Mu T, Richel A. Research Progress in 3D Printed Biobased and Biodegradable Polyester/Ceramic Composite Materials: Applications and Challenges in Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2791-2813. [PMID: 39760202 DOI: 10.1021/acsami.4c15719] [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: 01/07/2025]
Abstract
Transplantation of bone implants is currently recognized as one of the most effective means of treating bone defects. Biobased and biodegradable polyester composites combine the good mechanical and degradable properties of polyester, thereby providing an alternative for bone implant materials. Bone tissue engineering (BTE) accelerates bone defect repair by simulating the bone microenvironment. Composite scaffolds support bone formation and further accelerate the process of bone repair. The introduction of 3D printing technology enables the preparation of scaffolds to be more precise, reproducible, and flexible, which is a very promising development. This review presents the physical properties of BTE scaffolds and summarizes the strategies adopted by domestic and international scholars to improve the properties of scaffolds based on biobased and biodegradable polyester/ceramic composites in recent years. In addition, future development prospects in the field and the challenges of expanding production in clinical applications are presented.
Collapse
Affiliation(s)
- Shunshun Zhu
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
- Laboratory of Biomass and Green Technologies, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés, 2, 5030 Gembloux, Belgium
| | - Hongnan Sun
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
| | - Taihua Mu
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences; Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
| | - Aurore Richel
- Laboratory of Biomass and Green Technologies, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés, 2, 5030 Gembloux, Belgium
| |
Collapse
|
5
|
Karavasili C, Young T, Francis J, Blanco J, Mancini N, Chang C, Bernstock JD, Connolly ID, Shankar GM, Traverso G. Local drug delivery challenges and innovations in spinal neurosurgery. J Control Release 2024; 376:1225-1250. [PMID: 39505215 DOI: 10.1016/j.jconrel.2024.10.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The development of novel therapeutics in the field of spinal neurosurgery faces a litany of translational challenges. Achieving precise drug targeting within the confined spaces associated with the spinal cord, canal and vertebra requires the development of next generation delivery systems and devices. These must be capable of overcoming inherent barriers related to drug diffusion, whilst concurrently ensuring optimal drug distribution and retention. In this review, we provide an overview of the most recent advances in the therapeutic management of diseases and disorders affecting the spine, including systems and devices capable of releasing small molecules and biopharmaceuticals that help eliminate pain and restore the mechanical function and stability of the spine. We highlight material-based approaches and minimally invasive techniques that can be employed to provide control over drug release kinetics and improve retention. We also seek to explore how the newest advancements in nanotechnology, biomaterials, additive manufacturing technologies and imaging modalities can be employed in this translational pursuit. Finally, we discuss the landscape of clinical trials and recently approved products aimed at overcoming the complexities associated with drug delivery to the spine.
Collapse
Affiliation(s)
- Christina Karavasili
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Thomas Young
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua Francis
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Julianna Blanco
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Nicholas Mancini
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Charmaine Chang
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua D Bernstock
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ian D Connolly
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ganesh M Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
6
|
Yin M, Liu Z, Sun Z, Qu X, Chen Z, Diao Y, Cheng Y, Shen S, Wang X, Cai Z, Lu B, Tan S, Wang Y, Zhao X, Chen F. Biomimetic Scaffolds Regulating the Iron Homeostasis for Remolding Infected Osteogenic Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407251. [PMID: 39373362 PMCID: PMC11600272 DOI: 10.1002/advs.202407251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/12/2024] [Indexed: 10/08/2024]
Abstract
The treatment of infected bone defects (IBDs) needs simultaneous elimination of infection and acceleration of bone regeneration. One mechanism that hinders the regeneration of IBDs is the iron competition between pathogens and host cells, leading to an iron deficient microenvironment that impairs the innate immune responses. In this work, an in situ modification strategy is proposed for printing iron-active multifunctional scaffolds with iron homeostasis regulation ability for treating IBDs. As a proof-of-concept, ultralong hydroxyapatite (HA) nanowires are modified through in situ growth of a layer of iron gallate (FeGA) followed by incorporation in the poly(lactic-co-glycolic acid) (PLGA) matrix to print biomimetic PLGA based composite scaffolds containing FeGA modified HA nanowires (FeGA-HA@PLGA). The photothermal effect of FeGA endows the scaffolds with excellent antibacterial activity. The released iron ions from the FeGA-HA@PLGA help restore the iron homeostasis microenvironment, thereby promoting anti-inflammatory, angiogenesis and osteogenic differentiation. The transcriptomic analysis shows that FeGA-HA@PLGA scaffolds exert anti-inflammatory and pro-osteogenic differentiation by activating NF-κB, MAPK and PI3K-AKT signaling pathways. Animal experiments confirm the excellent bone repair performance of FeGA-HA@PLGA scaffolds for IBDs, suggesting the promising prospect of iron homeostasis regulation therapy in future clinical applications.
Collapse
Affiliation(s)
- Mengting Yin
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Zhiqing Liu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Zhongyi Sun
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
- Suzhou First People's HospitalSchool of MedicineAnhui University of Science and TechnologyAnhui232001P.R. China
| | - Xinyu Qu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Ziyan Chen
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Yuying Diao
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
| | - Yuxuan Cheng
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Sisi Shen
- Department of Plastic and Reconstructive SurgeryShanghai Key Laboratory of Tissue EngineeringShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Xiansong Wang
- Department of Plastic and Reconstructive SurgeryShanghai Key Laboratory of Tissue EngineeringShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Zhuyun Cai
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Bingqiang Lu
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Shuo Tan
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Yan Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
| | - Xinyu Zhao
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
| | - Feng Chen
- Center for Orthopaedic Science and Translational MedicineDepartment of OrthopaedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases Shanghai Stomatological Hospital & School of StomatologyFudan UniversityShanghai201102P. R. China
- Suzhou First People's HospitalSchool of MedicineAnhui University of Science and TechnologyAnhui232001P.R. China
| |
Collapse
|
7
|
Rajendran AK, Anthraper MSJ, Hwang NS, Rangasamy J. Osteogenesis and angiogenesis promoting bioactive ceramics. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2024; 159:100801. [DOI: 10.1016/j.mser.2024.100801] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
8
|
Lin X, Wang Y, Liu L, Du X, Wang W, Guo S, Zhang J, Ge K, Zhou G. Enhanced bone regeneration by osteoinductive and angiogenic zein/whitlockite composite scaffolds loaded with levofloxacin. RSC Adv 2024; 14:14470-14479. [PMID: 38708116 PMCID: PMC11063759 DOI: 10.1039/d4ra00772g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024] Open
Abstract
Promoting angiogenesis following biomaterial implantation is essential to bone tissue regeneration. Herein, the composite scaffolds composed of zein, whitlockite (WH), and levofloxacin (LEVO) were fabricated to augment bone repair by facilitating osteogenesis and angiogenesis. First, three-dimensional composite scaffolds containing zein and WH were prepared using the salt-leaching method. Then, as a model antibiotic drug, the LEVO was loaded into zein/WH scaffolds. Moreover, the addition of WH enhanced the adhesion, differentiation, and mineralization of osteoblasts. The zein/WH/LEVO composite scaffolds not only had significant osteoinductivity but also showed excellent antibacterial properties. The prepared composite scaffolds were then implanted into a calvarial defect model to evaluate their osteogenic induction effects in vivo. Micro-CT observation and histological analysis indicate that the scaffolds can accelerate bone regeneration with the contribution of endogenous cytokines. Based on amounts of data in vitro and in vivo, the scaffolds present profound effects on improving bone regeneration, especially for the favorable osteogenic, intensive angiogenic, and alleviated inflammation abilities. The results showed that the synthesized scaffolds could be a potential material for bone tissue engineering.
Collapse
Affiliation(s)
- Xue Lin
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
| | - Yu Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
- College of Basic Medical Science, Hebei University Baoding 071000 P. R. China
| | - Lingyu Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
- College of Basic Medical Science, Hebei University Baoding 071000 P. R. China
| | - Xiaomeng Du
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
| | - Wenying Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University Tianjin 300071 China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
| | - Kun Ge
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
| | - Guoqiang Zhou
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University Baoding 071002 China
- College of Basic Medical Science, Hebei University Baoding 071000 P. R. China
| |
Collapse
|
9
|
Jia B, Huang H, Dong Z, Ren X, Lu Y, Wang W, Zhou S, Zhao X, Guo B. Degradable biomedical elastomers: paving the future of tissue repair and regenerative medicine. Chem Soc Rev 2024; 53:4086-4153. [PMID: 38465517 DOI: 10.1039/d3cs00923h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Degradable biomedical elastomers (DBE), characterized by controlled biodegradability, excellent biocompatibility, tailored elasticity, and favorable network design and processability, have become indispensable in tissue repair. This review critically examines the recent advances of biodegradable elastomers for tissue repair, focusing mainly on degradation mechanisms and evaluation, synthesis and crosslinking methods, microstructure design, processing techniques, and tissue repair applications. The review explores the material composition and cross-linking methods of elastomers used in tissue repair, addressing chemistry-related challenges and structural design considerations. In addition, this review focuses on the processing methods of two- and three-dimensional structures of elastomers, and systematically discusses the contribution of processing methods such as solvent casting, electrostatic spinning, and three-/four-dimensional printing of DBE. Furthermore, we describe recent advances in tissue repair using DBE, and include advances achieved in regenerating different tissues, including nerves, tendons, muscle, cardiac, and bone, highlighting their efficacy and versatility. The review concludes by discussing the current challenges in material selection, biodegradation, bioactivation, and manufacturing in tissue repair, and suggests future research directions. This concise yet comprehensive analysis aims to provide valuable insights and technical guidance for advances in DBE for tissue engineering.
Collapse
Affiliation(s)
- Ben Jia
- School of Civil Aviation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Heyuan Huang
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Zhicheng Dong
- School of Civil Aviation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaoyang Ren
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Yanyan Lu
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Wenzhi Wang
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Shaowen Zhou
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
10
|
Nielsen JJ, Low SA, Chen C, Li X, Mbachu E, Trigg L, Sun S, Tremby M, Hadap R, Low PS. Targeted Delivery of Abaloparatide to Spinal Fusion Site Accelerates Fusion Process in Rats. Biomedicines 2024; 12:612. [PMID: 38540225 PMCID: PMC10967909 DOI: 10.3390/biomedicines12030612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 11/11/2024] Open
Abstract
Spinal fusions are performed to treat congenital skeletal malformations, spondylosis, degenerative disk diseases, and other pathologies of the vertebrae that can be resolved by reducing motion between neighboring vertebrae. Unfortunately, up to 100,000 fusion procedures fail per year in the United States, suggesting that efforts to develop new approaches to improve spinal fusions are justified. We have explored whether the use of an osteotropic oligopeptide to target an attached bone anabolic agent to the fusion site might be exploited to both accelerate the mineralization process and improve the overall success rate of spinal fusions. The data presented below demonstrate that subcutaneous administration of a modified abaloparatide conjugated to 20 mer of D-glutamic acid not only localizes at the spinal fusion site but also outperforms the standard of care (topically applied BMP2) in both speed of mineralization (p < 0.05) and overall fusion success rate (p < 0.05) in a posterior lateral spinal fusion model in male and female rats, with no accompanying ectopic mineralization. Because the bone-localizing conjugate can be administered ad libitum post-surgery, and since the procedure appears to improve on standard of care, we conclude that administration of a bone-homing anabolic agent for improvement of spinal fusion surgeries warrants further exploration.
Collapse
Affiliation(s)
- Jeffery J. Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA;
- Novosteo Inc., West Lafayette, IN 47906, USA
| | - Stewart A. Low
- Novosteo Inc., West Lafayette, IN 47906, USA
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | - Xinlan Li
- Novosteo Inc., West Lafayette, IN 47906, USA
| | | | - Lina Trigg
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Siyuan Sun
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Madeline Tremby
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Rahul Hadap
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S. Low
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA;
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
11
|
Dong J, Ding H, Wang Q, Wang L. A 3D-Printed Scaffold for Repairing Bone Defects. Polymers (Basel) 2024; 16:706. [PMID: 38475389 DOI: 10.3390/polym16050706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 11/04/2023] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
The treatment of bone defects has always posed challenges in the field of orthopedics. Scaffolds, as a vital component of bone tissue engineering, offer significant advantages in the research and treatment of clinical bone defects. This study aims to provide an overview of how 3D printing technology is applied in the production of bone repair scaffolds. Depending on the materials used, the 3D-printed scaffolds can be classified into two types: single-component scaffolds and composite scaffolds. We have conducted a comprehensive analysis of material composition, the characteristics of 3D printing, performance, advantages, disadvantages, and applications for each scaffold type. Furthermore, based on the current research status and progress, we offer suggestions for future research in this area. In conclusion, this review acts as a valuable reference for advancing the research in the field of bone repair scaffolds.
Collapse
Affiliation(s)
- Jianghui Dong
- Guangxi Engineering Research Center of Digital Medicine and Clinical Translation, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Hangxing Ding
- Guangxi Engineering Research Center of Digital Medicine and Clinical Translation, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Qin Wang
- Guangxi Engineering Research Center of Digital Medicine and Clinical Translation, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Liping Wang
- Guangxi Engineering Research Center of Digital Medicine and Clinical Translation, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| |
Collapse
|
12
|
Huang J, Park J, Jung N, Moon HS, Zong Z, Li G, Lin S, Cho SW, Park Y. Hydrothermally treated coral scaffold promotes proliferation of mesenchymal stem cells and enhances segmental bone defect healing. Front Bioeng Biotechnol 2023; 11:1332138. [PMID: 38173870 PMCID: PMC10761418 DOI: 10.3389/fbioe.2023.1332138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction: Synthetic hydroxyapatite (HAp) scaffolds have shown promising therapeutic outcomes in both animals and patients. In this study, we aim to evaluate the chemical and physical phenotype, biocompatibility, and bone repair effects of hydrothermally treated coral with natural coral and synthetic HAp. Methods: The phase composition, surface pattern, 3D structures, and porosity of the scaffolds were characterized, and cell viability, proliferation, and osteogenic differentiation of mesenchymal stem cells (MSCs) after seeding onto the scaffold were determined. The scaffolds were implanted into rats to assess their bone repair effects using micro-CT analysis, mechanical testing, and histological staining. Results: The results showed that the phase composition, porous structure, and porosity of hydrothermally treated coral were comparable to pure HAp scaffold. While only the natural coral happens to be dominantly calcium carbonate. Higher cell proliferation and osteogenic differentiation potential were observed in the hydrothermally treated coral scaffold compared to natural coral and pure HAp. Histological results also showed increased new bone formation in the hydrothermally treated coral group. Discussion: Overall, our study suggests that hydrothermal modification enhances the cytocompatibility and therapeutic capacity of coral without altering its physical properties, showing superior effectiveness in bone repair to synthetic HAp.
Collapse
Affiliation(s)
- Jianping Huang
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Jaehan Park
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Narae Jung
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Hong Seok Moon
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Zhixian Zong
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Sung-Won Cho
- Division of Anatomy and Developmental Biology, Department of Oral Biology, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Youngbum Park
- Department of Prosthodontics, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Ye Z, Liu Y, Song J, Gao Y, Fang H, Hu Z, Zhang M, Liao W, Cui L, Liu Y. Expanding the therapeutic potential of Salvia miltiorrhiza: a review of its pharmacological applications in musculoskeletal diseases. Front Pharmacol 2023; 14:1276038. [PMID: 38116081 PMCID: PMC10728493 DOI: 10.3389/fphar.2023.1276038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Salvia miltiorrhiz, commonly known as "Danshen" in Chinese medicine, has longstanding history of application in cardiovascular and cerebrovascular diseases. Renowned for its diverse therapeutic properties, including promoting blood circulation, removing blood stasis, calming the mind, tonifying the blood, and benefiting the "Qi", recent studies have revealed its significant positive effects on bone metabolism. This potential has garnered attention for its promising role in treating musculoskeletal disorders. Consequently, there is a high anticipation for a comprehensive review of the potential of Salvia miltiorrhiza in the treatment of various musculoskeletal diseases, effectively introducing an established traditional Chinese medicine into a burgeoning field. AIM OF THE REVIEW Musculoskeletal diseases (MSDs) present significant challenges to healthcare systems worldwide. Previous studies have demonstrated the high efficacy and prospects of Salvia miltiorrhiza and its active ingredients for treatment of MSDs. This review aims to illuminate the newfound applications of Salvia miltiorrhiza and its active ingredients in the treatment of various MSDs, effectively bridging the gap between an established medicine and an emerging field. METHODS In this review, previous studies related to Salvia miltiorrhiza and its active ingredients on the treatment of MSD were collected, the specific active ingredients of Salvia miltiorrhiza were summarized, the effects of Salvia miltiorrhiza and its active ingredients for the treatment of MSDs, as well as their potential molecular mechanisms were reviewed and discussed. RESULTS Based on previous publications, Salvianolic acid A, salvianolic acid B, tanshinone IIA are the representative active ingredients of Salvia miltiorrhiza. Their application has shown significant beneficial outcomes in osteoporosis, fractures, and arthritis. Salvia miltiorrhiza and its active ingredients protect against MSDs by regulating different signaling pathways, including ROS, Wnt, MAPK, and NF-κB signaling. CONCLUSION Salvia miltiorrhiza and its active ingredients demonstrate promising potential for bone diseases and have been explored across a wide variety of MSDs. Further exploration of Salvia miltiorrhiza's pharmacological applications in MSDs holds great promise for advancing therapeutic interventions and improving the lives of patients suffering from these diseases.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yuyu Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yin Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang, China
| | - Haiping Fang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Zilong Hu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Wenwei Liao
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| |
Collapse
|
14
|
Zhou J, See CW, Sreenivasamurthy S, Zhu D. Customized Additive Manufacturing in Bone Scaffolds-The Gateway to Precise Bone Defect Treatment. RESEARCH (WASHINGTON, D.C.) 2023; 6:0239. [PMID: 37818034 PMCID: PMC10561823 DOI: 10.34133/research.0239] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
In the advancing landscape of technology and novel material development, additive manufacturing (AM) is steadily making strides within the biomedical sector. Moving away from traditional, one-size-fits-all implant solutions, the advent of AM technology allows for patient-specific scaffolds that could improve integration and enhance wound healing. These scaffolds, meticulously designed with a myriad of geometries, mechanical properties, and biological responses, are made possible through the vast selection of materials and fabrication methods at our disposal. Recognizing the importance of precision in the treatment of bone defects, which display variability from macroscopic to microscopic scales in each case, a tailored treatment strategy is required. A patient-specific AM bone scaffold perfectly addresses this necessity. This review elucidates the pivotal role that customized AM bone scaffolds play in bone defect treatment, while offering comprehensive guidelines for their customization. This includes aspects such as bone defect imaging, material selection, topography design, and fabrication methodology. Additionally, we propose a cooperative model involving the patient, clinician, and engineer, thereby underscoring the interdisciplinary approach necessary for the effective design and clinical application of these customized AM bone scaffolds. This collaboration promises to usher in a new era of bioactive medical materials, responsive to individualized needs and capable of pushing boundaries in personalized medicine beyond those set by traditional medical materials.
Collapse
Affiliation(s)
- Juncen Zhou
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Carmine Wang See
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Sai Sreenivasamurthy
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Donghui Zhu
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
15
|
Ahmed S, Keniry M, Padilla V, Anaya-Barbosa N, Javed MN, Gilkerson R, Gomez K, Ashraf A, Narula AS, Lozano K. Development of pullulan/chitosan/salvianolic acid ternary fibrous membranes and their potential for chemotherapeutic applications. Int J Biol Macromol 2023; 250:126187. [PMID: 37558036 DOI: 10.1016/j.ijbiomac.2023.126187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/19/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
This study investigates the feasibility of centrifugal spinning for producing fibrous membranes containing pullulan, chitosan, and danshen extract. The danshen extract is composed of 20 wt% salvianolic acid B (SA). Citric acid was added to the mixture as a crosslinking agent to promote its use in the aqueous medium. The influence of the danshen concentration (25 wt% and 33 wt%) on fiber morphology, thermal behavior, and the biochemical effect was analyzed. Developed fiber-based membranes consist of long, continuous, and uniform fibers with a sparse scattering of beads. Fiber diameter analysis shows values ranging from 384 ± 123 nm to 644 ± 141 nm depending on the concentration of danshen. The nanofibers show adequate aqueous stability after crosslinking. Thermal analysis results prove that SA is loaded into nanofibers without compromising their structural integrity. Cell-based results indicate that the developed nanofiber membranes promote cell growth and are not detrimental to fibroblast cells. Anticancer studies reveal a promising inhibition to the proliferation of HCT116 colon cancer cells. The developed systems show potential as innovative systems to be used as a bioactive chemotherapeutic drug that could be placed on the removed tumor site to prevent development of colon cancer microdeposits.
Collapse
Affiliation(s)
- Salahuddin Ahmed
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Megan Keniry
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Victoria Padilla
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Narcedalia Anaya-Barbosa
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Md Noushad Javed
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Robert Gilkerson
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Kithzia Gomez
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Ali Ashraf
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Karen Lozano
- Department of Mechanical Engineering, The University of Texas Rio Grande Valley, Edinburg, TX, USA.
| |
Collapse
|
16
|
Saberi A, Kouhjani M, Mohammadi M, Hosta-Rigau L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: a cutting-edge approach. J Nanobiotechnology 2023; 21:351. [PMID: 37770928 PMCID: PMC10536787 DOI: 10.1186/s12951-023-02115-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Despite the recent advances in the development of bone graft substitutes, treatment of critical size bone defects continues to be a significant challenge, especially in the elderly population. A current approach to overcome this challenge involves the creation of bone-mimicking scaffolds that can simultaneously promote osteogenesis and angiogenesis. In this context, incorporating multiple bioactive agents like growth factors, genes, and small molecules into these scaffolds has emerged as a promising strategy. To incorporate such agents, researchers have developed scaffolds incorporating nanoparticles, including nanoparticulate carriers, inorganic nanoparticles, and exosomes. Current paper provides a summary of the latest advancements in using various bioactive agents, drugs, and cells to synergistically promote osteogenesis and angiogenesis in bone-mimetic scaffolds. It also discusses scaffold design properties aimed at maximizing the synergistic effects of osteogenesis and angiogenesis, various innovative fabrication strategies, and ongoing clinical studies.
Collapse
Affiliation(s)
- Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Kouhjani
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
17
|
He G, Chen G, Liu W, Ye D, Liu X, Liang X, Song J. Salvianolic Acid B: A Review of Pharmacological Effects, Safety, Combination Therapy, New Dosage Forms, and Novel Drug Delivery Routes. Pharmaceutics 2023; 15:2235. [PMID: 37765204 PMCID: PMC10538146 DOI: 10.3390/pharmaceutics15092235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Salvianolic acid B is extracted from the roots and rhizomes of Danshen (Salvia miltiorrhiza Bge., family Labiatae). It is a water-soluble, weakly acidic drug that has demonstrated antitumor and anti-inflammatory effects on various organs and tissues such as the lung, heart, kidney, intestine, bone, liver, and skin and protective effects in diseases such as depression and spinal cord injury. The mechanisms underlying the protective effects of salvianolic acid B are mainly related to its anti-inflammatory, antioxidant, anti- or pro-apoptotic, anti- or pro-autophagy, anti-fibrotic, and metabolism-regulating functions. Salvianolic acid B can regulate various signaling pathways, cells, and molecules to achieve maximum therapeutic effects. This review summarizes the safety profile, combination therapy potential, and new dosage forms and delivery routes of salvianolic acid B. Although significant research progress has been made, more in-depth pharmacological studies are warranted to identify the mechanism of action, related signaling pathways, more suitable combination drugs, more effective dosage forms, and novel routes of administration of salvianolic acid B.
Collapse
Affiliation(s)
- Guannan He
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Guangfeng Chen
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Weidong Liu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Dongxue Ye
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Xuehuan Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Xiaodong Liang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
| | - Jing Song
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (G.H.); (W.L.); (D.Y.)
- Shandong Yuze Pharmaceutical Industry Technology Research Institute Co., Ltd., Dezhou 251200, China
| |
Collapse
|
18
|
Sun T, Wang J, Huang H, Liu X, Zhang J, Zhang W, Wang H, Li Z. Low-temperature deposition manufacturing technology: a novel 3D printing method for bone scaffolds. Front Bioeng Biotechnol 2023; 11:1222102. [PMID: 37622000 PMCID: PMC10445654 DOI: 10.3389/fbioe.2023.1222102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
The application of three-dimensional printing technology in the medical field has great potential for bone defect repair, especially personalized and biological repair. As a green manufacturing process that does not involve liquefication through heating, low-temperature deposition manufacturing (LDM) is a promising type of rapid prototyping manufacturing and has been widely used to fabricate scaffolds in bone tissue engineering. The scaffolds fabricated by LDM have a multi-scale controllable pore structure and interconnected micropores, which are beneficial for the repair of bone defects. At the same time, different types of cells or bioactive factor can be integrated into three-dimensional structural scaffolds through LDM. Herein, we introduced LDM technology and summarize its applications in bone tissue engineering. We divide the scaffolds into four categories according to the skeleton materials and discuss the performance and limitations of the scaffolds. The ideas presented in this review have prospects in the development and application of LDM scaffolds.
Collapse
Affiliation(s)
- Tianze Sun
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
- Division of Energy Materials (DNL22), Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Wentao Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| | - Honghua Wang
- Division of Energy Materials (DNL22), Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| |
Collapse
|
19
|
Zhou S, Liu S, Wang Y, Li W, Wang J, Wang X, Wang S, Chen W, Lv H. Advances in the Study of Bionic Mineralized Collagen, PLGA, Magnesium Ionomer Materials, and Their Composite Scaffolds for Bone Defect Treatment. J Funct Biomater 2023; 14:406. [PMID: 37623651 PMCID: PMC10455784 DOI: 10.3390/jfb14080406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
The healing of bone defects after a fracture remains a key issue to be addressed. Globally, more than 20 million patients experience bone defects annually. Among all artificial bone repair materials that can aid healing, implantable scaffolds made from a mineralized collagen (MC) base have the strongest bionic properties. The MC/PLGA scaffold, created by adding Poly (lactic-co-glycolic acid) copolymer (PLGA) and magnesium metal to the MC substrate, plays a powerful role in promoting fracture healing because, on the one hand, it has good biocompatibility similar to that of MC; on the other hand, the addition of PLGA provides the scaffold with an interconnected porous structure, and the addition of magnesium allows the scaffold to perform anti-inflammatory, osteogenic, and angiogenic activities. Using the latest 3D printing technology for scaffold fabrication, it is possible to model the scaffold in advance according to the requirement and produce a therapeutic scaffold suitable for various bone-defect shapes with less time and effort, which can promote bone tissue healing and regeneration to the maximum extent. This study reviews the material selection and technical preparation of MC/PLGA scaffolds, and the progress of their research on bone defect treatment.
Collapse
Affiliation(s)
- Shuai Zhou
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Shihang Liu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Yan Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Wenjing Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Juan Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, No. 30 Shuangqing Road, Beijing 100084, China
| | - Shuo Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, No. 30 Shuangqing Road, Beijing 100084, China
| | - Wei Chen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| | - Hongzhi Lv
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China; (S.Z.); (S.L.); (Y.W.); (W.L.)
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, No. 139 Ziqiang Road, Shijiazhuang 050051, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang 050051, China
| |
Collapse
|
20
|
He Y, Liang L, Luo C, Zhang ZY, Huang J. Strategies for in situ tissue engineering of vascularized bone regeneration (Review). Biomed Rep 2023; 18:42. [PMID: 37325184 PMCID: PMC10265129 DOI: 10.3892/br.2023.1625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/03/2023] [Indexed: 06/17/2023] Open
Abstract
Numerous physiological processes occur following bone fracture, including inflammatory cell recruitment, vascularization, and callus formation and remodeling. In particular circumstances, such as critical bone defects or osteonecrosis, the regenerative microenvironment is compromised, rendering endogenous stem/progenitor cells incapable of fully manifesting their reparative potential. Consequently, external interventions, such as grafting or augmentation, are frequently necessary. In situ bone tissue engineering (iBTE) employs cell-free scaffolds that possess microenvironmental cues, which, upon implantation, redirect the behavior of endogenous stem/progenitor cells towards a pro-regenerative inflammatory response and reestablish angiogenesis-osteogenesis coupling. This process ultimately results in vascularized bone regeneration (VBR). In this context, a comprehensive review of the current techniques and modalities in VBR-targeted iBTE technology is provided.
Collapse
Affiliation(s)
- Yijun He
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Lin Liang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Cheng Luo
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Jiongfeng Huang
- Department of Osteoarthropathy and Sports Medicine, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 511400, P.R. China
| |
Collapse
|
21
|
Qiu E, Liu F. PLGA-based drug delivery systems in treating bone tumors. Front Bioeng Biotechnol 2023; 11:1199343. [PMID: 37324432 PMCID: PMC10267463 DOI: 10.3389/fbioe.2023.1199343] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Bone tumor has become a common disease that endangers human health. Surgical resection of bone tumors not only causes biomechanical defects of bone but also destroys the continuity and integrity of bone and cannot completely remove the local tumor cells. The remaining tumor cells in the lesion bring a hidden danger of local recurrence. To improve the chemotherapeutic effect and effectively clear tumor cells, traditional systemic chemotherapy often requires higher doses, and high doses of chemotherapeutic drugs inevitably cause a series of systemic toxic side effects, often intolerable to patients. PLGA-based drug delivery systems, such as nano delivery systems and scaffold-based local delivery systems, can help eliminate tumors and promote bone regeneration and therefore have more significant potential for application in bone tumor treatment. In this review, we summarize the research progress of PLGA nano drug delivery systems and PLGA scaffold-based local delivery systems in bone tumor treatment applications, expecting to provide a theoretical basis for developing novel bone tumor treatment strategies.
Collapse
Affiliation(s)
- Enduo Qiu
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | | |
Collapse
|
22
|
Alves-Silva JM, Pedreiro S, Cavaleiro C, Cruz MT, Figueirinha A, Salgueiro L. Effect of Thymbra capitata (L.) Cav. on Inflammation, Senescence and Cell Migration. Nutrients 2023; 15:nu15081930. [PMID: 37111149 PMCID: PMC10146686 DOI: 10.3390/nu15081930] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/30/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Aromatic plants are reported to display pharmacological properties, including anti-aging. This work aims to disclose the anti-aging effect of the essential oil (EO) of Thymbra capitata (L.) Cav., an aromatic and medicinal plant widely used as a spice, as well as of the hydrodistillation residual water (HRW), a discarded by-product of EO hydrodistillation. The phytochemical characterization of EO and HRW was assessed by GC-MS and HPLC-PDA-ESI-MSn, respectively. The DPPH, ABTS, and FRAP assays were used to disclose the antioxidant properties. The anti-inflammatory potential was evaluated using lipopolysaccharide-stimulated macrophages by assessing NO production, iNOS, and pro-IL-1β protein levels. Cell migration was evaluated using the scratch wound assay, and the etoposide-induced senescence was used to assess the modulation of senescence. The EO is mainly characterized by carvacrol, while the HRW is predominantly characterized by rosmarinic acid. The HRW exerts a stronger antioxidant effect in the DPPH and FRAP assays, whereas the EO was the most active sample in the ABTS assay. Both extracts reduce NO, iNOS, and pro-IL-1β. The EO has no effect on cell migration and presents anti-senescence effects. In opposition, HRW reduces cell migration and induces cellular senescence. Overall, our study highlights interesting pharmacological properties for both extracts, EO being of interest as an anti-aging ingredient and HRW relevant in cancer therapy.
Collapse
Affiliation(s)
- Jorge M Alves-Silva
- Institute for Clinical and Biomedical Research, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
| | - Sónia Pedreiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Carlos Cavaleiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), University of Porto, 4099-002 Porto, Portugal
- Chemical Process Engineering and Forest Products Research Centre, Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, 3030-790 Coimbra, Portugal
| |
Collapse
|
23
|
Lu Y, Cheng D, Niu B, Wang X, Wu X, Wang A. Properties of Poly (Lactic-co-Glycolic Acid) and Progress of Poly (Lactic-co-Glycolic Acid)-Based Biodegradable Materials in Biomedical Research. Pharmaceuticals (Basel) 2023; 16:ph16030454. [PMID: 36986553 PMCID: PMC10058621 DOI: 10.3390/ph16030454] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
In recent years, biodegradable polymers have gained the attention of many researchers for their promising applications, especially in drug delivery, due to their good biocompatibility and designable degradation time. Poly (lactic-co-glycolic acid) (PLGA) is a biodegradable functional polymer made from the polymerization of lactic acid (LA) and glycolic acid (GA) and is widely used in pharmaceuticals and medical engineering materials because of its biocompatibility, non-toxicity, and good plasticity. The aim of this review is to illustrate the progress of research on PLGA in biomedical applications, as well as its shortcomings, to provide some assistance for its future research development.
Collapse
Affiliation(s)
- Yue Lu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Baohua Niu
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Xiuzhi Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xiaxia Wu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Aiping Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Correspondence:
| |
Collapse
|
24
|
Broussolle T, Roux JP, Chapurlat R, Barrey C. Murine models of posterolateral spinal fusion: A systematic review. Neurochirurgie 2023; 69:101428. [PMID: 36871885 DOI: 10.1016/j.neuchi.2023.101428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Rodent models are commonly used experimentally to assess treatment effectiveness in spinal fusion. Certain factors are associated with better fusion rates. The objectives of the present study were to report the protocols most frequently used, to evaluate factors known to positively influence fusion rate, and to identify new factors. METHOD A systematic literature search of PubMed and Web of Science found 139 experimental studies of posterolateral lumbar spinal fusion in rodent models. Data for level and location of fusion, animal strain, sex, weight and age, graft, decortication, fusion assessment and fusion and mortality rates were collected and analyzed. RESULTS The standard murine model for spinal fusion was male Sprague Dawley rats of 295g weight and 13 weeks' age, using decortication, with L4-L5 as fusion level. The last two criteria were associated with significantly better fusion rates. On manual palpation, the overall mean fusion rate in rats was 58% and the autograft mean fusion rate was 61%. Most studies evaluated fusion as a binary on manual palpation, and only a few used CT and histology. Average mortality was 3.03% in rats and 1.56% in mice. CONCLUSIONS These results suggest using a rat model, younger than 10 weeks and weighing more than 300 grams on the day of surgery, to optimize fusion rates, with decortication before grafting and fusing the L4-L5 level.
Collapse
Affiliation(s)
- T Broussolle
- Department of Spine Surgery, P. Wertheimer University Hospital, GHE, hospices civils de Lyon, université Claude-Bernard Lyon 1, Lyon, France; Inserm UMR 1033, université Claude-Bernard Lyon 1, Lyon, France.
| | - Jean-Paul Roux
- Inserm UMR 1033, université Claude-Bernard Lyon 1, Lyon, France
| | - R Chapurlat
- Inserm UMR 1033, université Claude-Bernard Lyon 1, Lyon, France
| | - C Barrey
- Department of Spine Surgery, P. Wertheimer University Hospital, GHE, hospices civils de Lyon, université Claude-Bernard Lyon 1, Lyon, France; Arts et métiers ParisTech, ENSAM, 151, boulevard de l'Hôpital, 75013 Paris, France
| |
Collapse
|
25
|
Wang T, Liu K, Wang J, Xiang G, Hu X, Bai H, Lei W, Tao TH, Feng Y. Spatiotemporal Regulation of Injectable Heterogeneous Silk Gel Scaffolds for Accelerating Guided Vertebral Repair. Adv Healthc Mater 2023; 12:e2202210. [PMID: 36465008 DOI: 10.1002/adhm.202202210] [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: 08/30/2022] [Revised: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Osteoporotic vertebral fracture is jeopardizing the health of the aged population around the world, while the hypoxia microenvironment and oxidative damage of bone defect make it difficult to perform effective tissue regeneration. The balance of oxidative stress and the coupling of vessel and bone ingrowth are critical for bone regeneration. In this study, an injectable heterogeneous silk gel scaffold which can spatiotemporally and sustainedly release bone mesenchymal stem cell-derived small extracellular vesicles, HIF-1α pathway activator, and inhibitor is developed for bone repair and vertebral reinforcement. The initial enhancement of HIF-1α upregulates the expression of VEGF to promote angiogenesis, and the balance of reactive oxygen species level is regulated to effectively eliminate oxidative damage and abnormal microenvironment. The subsequent inhibition of HIF-1α avoids the overexpression of VEGF and vascular overgrowth. Meanwhile, complex macroporous structures and suitable mechanical support can be obtained within the silk gel scaffolds, which will promote in situ bone regeneration. These findings provide a new clinical translation strategy for osteoporotic vertebral augmentation on basis of hypoxia microenvironment improvement.
Collapse
Affiliation(s)
- Tianji Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Keyin Liu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Geng Xiang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaofan Hu
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Bai
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tiger H Tao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.,School of Physical Science and Technology, ShanghaiTech University, Shanghai, 200031, China.,Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, 200031, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 200031, China
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
26
|
Wang T, Zhao H, Jing S, Fan Y, Sheng G, Ding Q, Liu C, Wu H, Liu Y. Magnetofection of miR-21 promoted by electromagnetic field and iron oxide nanoparticles via the p38 MAPK pathway contributes to osteogenesis and angiogenesis for intervertebral fusion. J Nanobiotechnology 2023; 21:27. [PMID: 36694219 PMCID: PMC9875474 DOI: 10.1186/s12951-023-01789-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Magnetofection-mediated gene delivery shows great therapeutic potential through the regulation of the direction and degree of differentiation. Lumbar degenerative disc disease (DDD) is a serious global orthopaedic problem. However, even though intervertebral fusion is the gold standard for the treatment of DDD, its therapeutic effect is unsatisfactory. Here, we described a novel magnetofection system for delivering therapeutic miRNAs to promote osteogenesis and angiogenesis in patients with lumbar DDD. RESULTS Co-stimulation with electromagnetic field (EMF) and iron oxide nanoparticles (IONPs) enhanced magnetofection efficiency significantly. Moreover, in vitro, magnetofection of miR-21 into bone marrow mesenchymal stem cells (BMSCs) and human umbilical endothelial cells (HUVECs) influenced their cellular behaviour and promoted osteogenesis and angiogenesis. Then, gene-edited seed cells were planted onto polycaprolactone (PCL) and hydroxyapatite (HA) scaffolds (PCL/HA scaffolds) and evolved into the ideal tissue-engineered bone to promote intervertebral fusion. Finally, our results showed that EMF and polyethyleneimine (PEI)@IONPs were enhancing transfection efficiency by activating the p38 MAPK pathway. CONCLUSION Our findings illustrate that a magnetofection system for delivering miR-21 into BMSCs and HUVECs promoted osteogenesis and angiogenesis in vitro and in vivo and that magnetofection transfection efficiency improved significantly under the co-stimulation of EMF and IONPs. Moreover, it relied on the activation of p38 MAPK pathway. This magnetofection system could be a promising therapeutic approach for various orthopaedic diseases.
Collapse
Affiliation(s)
- Tianqi Wang
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hongqi Zhao
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shaoze Jing
- grid.470966.aThird Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032 China
| | - Yang Fan
- grid.412793.a0000 0004 1799 5032Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Gaohong Sheng
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qing Ding
- grid.412793.a0000 0004 1799 5032Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Chaoxu Liu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hua Wu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yang Liu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
27
|
Bassand C, Benabed L, Charlon S, Verin J, Freitag J, Siepmann F, Soulestin J, Siepmann J. 3D printed PLGA implants: APF DDM vs. FDM. J Control Release 2023; 353:864-874. [PMID: 36464064 DOI: 10.1016/j.jconrel.2022.11.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
3D Printing offers a considerable potential for personalized medicines. This is especially true for customized biodegradable implants, matching the specific needs of each patient. Poly(lactic-co-glycolic acid) (PLGA) is frequently used as matrix former in biodegradable implants. However, yet relatively little is known on the technologies, which can be used for the 3D printing of PLGA implants. The aim of this study was to compare: (i) Arburg Plastic Freeforming Droplet Deposition Modeling (APF DDM), and (ii) Fused Deposition Modeling (FDM) to print mesh-shaped, ibuprofen-loaded PLGA implants. During APF DDM, individual drug-polymer droplets are deposited, fusing together to form filaments, which build up the implants. During FDM, continuous drug-polymer filaments are deposited to form the meshes. The implants were thoroughly characterized before and after exposure to phosphate buffer pH 7.4 using optical and scanning electron microscopy, GPC, DSC, drug release measurements and monitoring dynamic changes in the systems' dry & wet mass and pH of the bulk fluid. Interestingly, the mesh structures were significantly different, although the device design (composition & theoretical geometry) were the same. This could be explained by the fact that the deposition of individual droplets during APF DDM led to curved and rather thick filaments, resulting in a much lower mesh porosity. In contrast, FDM printing generated straight and thinner filaments: The open spaces between them were much larger and allowed convective mass transport during drug release. Consequently, most of the drug was already released after 4 d, when substantial PLGA set on. In the case of APF DDM printed implants, most of the drug was still entrapped at that time point and substantial polymer swelling transformed the meshes into more or less continuous PLGA gels. Hence, the diffusion pathways became much longer and ibuprofen release was controlled over 2 weeks.
Collapse
Affiliation(s)
- C Bassand
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - L Benabed
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - S Charlon
- IMT Lille Douai, École Nationale Supérieure Mines-Télécom Lille Douai, Materials & Processes Center, Cité Scientifique, Villeneuve d'Ascq Cedex, France
| | - J Verin
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - J Freitag
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - F Siepmann
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - J Soulestin
- IMT Lille Douai, École Nationale Supérieure Mines-Télécom Lille Douai, Materials & Processes Center, Cité Scientifique, Villeneuve d'Ascq Cedex, France
| | - J Siepmann
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France.
| |
Collapse
|
28
|
Gai K, Ren X, Chen J, Zhou X, Wan Q, Wang Q, Li Y. Construction of Helically Oriented Syndiotactic Polypropylene/Isotactic Polypropylene Composites for Medical Interventional Tubes via Rotation Extrusion. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c03446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Kuo Gai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Xiangfeng Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Xiling Zhou
- National Engineering Research Center of Novel Equipment for Polymer Processing; Key Laboratory of Polymer Processing Engineering, Ministry of Education; Guangdong Provincial Key Laboratory of Technique and Equipment for Macromolecular Advanced Manufacturing; Department of Mechanical and Automotive Engineering, South China University of Technology, Guangzhou510641, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yijun Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| |
Collapse
|
29
|
Liu S, Xu Z, Hu J, Wu Z, Zheng Y. Preparation and sustained-release properties of poly(lactic acid)/graphene oxide porous biomimetic composite scaffolds loaded with salvianolic acid B. RSC Adv 2022; 12:28867-28877. [PMID: 36329763 PMCID: PMC9585927 DOI: 10.1039/d2ra05371c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/01/2022] [Indexed: 01/24/2023] Open
Abstract
Biomimetic scaffolds loaded with drugs can improve the osteogenesis and neovascularisation of scaffolds. A series of PLA/GO/Sal-B drug-loaded scaffolds was prepared by thermally induced phase separation. The addition of Sal-B increased the diameter of the fibres, but the scaffold showed a porous nanofibrous structure after drug release. X-ray diffraction results showed that the addition of Sal-B did not affect the formation of the nanofibre biomimetic structure of the scaffold. FTIR results indicated a certain interaction between Sal-B and PLA/GO. Water absorption and porosity test results revealed that the scaffolds had good hydrophilicity and appropriate porosity. The addition of Sal-B was also conducive to the formation of sediments possibly due to the good water solubility of Sal-B itself. The prepared scaffolds had good blood compatibility and cytocompatibility, and a small additional amount of Sal-B could significantly promote cell proliferation and alkaline phosphatase activity. Their sustained release performance indicated that the biomimetic scaffolds had controlled the release of Sal-B. The kinetic model showed that the PLA/GO/Sal-B drug-loaded biomimetic scaffolds followed the diffusion mechanism.
Collapse
Affiliation(s)
- Shuqiong Liu
- College of Ecology and Resource Engineering, Wuyi University Wuyishan 354300 People's Republic of China
| | - Zhenyi Xu
- College of Ecology and Resource Engineering, Wuyi University Wuyishan 354300 People's Republic of China
| | - Jiapeng Hu
- College of Ecology and Resource Engineering, Wuyi University Wuyishan 354300 People's Republic of China
| | - Zhenzeng Wu
- College of Ecology and Resource Engineering, Wuyi University Wuyishan 354300 People's Republic of China
| | - Yuying Zheng
- College of Materials Science and Engineering, Fuzhou University Fuzhou 350108 People's Republic of China +86-591-22866524
| |
Collapse
|
30
|
Zhang X, Wang X, Lee YW, Feng L, Wang B, Pan Q, Meng X, Cao H, Li L, Wang H, Bai S, Kong L, Chow DHK, Qin L, Cui L, Lin S, Li G. Bioactive Scaffold Fabricated by 3D Printing for Enhancing Osteoporotic Bone Regeneration. Bioengineering (Basel) 2022; 9:525. [PMID: 36290493 PMCID: PMC9598556 DOI: 10.3390/bioengineering9100525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 10/27/2023] Open
Abstract
We develop a poly (lactic-co-glycolic acid)/β-calcium phosphate (PLGA/TCP)-based scaffold through a three-dimensional (3D) printing technique incorporating icaritin (ICT), a unique phytomolecule, and secretome derived from human fetal mesenchymal stem cells (HFS), to provide mechanical support and biological cues for stimulating bone defect healing. With the sustained release of ICT and HFS from the composite scaffold, the cell-free scaffold efficiently facilitates the migration of MSCs and promotes bone regeneration at the femoral defect site in the ovariectomy (OVX)-induced osteoporotic rat model. Furthermore, mechanism study results indicate that the combination of ICT and HFS additively activates the Integrin-FAK (focal adhesion kinase)-ERK1/2 (extracellular signal-regulated kinase 1/2)-Runx2 (Runt-related transcription factor 2) axis, which could be linked to the beneficial recruitment of MSCs to the implant and subsequent osteogenesis enhancement. Collectively, the PLGA/TCP/ICT/HFS (P/T/I/S) bioactive scaffold is a promising biomaterial for repairing osteoporotic bone defects, which may have immense implications for their translation to clinical practice.
Collapse
Affiliation(s)
- Xiaoting Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xinluan Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuk-wai Lee
- SH Ho Scoliosis Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lu Feng
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Bin Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Qi Pan
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xiangbo Meng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huijuan Cao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linlong Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Haixing Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Shanshan Bai
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Lingchi Kong
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Liao Cui
- School of Pharmacy and Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
31
|
Cho S, Choi H, Jeong H, Kwon SY, Roh EJ, Jeong KH, Baek I, Kim BJ, Lee SH, Han I, Cha JM. Preclinical Study of Human Bone Marrow-Derived Mesenchymal Stem Cells Using a 3-Dimensional Manufacturing Setting for Enhancing Spinal Fusion. Stem Cells Transl Med 2022; 11:1072-1088. [PMID: 36180050 PMCID: PMC9585955 DOI: 10.1093/stcltm/szac052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/12/2022] [Indexed: 11/29/2022] Open
Abstract
Spinal fusion surgery is a surgical technique that connects one or more vertebrae at the same time to prevent movement between the vertebrae. Although synthetic bone substitutes or osteogenesis-inducing recombinant proteins were introduced to promote bone union, the rate of revision surgery is still high due to pseudarthrosis. To promote successful fusion after surgery, stem cells with or without biomaterials were introduced; however, conventional 2D-culture environments have resulted in a considerable loss of the innate therapeutic properties of stem cells. Therefore, we conducted a preclinical study applying 3D-spheroids of human bone marrow-dewrived mesenchymal stem cells (MSCs) to a mouse spinal fusion model. First, we built a large-scale manufacturing platform for MSC spheroids, which is applicable to good manufacturing practice (GMP). Comprehensive biomolecular examinations, which include liquid chromatography-mass spectrometry and bioinformatics could suggest a framework of quality control (QC) standards for the MSC spheroid product regarding the identity, purity, viability, and potency. In our animal study, the mass-produced and quality-controlled MSC spheroids, either undifferentiated or osteogenically differentiated were well-integrated into decorticated bone of the lumbar spine, and efficiently improved angiogenesis, bone regeneration, and mechanical stability with statistical significance compared to 2D-cultured MSCs. This study proposes a GMP-applicable bioprocessing platform and QC directions of MSC spheroids aiming for their clinical application in spinal fusion surgery as a new bone graft substitute.
Collapse
Affiliation(s)
- Sumin Cho
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyundoo Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Ji Roh
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwang-Hun Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Inho Baek
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Byoung Ju Kim
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae Min Cha
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| |
Collapse
|
32
|
Xia D, Chen J, Zhang Z, Dong M. Emerging polymeric biomaterials and manufacturing techniques in regenerative medicine. AGGREGATE 2022; 3. [DOI: 10.1002/agt2.176] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
AbstractThe current demand for patients’ organ and tissue repair and regeneration is continually increasing, where autologous or allograft is the golden standard treatment in the clinic. However, due to the shortage of donors, mismatched size and modality, functional loss of the donor region, possible immune rejection, and so forth, the application of auto‐/allo‐grafts is frequently hindered in many cases. In order to solve these problems, artificial constructs structurally and functionally imitating the extracellular matrix have been developed as substitutes to promoting cell attachment, proliferation, and differentiation, and ultimately forming functional tissues or organs for better tissue regeneration. Particularly, polymeric materials have been widely utilized in regenerative medicine because of their ease of manufacturing, flexibility, biocompatibility, as well as good mechanical, chemical, and thermal properties. This review presents a comprehensive overview of a variety of polymeric materials, their fabrication methods as well applications in regenerative medicine. Finally, we discussed the future challenges and perspectives in the development and clinical transformation of polymeric biomaterials.
Collapse
Affiliation(s)
- Dan Xia
- Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, School of Materials Science and Engineering Hebei University of Technology Tianjin China
| | - Jiatian Chen
- Tianjin Key Laboratory of Materials Laminating Fabrication and Interface Control Technology, School of Materials Science and Engineering Hebei University of Technology Tianjin China
| | - Zhongyang Zhang
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Aarhus Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Aarhus Denmark
| |
Collapse
|
33
|
Xu Z, Ma Y, Dai H, Tan S, Han B. Advancements and Applications in the Composites of Silk Fibroin and Graphene-Based Materials. Polymers (Basel) 2022; 14:polym14153110. [PMID: 35956625 PMCID: PMC9370577 DOI: 10.3390/polym14153110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Silk fibroin and three kinds of graphene-based materials (graphene, graphene oxide, and reduced graphene oxide) have been widely investigated in biomedical fields. Recently, the hybrid composites of silk fibroin and graphene-based materials have attracted much attention owing to their combined advantages, i.e., presenting outstanding biocompatibility, mechanical properties, and excellent electrical conductivity. However, maintaining bio-toxicity and biodegradability at a proper level remains a challenge for other applications. This report describes the first attempt to summarize the hybrid composites’ preparation methods, properties, and applications to the best of our knowledge. We strongly believe that this review will open new doors for coming researchers.
Collapse
|
34
|
Sun F, Sun X, Wang H, Li C, Zhao Y, Tian J, Lin Y. Application of 3D-Printed, PLGA-Based Scaffolds in Bone Tissue Engineering. Int J Mol Sci 2022; 23:ijms23105831. [PMID: 35628638 PMCID: PMC9143187 DOI: 10.3390/ijms23105831] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023] Open
Abstract
Polylactic acid–glycolic acid (PLGA) has been widely used in bone tissue engineering due to its favorable biocompatibility and adjustable biodegradation. 3D printing technology can prepare scaffolds with rich structure and function, and is one of the best methods to obtain scaffolds for bone tissue repair. This review systematically summarizes the research progress of 3D-printed, PLGA-based scaffolds. The properties of the modified components of scaffolds are introduced in detail. The influence of structure and printing method change in printing process is analyzed. The advantages and disadvantages of their applications are illustrated by several examples. Finally, we briefly discuss the limitations and future development direction of current 3D-printed, PLGA-based materials for bone tissue repair.
Collapse
Affiliation(s)
- Fengbo Sun
- State Key Laboratory of Advanced Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China; (X.S.); (H.W.)
- Correspondence: (F.S.); (Y.L.); Tel.: +86-010-62773741 (Y.L.)
| | - Xiaodan Sun
- State Key Laboratory of Advanced Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China; (X.S.); (H.W.)
| | - Hetong Wang
- State Key Laboratory of Advanced Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China; (X.S.); (H.W.)
| | - Chunxu Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China; (C.L.); (Y.Z.); (J.T.)
| | - Yu Zhao
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China; (C.L.); (Y.Z.); (J.T.)
| | - Jingjing Tian
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China; (C.L.); (Y.Z.); (J.T.)
| | - Yuanhua Lin
- State Key Laboratory of Advanced Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China; (X.S.); (H.W.)
- Correspondence: (F.S.); (Y.L.); Tel.: +86-010-62773741 (Y.L.)
| |
Collapse
|
35
|
Wang C, Ma Z, Yuan K, Ji T. Using scaffolds as drug delivery systems to treat bone tumor. NANOTECHNOLOGY 2022; 33:212002. [PMID: 35092950 DOI: 10.1088/1361-6528/ac5017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Surgery is the principal strategy to treat osteosarcoma and other types of bone tumors, but it causes bone defects that cannot be healed spontaneously. After surgery, patients still need to receive radiotherapy and/or chemotherapy to prevent tumor recurrence and metastasis, which leads to systemic side effects. Bone scaffolds exhibit the potentials to load cargos (drugs or growth factors) and act as drug delivery systems (DDSs) in the osteosarcoma postoperative treatment. This review introduces current types of bone scaffolds and highlights representative works using scaffolds as DDSs to treat osteosarcomas. Challenges and perspectives in the scaffold-based DDSs are also discussed. This review may provide references to develop effective and safe strategies for osteosarcoma postoperative treatment.
Collapse
Affiliation(s)
- Caifeng Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zijiu Ma
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Kemeng Yuan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
36
|
Shi G, Yang C, Wang Q, Wang S, Wang G, Ao R, Li D. Traditional Chinese Medicine Compound-Loaded Materials in Bone Regeneration. Front Bioeng Biotechnol 2022; 10:851561. [PMID: 35252158 PMCID: PMC8894853 DOI: 10.3389/fbioe.2022.851561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/01/2023] Open
Abstract
Bone is a dynamic organ that has the ability to repair minor injuries via regeneration. However, large bone defects with limited regeneration are debilitating conditions in patients and cause a substantial clinical burden. Bone tissue engineering (BTE) is an alternative method that mainly involves three factors: scaffolds, biologically active factors, and cells with osteogenic potential. However, active factors such as bone morphogenetic protein-2 (BMP-2) are costly and show an unstable release. Previous studies have shown that compounds of traditional Chinese medicines (TCMs) can effectively promote regeneration of bone defects when administered locally and systemically. However, due to the low bioavailability of these compounds, many recent studies have combined TCM compounds with materials to enhance drug bioavailability and bone regeneration. Hence, the article comprehensively reviewed the local application of TCM compounds to the materials in the bone regeneration in vitro and in vivo. The compounds included icariin, naringin, quercetin, curcumin, berberine, resveratrol, ginsenosides, and salvianolic acids. These findings will contribute to the potential use of TCM compound-loaded materials in BTE.
Collapse
Affiliation(s)
- Guiwen Shi
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chaohua Yang
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Wang
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Qing Wang, ; Rongguang Ao, ; Dejian Li,
| | - Song Wang
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gaoju Wang
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rongguang Ao
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- *Correspondence: Qing Wang, ; Rongguang Ao, ; Dejian Li,
| | - Dejian Li
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- *Correspondence: Qing Wang, ; Rongguang Ao, ; Dejian Li,
| |
Collapse
|
37
|
Chen J, Wang Y, Wang S, Zhao X, Zhao L, Wang Y. Salvianolic acid B and ferulic acid synergistically promote angiogenesis in HUVECs and zebrafish via regulating VEGF signaling. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114667. [PMID: 34597652 DOI: 10.1016/j.jep.2021.114667] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/09/2021] [Accepted: 09/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Induced vascular growth in the myocardium has been widely acknowledged as a promising intervention strategy for patients with ischemic coronary artery disease. Yet despite long-term efforts on gene, protein or cell-based pro-angiogenic therapies, the clinical translation remains challenging. Noticeably, multiple medicinal herbs have long-term documented effects in promoting blood circulation. Salvia miltiorrhiza and Ligusticum stratum are two representative traditional Chinese medicine herbs with suggested roles in enhancing organ blood supply, and Guanxinning Tablet (GXNT), a botanical drug which is formulated with these two herbs, exhibited significant efficacy against angina pectoris in clinical practices. AIM OF THE STUDY This study aimed to examine the pro-angiogenic activity of GXNT and its major components, as well as to explore their pharmacological mechanism in promoting angiogenesis. MATERIALS AND METHODS In vitro, the pro-angiogenic effects of GXNT and its major components were examined on human umbilical vein endothelial cells by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT), scratch assay, and endothelial cell tube formation assay. In vivo, the pro-angiogenic effects were examined on the ponatinib-induced angiogenesis defective zebrafish model. The active compounds were identified through phenotype-based screening in zebrafish, and their pharmacological mechanism was explored in both in vitro and in vivo models by immunofluorescent staining, cell cycle analysis, quantitative PCR and whole embryo in-situ hybridization. RESULTS We demonstrated strong pro-angiogenic effects of GXNT in both human umbilical vein endothelial cells and zebrafish model. Moreover, through phenotype-based screening in zebrafish for active compounds, pro-angiogenic effects was discovered for salvianolic acid B (Sal B), a major component of Salvia miltiorrhiza, and its activity was further enhanced when co-administered with ferulic acid (FA), which is contained in Ligusticum stratum. On the cellular level, Sal B and FA cotreatment increased endothelial cell proliferation of sprouting arterial intersomitic vessels in zebrafish, as well as largely restored G1-S cell cycle progression and cyclin D1 expression in angiogenic defective HUVECs. Through quantitative transcriptional analysis, increased expression of vegfr2 (kdr, kdrl) and vegfr1 was detected after GXNT or SalB/FA treatment, together with upregulated transcription of their ligands including vegf-a, vegf-b, and pgfb. Bevacizumab, an anti-human VEGF-A monoclonal antibody, was able to significantly, but not completely, block the pro-angiogenic effects of GXNT or SalB/FA, suggesting their multi-targeting properties. CONCLUSIONS In conclusion, from a traditional Chinese medicine with effects in enhancing blood circulation, we demonstrated the synergistic pro-angiogenic effects of Sal B and FA via both in vitro and in vivo models, which function at least partially through regulating the expression of VEGF receptors and ligands. Future studies are warranted to further elaborate the molecular interaction between these two compounds and the key regulators in the process of neovascularization.
Collapse
Affiliation(s)
- Jing Chen
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shufang Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoping Zhao
- College of Preclinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617, China.
| |
Collapse
|
38
|
Alvarez Echazú MI, Perna O, Olivetti CE, Antezana PE, Municoy S, Tuttolomondo MV, Galdopórpora JM, Alvarez GS, Olmedo DG, Desimone MF. Recent Advances in Synthetic and Natural Biomaterials-Based Therapy for Bone Defects. Macromol Biosci 2022; 22:e2100383. [PMID: 34984818 DOI: 10.1002/mabi.202100383] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/04/2021] [Indexed: 12/31/2022]
Abstract
Synthetic and natural biomaterials are a promising alternative for the treatment of critical-sized bone defects. Several parameters such as their porosity, surface, and mechanical properties are extensively pointed out as key points to recapitulate the bone microenvironment. Many biomaterials with this pursuit are employed to provide a matrix, which can supply the specific environment and architecture for an adequate bone growth. Nevertheless, some queries remain unanswered. This review discusses the recent advances achieved by some synthetic and natural biomaterials to mimic the native structure of bone and the manufacturing technology applied to obtain biomaterial candidates. The focus of this review is placed in the recent advances in the development of biomaterial-based therapy for bone defects in different types of bone. In this context, this review gives an overview of the potentialities of synthetic and natural biomaterials: polyurethanes, polyesters, hyaluronic acid, collagen, titanium, and silica as successful candidates for the treatment of bone defects.
Collapse
Affiliation(s)
- María I Alvarez Echazú
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina.,Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Anatomía Patológica, Marcelo T. de Alvear 2142 (1122), CABA, Argentina
| | - Oriana Perna
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Christian E Olivetti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Pablo E Antezana
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Sofia Municoy
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - María V Tuttolomondo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Juan M Galdopórpora
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Gisela S Alvarez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| | - Daniel G Olmedo
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Anatomía Patológica, Marcelo T. de Alvear 2142 (1122), CABA, Argentina.,CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, Buenos Aires, 1425, Argentina
| | - Martín F Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Junín 956, Piso 3°, (1113) Buenos Aires, Argentina., Universidad de Buenos Aires, Junín 956, Piso 3°, Buenos Aires, 1113, Argentina
| |
Collapse
|
39
|
Functional Graphene Nanomaterials-Based Hybrid Scaffolds for Osteogenesis and Chondrogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1351:65-87. [DOI: 10.1007/978-981-16-4923-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
Lee SS, Laganenka L, Du X, Hardt WD, Ferguson SJ. Silicon Nitride, a Bioceramic for Bone Tissue Engineering: A Reinforced Cryogel System With Antibiofilm and Osteogenic Effects. Front Bioeng Biotechnol 2021; 9:794586. [PMID: 34976982 PMCID: PMC8714913 DOI: 10.3389/fbioe.2021.794586] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023] Open
Abstract
Silicon nitride (SiN [Si3N4]) is a promising bioceramic for use in a wide variety of orthopedic applications. Over the past decades, it has been mainly used in industrial applications, such as space shuttle engines, but not in the medical field due to scarce data on the biological effects of SiN. More recently, it has been increasingly identified as an emerging material for dental and orthopedic implant applications. Although a few reports about the antibacterial properties and osteoconductivity of SiN have been published to date, there have been limited studies of SiN-based scaffolds for bone tissue engineering. Here, we developed a silicon nitride reinforced gelatin/chitosan cryogel system (SiN-GC) by loading silicon nitride microparticles into a gelatin/chitosan cryogel (GC), with the aim of producing a biomimetic scaffold with antibiofilm and osteogenic properties. In this scaffold system, the GC component provides a hydrophilic and macroporous environment for cells, while the SiN component not only provides antibacterial properties and osteoconductivity but also increases the mechanical stiffness of the scaffold. This provides enhanced mechanical support for the defect area and a better osteogenic environment. First, we analyzed the scaffold characteristics of SiN-GC with different SiN concentrations, followed by evaluation of its apatite-forming capacity in simulated body fluid and protein adsorption capacity. We further confirmed an antibiofilm effect of SiN-GC against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) as well as enhanced cell proliferation, mineralization, and osteogenic gene upregulation for MC3T3-E1 pre-osteoblast cells. Finally, we developed a bioreactor to culture cell-laden scaffolds under cyclic compressive loading to mimic physiological conditions and were able to demonstrate improved mineralization and osteogenesis from SiN-GC. Overall, we confirmed the antibiofilm and osteogenic effect of a silicon nitride reinforced cryogel system, and the results indicate that silicon nitride as a biomaterial system component has a promising potential to be developed further for bone tissue engineering applications.
Collapse
Affiliation(s)
- Seunghun S. Lee
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Leanid Laganenka
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Xiaoyu Du
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Stephen J. Ferguson
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Li J, Wang C, Gao G, Yin X, Pu X, Shi B, Liu Y, Huang Z, Wang J, Li J, Yin G. MBG/ PGA-PCL composite scaffolds provide highly tunable degradation and osteogenic features. Bioact Mater 2021; 15:53-67. [PMID: 35386352 PMCID: PMC8941175 DOI: 10.1016/j.bioactmat.2021.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 11/02/2022] Open
|
42
|
Purification of Colon Carcinoma Cells from Primary Colon Tumor Using a Filtration Method via Porous Polymeric Filters. Polymers (Basel) 2021; 13:polym13193411. [PMID: 34641226 PMCID: PMC8513025 DOI: 10.3390/polym13193411] [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: 09/13/2021] [Revised: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Cancer stem cells (CSCs) or cancer-initiating cells (CICs) are key factors for tumor generation and metastasis. We investigated a filtration method to enhance CSCs (CICs) from colon carcinoma HT-29 cells and primary colon carcinoma cells derived from patient colon tumors using poly(lactide-co-glycolic acid)/silk screen (PLGA/SK) filters. The colon carcinoma cell solutions were permeated via porous filters to obtain a permeation solution. Then, the cell cultivation media were permeated via the filters to obtain the recovered solution, where the colon carcinoma cells that adhered to the filters were washed off into the recovered solution. Subsequently, the filters were incubated in the culture media to obtain the migrated cells via the filters. Colon carcinoma HT-29 cells with high tumorigenicity, which might be CSCs (CICs), were enhanced in the cells in the recovered solution and in the migrated cells based on the CSC (CIC) marker expression, colony-forming unit assay, and carcinoembryonic antigen (CEA) production. Although primary colon carcinoma cells isolated from colon tumor tissues contained fibroblast-like cells, the primary colon carcinoma cells were purified from fibroblast-like cells by filtration through PLGA/SK filters, indicating that the filtration method is effective in purifying primary colon carcinoma cells.
Collapse
|
43
|
Comprehensive in vitro comparison of cellular and osteogenic response to alternative biomaterials for spinal implants. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112251. [PMID: 34225890 DOI: 10.1016/j.msec.2021.112251] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 02/02/2023]
Abstract
A variety of novel biomaterials are emerging as alternatives to conventional metals and alloys, for use in spinal implants. These promise potential advantages with respect to e.g. elastic modulus compatibility with the host bone, improved radiological imaging or enhanced cellular response to facilitate osseointegration. However, to date there is scarce comparative data on the biological response to many of these biomaterials that would give insights into the relative level of bone formation, resorption inhibition and inflammation. Thus, in this study, we aimed to evaluate and compare the in vitro biological response to standard discs of four alternative biomaterials: polyether ether ketone (PEEK), zirconia toughened alumina (ZTA), silicon nitride (SN) and surface-textured silicon nitride (ST-SN), and the reference titanium alloy Ti6Al4V (TI). Material-specific characteristics of these biomaterials were evaluated, such as surface roughness, wettability, protein adsorption (BSA) and apatite forming capacity in simulated body fluid. The activity of pre-osteoblasts seeded on the discs was characterized, by measuring viability, proliferation, attachment and morphology. Then, the osteogenic differentiation of pre-osteoblasts was compared in vitro from early to late stage by Alizarin Red S staining and real-time PCR analysis. Finally, osteoclast activity and inflammatory response were assessed by real-time PCR analysis. Compared to TI, all other materials generally demonstrated a lower osteoclastic activity and inflammatory response. ZTA and SN showed generally an enhanced osteogenic differentiation and actin length. Overall, we could show that SN and ST-SN showed a higher osteogenic effect than the other reference groups, an inhibitive effect against bone resorption and low inflammation, and the results indicate that silicon nitride has a promising potential to be developed further for spinal implants that require enhanced osseointegration.
Collapse
|
44
|
Jin S, Xia X, Huang J, Yuan C, Zuo Y, Li Y, Li J. Recent advances in PLGA-based biomaterials for bone tissue regeneration. Acta Biomater 2021; 127:56-79. [PMID: 33831569 DOI: 10.1016/j.actbio.2021.03.067] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
Bone regeneration is an interdisciplinary complex lesson, including but not limited to materials science, biomechanics, immunology, and biology. Having witnessed impressive progress in the past decades in the development of bone substitutes; however, it must be said that the most suitable biomaterial for bone regeneration remains an area of intense debate. Since its discovery, poly (lactic-co-glycolic acid) (PLGA) has been widely used in bone tissue engineering due to its good biocompatibility and adjustable biodegradability. This review systematically covers the past and the most recent advances in developing PLGA-based bone regeneration materials. Taking the different application forms of PLGA-based materials as the starting point, we describe each form's specific application and its corresponding advantages and disadvantages with many examples. We focus on the progress of electrospun nanofibrous scaffolds, three-dimensional (3D) printed scaffolds, microspheres/nanoparticles, hydrogels, multiphasic scaffolds, and stents prepared by other traditional and emerging methods. Finally, we briefly discuss the current limitations and future directions of PLGA-based bone repair materials. STATEMENT OF SIGNIFICANCE: As a key synthetic biopolymer in bone tissue engineering application, the progress of PLGA-based bone substitute is impressive. In this review, we summarized the past and the most recent advances in the development of PLGA-based bone regeneration materials. According to the typical application forms and corresponding crafts of PLGA-based substitutes, we described the development of electrospinning nanofibrous scaffolds, 3D printed scaffolds, microspheres/nanoparticles, hydrogels, multiphasic scaffolds and scaffolds fabricated by other manufacturing process. Finally, we briefly discussed the current limitations and proposed the newly strategy for the design and fabrication of PLGA-based bone materials or devices.
Collapse
|
45
|
Sung TC, Huang WL, Ban LK, Lee HHC, Wang JH, Su HY, Jen SH, Chang YH, Yang JM, Higuchi A, Ye Q. Enrichment of cancer-initiating cells from colon cancer cells through porous polymeric membranes by a membrane filtration method. J Mater Chem B 2021; 8:10577-10585. [PMID: 33124643 DOI: 10.1039/d0tb02312d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer-initiating cells (CICs) or cancer stem cells (CSCs) are primarily responsible for tumor initiation, growth, and metastasis and represent a few percent of the total tumor cell population. We designed a membrane filtration protocol to enrich CICs (CSCs) from the LoVo colon cancer cell line via nylon mesh filter membranes with 11 and 20 μm pore sizes and poly(lactide-co-glycolic acid)/silk screen (PLGA/silk screen) porous membranes (pore sizes of 20-30 μm). The colon cancer cell solution was filtered through the membranes to obtain a permeate solution. Subsequently, the cell culture medium was filtered through the membranes to collect the recovery solution where the cells attached to the membranes were rinsed off into the recovery solution. Then, the membranes were cultivated in the cultivation medium to collect the migrated cells from the membranes. The cells migrated from any membrane had higher expression of the CSC surface markers CD44 and CD133, had higher colony formation levels, and produced more carcinoembryonic antigen (CEA) than the colon cancer cells cultivated on conventional tissue culture plates (control). We established a method to enrich the CICs (CSCs) of colon cancer cells from migrated cells through porous polymeric membranes by the membrane filtration protocol developed in this study.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan. and School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Wei-Lun Huang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan.
| | - Lee-Kiat Ban
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan
| | - Henry Hsin-Chung Lee
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan and Graduate Institute of Translational and Interdisciplinary Medicine, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan
| | - Jia-Hua Wang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan.
| | - Her-Young Su
- Department of Obstetrics and Gynecology, Bobson Yuho Women and Children's Clinic, No. 182, Zhuangjing S. Rd., Zhubei City, Hsinchu 302, Taiwan
| | - Shih Hsi Jen
- Department of Obstetrics and Gynecology, Taiwan Landseed Hospital, 77, Kuangtai Road, Pingjen City, Taoyuan 32405, Taiwan
| | - Yen-Hsiang Chang
- Department of General Dentistry, Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
| | - Jen-Ming Yang
- Department of Chemical and Materials Engineering, Chang Gung University, Guishan, Taoyuan 333, Taiwan.
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd., Jhongli, Taoyuan, 32001, Taiwan. and School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China and Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan and Center for Emergent Matter Science, Riken, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan and Wenzhou Institute, University of Chinese Academy of Science, No. 16, Xinsan Road, Hi-tech Industry Park, Wenzhou, Zhejiang, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China and Skeletal Biology Research Center, OMFS, Massachusetts General Hospital & Harvard School of Dental Medicine, Boston, MA02114, USA and School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
46
|
Uniaxially fixed mechanical boundary condition elicits cellular alignment in collagen matrix with induction of osteogenesis. Sci Rep 2021; 11:9009. [PMID: 33907271 PMCID: PMC8079399 DOI: 10.1038/s41598-021-88505-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Osteocytes differentiated from osteoblasts play significant roles as mechanosensors in modulating the bone remodeling process. While the well-aligned osteocyte network along the trabeculae with slender cell processes perpendicular to the trabeculae surface is known to facilitate the sensing of mechanical stimuli by cells and the intracellular communication in the bone matrix, the mechanisms underlying osteocyte network formation remains unclear. Here, we developed a novel in vitro collagen matrix system exerting a uniaxially-fixed mechanical boundary condition on which mouse osteoblast-like MC3T3-E1 cells were subcultured, evoking cellular alignment along the uniaxial boundary condition. Using a myosin II inhibitor, blebbistatin, we showed that the intracellular tension via contraction of actin fibers contributed to the cellular alignment under the influence of isometric matrix condition along the uniaxially-fixed mechanical boundary condition. Furthermore, the cells actively migrated inside the collagen matrix and promoted the expression of osteoblast and osteocyte genes with their orientations aligned along the uniaxially-fixed boundary condition. Collectively, our results suggest that the intracellular tension of osteoblasts under a uniaxially-fixed mechanical boundary condition is one of the factors that determines the osteocyte alignment inside the bone matrix.
Collapse
|
47
|
Kayalar E, Deynek GT, Tok OE, Kucuk S. Effect of salvianolic acid B on new bone formation in the orthopedically expanded suture. Angle Orthod 2021; 91:248-254. [PMID: 33367542 DOI: 10.2319/042620-360.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To determine the effects of Salvianolic acid B (Sal B) on new bone formation in the orthopedically expanded premaxillary sutures in rats. MATERIALS AND METHODS The sample consisting of Sprague Dawley rats (male, n = 14) was split in half by random selection: the experiment group (Sal B) and the control group. The premaxillary suture of each rat was expanded by bonding an open-loop spring to two maxillary incisors, each end to one tooth. A 5-day expansion period followed by a 12-day retention period was conducted. The 17-day intraperitoneal administration of Sal B was performed daily for the experiment group at a dose of 40 mg/kilo. The trial was completed after sacrificing the rats and dissection of the premaxillae for histological analysis. The amount of new bone, quantity of capillaries and intensity of inflammatory cells were histomorphometrically determined while the quantities of osteoblasts and osteoclasts were determined immunohistochemically. RESULTS The Sal B group was significantly different from the control group and had greater quantities of new bone, capillaries, inflammatory cells, osteoblasts, and osteoclasts. CONCLUSIONS Salvianolic acid B displays a positive effect during premaxillary expansion with a greater number of capillaries potentially in association with higher bone formation and improved angiogenesis in rats.
Collapse
|
48
|
Xiang G, Liu K, Wang T, Hu X, Wang J, Gao Z, Lei W, Feng Y, Tao TH. In Situ Regulation of Macrophage Polarization to Enhance Osseointegration Under Diabetic Conditions Using Injectable Silk/Sitagliptin Gel Scaffolds. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002328. [PMID: 33552858 PMCID: PMC7856907 DOI: 10.1002/advs.202002328] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/13/2020] [Indexed: 05/03/2023]
Abstract
As a chronic inflammatory disease, diabetes mellitus creates a proinflammatory microenvironment around implants, resulting in a high rate of implant loosening or failure in osteological therapies. In this study, macroporous silk gel scaffolds are injected at the bone-implant interface for in situ release of sitagliptin that can regulate macrophage response to create a prohealing microenvironment in diabetes mellitus disease. Notably, it is discovered that sitagliptin induces macrophage polarization to the M2 phenotype and alleviates the impaired behaviors of osteoblasts on titanium (Ti) implants under diabetic conditions in a dose-dependent manner. The silk gel scaffolds loaded with sitagliptin elicite a stronger recruitment of M2 macrophages to the sites of Ti implants and a significant promotion of osteointegration, as compared to oral sitagliptin administration. The results suggest that injectable silk/sitagliptin gel scaffolds can be utilized to modulate the immune responses at the bone-implant interface, thus enhancing bone regeneration required for successful implantation of orthopedic and dental devices in diabetic patients.
Collapse
Affiliation(s)
- Geng Xiang
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Keyin Liu
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information TechnologyChinese Academy of SciencesShanghai200050China
| | - Tianji Wang
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Xiaofan Hu
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Jing Wang
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Zhiheng Gao
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information TechnologyChinese Academy of SciencesShanghai200050China
| | - Wei Lei
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Yafei Feng
- Department of OrthopedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032China
| | - Tiger H. Tao
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information TechnologyChinese Academy of SciencesShanghai200050China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
- School of Physical Science and TechnologyShanghaiTech UniversityShanghai200031China
- Institute of Brain‐Intelligence TechnologyZhangjiang LaboratoryShanghai200031China
- Shanghai Research Center for Brain Science and Brain‐Inspired IntelligenceShanghai200031China
| |
Collapse
|
49
|
Jiang L, Sheng K, Wang C, Xue D, Pan Z. The Effect of MMP-2 Inhibitor 1 on Osteogenesis and Angiogenesis During Bone Regeneration. Front Cell Dev Biol 2021; 8:596783. [PMID: 33553142 PMCID: PMC7862568 DOI: 10.3389/fcell.2020.596783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/28/2020] [Indexed: 01/03/2023] Open
Abstract
Bone regeneration is a popular research focus around the world. Recent studies have suggested that the formation of a vascular network as well as intrinsic osteogenic ability is important for bone regeneration. Here, we show for the first time that matrix metalloproteinase (MMP) 2 inhibitor 1 (MMP2-I1) has a positive role in the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs) and angiogenesis of human vascular endothelial cells (HUVECs). MMP2-I1 activated the p38/mitogen-activated protein kinase signaling pathway to promote the osteogenesis of hBMSCs, and promoted the angiogenesis of HUVECs via the hypoxia-inducible factor-1α signaling pathway. We also found that MMP2-I1 enhanced bone formation using a rat tibial defect model and prevented bone loss using an ovariectomy-induced mouse model of osteoporosis. Data from the mouse model demonstrated that MMP2-I1 generated more type H vessels (CD31hiEmcnhi) when preventing bone loss. These results provide important insights into the regulatory effects of MMP2-I1 on bone regeneration.
Collapse
Affiliation(s)
- Liangjun Jiang
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kunkun Sheng
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Wang
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
50
|
Chocarro‐Wrona C, de Vicente J, Antich C, Jiménez G, Martínez‐Moreno D, Carrillo E, Montañez E, Gálvez‐Martín P, Perán M, López‐Ruiz E, Marchal JA. Validation of the 1,4-butanediol thermoplastic polyurethane as a novel material for 3D bioprinting applications. Bioeng Transl Med 2021; 6:e10192. [PMID: 33532591 PMCID: PMC7823129 DOI: 10.1002/btm2.10192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022] Open
Abstract
Tissue engineering (TE) seeks to fabricate implants that mimic the mechanical strength, structure, and composition of native tissues. Cartilage TE requires the development of functional personalized implants with cartilage-like mechanical properties capable of sustaining high load-bearing environments to integrate into the surrounding tissue of the cartilage defect. In this study, we evaluated the novel 1,4-butanediol thermoplastic polyurethane elastomer (b-TPUe) derivative filament as a 3D bioprinting material with application in cartilage TE. The mechanical behavior of b-TPUe in terms of friction and elasticity were examined and compared with human articular cartilage, PCL, and PLA. Moreover, infrapatellar fat pad-derived human mesenchymal stem cells (MSCs) were bioprinted together with scaffolds. in vitro cytotoxicity, proliferative potential, cell viability, and chondrogenic differentiation were analyzed by Alamar blue assay, SEM, confocal microscopy, and RT-qPCR. Moreover, in vivo biocompatibility and host integration were analyzed. b-TPUe demonstrated a much closer compression and shear behavior to native cartilage than PCL and PLA, as well as closer tribological properties to cartilage. Moreover, b-TPUe bioprinted scaffolds were able to maintain proper proliferative potential, cell viability, and supported MSCs chondrogenesis. Finally, in vivo studies revealed no toxic effects 21 days after scaffolds implantation, extracellular matrix deposition and integration within the surrounding tissue. This is the first study that validates the biocompatibility of b-TPUe for 3D bioprinting. Our findings indicate that this biomaterial can be exploited for the automated biofabrication of artificial tissues with tailorable mechanical properties including the great potential for cartilage TE applications.
Collapse
Affiliation(s)
- Carlos Chocarro‐Wrona
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Juan de Vicente
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Applied PhysicsFaculty of Sciences, University of GranadaGranadaSpain
| | - Cristina Antich
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Gema Jiménez
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Daniel Martínez‐Moreno
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Esmeralda Carrillo
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Elvira Montañez
- Biomedical Research Institute of Málaga (IBIMA)Málaga
- Department of Orthopedic Surgery and TraumatologyVirgen de la Victoria University HospitalMálagaSpain
| | - Patricia Gálvez‐Martín
- Department of Pharmacy and Pharmaceutical TechnologySchool of Pharmacy, University of GranadaGranadaSpain
- Advanced Therapies AreaBioibérica S.A.UBarcelonaSpain
| | - Macarena Perán
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Health SciencesUniversity of JaénJaénSpain
| | - Elena López‐Ruiz
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Health SciencesUniversity of JaénJaénSpain
| | - Juan Antonio Marchal
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| |
Collapse
|