1
|
Han Q, He J, Bai L, Huang Y, Chen B, Li Z, Xu M, Liu Q, Wang S, Wen N, Zhang J, Guo B, Yin Z. Injectable Bioadhesive Photocrosslinkable Hydrogels with Sustained Release of Kartogenin to Promote Chondrogenic Differentiation and Partial-Thickness Cartilage Defects Repair. Adv Healthc Mater 2024; 13:e2303255. [PMID: 38253413 DOI: 10.1002/adhm.202303255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/05/2024] [Indexed: 01/24/2024]
Abstract
Partial-thickness cartilage defect (PTCD) is a common and formidable clinical challenge without effective therapeutic approaches. The inherent anti-adhesive characteristics of the extracellular matrix within cartilage pose a significant impediment to the integration of cells or biomaterials with the native cartilage during cartilage repair. Here, an injectable photocrosslinked bioadhesive hydrogel, consisting of gelatin methacryloyl (GM), acryloyl-6-aminocaproic acid-g-N-hydroxysuccinimide (AN), and poly(lactic-co-glycolic acid) microspheres loaded with kartogenin (KGN) (abbreviated as GM/AN/KGN hydrogel), is designed to enhance interfacial integration and repair of PTCD. After injected in situ at the irregular defect, a stable and robust hydrogel network is rapidly formed by ultraviolet irradiation, and it can be quickly and tightly adhered to native cartilage through amide bonds. The hydrogel exhibits good adhesion strength up to 27.25 ± 1.22 kPa by lap shear strength experiments. The GM/AN/KGN hydrogel demonstrates good adhesion, low swelling, resistance to fatigue, biocompatibility, and chondrogenesis properties in vitro. A rat model with PTCD exhibits restoration of a smoother surface, stable seamless integration, and abundant aggrecan and type II collagen production. The injectable stable adhesive hydrogel with long-term chondrogenic differentiation capacity shows great potential to facilitate repair of PTCD.
Collapse
Affiliation(s)
- Qian Han
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, and State Key Laboratory for Mechanical Behavior of Materials and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jiahui He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, and State Key Laboratory for Mechanical Behavior of Materials and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Huang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, and State Key Laboratory for Mechanical Behavior of Materials and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Baojun Chen
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Zhenlong Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, and State Key Laboratory for Mechanical Behavior of Materials and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Meiguang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qiaonan Liu
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuai Wang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Nuanyang Wen
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jing Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University, Xi'an, 710069, China
| | - Baolin Guo
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, and State Key Laboratory for Mechanical Behavior of Materials and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhanhai Yin
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
2
|
Tang Y, Wang K, Wu B, Yao K, Feng S, Zhou X, Xiang L. Photoelectrons Sequentially Regulate Antibacterial Activity and Osseointegration of Titanium Implants. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307756. [PMID: 37974525 DOI: 10.1002/adma.202307756] [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: 08/02/2023] [Revised: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Titanium implants are widely used ; however, implantation occasionally fails due to infections during the surgery or poor osseointegration after the surgery. To solve the problem, an intelligent functional surface on titanium implant that can sequentially eradicate bacteria biofilm at the initial period and promote osseointegration at the late period of post-surgery time is designed. Such surfaces can be excited by near infrared light (NIR), with rare earth nanoparticles to upconvert the NIR light to visible range and adsorb by Au nanoparticles, supported by titanium oxide porous film on titanium implants. Under NIR irradiation, the implant converts the energy of phonon to hot electrons and lattice vibrations, while the former flows directly to the contact substance or partially reacts with the surrounding to generate reactive oxygen species, and the latter leads to the local temperature increase. The biofilm or microbes on the implant surface can be eradicated by NIR treatment in vitro and in vivo. Additionally, the surface exhibits superior biocompatibility for cell survival, adhesion, proliferation, and osteogenic differentiation, which provides the foundation for osseointegration. In vivo implantation experiments demonstrate osseointegration is also promoted. This work thus demonstrates NIR-generated electrons can sequentially eradicate biofilms and regulate the osteogenic process, providing new solutions to fabricate efficient implant surfaces.
Collapse
Affiliation(s)
- Yufei Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kai Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Bingfeng Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Keyi Yao
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Shuqi Feng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuemei Zhou
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Zhang Q, Zhou X, Du H, Ha Y, Xu Y, Ao R, He C. Bifunctional Hydrogel-Integrated 3D Printed Scaffold for Repairing Infected Bone Defects. ACS Biomater Sci Eng 2023; 9:4583-4596. [PMID: 37318182 DOI: 10.1021/acsbiomaterials.3c00564] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The clinical treatment of infectious bone defects is difficult and time-consuming due to the coexistence of infection and bone defects, and the simultaneous control of infection and repair of bone defects is considered a promising therapy. In this study, a dual-drug delivery scaffold system was fabricated by the combination of a three-dimensional (3D) printed scaffold with hydrogel for infected bone defects repair. The 3D printed polycaprolactone scaffold was incorporated with biodegradable mesoporous silica nanoparticles containing the small molecular drug fingolimod (FTY720) to provide structural support and promote angiogenesis and osteogenesis. The vancomycin (Van)-loaded hydrogel was prepared from aldehyde hyaluronic acid (AHA) and carboxymethyl chitosan (NOCC) by the Schiff base reaction, which can fill the pores of the 3D-printed scaffold to produce a bifunctional composite scaffold. The in vitro results demonstrated that the composite scaffold had Van concentration-dependent antimicrobial properties. Furthermore, the FTY720-loaded composite scaffold demonstrated excellent biocompatibility, vascularization, and osteogenic ability in vitro. In the rat femoral defect model with bacterial infection, the dual-drug composite scaffold showed a better outcome in both infection control and bone regeneration compared to other groups. Therefore, the prepared bifunctional composite scaffold has potential application in the treatment of infected bone defects.
Collapse
Affiliation(s)
- Qianqian Zhang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Xiaojun Zhou
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Haibo Du
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Yujie Ha
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Yao Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Rongguang Ao
- Department of Trauma Orthopaedics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P. R. China
| | - Chuanglong He
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine; College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| |
Collapse
|
4
|
Bian Y, Cai X, Lv Z, Xu Y, Wang H, Tan C, Liang R, Weng X. Layered Double Hydroxides: A Novel Promising 2D Nanomaterial for Bone Diseases Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301806. [PMID: 37329200 PMCID: PMC10460877 DOI: 10.1002/advs.202301806] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/07/2023] [Indexed: 06/18/2023]
Abstract
Bone diseases including bone defects, bone infections, osteoarthritis, and bone tumors seriously affect life quality of the patient and bring serious economic burdens to social health management, for which the current clinical treatments bear dissatisfactory therapeutic effects. Biomaterial-based strategies have been widely applied in the treatment of orthopedic diseases but are still plagued by deficient bioreactivity. With the development of nanotechnology, layered double hydroxides (LDHs) with adjustable metal ion composition and alterable interlayer structure possessing charming physicochemical characteristics, versatile bioactive properties, and excellent drug loading and delivery capabilities arise widespread attention and have achieved considerable achievements for bone disease treatment in the last decade. However, to the authors' best knowledge, no review has comprehensively summarized the advances of LDHs in treating bone disease so far. Herein, the advantages of LDHs for orthopedic disorders treatment are outlined and the corresponding state-of-the-art achievements are summarized for the first time. The potential of LDHs-based nanocomposites for extended therapeutics for bone diseases is highlighted and perspectives for LDHs-based scaffold design are proposed for facilitated clinical translation.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Xuejie Cai
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| | - Chaoliang Tan
- Department of Chemistry and Center of Super‐Diamond and Advanced Films (COSDAF)City University of Hong KongKowloonHong KongP. R. China
- Shenzhen Research InstituteCity University of Hong KongShenzhen518057P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100730P. R. China
| |
Collapse
|
5
|
Baei P, Daemi H, Aramesh F, Baharvand H, Eslaminejad MB. Advances in mechanically robust and biomimetic polysaccharide-based constructs for cartilage tissue engineering. Carbohydr Polym 2023; 308:120650. [PMID: 36813342 DOI: 10.1016/j.carbpol.2023.120650] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The purpose of cartilage tissue engineering is to provide artificial constructs with biological functions and mechanical features that resemble native tissue to improve tissue regeneration. Biochemical characteristics of the cartilage extracellular matrix (ECM) microenvironment provide a platform for researchers to develop biomimetic materials for optimal tissue repair. Due to the structural similarity of polysaccharides into physicochemical characteristics of cartilage ECM, these natural polymers capture special attention for developing biomimetic materials. The mechanical properties of constructs play a crucial influence in load-bearing cartilage tissues. Moreover, the addition of appropriate bioactive molecules to these constructs can promote chondrogenesis. Here, we discuss polysaccharide-based constructs that can be used to create substitutes for cartilage regeneration. We intend to focus on newly developed bioinspired materials, fine-tuning the mechanical properties of constructs, the design of carriers loaded by chondroinductive agents, and development of appropriate bioinks as a bioprinting approach for cartilage regeneration.
Collapse
Affiliation(s)
- Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran.
| | - Fatemeh Aramesh
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University ofTehran, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
6
|
Khayati M, Manjili HK, Soleimani M, Hosseinzadeh S, Akrami M, Haririan I, Tafti SHA. Microfluidic synthesis of zoledronic acid loaded chitosan nanoparticles used for osteogenic differentiation of mesenchymal cells. Int J Biol Macromol 2023; 234:123056. [PMID: 36587647 DOI: 10.1016/j.ijbiomac.2022.12.275] [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: 08/10/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022]
Abstract
Zoledronic acid (ZA) is known as a potent bisphosphonate in osteogenic differentiation, but at high doses, it possesses toxic effects and causes decreased proliferation and differentiation of osteoblasts. Therefore, encapsulation of ZA into nanoparticles and control of its release is expected to promote differentiation of stem cells into osteoblasts. The present work aimed to develop a simple method for synthesis of monodisperse ZA-loaded chitosan (CS) nanoparticles. In this regard, we proposed a microfluidic synthesis of nanoparticles through the ionic cross-linking of CS in the presence of ZA without a crosslinker. The main advantages of these microfluidic generated nanoparticles were narrow size distribution and fine spherical shape. Conversely, the nanoparticles that were synthesized using a bulk mixing method had an irregular shape with a broad size distribution. Real-time PCR assay as well as alizarin red staining were used to evaluate the in-vitro osteogenic potential of the nanoparticles. The results indicated that the controlled release of ZA from the microfluidic system generated uniform nanoparticles, improving the osteogenic differentiation of mesenchymal stem cells. Additionally, this microfluidic device provided the well-controlled synthesis of novel nanoparticles with a modified CS macromolecular polymer for targeted drug delivery systems.
Collapse
Affiliation(s)
- Maryam Khayati
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamidreza Kheiri Manjili
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Masoud Soleimani
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simzar Hosseinzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Institute of Biomaterials, University of Tehran & Tehran University of Medical Sciences (IBUTUMS), Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Institute of Biomaterials, University of Tehran & Tehran University of Medical Sciences (IBUTUMS), Tehran, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center Hospital, Tehran University of Medical Sciences, Tehran 14535, Iran
| |
Collapse
|
7
|
Gurgul SJ, Moreira A, Xiao Y, Varma SN, Liu C, Costa PF, Williams GR. Electrosprayed Particles Loaded with Kartogenin as a Potential Osteochondral Repair Implant. Polymers (Basel) 2023; 15:polym15051275. [PMID: 36904516 PMCID: PMC10007262 DOI: 10.3390/polym15051275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The restoration of cartilage damage is a slow and not always successful process. Kartogenin (KGN) has significant potential in this space-it is able to induce the chondrogenic differentiation of stem cells and protect articular chondrocytes. In this work, a series of poly(lactic-co-glycolic acid) (PLGA)-based particles loaded with KGN were successfully electrosprayed. In this family of materials, PLGA was blended with a hydrophilic polymer (either polyethyleneglycol (PEG) or polyvinylpyrrolidone (PVP)) to control the release rate. Spherical particles with sizes in the range of 2.4-4.1 µm were fabricated. They were found to comprise amorphous solid dispersions, with high entrapment efficiencies of >93%. The various blends of polymers had a range of release profiles. The PLGA-KGN particles displayed the slowest release rate, and blending with PVP or PEG led to faster release profiles, with most systems giving a high burst release in the first 24 h. The range of release profiles observed offers the potential to provide a precisely tailored profile via preparing physical mixtures of the materials. The formulations are highly cytocompatible with primary human osteoblasts.
Collapse
Affiliation(s)
| | | | - Yi Xiao
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Swastina Nath Varma
- Institute of Orthopaedic and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore HA7 4AP, UK
| | - Chaozong Liu
- Institute of Orthopaedic and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore HA7 4AP, UK
| | | | - Gareth R. Williams
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
- Correspondence: ; Tel.: +44-0203-987-2817
| |
Collapse
|
8
|
Mohsenifard S, Mashayekhan S, Safari H. A hybrid cartilage extracellular matrix-based hydrogel/poly (ε-caprolactone) scaffold incorporated with Kartogenin for cartilage tissue engineering. J Biomater Appl 2023; 37:1243-1258. [PMID: 36217954 DOI: 10.1177/08853282221132987] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite extensive studies, hydrogels are unable to meet the mechanical and biological requirements for successful outcomes in cartilage tissue engineering. In the present study, beta cyclodextrin (β-CD)-modified alginate/cartilage extracellular matrix (ECM)-based interpenetrating polymer network (IPN) hydrogel was developed for sustained release of Kartogenin (KGN). Furthermore, the hydrogel was incorporated within a 3D-printed poly (ε-caprolactone) (PCL)/starch microfiber network in order to reinforce the construct for cartilage tissue engineering. All the synthesized compounds were characterized by H1-NMR spectroscopy. The hydrogel/microfiber composite with a microfiber strand size and strand spacing of 300 μm and 2 mm, respectively showed a compressive modulus of 17.2 MPa, resembling the properties of the native cartilage tissue. Considering water uptake capacity, degradation rate, mechanical property, cell cytotoxicity and glycosaminoglycan secretions, β-CD-modified hydrogel reinforced with printed PCL/starch microfibers with controlled release of KGN may be considered as a promising candidate for using in articular cartilage defects.
Collapse
Affiliation(s)
- Sadaf Mohsenifard
- Chemical and Petroleum Engineering Department, 68260Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Chemical and Petroleum Engineering Department, 68260Sharif University of Technology, Tehran, Iran
| | - Hanieh Safari
- Chemical and Petroleum Engineering Department, 68260Sharif University of Technology, Tehran, Iran
| |
Collapse
|
9
|
Zeng C, Wang S, Chen F, Wang Z, Li J, Xie Z, Ma M, Wang P, Shen H, Wu Y. Alpinetin alleviates osteoporosis by promoting osteogenic differentiation in BMSCs by triggering autophagy via PKA/mTOR/ULK1 signaling. Phytother Res 2023; 37:252-270. [PMID: 36104214 PMCID: PMC10087978 DOI: 10.1002/ptr.7610] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Abstract
Osteoporosis, a systemic bone disease that is characterized by a reduction in bone mass and destruction of bone microstructure, is becoming a serious problem worldwide. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into bone-forming osteoblasts, and play an important role in maintaining homeostasis of bone metabolism, thus being a potential therapeutic target for osteoporosis. Although the phytochemical alpinetin (APT) has been reported to possess a variety of pharmacological activities, it is still unclear whether APT can influence the osteogenic differentiation of on BMSCs and if it can improve osteoporosis. In this study, we found that APT treatment was able to enhance osteogenic differentiation levels of human BMSCs in vitro and mouse ones in vivo as revealed by multiple osteogenic markers including increased alkaline phosphatase activity and osteocalcin expression. Mechanistically, the protein kinase A (PKA)/mTOR/ULK1 signaling was involved in the action of APT to enhance the osteogenic differentiation of BMSCs. In addition, oral administration of APT significantly mitigated the bone loss in a dexamethasone-induced mouse model of osteoporosis through strengthening PKA signaling and autophagy. Altogether, these data demonstrate that APT promotes osteogenic differentiation in BMSCs by augmenting the PKA/mTOR/ULK1 autophagy signaling, highlighting its potential therapeutic application for treating osteoporotic diseases.
Collapse
Affiliation(s)
- Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jinteng Li
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| |
Collapse
|
10
|
Xing Y, Zhang MS, Xiao JH, Liu RM. Galangin induces the osteogenic differentiation of human amniotic mesenchymal stem cells via the JAK2/STAT3 signaling pathway. Eur J Pharmacol 2022; 935:175326. [PMID: 36257381 DOI: 10.1016/j.ejphar.2022.175326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/03/2022]
|
11
|
Osteogenic Differentiation Effect of BMP-9 with Phenamil and Simvastatin on Intact Human Amniotic Epithelial Stem Cells. IRANIAN BIOMEDICAL JOURNAL 2022; 26:463-74. [PMID: 36437797 PMCID: PMC9841223 DOI: 10.52547/ibj.3748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Background: Bone tissue engineering has shown to be a promising strategy for repairing bone defects without causing harmful side effects to the patient. Three main building blocks of tissue engineering, including seeding cells, scaffold, and signaling molecules, are required for adequate bone regeneration. The human amniotic membrane (hAM) is the innermost of the placental membranes. In addition to providing a source of stem cells and growth factors, hAM has several features that make it an appropriate scaffold containing stem cells for use in tissue engineering purposes. The present investigation aimed to assess the effect of bone morphogenetic protein-9 (BMP-9) combined with phenamil and simvastatin on osteogenic induction of hAM with its human amniotic membrane epithelial cells (hAECs). Method Methods: Using six different osteogenic medium (OMs), we cultured hAM for 14 days. The basic OMs were chosen as the first group and other media were made by adding BMP-9, phenamil, simvastatin, BMP-9 alongside phenamil, and BMP-9 alongside simvastatin to the basic OMs. Finally, viability assay, tissue mineralization, calcium and phosphate content determination, and measurement of lactic acid dehydrogenase (LDH), and alkaline phosphatase (ALP) activity were performed. Results Results: Among all study groups, groups containing simvastatin showed a significantly lower level of viability. Although all media could induce osteogenic features, the hAECs cultured in media containing BMP-9 and phenamil demonstrated a wider area of mineralization and a significantly higher level of calcium and phosphate content, LDH, and ALP activity. Conclusion Conclusion: Our findings indicated that the use of phenamil together with BMP-9 could synergistically show in situ osteogenic induction in hAECs, which could be a new insight into translational medicine.
Collapse
|
12
|
Cui X, Wang X, Wen J, Li X, Li N, Hao X, Zhao B, Wu X, Miao J. Identification of a new way to induce differentiation of dermal fibroblasts into vascular endothelial cells. STEM CELL RESEARCH & THERAPY 2022; 13:501. [PMID: 36210433 PMCID: PMC9549676 DOI: 10.1186/s13287-022-03185-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 09/04/2021] [Indexed: 12/03/2022]
Abstract
Background Human dermal fibroblasts (HDFs) have the potential to differentiate into vascular endothelial cells (VECs), but their differentiation rate is low and the mechanism involved is not clear. The small molecule pathway controls the phenotype of fibroblasts by activating cellular signaling pathways, which is a more convenient method in the differentiation strategy of HDFs into VECs. Methods In this study, HDFs were treated with the different doses of CPP ((E)-4-(4-(4-(7-(diethylamino)-2-oxo-2H-chromene-3-carbonyl) piperazin-1-yl) styryl)-1-methylpyridin-1-ium iodide), and the mRNA and protein levels of HDFs were detected by qPCR, Western blot, flow cytometry and immunofluorescent staining. The matrigel assays, acetylated-LDL uptake and angiogenesis assays of chick embryo chorioallantoic membrane (CAM) and hindlimb ischemia model of nude mice were performed to evaluate the functions of VECs derived from HDFs. Results Here, we report that the small chemical molecule, CPP, can effectively induce HDFs to differentiate into VECs. First, we observed the morphological changes of HDFS treated with CPP. Flow cytometry, Western blot and qRT-PCR analyses showed that CPP effectively decreased the level of the HDFs-marker Vimentin and increased levels of the VEC-markers CD31, CD133, TEK, ERG, vWF, KDR and CDH5. Detection of the percentage of CD31-positive cells by immunofluorescent staining confirmed that CPP can effectively induce HDFs to differentiate into VECs. The results of Matrigel assays, DiI-ac-LDL uptake, angiogenesis assays on CAM and hindlimb ischemia model of nude mice showed that CPP-induced HDFs have the functions of VECs in vitro and in vivo. Western blot and qRT-PCR analysis showed that CPP induces HDFs to differentiate into VECs by promoting the expression of pro-angiogenic factors (VEGF, FGF-2 and PDGF-BB). Conclusions Our data suggest that the small chemical molecule CPP efficiently induces the differentiation of HDFs into VECs. Simultaneously, this new inducer provides a potential to develop new approaches to restore vascular function for the treatment of ischemic vascular diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03185-4.
Collapse
|
13
|
Chang S, Li C, Xu N, Wang J, Jing Z, Cai H, Tian Y, Wang S, Liu Z, Wang X. A sustained release of alendronate from an injectable tetra-PEG hydrogel for efficient bone repair. Front Bioeng Biotechnol 2022; 10:961227. [PMID: 36177182 PMCID: PMC9513246 DOI: 10.3389/fbioe.2022.961227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023] Open
Abstract
Significant efforts on construction of smart drug delivery for developing minimally invasive gelling system to prolong local delivery of bisphosphonates are considered as promising perspectives for the bone-related diseases, which provide the hydrogels with unique bioactivities for bone repair in clinic. Herein, we have constructed an alendronate (ALN)-conjoined injectable tetra-PEG hydrogel with excellent biocompatibility, uniform network, and favorable mechanical properties in one-pot strategy. In views of the quick ammonolysis reaction between N-hydroxysuccinimide (NHS)-ester of tetra-PEG-SG and amine groups of tetra-PEG-NH2 polymer and ALN molecules, the uniform networks were formed within seconds along with the easy injection, favorable biocompatibility and mechanical properties for hydrogel scaffolds. On account of the simultaneous physical encapsulation and chemical linkage of the ALN within the hydrogels, the ALN-conjoined tetra-PEG hydrogel exhibited a sustained drug release delivery that could persistently and effectively facilitate viability, growth, proliferation, and osteogenesis differentiation of stem cells, thereby allowing the consequent adaptation of hydrogels into the bone defects with irregular shapes, which endowed the ALN-conjoined tetra-PEG hydrogel with depot formulation capacity for governing the on-demand release of ALN drugs. Consequently, the findings imply that these drug-based tetra-PEG hydrogels mediate optimal release of therapeutic cargoes and effective promotion of in situ bone regeneration, which will be broadly utilized as therapeutic scaffolds in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shuai Chang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Chao Li
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Nanfang Xu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Jiedong Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Zehao Jing
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Hong Cai
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Shaobo Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
- *Correspondence: Zhongjun Liu, ; Xing Wang,
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Zhongjun Liu, ; Xing Wang,
| |
Collapse
|
14
|
Fallahi H, Daemi H, Bagheri F, Baghaban Eslaminejad M. A supramolecular injectable hydrogel based on β-cyclodextrin-grafted alginate and pluronic-amine loaded with kartogenin for chondrogenic differentiation of mesenchymal stem cells. Biomed Mater 2022; 17. [PMID: 35995044 DOI: 10.1088/1748-605x/ac8bbd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Owing to the similarity of hydrogels to cartilage ECM, they have been extensively utilized in the chondral lesions. Moreover, their tunable administration properties are desirable for reducing injuries in lesion sites. Generally, injectable hydrogels are mechanically weak, requiring some modifications for being used as a cell carrier in place of articular cartilage. In this study, a combination of β-cyclodextrin-grafted alginate (Alg-β-CD) and pluronic-amine (PA) with multiple physical crosslinking was used for the first time. Supramolecular interactions, including electrostatic forces, host-guest interaction, and hydrophobic interaction with increasing temperature maintain injectability of hydrogels while these interactions boost mechanical properties to the extent that shear modulus surpassed 40 kPa. Vacant β-CD cavities in conjunction with gel network was exploited for kartogenin (KGN) loading. All groups had gel time of less than one minute and gel temperature was 28 ℃. No toxic effect of hydrogels on encapsulated cells was observed. While the optimum combination of polymers provided a sustainable release for KGN, it also extended the in vitro degradation time of hydrogels from 6 days to 2 weeks. KGN facilitated encapsulated mesenchymal stem cells (MSCs) differentiation towards chondrocytes. Taken together, the synthesized hydrogel proved to be a promising candidate for being utilized in cartilage regeneration.
Collapse
Affiliation(s)
- Hooman Fallahi
- Department of Biomedical Engineering, Tarbiat Modares University, jalale al ahmad, Tehran, 0098, Iran (the Islamic Republic of)
| | - Hamed Daemi
- Cell Enginerring, Royan institute, Banihashem street, Tehran, 0098, Iran (the Islamic Republic of)
| | - Fatemeh Bagheri
- Department of Biotechnology, Tarbiat Modares University, Jallale al ahmad, Tehran, 0098, Iran (the Islamic Republic of)
| | | |
Collapse
|
15
|
Shokri M, Dalili F, Kharaziha M, Baghaban Eslaminejad M, Ahmadi Tafti H. Strong and bioactive bioinspired biomaterials, next generation of bone adhesives. Adv Colloid Interface Sci 2022; 305:102706. [PMID: 35623113 DOI: 10.1016/j.cis.2022.102706] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/20/2022] [Accepted: 05/15/2022] [Indexed: 12/29/2022]
Abstract
The bone adhesive is a clinical requirement for complicated bone fractures always articulated by surgeons. Applying glue is a quick and easy way to fix broken bones. Adhesives, unlike conventional fixation methods such as wires and sutures, improve healing conditions and reduce postoperative pain by creating a complete connection at the fractured joint. Despite many efforts in the field of bone adhesives, the creation of a successful adhesive with robust adhesion and appropriate bioactivity for the treatment of bone fractures is still in its infancy. Because of the resemblance of the body's humid environment to the underwater environment, in the latest decades, researchers have pursued inspiration from nature to develop strong bioactive adhesives for bone tissue. The aim of this review article is to discuss the recent state of the art in bone adhesives with a specific focus on biomimetic adhesives, their action mechanisms, and upcoming perspective. Firstly, the adhesive biomaterials with specific affinity to bone tissue are introduced and their rational design is studied. Consequently, various types of synthetic and natural bioadhesives for bone tissue are comprehensively overviewed. Then, bioinspired-adhesives are described, highlighting relevant structures and examples of biomimetic adhesives mainly made of DOPA and the complex coacervates inspired by proteins secreted in mussel and sandcastle worms, respectively. Finally, this article overviews the challenges of the current bioadhesives and the future research for the improvement of the properties of biomimetic adhesives for use as bone adhesives.
Collapse
Affiliation(s)
- Mahshid Shokri
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Faezeh Dalili
- School of Metallurgy & Materials Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Hossein Ahmadi Tafti
- Tehran Heart Hospital Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Jiang Y, Tao Y, Chen Y, Xue X, Ding G, Wang S, Liu G, Li M, Su J. Role of Phosphorus-Containing Molecules on the Formation of Nano-Sized Calcium Phosphate for Bone Therapy. Front Bioeng Biotechnol 2022; 10:875531. [PMID: 35813995 PMCID: PMC9257216 DOI: 10.3389/fbioe.2022.875531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/16/2022] [Indexed: 12/11/2022] Open
Abstract
Calcium phosphate (CaP) is the principal inorganic constituent of bone and teeth in vertebrates and has various applications in biomedical areas. Among various types of CaPs, amorphous calcium phosphate (ACP) is considered to have superior bioactivity and biodegradability. With regard to the instability of ACP, the phosphorus-containing molecules are usually adopted to solve this issue, but the specific roles of the molecules in the formation of nano-sized CaP have not been clearly clarified yet. Herein, alendronate, cyclophosphamide, zoledronate, and foscarnet are selected as the model molecules, and theoretical calculations were performed to elucidate the interaction between calcium ions and different model molecules. Subsequently, CaPs were prepared with the addition of the phosphorus-containing molecules. It is found that cyclophosphamide has limited influence on the generation of CaPs due to their weak interaction. During the co-precipitation process of Ca2+ and PO43-, the competitive relation among alendronate, zoledronate, and foscarnet plays critical roles in the produced inorganic-organic complex. Moreover, the biocompatibility of CaPs was also systematically evaluated. The DFT calculation provides a convincing strategy for predicting the structure of CaPs with various additives. This work is promising for designing CaP-based multifunctional drug delivery systems and tissue engineering materials.
Collapse
Affiliation(s)
- Yingying Jiang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yali Tao
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Yutong Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xu Xue
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Gangyi Ding
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai, China
| | - Guodong Liu
- Wound Care Center, Daping Hospital, Army Medical Center of PLA, Chongqing, China
- *Correspondence: Guodong Liu, ; Mengmeng Li, ; Jiacan Su,
| | - Mengmeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- *Correspondence: Guodong Liu, ; Mengmeng Li, ; Jiacan Su,
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Guodong Liu, ; Mengmeng Li, ; Jiacan Su,
| |
Collapse
|
17
|
Rastegar Adib F, Bagheri F, Sharifi AM. Osteochondral regeneration in rabbit using xenograft decellularized ECM in combination with different biological products; platelet-rich fibrin, amniotic membrane extract, and mesenchymal stromal cells. J Biomed Mater Res B Appl Biomater 2022; 110:2089-2099. [PMID: 35383398 DOI: 10.1002/jbm.b.35063] [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: 09/30/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/11/2022]
Abstract
This study aimed to investigate the regenerative effect of decellularized osteochondral ECM xenograft in combination with various biological products in an osteochondral (OC) defect. OC tissue from the sheep femur were obtained and decellularized. The decellularized ECM (dECM) was combined with either platelet-rich fibrin (PRF), amniotic membrane extract (AME), or rabbit bone marrow-derived mesenchymal stromal cells (rBMSCs). The hybrid dECM-biological products were then utilized for the treatment of rabbit OC critical size defects. The regenerative potential of different groups was compared using; MRI, macroscopic assessment, histopathology, and histomorphometry. All characterizations analysis verified successful decellularization. Three months post-surgery, macroscopic findings indicated that dECM was better compared to controls. Also, dECM in combination with AME, PRF, and rBMSCs showed enhanced OC regeneration compared to only dECM (AME: +100%, PRF: +61%, rBMSCs: +28%). In particular, the dECM+AME group results in the best integration of new cartilage into surrounding cartilage tissue. The histomorphometric evaluations demonstrated enhancement in new cartilage formation and bone tissue (86.5 ± 5.9% and 90 ± 7.7%, respectively) for the dECM+AME group compared to other groups. Furthermore, histological results for the dECM+AME elucidated a mature hyaline cartilage tissue that covered the new and symmetrically formed subchondral bone, exhibiting a significantly higher regenerative effect compared to all other treated groups. This finding was also confirmed with MRI images. The current study revealed that in addition to the benefits of dECM alone, its combination with AME indicated to have a superior regenerative effect on OC regeneration. Overall, dECM+AME may be considered a suitable construct for treating knee OC injuries.
Collapse
Affiliation(s)
- Fatemeh Rastegar Adib
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Group, (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
18
|
Chan YH, Ho KN, Lee YC, Chou MJ, Lew WZ, Huang HM, Lai PC, Feng SW. Melatonin enhances osteogenic differentiation of dental pulp mesenchymal stem cells by regulating MAPK pathways and promotes the efficiency of bone regeneration in calvarial bone defects. Stem Cell Res Ther 2022; 13:73. [PMID: 35183254 PMCID: PMC8858457 DOI: 10.1186/s13287-022-02744-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stem cell (MSC)-based tissue engineering plays a major role in regenerative medicine. However, the efficiency of MSC transplantation and survival of engrafted stem cells remain challenging. Melatonin can regulate MSC biology. However, its function in the osteogenic differentiation of dental pulp-derived MSCs (DPSCs) remains unclear. We investigated the effects and mechanisms of melatonin on the osteogenic differentiation and bone regeneration capacities of DPSCs. Methods The biological effects and signaling mechanisms of melatonin with different concentrations on DPSCs were evaluated using a proliferation assay, the quantitative alkaline phosphatase (ALP) activity, Alizarin red staining, a real-time polymerase chain reaction, and a western blot in vitro cell culture model. The in vivo bone regeneration capacities were assessed among empty control, MBCP, MBCP + DPSCs, and MBCP + DPSCs + melatonin preconditioning in four-created calvarial bone defects by using micro-computed tomographic, histological, histomorphometric, and immunohistochemical analyses after 4 and 8 weeks of healing. Results In vitro experiments revealed that melatonin (1, 10, and 100 μM) significantly and concentration-dependently promoted proliferation, surface marker expression (CD 146), ALP activity and extracellular calcium deposition, and osteogenic gene expression of DPSCs (p < 0.05). Melatonin activated the protein expression of ALP, OCN, and RUNX-2 and inhibited COX-2/NF-κB expression. Furthermore, the phosphorylation of mitogen-activated protein kinase (MAPK) p38/ERK signaling was significantly increased in DPSCs treated with 100 μM melatonin, and their inhibitors significantly decreased osteogenic differentiation. In vivo experiments demonstrated that bone defects implanted with MBCP bone-grafting materials and melatonin-preconditioned DPSCs exhibited significantly greater bone volume fraction, trabecular bone structural modeling, new bone formation, and osteogenesis-related protein expression than the other three groups at 4 and 8 weeks postoperatively (p < 0.05). Conclusions These results suggest that melatonin promotes the proliferation and osteogenic differentiation of DPSCs by regulating COX-2/NF-κB and p38/ERK MAPK signaling pathways. Preconditioning DPSCs with melatonin before transplantation can efficiently enhance MSCs function and regenerative capacities.
Collapse
|
19
|
Woo Y, Patel M, Kim H, Park JK, Jung YJ, Cha SS, Jeong B. Pralatrexate Sustainably Released from Polypeptide Thermogel Is Effective for Chondrogenic Differentiation of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3773-3783. [PMID: 35014790 DOI: 10.1021/acsami.1c20585] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Folic acid was reported to significantly improve chondrogenic differentiation of mesenchymal stem cells. In a similar mechanism of action, we investigated clinically approved antifolates by the U.S. Food and Drug Administration as chondrogenic-promoting compounds for tonsil-derived mesenchymal stem cells. A poly(ethylene glycol)-poly(l-alanine) thermogelling system was used as a three-dimensional cell culture matrix, where stem cells and antifolates could be incorporated simultaneously during a heat-induced in situ sol-to-gel transition. The antifolates could be supplied over several days by the sustained release of the drug from the thermogel. Initially, seven antifolates were prescreened based on cell viability and expression of a typical chondrogenic biomarker of type II collagen (COL II) at the mRNA level. Then, dapsone, pralatrexate, and trimethoprim were selected as candidate compounds in the second round screening, and detailed studies were carried out on the mRNA and protein expression of various chondrogenic biomarkers including COL II, SRY box transcription factor 9, and aggrecan. Three-dimensional cultures of stem cells in the thermogel in the absence of a chondrogenic promoter compound and in the presence of kartogenin (KGN) were performed as a negative control and positive control, respectively. The chondrogenic biomarkers were significantly increased in the selected antifolate-incorporating systems compared to the negative control system, without an increase in type I collagen (an osteogenic biomarker) expression. Pralatrexate was the best compound for inducing chondrogenic differentiation of the stem cells, even better than the positive control (KGN). Nuclear translocation of the core-binding factor β subunit (CBFβ) and enhanced nuclear runt-related transcription factor 1 (RUNX1) by antifolate treatment suggested that the chondrogenesis-enhancing mechanism is mediated by CBFβ and RUNX1. An in silico modeling study confirmed the mechanism by proving the high binding affinity of pralatrexate to a target protein of filamin A compared with other antifolate candidates. To conclude, pralatrexate was rediscovered as a lead compound, and the polypeptide thermogel incorporating pralatrexate and mesenchymal stem cells can be a very effective system in promoting chondrogenic differentiation of stem cells and might be used in injectable tissue engineering for cartilage repair.
Collapse
Affiliation(s)
- Yejin Woo
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Hyelin Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Jin Kyung Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Yeon-Ju Jung
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| |
Collapse
|
20
|
Sun Z, Xu X, Lv Z, Li J, Shi T, Sun H, Sun K, Tan G, Yan W, Yang YX, Wu R, Xu J, Guo H, Jiang Q, Shi D. Intraarticular injection of SHP2 inhibitor SHP099 promotes the repair of rabbit full-thickness cartilage defect. J Orthop Translat 2022; 32:112-120. [PMID: 35228993 PMCID: PMC8857578 DOI: 10.1016/j.jot.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/06/2022] [Accepted: 01/13/2022] [Indexed: 11/19/2022] Open
Abstract
Background Cartilage repair has been a challenge in the field of orthopaedics for decades, highlighting the significance of investigating potential therapeutic drugs. In this study, we explored the effect of the SHP2 inhibitor SHP099, a small-molecule drug, on cartilage repair. Methods Human synovial mesenchymal stem cells (SMSCs) were isolated, and their three-way differentiation potential was examined. After treatment with chondrogenic medium, the chondrogenic effect of SHP099 on SMSCs was examined by western blot, qPCR, and immunofluorescence (IF). Micro-mass culture was also used to detect the effect of SHP099. To explore the chondrogenic effects of SHP099 in vivo, full-thickness cartilage defects with microfractures were constructed in the right femoral trochlea of New Zealand White rabbits. Intraarticular injection of SHP099 or normal saline was performed twice a week for 6 weeks. Cartilage repair was evaluated by haematoxylin and eosin (HE) staining and safranin O/fast green staining. Immunohistochemistry (IHC) for collagen II (COL2) was also conducted to verify the abundance of cartilage extracellular matrix after SHP099 treatment. The mechanism involving yes-associated protein (YAP) and WNT signalling was investigated in vitro. Results SMSCs isolated from human synovium have optimal multi-differentiation potential. SHP099 increased chondrogenic marker (SOX9, COL2) expression and decreased hypertrophic marker (COL10, RUNX2) expression in SMSCs. In micro-mass culture, the SHP099-induced cartilage tissues had a better result of Safranin O and Toluidine blue staining and are enriched in cartilage-specific collagen II. Inhibition of YAP and WNT signalling was also observed. Moreover, compared to the normal saline group at 6 weeks, intraarticular injection of SHP099 resulted in better defect filling, forming increased hyaline cartilage-like tissue with higher levels of glycosaminoglycan (GAG) and COL2. Conclusion SHP099 promotes the repair of rabbit full-thickness cartilage defects, representing a potential therapeutic drug for cartilage repair. The Translational potential of this article This study provides evidence that SHP2 inhibition promotes chondrogenesis and the repair of cartilage in defect area, which could be a novel therapeutic approach for cartilage repair.
Collapse
Affiliation(s)
- Ziying Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Xingquan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Zhongyang Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Jiawei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Heng Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Kuoyang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Guihua Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Wenqiang Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Yannick Xiaofan Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing, 210008, Jiangsu, PR China
| | - Rui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Jia Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing, 210008, Jiangsu, PR China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Dongquan Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Corresponding author. Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
21
|
Berent ZT, Wagoner Johnson AJ. Morphological switch is associated with increase in cell-cell contacts, ALP, and confluence above a minimum island area to perimeter ratio. J Biomed Mater Res A 2021; 110:164-180. [PMID: 34331408 DOI: 10.1002/jbm.a.37274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/21/2021] [Accepted: 07/07/2021] [Indexed: 11/06/2022]
Abstract
During osteogenic differentiation in vitro, stem-like cells seeded at a low-density spread and are isolated. As the cells proliferate and mature, they become more cuboidal in shape with more cell-cell contacts. However, the coordination of this switch in cell morphology from elongated to cuboidal, cell-cell contacts, and differentiation is not known. In this work, we present results from experiments and a simulation of cell proliferation on protein-micropatterned islands that, independent of island size (25-1,000 μm) or shape (circles, squares, and hollow squares), shows a distinct morphological switch that is better described as a function of island confluence than time in culture, the standard measure in cell culture experiments. The simulation and experiments show cell morphology and island cell density versus confluence collapse to a single curve for all islands if the island area to perimeter ratio is ≥25 μm. Cell-cell contacts in the simulation and alkaline phosphatase (ALP) expression in experiments, a common marker for osteogenic differentiation, show exponential growth with confluence, rapidly increasing after the switch at ≈0.5 confluence. Furthermore, cell morphology, density, contacts, and ALP are better predicted by confluence than time in culture. The variability with time in culture leads to challenges in not only interpreting data but also in comparing data across research laboratories. This simulation can be used to predict cell behavior on different size and shape islands and to plan and optimize experiments that explore cell behavior as a function of a wide range of island geometries.
Collapse
Affiliation(s)
- Zachary T Berent
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Amy J Wagoner Johnson
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| |
Collapse
|
22
|
Wang B, Liu J, Niu D, Wu N, Yun W, Wang W, Zhang K, Li G, Yan S, Xu G, Yin J. Mussel-Inspired Bisphosphonated Injectable Nanocomposite Hydrogels with Adhesive, Self-Healing, and Osteogenic Properties for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32673-32689. [PMID: 34227792 DOI: 10.1021/acsami.1c06058] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Injectable hydrogels have received much attention because of the advantages of simulation of the natural extracellular matrix, microinvasive implantation, and filling and repairing of complex shape defects. Yet, for bone repair, the current injectable hydrogels have shown significant limitations such as the lack of tissue adhesion, deficiency of self-healing ability, and absence of osteogenic activity. Herein, a strategy to construct mussel-inspired bisphosphonated injectable nanocomposite hydrogels with adhesive, self-healing, and osteogenic properties is developed. The nano-hydroxyapatite/poly(l-glutamic acid)-dextran (nHA/PLGA-Dex) dually cross-linked (DC) injectable hydrogels are fabricated via Schiff base cross-linking and noncovalent nHA-BP chelation. The chelation between bisphosphonate ligands (alendronate sodium, BP) and nHA favors the uniform dispersion of the latter. Moreover, multiple adhesion ligands based on catechol motifs, BP, and aldehyde groups endow the hydrogels with good tissue adhesion. The hydrogels possess excellent biocompatibility and the introduction of BP and nHA both can effectively promote viability, proliferation, migration, and osteogenesis differentiation of MC3T3-E1 cells. The incorporation of BP groups and HA nanoparticles could also facilitate the angiogenic property of endothelial cells. The nHA/PLGA-Dex DC hydrogels exhibited considerable biocompatibility despite the presence of a certain degree of inflammatory response in the early stage. The successful healing of a rat cranial defect further proves the bone regeneration ability of nHA/PLGA-Dex DC injectable hydrogels. The developed tissue adhesive osteogenic injectable nHA/PLGA-Dex hydrogels show significant potential for bone regeneration application.
Collapse
Affiliation(s)
- Bo Wang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Jia Liu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Dongyang Niu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Nianqi Wu
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Wentao Yun
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Weidong Wang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Kunxi Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Guifei Li
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Shifeng Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| | - Guohua Xu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, PR China
| |
Collapse
|
23
|
Gonçalves AM, Moreira A, Weber A, Williams GR, Costa PF. Osteochondral Tissue Engineering: The Potential of Electrospinning and Additive Manufacturing. Pharmaceutics 2021; 13:983. [PMID: 34209671 PMCID: PMC8309012 DOI: 10.3390/pharmaceutics13070983] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
The socioeconomic impact of osteochondral (OC) damage has been increasing steadily over time in the global population, and the promise of tissue engineering in generating biomimetic tissues replicating the physiological OC environment and architecture has been falling short of its projected potential. The most recent advances in OC tissue engineering are summarised in this work, with a focus on electrospun and 3D printed biomaterials combined with stem cells and biochemical stimuli, to identify what is causing this pitfall between the bench and the patients' bedside. Even though significant progress has been achieved in electrospinning, 3D-(bio)printing, and induced pluripotent stem cell (iPSC) technologies, it is still challenging to artificially emulate the OC interface and achieve complete regeneration of bone and cartilage tissues. Their intricate architecture and the need for tight spatiotemporal control of cellular and biochemical cues hinder the attainment of long-term functional integration of tissue-engineered constructs. Moreover, this complexity and the high variability in experimental conditions used in different studies undermine the scalability and reproducibility of prospective regenerative medicine solutions. It is clear that further development of standardised, integrative, and economically viable methods regarding scaffold production, cell selection, and additional biochemical and biomechanical stimulation is likely to be the key to accelerate the clinical translation and fill the gap in OC treatment.
Collapse
Affiliation(s)
| | - Anabela Moreira
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| | - Achim Weber
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstrasse 12, 70569 Stuttgart, Germany;
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| |
Collapse
|
24
|
Recent Developed Strategies for Enhancing Chondrogenic Differentiation of MSC: Impact on MSC-Based Therapy for Cartilage Regeneration. Stem Cells Int 2021; 2021:8830834. [PMID: 33824665 PMCID: PMC8007380 DOI: 10.1155/2021/8830834] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022] Open
Abstract
Articular cartilage is susceptible to damage, but its self-repair is hindered by its avascular nature. Traditional treatment methods are not able to achieve satisfactory repair effects, and the development of tissue engineering techniques has shed new light on cartilage regeneration. Mesenchymal stem cells (MSCs) are one of the most commonly used seed cells in cartilage tissue engineering. However, MSCs tend to lose their multipotency, and the composition and structure of cartilage-like tissues formed by MSCs are far from those of native cartilage. Thus, there is an urgent need to develop strategies that promote MSC chondrogenic differentiation to give rise to durable and phenotypically correct regenerated cartilage. This review provides an overview of recent advances in enhancement strategies for MSC chondrogenic differentiation, including optimization of bioactive factors, culture conditions, cell type selection, coculture, gene editing, scaffolds, and physical stimulation. This review will aid the further understanding of the MSC chondrogenic differentiation process and enable improvement of MSC-based cartilage tissue engineering.
Collapse
|
25
|
Jarrar H, Çetin Altındal D, Gümüşderelioğlu M. Scaffold-based osteogenic dual delivery system with melatonin and BMP-2 releasing PLGA microparticles. Int J Pharm 2021; 600:120489. [PMID: 33744449 DOI: 10.1016/j.ijpharm.2021.120489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/27/2021] [Accepted: 03/10/2021] [Indexed: 01/07/2023]
Abstract
The growing safety problems about the use of bone morphogenetic protein 2 (BMP-2) is one of the recent issues that was improved by using low doses of BMP-2 with the support of other osteoinductive agents and/or using appropriate carriers. The aim of the present study is to investigate the effect of scaffold-based dual release system including melatonin (MEL) and BMP-2 loaded polylactic-co-glycolic acid (PLGA) microparticles on the osteogenic activity of pre-osteoblastic MC3T3-E1 cells. MEL and BMP-2 loaded microparticles were prepared by double emulsion solvent evaporation method in the average diameters of ~2 µm and ~11 µm, respectively and loaded into chitosan/hydroxyapatite (HAp) scaffolds. In vitro MC3T3-E1 culture studies were carried out comparatively with blank scaffolds, single (BMP-2 or MEL) releasing groups and dual (BMP-2 and MEL) releasing group. Microscopic observations and hematoxylin/eosin staining showed enhanced number of cells and dense ECM in dual release group. The expressions of differentiation markers, Runt-related transcription factor 2 (RUNX2) and alkaline phosphatase (ALP) and also mineralization were higher in dual release group than that of the other groups. Our findings showed that BMP-2 at low doses (~20 ng per scaffold) was sufficient in terms of osteogenic activity with controlled release systems where it was used in combination with MEL (~10 µg per scaffold).
Collapse
Affiliation(s)
- Hala Jarrar
- Hacettepe University, Bioengineering Department, 06800 Beytepe, Ankara, Turkey
| | | | | |
Collapse
|
26
|
Lin H, Zhou Y, Lei Q, Lin D, Chen J, Wu C. Effect of inorganic phosphate on migration and osteogenic differentiation of bone marrow mesenchymal stem cells. BMC DEVELOPMENTAL BIOLOGY 2021; 21:1. [PMID: 33407089 PMCID: PMC7788862 DOI: 10.1186/s12861-020-00229-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
Background Phosphate is the major ingredient of bone tissue, and is also an important component of commercial bone substitute materials, bone scaffolds, and implant surface coatings. With the dissolution of the bone substitute materials and the degradation by cells, local ion concentrations will change and affect bone tissue reconstruction. Bone marrow -derived mesenchymal stem cells (BM-MSCs) are main autologous cells to repair injured bone. When bone injure occurs, BM-MSCs migrate to the damaged area, differentiate into osteoblasts, and secrete bioactive factors to promote bone tissue repaired. This study aimed to investigate the effect of inorganic phosphate (Pi) at a series of concentration on migration and osteogenic differentiation of human bone marrow -derived mesenchymal stem cells(hBM-MSCs). Methods The culture of hBM-MSCs in mediums with different concentration of Pi from 2 mM to 10 mM were performed. HBM-MSCs migration were examined with transwell assays. HBM-MSCs proliferation were evaluated by cell counting kit-8 colorimetric method. Osteogenic genes expression were analyzed by real-time reverse transcriptase polymerase chain reaction. Mineralized nodules formation were demonstrated by Alizarin red staining. Result 4–10 mM Pi could effectively promote the migration of hBM-MSCs at 12 h and 18 h. There was no significant difference in the migration number of hBM-MSCs in Pi culture mediums at a concentration of 6, 8, and10mM. 2–10 mM Pi could promote the proliferation of hBM-MSCs to varying degrees in the observation period, while 4–10 mM Pi could promote the osteogenic differentiation and mineralization of hBM-MSCs. Conclusion The findings in our study showed 4-10 mM Pi could promote the migration, osteogenic differentiation, and mineralization of hBM-MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-020-00229-x.
Collapse
Affiliation(s)
- Hengzhang Lin
- Department of Stomatology, Fujian Provincial Governmental Hospital, Fuzhou, China
| | - Yong Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Qun Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| | - Dong Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jiang Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Chuhuo Wu
- Fujian Medical University, Fuzhou, China
| |
Collapse
|
27
|
Jarrar H, Çetİn Altindal D, GÜmÜŞderelİoĞlu M. The inhibitory effect of melatonin on osteoclastogenesis of RAW 264.7 cells in low concentrations of RANKL and MCSF. ACTA ACUST UNITED AC 2020; 44:427-436. [PMID: 33402869 PMCID: PMC7759193 DOI: 10.3906/biy-2007-85] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022]
Abstract
RAW 264.7 cells are one of the most recommended cell lines for investigating the activity and differentiation of osteoclasts. These cells differentiate into osteoclasts in the presence of two critical components: receptor activator of nuclear factor kappa B ligand (RANKL) and macrophage colony stimulating factor (MCSF). Melatonin (MEL) hormone has recently become one of the small molecules used in the field of bone regeneration and bone disease treatment, as it has the ability to inhibit the differentiation of osteoclasts directly by suppression of the NF-κB signaling pathway. The main aim of the current study is to determine sufficient RANKL/MCSF concentrations for differentiation of the cells to osteoclasts and to describe the repressive effect of MEL on the osteoclastogenesis of these cells. In this regard, it was found that 10 ng/mL of RANKL- and MCSF-containing medium is suitable for inducing osteoclastogenesis of the cells. In addition, melatonin at doses in the range of 100-1000 µM does not have a cytotoxic effect. Subsequently, results of tartrate resistant acid phosphatase (TRAP) activity, TRAP staining, and relative expressions of cathepsin K, nuclear factor of activated T cells one (NFATC1), and TRAP genes showed a suppressive effect of MEL -especially 800 µM- on RANKL-induced osteoclastogenesis of these cells.
Collapse
Affiliation(s)
- Hala Jarrar
- Bioengineering Division, Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara Turkey
| | - Damla Çetİn Altindal
- Bioengineering Division, Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara Turkey
| | - Menemşe GÜmÜŞderelİoĞlu
- Bioengineering Division, Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara Turkey
| |
Collapse
|
28
|
Effects of icariin on the proliferation and osteogenic differentiation of human amniotic mesenchymal stem cells. J Orthop Surg Res 2020; 15:578. [PMID: 33267896 PMCID: PMC7709318 DOI: 10.1186/s13018-020-02076-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background Tissue engineering technology has been applied extensively for clinical research and human amnion mesenchymal stem cells (hAMSCs) could cause mesenchymal stem cells to differentiate into the bone tissue. However, it is necessary to develop and identify the safer appropriate amount of osteogenic inducer. The objective of this study is to investigate the effect of icariin (ICA) on the proliferation and osteogenic differentiation of hAMSCs. Methods The morphology and phenotype of hAMSCs were discovered by flow cytometry and immunocytochemical staining. The osteogenic differentiation of hAMSCs under the influence of different concentrations of ICA were assessed by alkaline phosphatase (ALP) activity substrate assay and alizarin red staining. Results MTT assay revealed that the hAMSCs pretreated with ICA exhibited increased proliferation when compared with the control group, and the most optimum concentration of ICA was 1 × 10− 6 mol/L. The combined analysis of ALP activity and ARS staining showed that ICA could significantly promote the osteogenic differentiation of hAMSCs, and the effect was most significant when the concentration of ICA was 1 × 10− 6 mol/L. Conclusion All the above results implied that ICA could significantly increase proliferation and enhance the osteogenic differentiation of hAMSCs, especially when the concentration of ICA was 1 × 10− 6 mol/L.
Collapse
|
29
|
Osteogenic effects of the bioactive small molecules and minerals in the scaffold-based bone tissue engineering. Colloids Surf B Biointerfaces 2020; 198:111462. [PMID: 33239252 DOI: 10.1016/j.colsurfb.2020.111462] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/30/2020] [Accepted: 11/08/2020] [Indexed: 12/26/2022]
Abstract
Reconstruction of the damaged bone is a striking challenge in the medical field. The bone grafts as a current treatment is associated with inherent limitations; hence, the bone tissue engineering as an alternative therapeutic approach has been considered in the recent decades. Bone tissue engineering aims at replacing the lost tissue and restoring its function by recapitulating the natural regeneration process. Concerted participation and combination of the biocompatible materials, osteoprogenitor/ stem cells and bioactive factors closely mimic the bone microenvironment. The bioactive factors regulate the cell behavior and they induce the stem cells to osteogenic differentiation by activating specific signaling cascades. Growth factors (GFs) are the most important bioactive molecules and mediators of the natural bone repair process. Although these soluble factors have approved applications in the bone regeneration, however, there are several limitations such as the instability, high dose requirements, and serious side effects which could restrict their clinical usage. Alternatively, a new generation of bioactive molecules with the osteogenic properties are used. The non-peptide organic or inorganic molecules are physiologically stable and non-immunogenic due to their small size. Many of them are obtained from the natural resources and some are synthesized through the chemical methods. As a result, these molecules have been introduced as the cost-effective osteogenic agents in the bone tissue regeneration. In this paper, three groups of these bioactive agents including the organic small molecules, minerals and metallic nanoparticles have been investigated, considering their function in accelerating the bone regeneration. We review the recent in vitro and in vivo studies that utilized the osteogenic molecules to promote the bone formation in the scaffold-based bone tissue engineering systems.
Collapse
|
30
|
Ghorbaninejad M, Khademi-Shirvan M, Hosseini S, Baghaban Eslaminejad M. Epidrugs: novel epigenetic regulators that open a new window for targeting osteoblast differentiation. Stem Cell Res Ther 2020; 11:456. [PMID: 33115508 PMCID: PMC7594482 DOI: 10.1186/s13287-020-01966-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/05/2020] [Indexed: 01/01/2023] Open
Abstract
Efficient osteogenic differentiation of mesenchymal stem cells (MSCs) is a critical step in the treatment of bone defects and skeletal disorders, which present challenges for cell-based therapy and regenerative medicine. Thus, it is necessary to understand the regulatory agents involved in osteogenesis. Epigenetic mechanisms are considered to be the primary mediators that regulate gene expression during MSC differentiation. In recent years, epigenetic enzyme inhibitors have been used as epidrugs in cancer therapy. A number of studies mentioned the role of epigenetic inhibitors in the regulation of gene expression patterns related to osteogenic differentiation. This review attempts to provide an overview of the key regulatory agents of osteogenesis: transcription factors, signaling pathways, and, especially, epigenetic mechanisms. In addition, we propose to introduce epigenetic enzyme inhibitors (epidrugs) and their applications as future therapeutic approaches for bone defect regeneration.
Collapse
Affiliation(s)
- Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maliheh Khademi-Shirvan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
31
|
Small Molecule–Mediated Enhanced Osteogenesis of Human Mesenchymal Stem Cells: a Probable Alternate for BMP-2. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00179-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
32
|
Hong Y, Liu N, Zhou R, Zhao X, Han Y, Xia F, Cheng J, Duan M, Qian Q, Wang X, Cai W, Zreiqat H, Feng D, Xu J, Cui D. Combination Therapy Using Kartogenin-Based Chondrogenesis and Complex Polymer Scaffold for Cartilage Defect Regeneration. ACS Biomater Sci Eng 2020; 6:6276-6284. [PMID: 33449656 DOI: 10.1021/acsbiomaterials.0c00724] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Articular cartilage has a highly organized structure, responsible for supporting tremendous mechanical loads. How to repair defected articular cartilage has become a great challenge as the avascular nature of cartilage limits its regenerative ability. Aiming to facilitate chondrogenic differentiation and cartilage regeneration, we recently explored a novel combination therapy using soluble poly-l-lysine/Kartogenin (L-K) nanoparticles and a poly(lactic-co-glycolic acid) PLGA/methacrylated hyaluronic acid (PLHA) complex scaffold. The potential use for joint cartilage reconstruction was investigated through L-K nanoparticles stimulating adipose-derived stem cells (ADSCs) on PLHA scaffolding, which ultimately differentiated into cartilage in vivo. In this study, on one hand, an effective method was established for obtaining uniform L-K nanoparticles by self-assembly. They were further proved to be biocompatible to ADSCs via cytotoxicity assays in vitro and to accelerate ADSCs secreting type 2 collagen in a dose-dependent manner by immunofluorescence. On the other hand, the porous PLHA scaffold was manufactured by the combination of coprecipitation and ultraviolet (UV) cross-linking. Nanoindentation technology-verified PLHA had an appropriate stiffness close to actual cartilage tissue. Additional microscopic observation confirmed that the PLHA platform supported proliferation and chondrogenesis for ADSCs in vitro. In the presence of ADSCs, a 12-week osteochondral defect regeneration by the combination therapy showed that smooth and intact cartilage tissue successfully regenerated. Furthermore, the results of combination therapy were superior to those of phosphate-buffered saline (PBS) only, KGN, or KGN/PLHA treatment. The results of magnetic resonance imaging (MRI) and histological assessment indicated that the renascent tissue gradually regenerated while the PLHA scaffold degraded. In conclusion, we have developed a novel multidimensional combination therapy of cartilage defect repair that facilitated cartilage regeneration. This strategy has a great clinical translational potential for articular cartilage repair in the near future.
Collapse
Affiliation(s)
- Yuping Hong
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Ning Liu
- Department of Joint Surgery and Sports Medicine, Changzheng Hospital, Second Naval Military University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Rong Zhou
- Department of Joint Surgery and Sports Medicine, Changzheng Hospital, Second Naval Military University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Xinxin Zhao
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong Univesity, 160 Pujian Road, Sahnghai 200127, P. R. China
| | - Yaguang Han
- Department of Joint Surgery and Sports Medicine, Changzheng Hospital, Second Naval Military University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Fangfang Xia
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Jin Cheng
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Meng Duan
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Qirong Qian
- Department of Joint Surgery and Sports Medicine, Changzheng Hospital, Second Naval Military University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Xiuying Wang
- School of Computer Science, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Weidong Cai
- School of Computer Science, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Hala Zreiqat
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW 2065, Australia
| | - Dagan Feng
- School of Computer Science, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Jianrong Xu
- Department of Radiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong Univesity, 160 Pujian Road, Sahnghai 200127, P. R. China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
33
|
Li T, Liu B, Chen K, Lou Y, Jiang Y, Zhang D. Small molecule compounds promote the proliferation of chondrocytes and chondrogenic differentiation of stem cells in cartilage tissue engineering. Biomed Pharmacother 2020; 131:110652. [PMID: 32942151 DOI: 10.1016/j.biopha.2020.110652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 02/09/2023] Open
Abstract
The application of tissue engineering to generate cartilage is limited because of low proliferative ability and unstable phenotype of chondrocytes. The sources of cartilage seed cells are mainly chondrocytes and stem cells. A variety of methods have been used to obtain large numbers of chondrocytes, including increasing chondrocyte proliferation and stem cell chondrogenic differentiation via cytokines, genes, and proteins. Natural or synthetic small molecule compounds can provide a simple and effective method to promote chondrocyte proliferation, maintain a stable chondrocyte phenotype, and promote stem cell chondrogenic differentiation. Therefore, the study of small molecule compounds is of great importance for cartilage tissue engineering. Herein, we review a series of small molecule compounds and their mechanisms that can promote chondrocyte proliferation, maintain chondrocyte phenotype, or induce stem cell chondrogenesis. The studies in this field represent significant contributions to the research in cartilage tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tian Li
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Bingzhang Liu
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Kang Chen
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yuhan Jiang
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Bethune Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
34
|
Yuan J, Hou Q, Zhong L, Dai X, Lu Q, Li M, Fu X. Sustained release of inhibitor from bionic scaffolds for wound healing and functional regeneration. Biomater Sci 2020; 8:5647-5655. [PMID: 33049013 DOI: 10.1039/d0bm00929f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Small molecules play remarkable roles in promoting tissue regeneration, but are limited by their burst release. Small molecules such as deferoxamine (DFO) have been released slowly from silk hydrogels and stimulated angiogenesis and wound healing, but failed to achieve functional recovery of skin. Various bioactive molecules are required to create a suitable niche for better skin regeneration by controlling their release behaviors. Herein, a small molecule SB216763, a GSK-3 inhibitor, was loaded on silk fibroin nanofibers (SNF), and then mixed with chitosan (CS) to prepare the small molecule-loaded composite bionic scaffolds (CSNF-SB). Given the interaction of SNF and SB216763, the sustained release of SB216763 for more than 21 days was achieved for SNF and CSNF-SB composite scaffolds. Compared to drug-free CSNF scaffolds, CSNF-SB showed better cell adhesion and proliferation capacity, suggesting bioactivity. The upregulated expression of β-catenin in fibroblasts in vitro revealed that the released small molecules maintained their function in composite scaffolds. Quicker and better wound healing was realized with the drug-loaded scaffolds, which was significantly superior to that treated with drug-free scaffolds. Unlike the DFO-loaded silk hydrogel system, hair follicle neogenesis was also found in the drug-loaded-scaffold treatment wounds, demonstrating functional recovery. Therefore, silk nanofibers as versatile carriers for different small bioactive molecules could be used to fabricate scaffolds with optimized niches and then achieve functional recovery of tissues. The small molecule-loaded bionic scaffolds have a promising future in skin tissue regeneration.
Collapse
Affiliation(s)
- Jifang Yuan
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China.
| | | | | | | | | | | | | |
Collapse
|
35
|
Breathwaite E, Weaver J, Odanga J, dela Pena-Ponce M, Lee JB. 3D Bioprinted Osteogenic Tissue Models for In Vitro Drug Screening. Molecules 2020; 25:E3442. [PMID: 32751124 PMCID: PMC7435717 DOI: 10.3390/molecules25153442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023] Open
Abstract
Metabolic bone disease affects hundreds of millions of people worldwide, and as a result, in vitro models of bone tissue have become essential tools to help analyze bone pathogenesis, develop drug screening, and test potential therapeutic strategies. Drugs that either promote or impair bone formation are in high demand for the treatment of metabolic bone diseases. These drugs work by targeting numerous signaling pathways responsible for regulating osteogenesis such as Hedgehog, Wnt/β-catenin, and PI3K-AKT. In this study, differentiated bone marrow-derived mesenchymal stem cell (BM-MSC) scaffold-free 3D bioprinted constructs and 2D monolayer cultures were utilized to screen four drugs predicted to either promote (Icariin and Purmorphamine) or impair osteogenesis (PD98059 and U0126). Osteogenic differentiation capacity was analyzed over a four week culture period by evaluating mineralization, alkaline phosphatase (ALP) activity, and osteogenesis related gene expression. Responses to drug treatment were observed in both 3D differentiated constructs and 2D monolayer cultures. After four weeks in culture, 3D differentiated constructs and 2D monolayer cultures treated with Icariin or Purmorphamine showed increased mineralization, ALP activity, and the gene expression of bone formation markers (BGLAP, SSP1, and COL1A1), signaling molecules (MAPK1, WNT1, and AKT1), and transcription factors (RUNX2 and GLI1) that regulate osteogenic differentiation relative to untreated. 3D differentiated constructs and 2D monolayer cultures treated with PD98059 or U0126 showed decreased mineralization, ALP activity, and the expression of the aforementioned genes BGLAP, SPP1, COL1A1, MAPK1, AKT1, RUNX2, and GLI1 relative to untreated. Differences in ALP activity and osteogenesis related gene expression relative to untreated cells cultured in a 2D monolayer were greater in 3D constructs compared to 2D monolayer cultures. These findings suggest that our bioprinted bone model system offers a more sensitive, biologically relevant drug screening platform than traditional 2D monolayer in vitro testing platforms.
Collapse
Affiliation(s)
| | | | | | | | - Jung Bok Lee
- Institute of Regenerative Medicine, LifeNet Health, 1864 Concert Drive, Virginia Beach, VA 23453, USA; (E.B.); (J.W.); (J.O.); (M.d.P.-P.)
| |
Collapse
|
36
|
Asgari N, Bagheri F, Eslaminejad MB, Ghanian MH, Sayahpour FA, Ghafari AM. Dual functional construct containing kartogenin releasing microtissues and curcumin for cartilage regeneration. Stem Cell Res Ther 2020; 11:289. [PMID: 32678019 PMCID: PMC7367357 DOI: 10.1186/s13287-020-01797-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/15/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Regeneration of articular cartilage poses a tremendous challenge due to its limited self-repair capability and inflammation at the damaged site. To generate the desired structures that mimic the structure of native tissue, microtissues with repeated functional units such as cell aggregates have been developed. Multicellular aggregates of mesenchymal stem cells (MSCs) can be used as microscale building blocks of cartilage due to their potential for cell-cell contact, cell proliferation, and differentiation. METHODS Chondrogenic microtissues were developed through incorporation of kartogenin-releasing poly (lactic-co-glycolic acid) (PLGA) microparticles (KGN-MP) within the MSC aggregates. The chondrogenic potential of KGN-MP treated MSC aggregates was proven in vitro by studying the chondrogenic markers at the RNA level and histological analysis. In order to address the inflammatory responses at the defect site, the microtissues were delivered in vivo via an injectable, anti-inflammatory hydrogel that contained gelatin methacryloyl (GelMA) loaded with curcumin (Cur). RESULTS The KGN-MPs were fabricated to support MSCs during cartilage differentiation. According to real-time RT-PCR analysis, the presence of KGN in the aggregates led to the expression of cartilage markers by the MSCs. Both toluidine blue (TB) and safranin O (SO) staining demonstrated homogeneous glycosaminoglycan production throughout the KGN-MP incorporated MSC aggregates. The curcumin treatment efficiently reduced the expressions of hypertrophy markers by MSCs in vitro. The in vivo results showed that implantation of chondrogenic microtissues (KGN-MP incorporated MSC aggregates) using the curcumin loaded GelMA hydrogel resulted in cartilage tissue regeneration that had characteristic features close to the natural hyaline cartilage according to observational and histological results. CONCLUSIONS The use of this novel construct that contained chondrogenic cell blocks and curcumin is highly desired for cartilage regeneration.
Collapse
Affiliation(s)
- Negin Asgari
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Jalal Ale Ahmad Street, P.O.Box: 14115-111, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat Highway, P.O. Box 16635-148, Tehran, Iran.
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forogh Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat Highway, P.O. Box 16635-148, Tehran, Iran
| | - Amir Mohammad Ghafari
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Center for Functional Materials, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| |
Collapse
|
37
|
Zhao Y, Teng B, Sun X, Dong Y, Wang S, Hu Y, Wang Z, Ma X, Yang Q. Synergistic Effects of Kartogenin and Transforming Growth Factor-β3 on Chondrogenesis of Human Umbilical Cord Mesenchymal Stem Cells In Vitro. Orthop Surg 2020; 12:938-945. [PMID: 32462800 PMCID: PMC7307229 DOI: 10.1111/os.12691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/29/2020] [Accepted: 04/03/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE To explore the effect of kartogenin (KGN) on proliferation and chondrogenic differentiation of human umbilical cord mesenchymal stem cells (hUCMSC) in vitro, and the synergistic effects of KGN and transforming growth factor (TGF)-β3 on hUCMSC. METHODS Human umbilical cord mesenchymal stem cells were isolated and cultured. Then the differentiation properties were identified by flow cytometry analysis. HUCMSC were divided into four groups: control group, KGN group, TGF-β3 group, and TK group (with TGF-β3 and KGN added into the medium simultaneously). Cells in all groups were induced for 21 days using the suspension ball culture method. Hematoxylin and eosin, immunofluorescence, and Alcian blue staining were used to analyze chondrogenic differentiation. Real-time reverse transcriptase polymerase chain reaction was performed to investigate genes associated with chondrogenic differentiation. RESULT Hematoxylin and eosin staining showed that cells in the TGF-β3 group and the TK group had formed cartilage-like tissue after 21 days of culture. The results of immunofluorescence and Alcian blue staining showed that compared with the control group, cells in the KGN and TGF-β3 groups demonstrated increased secretion of aggrecan after 21 days of culture. In addition, cells in the group combining KGN with TGF-β3 (5.587 ± 0.27, P < 0.01) had more collagen II secretion than cells in the TGF-β3 alone group (2.86 ± 0.141, P < 0.01) or the KGN group (1.203 ± 0.215, P < 0.01). The expression of aggrecan (2.468 ± 0.097, P < 0.05) and SRY-Box 9 (4.08 ± 0.13, P < 0.05) in cells in the group combining KGN with TGF-β3 was significantly higher than those in the TGF-β3 group (2.216 ± 0.09, 3.02 ± 0.132, P < 0.05).' CONCLUSION The combination of KGN and TGF-β3 had synergistic effects and induced hUCMSC chondrogenesis. This could represent a new approach for clinical application and studies on cartilage repair and regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical UniversityTianjinChina
| | - Binhong Teng
- Stomatological Hospital of Tianjin Medical UniversityTianjinChina
- Department of Oral and Maxillofacial SurgerySchool and Hospital of Stomatology, Peking UniversityBeijingChina
| | - Xun Sun
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai UniversityTianjinChina
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai UniversityTianjinChina
| | - Yongcheng Hu
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Zheng Wang
- Department of OrthopedicsNo. 1 Medical Center of Chinese PLA General HospitalBeijingChina
| | - Xinlong Ma
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| | - Qiang Yang
- Department of Spine SurgeryTianjin Hospital, Tianjin UniversityTianjinChina
| |
Collapse
|
38
|
Patil S, Dhyani V, Kaur T, Singh N. Spatiotemporal Control over Cell Proliferation and Differentiation for Tissue Engineering and Regenerative Medicine Applications Using Silk Fibroin Scaffolds. ACS APPLIED BIO MATERIALS 2020; 3:3476-3493. [DOI: 10.1021/acsabm.0c00305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Smita Patil
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Vartika Dhyani
- Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Tejinder Kaur
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
39
|
Wang YQ, Wang NX, Luo Y, Yu CY, Xiao JH. Ganoderal A effectively induces osteogenic differentiation of human amniotic mesenchymal stem cells via cross-talk between Wnt/β-catenin and BMP/SMAD signaling pathways. Biomed Pharmacother 2019; 123:109807. [PMID: 31896066 DOI: 10.1016/j.biopha.2019.109807] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/12/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Osteogenic inducers play central roles in effective stem cell-based treatment of bone defects/losses. However, the current routine osteogenic inducer is a cocktail comprising three components that must be improved due to low induction efficiency and side effects. Therefore, there is an urgent need to develop safer and more effective osteoinducers. Herein, we demonstrated the osteogenic effect of Ganoderal A (GD-A), a tetracyclic triterpenoid compound from Ganoderma lucidum. GD-A showed no cytotoxicity toward human amniotic mesenchymal stem cells (hAMSCs) at doses of 0.001-10 μM; furthermore, 0.01 μM GD-A significantly induced the generation of osteoblast-specific markers, such as alkaline phosphatase, and calcium deposition in hAMSCs. At molecular levels, GD-A promoted the expression of multiple osteoblast differentiation markers, such as RUNX2, OSX, OPN, ALP, OCN, and COL1α1. Both Wnt/β-catenin and BMP/SMAD signaling were shown as active during hAMSC osteodifferentiation. Furthermore, specific blocking of both signals by KYA1797K and SB431542 significantly inhibited alkaline phosphatase secretion and RUNX2 and ALP expression when used alone or in combination. Meanwhile, both signals were also blocked. These findings suggest that GD-A induces hAMSC differentiation into osteoblasts through signaling cross-talk between Wnt/β-catenin and BMP/SMAD. Taken together, GD-A is a safe, effective, and novel osteoinducer and might be used for stem cell-based therapy for bone defects/losses.
Collapse
Affiliation(s)
- Yi-Qing Wang
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Nuo-Xin Wang
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yi Luo
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Chang-Yin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China.
| | - Jian-Hui Xiao
- Zunyi Municiptal Key Laboratory of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China.
| |
Collapse
|
40
|
Wang SJ, Qin JZ, Zhang TE, Xia C. Intra-articular Injection of Kartogenin-Incorporated Thermogel Enhancing Osteoarthritis Treatment. Front Chem 2019; 7:677. [PMID: 31681730 PMCID: PMC6813204 DOI: 10.3389/fchem.2019.00677] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
To provide a vehicle for sustained release of cartilage-protective agent for the potential application of osteoarthritis (OA) treatment, we developed a kartogenin (KGN)-incorporated thermogel for intra-articular injection. We fabricated a poly(lactide-co-glycolide)-block-poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA–PEG–PLGA) thermogel as a KGN carrier for IA injection. OA chondrocytes were cultured in thermogel with or with no KGN to investigate the effect of KGN thermogel on cartilage matrix. The in vivo effect of KGN thermogel on OA was examined in a rabbit OA model. The KGN thermogel showed a sustained in vitro release of KGN for 3 weeks. OA chondrocytes proliferated well both in thermogel and KGN thermogel. In addition, OA chondrocytes produced higher amount of [type 2 collagen (COL-2) and glycosaminoglycan (GAG)], as well as lower level of matrix metalloproteinase 13 (MMP-13) in KGN thermogel that those in thermogel with no addition of KGN. The gene analysis supported that KGN thermogel enhanced expression of hyaline-cartilage specific genes Col 2 and AGC, and inhibited the expression of MMP-13. Compared with intra-articular injection of saline or thermogel containing no KGN, KGN thermogel can enhance cartilage regeneration and inhibit joint inflammation of arthritic knees in a rabbit ACLT-induced OA model at 3 weeks after the injection. Therefore, the KGN-incorporated PLGA–PEG–PLGA thermogel may provide a novel treatment modality for OA treatment with IA injection.
Collapse
Affiliation(s)
- Shao-Jie Wang
- Department of Joint Surgery and Sports Medicine, Xiamen University Zhongshan Hospital, Xiamen, China
| | - Ji-Zheng Qin
- Department of Joint Surgery and Sports Medicine, Xiamen University Zhongshan Hospital, Xiamen, China
| | | | - Chun Xia
- Department of Joint Surgery and Sports Medicine, Xiamen University Zhongshan Hospital, Xiamen, China
| |
Collapse
|
41
|
How can microsphere-mediated delivery of small molecules serve as a novel tool for engineering tissues from stem cells? Ther Deliv 2019; 10:671-674. [PMID: 31608826 DOI: 10.4155/tde-2019-0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
42
|
Song W, Ma Z, Wang C, Li H, He Y. Pro-chondrogenic and immunomodulatory melatonin-loaded electrospun membranes for tendon-to-bone healing. J Mater Chem B 2019; 7:6564-6575. [PMID: 31588948 DOI: 10.1039/c9tb01516g] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Reconstructing the native structure of the tendon-to-bone insertion site (enthesis) in rotator cuff repair has always been a great challenge for orthopedic surgeons. Difficulty arises mainly due to the limited enthesis regenerative capability and severe inflammatory cell infiltration, which result in fibrovascular scar formation instead of native cartilage-like enthesis. Therefore, tissue engineering scaffolds with pro-chondrogenic and immunomodulatory capabilities may offer a new strategy for native enthesis regeneration. In this study, melatonin-loaded aligned polycaprolactone (PCL) electrospun fibrous membranes were fabricated. The sustained release of melatonin from this membrane significantly promoted the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in a long-term chondroid pellet model. After the membranes were implanted in a rat acute rotator cuff tear model, melatonin-loaded PCL membranes inhibited macrophage infiltration in the tendon-to-bone interface at the early healing phase, increasing chondroid zone formation, promoting collagen maturation, decreasing fibrovascular tissue formation and eventually improving the biomechanical strength of the regenerated enthesis. Taken together, melatonin-loaded PCL membranes possess great clinical application potential for tendon-to-bone healing.
Collapse
Affiliation(s)
- Wei Song
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Zhijie Ma
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Chongyang Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Haiyan Li
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.
| | - Yaohua He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China. and Department of Orthopedics, Shanghai Sixth People's Hospital, Jinshan Branch, 147 Jiankang Road, Shanghai 201599, China
| |
Collapse
|
43
|
Bao Q, Yang D, Hong F, Yang J, Li L, Jin Y, Ma C. αB-crystallin (CRYAB) regulates the proliferation, apoptosis, synthesis and degradation of extracellular matrix of chondrocytes in osteoarthritis. Exp Cell Res 2019; 382:111459. [PMID: 31226261 DOI: 10.1016/j.yexcr.2019.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/14/2019] [Accepted: 06/08/2019] [Indexed: 01/01/2023]
Abstract
Osteoarthritis (OA) is a chronic joint disease and hard to cure at present. Alpha B-crystallin (CRYAB) has been identified as a downregulated gene in OA cartilage. However, the precise roles and underlying molecular mechanisms of CRYAB in OA progression have not been elucidated. In the present study, we found that the expression of CRYAB in cartilages from patients with OA was significantly lower than that in the cartilages from patients with no prior medical history of OA. We established mouse models with OA by destabilization of the medial meniscus (DMM) surgery and found that the expression of CRYAB in OA cartilage was lower than that in the normal cartilages, too. Moreover, we demonstrated that the expression of CRYAB was increased during chondrogenic differentiation and cartilage development. Functional assays revealed that overexpression of CRYAB promoted the proliferation of chondrocytes and inhibited apoptosis, while knockdown of CRYAB presented opposite results. In addition, overexpression of CRYAB upregulated the expression of anabolic markers, Col2a1 and ACAN, and reduced the expression of catabolic markers, MMP13 and ADAMTS5. Conversely, knockdown of CRYAB blocked the expression of the anabolic markers and increased the expression of catabolic markers. Collectively, the results suggest that CRYAB promoted the proliferation and extracellular matrix production of chondrocytes, and inhibited chondrocytes apoptosis and cartilage degradation simultaneously. Thus, CRYAB might be a potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Qianyi Bao
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China; Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| | - Dawei Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China; Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, 211166, PR China.
| | - Fangling Hong
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| | - Lingyun Li
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| | - Yucui Jin
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| | - Changyan Ma
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China; Department of Medical Genetics, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, PR China.
| |
Collapse
|
44
|
Kalani MM, Nourmohammadi J, Negahdari B. Osteogenic potential of Rosuvastatin immobilized on silk fibroin nanofibers using argon plasma treatment. Biomed Mater 2018; 14:025002. [DOI: 10.1088/1748-605x/aaec26] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
45
|
Cai G, Liu W, He Y, Huang J, Duan L, Xiong J, Liu L, Wang D. Recent advances in kartogenin for cartilage regeneration. J Drug Target 2018; 27:28-32. [PMID: 29772932 DOI: 10.1080/1061186x.2018.1464011] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Either osteoarthritis or sports-related injuries can lead to cartilage defects, whereas both chondrocyte self-renewal and conventional treatments face limitations. In cartilage regenerative medicine, growth factors are commonly used to induce chondrogenic differentiation of stem cells. However, application of growth factors is confined by some drawbacks. Emerging small molecules are regarded as an alternative for cartilage regeneration. A recently discovered small-molecule compound, kartogenin (KGN), has been proven to be a chondrogenic and chondroprotective agent and is more effective in inducing cartilage regeneration when compared with growth factors. KGN has been processed and applied in many forms, such as in intra-articular injection, in collaboration with growth factors, in incorporation in drug delivery systems, and in combination with scaffolds. Fortunately, progress has been achieved in KGN applications. The current review discusses the recent advances in KGN for cartilage regeneration and thus presents new concepts in cartilage repair in clinical settings.
Collapse
Affiliation(s)
- Gaorui Cai
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China.,b Shenzhen Key Laboratory of Tissue Engineering , Shenzhen , Guangdong Province , China
| | - Wei Liu
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China.,c Department of Sports Medicine , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China
| | - Yong He
- b Shenzhen Key Laboratory of Tissue Engineering , Shenzhen , Guangdong Province , China
| | - Jianghong Huang
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China.,c Department of Sports Medicine , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China
| | - Li Duan
- b Shenzhen Key Laboratory of Tissue Engineering , Shenzhen , Guangdong Province , China
| | - Jianyi Xiong
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China.,b Shenzhen Key Laboratory of Tissue Engineering , Shenzhen , Guangdong Province , China.,c Department of Sports Medicine , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China
| | - Lijun Liu
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China
| | - Daping Wang
- a Department of Traumatic Orthopedics , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China.,b Shenzhen Key Laboratory of Tissue Engineering , Shenzhen , Guangdong Province , China.,c Department of Sports Medicine , the First Affiliated Hospital of Shenzhen University , Shenzhen , Guangdong Province , China
| |
Collapse
|
46
|
Gao L, Orth P, Cucchiarini M, Madry H. Effects of solid acellular type-I/III collagen biomaterials on in vitro and in vivo chondrogenesis of mesenchymal stem cells. Expert Rev Med Devices 2018; 14:717-732. [PMID: 28817971 DOI: 10.1080/17434440.2017.1368386] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Type-I/III collagen membranes are advocated for clinical use in articular cartilage repair as being able of inducing chondrogenesis, a technique termed autologous matrix-induced chondrogenesis (AMIC). Area covered: The current in vitro and translational in vivo evidence for chondrogenic effects of solid acellular type-I/III collagen biomaterials. Expert commentary: In vitro, mesenchymal stem cells (MSCs) adhere to the fibers of the type-I/III collagen membrane. No in vitro study provides evidence that a type-I/III collagen matrix alone may induce chondrogenesis. Few in vitro studies compare the effects of type-I and type-II collagen scaffolds on chondrogenesis. Recent investigations suggest better chondrogenesis with type-II collagen scaffolds. A systematic review of the translational in vivo data identified one long-term study showing that covering of cartilage defects treated by microfracture with a type-I/III collagen membrane significantly enhanced the repair tissue volume compared with microfracture alone. Other in vivo evidence is lacking to suggest either improved histological structure or biomechanical function of the repair tissue. Taken together, there is a paucity of in vitro and preclinical in vivo evidence supporting the concept that solid acellular type-I/III collagen scaffolds may be superior to classical approaches to induce in vitro or in vivo chondrogenesis of MSCs.
Collapse
Affiliation(s)
- Liang Gao
- a Lehrstuhl für Experimentelle Orthopädie und Arthroseforschung , Saarland University , Homburg/Saar , Germany
| | - Patrick Orth
- a Lehrstuhl für Experimentelle Orthopädie und Arthroseforschung , Saarland University , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Lehrstuhl für Experimentelle Orthopädie und Arthroseforschung , Saarland University , Homburg/Saar , Germany
| | - Henning Madry
- a Lehrstuhl für Experimentelle Orthopädie und Arthroseforschung , Saarland University , Homburg/Saar , Germany
| |
Collapse
|
47
|
Spakova T, Plsikova J, Harvanova D, Lacko M, Stolfa S, Rosocha J. Influence of Kartogenin on Chondrogenic Differentiation of Human Bone Marrow-Derived MSCs in 2D Culture and in Co-Cultivation with OA Osteochondral Explant. Molecules 2018; 23:molecules23010181. [PMID: 29337871 PMCID: PMC6017512 DOI: 10.3390/molecules23010181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/10/2018] [Accepted: 01/14/2018] [Indexed: 12/18/2022] Open
Abstract
Articular cartilage has limited capacity for natural regeneration and repair. In the present study, we evaluated kartogenin (KGN), a bioactive small heterocyclic molecule, for its effect on in vitro proliferation and chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells (hBMSCs) in monolayer culture and in co-culture models in vitro. OA osteochondral cylinders and hBMSCs were collected during total knee replacement. The effect of KGN on hBMSCs during 21 days of culture was monitored by real-time proliferation assay, immunofluorescence staining, histological assay, scanning electron microscopy (SEM) (imaging and multiplex enzyme-linked immunosorbent assay) ELISA assay. The rate of proliferation of hBMSCs was significantly increased by treatment with 10 µM KGN during nine days of culture. Histological and SEM analyses showed the ability of hBMSCs in the presence of KGN to colonize the surface of OA cartilage and to produce glycosaminoglycans and proteoglycans after 21 days of co-culture. KGN treated hBMSCs secreted higher concentrations of TIMPs and the secretion of pro-inflammatory molecules (MMP 13, TNF-α) were significantly suppressed in comparison with control without hBMSCs. Our preliminary results support the concept that 10 µM KGN enhances proliferation and chondrogenic differentiation of hBMSCs and suggest that KGN is a potential promoter for cell-based therapeutic application for cartilage regeneration.
Collapse
Affiliation(s)
- Timea Spakova
- Associated Tissue Bank of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| | - Jana Plsikova
- Associated Tissue Bank of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| | - Denisa Harvanova
- Associated Tissue Bank of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| | - Marek Lacko
- Department of Orthopaedics and Traumatology of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| | - Stefan Stolfa
- Department of Orthopaedics and Traumatology of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| | - Jan Rosocha
- Associated Tissue Bank of Faculty of Medicine of P. J. Safarik University and L. Pasteur University Hospital, Trieda SNP 1, 04011 Kosice, Slovakia.
| |
Collapse
|
48
|
Zhang C, Wang X, Zhang E, Yang L, Yuan H, Tu W, Zhang H, Yin Z, Shen W, Chen X, Zhang Y, Ouyang H. An epigenetic bioactive composite scaffold with well-aligned nanofibers for functional tendon tissue engineering. Acta Biomater 2018; 66:141-156. [PMID: 28963019 DOI: 10.1016/j.actbio.2017.09.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022]
Abstract
Poor tendon repair is often a clinical challenge due to the lack of ideal biomaterials. Electrospun aligned fibers, resembling the ultrastructure of tendon, have been previously reported to promote tenogenesis. However, the underlying mechanism is unclear and the aligned fibers alone are not capable enough to commit teno-differentiation of stem cells. Here, based on our observation of reduced expression of histone deacetylases (HDACs) in tendon stem/progenitor cells (TSPCs) cultured on aligned fibers, we proposed a strategy to enhance the tenogenesis effect of aligned fibers by using a small molecule Trichostatin A (TSA), an HDAC inhibitor. Such a TSA-laden poly (l-lactic acid) (PLLA) aligned fiber (A-TSA) scaffold was successfully fabricated by a stable jet electrospinning method, and demonstrated its sustained capability in releasing TSA. We found that TSA incorporated aligned fibers of PLLA had an additive effect in directing tenogenic differentiation. Moreover, the in situ implantation study in rat model further confirmed that A-TSA scaffold promoted the structural and mechanical properties of the regenerated Achilles tendon. This study demonstrated that HDAC was involved in the teno-differentiation with aligned fiber topography, and the combination of HDAC with aligned topography might be a more efficient strategy to promote tenogenesis of stem cells. STATEMENT OF SIGNIFICANCE Electrospun aligned fibers, resembling the ultrastructure of tendon, have been previously reported to promote tenogenesis. However, the underlying mechanism is unclear and the aligned fibers alone are not capable enough to commit teno-differentiation of stem cells. The uniqueness of our studies are as follows, based on our observation of reduced expression of histone deacetylases (HDACs) in tendon stem/progenitor cells (TSPCs) cultured on aligned fibers, we proposed a strategy to enhance the tenogenesis effect of aligned fibers by using a small molecule Trichostatin A (TSA), a HDAC inhibitor. Such a TSA-laden poly (l-lactic acid) (PLLA) aligned fiber (A-TSA) scaffold was successfully fabricated by a stable jet electrospinning method, and demonstrated its sustained capability in releasing TSA. The incorporation and subsequent release of bioactive small molecule TSA into electrospun aligned fibers allows a controllable manner for both biochemical and physical regulation of tenogenesis of stem cells both in vitro and in vivo. Collectively, the present study provides a model of "translating the biological knowledge learned from cell-material interaction into optimizing biomaterials (from Biomat-to-Biomat)".
Collapse
Affiliation(s)
- Can Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China; Institute of Bionanotechnology and Tissue Engineering, College of Biology, Hunan University, Changsha 410082, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Erchen Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Long Yang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Huihua Yuan
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Wenjing Tu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Huilan Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China.
| | - Yanzhong Zhang
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China; College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou 310058, China; Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
49
|
Liu L, Li X, Shi X, Wang Y. Injectable alendronate-functionalized GelMA hydrogels for mineralization and osteogenesis. RSC Adv 2018; 8:22764-22776. [PMID: 35539745 PMCID: PMC9081581 DOI: 10.1039/c8ra03550d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/11/2018] [Indexed: 12/19/2022] Open
Abstract
Injectable alendronate-modified GelMA hydrogel greatly improved mineralization and in vitro osteogenesis both at the surface and inside of the hydrogel, which have potential in treatment of irregular bone defects.
Collapse
Affiliation(s)
- Lei Liu
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- PR China
- School of Material Science and Engineering
| | - Xiaoyu Li
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- PR China
- School of Material Science and Engineering
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- PR China
- School of Material Science and Engineering
| | - Yingjun Wang
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- South China University of Technology
- Guangzhou 510006
- PR China
- School of Material Science and Engineering
| |
Collapse
|
50
|
He Y, Zhu T, Liu L, Shi X, Lin Z. Modifying collagen with alendronate sodium for bone regeneration applications. RSC Adv 2018; 8:16762-16772. [PMID: 35540497 PMCID: PMC9080299 DOI: 10.1039/c8ra01872c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/26/2018] [Indexed: 11/21/2022] Open
Abstract
Phosphorylated materials are attractive candidates for bone regeneration because they may facilitate the construction of a phosphorylated bone extracellular matrix (ECM) to build a beneficial environment for bone formation. Here, we designed and synthesized a new phosphorylated material, collagen type I phosphorylated with alendronate sodium (Col-Aln), based on the biodegradable osteoconductive collagen backbone. Col-Aln can distinctly accelerate in vitro mineralization in simulated body fluid. Col-Aln showed good biocompatibility with bone marrow mesenchymal stem cells (BMSCs) and promoted their adhesion as well as the osteogenic differentiation of BMSCs more effectively than did pure collagen. Furthermore, collagen and Col-Aln scaffolds implanted into a critical-sized rat cranial defect for 4 and 8 weeks were shown to degrade in vivo and helped to facilitate bone growth in the defect, while the phosphate-containing Col-Aln scaffold significantly promoted new bone formation. Col-Aln provides a new strategy to integrate bioactive phosphate molecules via covalent grafting onto biopolymers and has promise for bone regeneration applications. Efficient covalent bonding with phosphate-containing alendronate prompts the fast mineralization and osteoinduction of the collagen scaffold.![]()
Collapse
Affiliation(s)
- Yingcong He
- Department of Operative Dentistry and Endodontics
- Guanghua School of Stomatology
- Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Stomatology
- Guangzhou
| | - Ting Zhu
- Department of Operative Dentistry and Endodontics
- Guanghua School of Stomatology
- Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Stomatology
- Guangzhou
| | - Lei Liu
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- School of Material Science and Engineering
- South China University of Technology
- Guangzhou
- PR China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction
- School of Material Science and Engineering
- South China University of Technology
- Guangzhou
- PR China
| | - Zhengmei Lin
- Department of Operative Dentistry and Endodontics
- Guanghua School of Stomatology
- Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Stomatology
- Guangzhou
| |
Collapse
|