1
|
Li Z, Ren K, Chen J, Zhuang Y, Dong S, Wang J, Liu H, Ding J. Bioactive hydrogel formulations for regeneration of pathological bone defects. J Control Release 2025; 380:686-714. [PMID: 39880040 DOI: 10.1016/j.jconrel.2025.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Bone defects caused by osteoporosis, infection, diabetes, post-tumor resection, and nonunion often cause severe pain and markedly increase morbidity and mortality, which remain a significant challenge for orthopedic surgeons. The precise local treatments for these pathological complications are essential to avoid poor or failed bone repair. Hydrogel formulations serve as injectable innovative platforms that overcome microenvironmental obstacles and as delivery systems for controlled release of various bioactive substances to bone defects in a targeted manner. Additionally, hydrogel formulations can be tailored for specific mechanical strengths and degradation profiles by adjusting their physical and chemical properties, which are crucial for prolonged drug retention and effective bone repair. This review summarizes recent advances in bioactive hydrogel formulations as three-dimensional scaffolds that support cell proliferation and differentiation. It also highlights their role as smart drug-delivery systems with capable of continuously releasing antibacterial agents, anti-inflammatory drugs, chemotherapeutic agents, and osteogenesis-related factors to enhance bone regeneration in pathological areas. Furthermore, the limitations of hydrogel formulations in pathological bone repair are discussed, and future development directions are proposed, which is expected to pave the way for the repair of pathological bone defects.
Collapse
Affiliation(s)
- Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - Kaixuan Ren
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Jiajia Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Yaling Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
2
|
Yu K, Zhu M, Dai Z, Zhang Q, Xiao L, Li X, Dai J, Jia Z, Li J, Bai Y, Zhang K. Dental resin for periodontal and tooth root regeneration via metformin to enhance osteogenic and cementogenic differentiation of human periodontal ligament stem cells. J Dent 2025; 153:105507. [PMID: 39643264 DOI: 10.1016/j.jdent.2024.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024] Open
Abstract
OBJECTIVES This study developed a novel dental resin incorporating metformin to repair root caries and periodontitis defects. The objectives were to: (1) Develop a novel dental resin with metformin release to fulfill the clinical requirements for mechanical properties; and (2) investigate the metformin release pattern and the effects on osteogenic and cementogenic differentiation of human periodontal ligament stem cells (hPDLSCs). METHODS Resin specimens with different concentrations of metformin were fabricated. Metformin release was measured using high-performance liquid chromatography. Cellular growth and proliferation on resin were assessed. Alizarin red S staining and an alkaline phosphatase (ALP) activity were determined. Quantitative real-time reverse transcription PCR was employed to determine osteogenic and cementogenic differentiation. RESULTS The resin with 7.5 % metformin had mechanical properties comparable to those of a commercial control, thus satisfying the clinical requirements (mean ± SD; n = 10). hPDLSCs showed robust growth and proliferation on resin. At 14 days, the number of hPDLSCs increased by four-fold (p > 0.1). ALP activity in the metformin group was 1.8 times higher than that of control (p < 0.05). At 21 days, the metformin group showed a significant increase in mineral synthesis. Metformin group had cementogenic and osteogenic differentiation values that were 2.5 and 2.3 times higher, respectively, than control without metformin (p < 0.05). SIGNIFICANCE A metformin-resin was formulated that greatly promoted both cementogenic and osteogenic differentiation for the first time. This resin was mechanically strong and supported hPDLSC adhesion, growth and proliferation. This resin substantially enhanced hPDLSC osteogenic and cementogenic differentiation, yielding mineral synthesis of hPDLSCs more than 2-fold that of control without metformin. This new metformin-resin shows promise for the restoration of tooth root caries and the regeneration of periodontal tissues including alveolar bone and cementum.
Collapse
Affiliation(s)
- Kan Yu
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Minjia Zhu
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Zixiang Dai
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| | - Qinrou Zhang
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Le Xiao
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Xinyi Li
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Jialiang Dai
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Zihan Jia
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Jingyi Li
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Yuxing Bai
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Ke Zhang
- Department of Orthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
3
|
Zhang X, Zhou X, Zhai W, Cui J, Pan Z, Du L, Wen L, Ye R, Zhang B, Huang L, Li D, Wang C, Sun H. Novel L-(CaP-ZnP)/SA Nanocomposite Hydrogel with Dual Anti-Inflammatory and Mineralization Effects for Efficient Vital Pulp Therapy. Int J Nanomedicine 2024; 19:6659-6676. [PMID: 38975320 PMCID: PMC11227880 DOI: 10.2147/ijn.s464871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/26/2024] [Indexed: 07/09/2024] Open
Abstract
Background Vital pulp therapy (VPT) is considered a conservative treatment for preserving pulp viability in caries and trauma-induced pulpitis. However, Mineral trioxide aggregate (MTA) as the most frequently used repair material, exhibits limited efficacy under inflammatory conditions. This study introduces an innovative nanocomposite hydrogel, tailored to simultaneously target anti-inflammation and dentin mineralization, aiming to efficiently preserve vital pulp tissue. Methods The L-(CaP-ZnP)/SA nanocomposite hydrogel was designed by combining L-Arginine modified calcium phosphate/zinc phosphate nanoparticles (L-(CaP-ZnP) NPs) with sodium alginate (SA), and was characterized with TEM, SEM, FTIR, EDX, ICP-AES, and Zeta potential. In vitro, we evaluated the cytotoxicity and anti-inflammatory properties. Human dental pulp stem cells (hDPSCs) were cultured with lipopolysaccharide (LPS) to induce an inflammatory response, and the cell odontogenic differentiation was measured and possible signaling pathways were explored by alkaline phosphatase (ALP)/alizarin red S (ARS) staining, qRT-PCR, immunofluorescence staining, and Western blotting, respectively. In vivo, a pulpitis model was utilized to explore the potential of the L-(CaP-ZnP)/SA nanocomposite hydrogel in controlling pulp inflammation and enhancing dentin mineralization by Hematoxylin and eosin (HE) staining and immunohistochemistry staining. Results In vitro experiments revealed that the nanocomposite hydrogel was synthesized successfully and presented desirable biocompatibility. Under inflammatory conditions, compared to MTA, the L-(CaP-ZnP)/SA nanocomposite hydrogel demonstrated superior anti-inflammatory and pro-odontogenesis effects. Furthermore, the nanocomposite hydrogel significantly augmented p38 phosphorylation, implicating the involvement of the p38 signaling pathway in pulp repair. Significantly, in a rat pulpitis model, the L-(CaP-ZnP)/SA nanocomposite hydrogel downregulated inflammatory markers while upregulating mineralization-related markers, thereby stimulating the formation of robust reparative dentin. Conclusion The L-(CaP-ZnP)/SA nanocomposite hydrogel with good biocompatibility efficiently promoted inflammation resolution and enhanced dentin mineralization by activating p38 signal pathway, as a pulp-capping material, offering a promising and advanced solution for treatment of pulpitis.
Collapse
Affiliation(s)
- Xu Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Xue Zhou
- Department of Chemistry, Northeast Normal University, Changchun, 130024, People’s Republic of China
| | - Wenhao Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Ziyi Pan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Liuyi Du
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Linlin Wen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Rongrong Ye
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Boya Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Chungang Wang
- Department of Chemistry, Northeast Normal University, Changchun, 130024, People’s Republic of China
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
4
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [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: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
5
|
Cao Z, Qin Z, Duns GJ, Huang Z, Chen Y, Wang S, Deng R, Nie L, Luo X. Repair of Infected Bone Defects with Hydrogel Materials. Polymers (Basel) 2024; 16:281. [PMID: 38276689 PMCID: PMC10820481 DOI: 10.3390/polym16020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Infected bone defects represent a common clinical condition involving bone tissue, often necessitating surgical intervention and antibiotic therapy. However, conventional treatment methods face obstacles such as antibiotic resistance and susceptibility to postoperative infections. Hydrogels show great potential for application in the field of tissue engineering due to their advantageous biocompatibility, unique mechanical properties, exceptional processability, and degradability. Recent interest has surged in employing hydrogels as a novel therapeutic intervention for infected bone repair. This article aims to comprehensively review the existing literature on the anti-microbial and osteogenic approaches utilized by hydrogels in repairing infected bones, encompassing their fabrication techniques, biocompatibility, antimicrobial efficacy, and biological activities. Additionally, the potential opportunities and obstacles in their practical implementation will be explored. Lastly, the limitations presently encountered and the prospective avenues for further investigation in the realm of hydrogel materials for the management of infected bone defects will be deliberated. This review provides a theoretical foundation and advanced design strategies for the application of hydrogel materials in the treatment of infected bone defects.
Collapse
Affiliation(s)
- Zhenmin Cao
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Zuodong Qin
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Gregory J. Duns
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| | - Zhao Huang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Yao Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Sheng Wang
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Ruqi Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Libo Nie
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
| | - Xiaofang Luo
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, China; (Z.C.); (Z.Q.); (Z.H.); (Y.C.); (S.W.); (R.D.)
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China;
| |
Collapse
|
6
|
Christie B, Musri N, Djustiana N, Takarini V, Tuygunov N, Zakaria M, Cahyanto A. Advances and challenges in regenerative dentistry: A systematic review of calcium phosphate and silicate-based materials on human dental pulp stem cells. Mater Today Bio 2023; 23:100815. [PMID: 37779917 PMCID: PMC10539671 DOI: 10.1016/j.mtbio.2023.100815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
Conventional dentistry faces limitations in preserving tooth health due to the finite lifespan of restorative materials. Regenerative dentistry, utilizing stem cells and bioactive materials, offers a promising approach for regenerating dental tissues. Human dental pulp stem cells (hDPSCs) and bioactive materials like calcium phosphate (CaP) and silicate-based materials have shown potential for dental tissue regeneration. This systematic review aims to investigate the effects of CaP and silicate-based materials on hDPSCs through in vitro studies published since 2015. Following the PRISMA guidelines, a comprehensive search strategy was implemented in PubMed MedLine, Cochrane, and ScienceDirect databases. Eligibility criteria were established using the PICOS scheme. Data extraction and risk of bias (RoB) assessment were conducted, with the included studies assessed for bias using the Office of Health and Translation (OHAT) RoB tool. The research has been registered at OSF Registries. Ten in vitro studies met the eligibility criteria out of 1088 initial studies. Methodological heterogeneity and the use of self-synthesized biomaterials with limited generalizability were observed in the included study. The findings highlight the positive effect of CaP and silicate-based materials on hDPSCs viability, adhesion, migration, proliferation, and differentiation. While the overall RoB assessment indicated satisfactory credibility of the reviewed studies, the limited number of studies and methodological heterogeneity pose challenges for quantitative research. In conclusion, this systematic review provides valuable insights into the effects of CaP and silicate-based materials on hDPSCs. Further research is awaited to enhance our understanding and optimize regenerative dental treatments using bioactive materials and hDPSCs, which promise to improve patient outcomes.
Collapse
Affiliation(s)
- B. Christie
- Faculty of Dentistry, Universitas Padjadjaran, Jalan Sekeloa Selatan 1, Bandung, 40134, Indonesia
| | - N. Musri
- Faculty of Dentistry, Universitas Padjadjaran, Jalan Sekeloa Selatan 1, Bandung, 40134, Indonesia
| | - N. Djustiana
- Department of Dental Materials Science and Technology, Faculty of Dentistry, Universitas Padjadjaran, Jalan Raya Bandung Sumedang Km 21, Jatinangor, 45363, Indonesia
- Oral Biomaterials Study Center, Faculty of Dentistry, Universitas Padjadjaran, Jalan Sekeloa Selatan 1, Bandung, 40134, Indonesia
| | - V. Takarini
- Department of Dental Materials Science and Technology, Faculty of Dentistry, Universitas Padjadjaran, Jalan Raya Bandung Sumedang Km 21, Jatinangor, 45363, Indonesia
- Oral Biomaterials Study Center, Faculty of Dentistry, Universitas Padjadjaran, Jalan Sekeloa Selatan 1, Bandung, 40134, Indonesia
| | - N. Tuygunov
- Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - M.N. Zakaria
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - A. Cahyanto
- Department of Dental Materials Science and Technology, Faculty of Dentistry, Universitas Padjadjaran, Jalan Raya Bandung Sumedang Km 21, Jatinangor, 45363, Indonesia
- Oral Biomaterials Study Center, Faculty of Dentistry, Universitas Padjadjaran, Jalan Sekeloa Selatan 1, Bandung, 40134, Indonesia
- Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jalan Raya Bandung-Sumedang Km 21, Jatinangor, 45363, Indonesia
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| |
Collapse
|
7
|
Sun Y, Zhao Z, Qiao Q, Li S, Yu W, Guan X, Schneider A, Weir MD, Xu HHK, Zhang K, Bai Y. Injectable periodontal ligament stem cell-metformin-calcium phosphate scaffold for bone regeneration and vascularization in rats. Dent Mater 2023; 39:872-885. [PMID: 37574338 DOI: 10.1016/j.dental.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
OBJECTIVES Injectable and self-setting calcium phosphate cement scaffold (CPC) capable of encapsulating and delivering stem cells and bioactive agents would be highly beneficial for dental and craniofacial repairs. The objectives of this study were to: (1) develop a novel injectable CPC scaffold encapsulating human periodontal ligament stem cells (hPDLSCs) and metformin (Met) for bone engineering; (2) test bone regeneration efficacy in vitro and in vivo. METHODS hPDLSCs were encapsulated in degradable alginate fibers, which were then mixed into CPC paste. Five groups were tested: (1) CPC control; (2) CPC + hPDLSC-fibers + 0% Met (CPC + hPDLSCs + 0%Met); (3) CPC + hPDLSC-fibers + 0.1% Met (CPC + hPDLSCs + 0.1%Met); (4) CPC + hPDLSC-fibers + 0.2% Met (CPC + hPDLSCs + 0.2%Met); (5) CPC + hPDLSC-fibers + 0.4% Met (CPC + hPDLSCs + 0.4%Met). The injectability, mechanical properties, metformin release, and hPDLSC osteogenic differentiation and bone mineral were determined in vitro. A rat cranial defect model was used to evaluate new bone formation. RESULTS The novel construct had good injectability and physical properties. Alginate fibers degraded in 7 days and released hPDLSCs, with 5-fold increase of proliferation (p<0.05). The ALP activity and mineral synthesis of hPDLSCs were increased by Met delivery (p<0.05). Among all groups, CPC+hPDLSCs+ 0.1%Met showed the greatest cell mineralization and osteogenesis, which were 1.5-10 folds those without Met (p<0.05). Compared to CPC control, CPC+hPDLSCs+ 0.1%Met enhanced bone regeneration in rats by 9 folds, and increased vascularization by 3 folds (p<0.05). CONCLUSIONS The novel injectable construct with hPDLSC and Met encapsulation demonstrated excellent efficacy for bone regeneration and vascularization in vivo in an animal model. CPC+hPDLSCs+ 0.1%Met is highly promising for dental and craniofacial applications.
Collapse
Affiliation(s)
- Yaxi Sun
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Shengnan Li
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Wenting Yu
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, USA
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Ma M, Shen W, Li B, Sun M, Lin D, Meng L. Optimization of a concentrated growth factor/mesoporous bioactive glass composite scaffold and its application in rabbit mandible defect regeneration. Biomater Sci 2023; 11:6357-6372. [PMID: 37584200 DOI: 10.1039/d3bm00805c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Maxillofacial bone defect repair and regeneration remains a tremendous challenge in the field of stomatology. However, the limited osteoinductivity of artificial materials and the high cost of bioactive agents restrain their clinical translation. This study aimed to construct an economical and efficient concentrated growth factor/mesoporous bioactive glass (CGF/MBG) composite scaffold for bone regeneration. The biochemical composition and biological effects of different forms of CGFs were systematically compared, and the results showed that CGF-conditioned medium effectively promoted proliferation, migration and osteogenesis of allogenic BMSCs. Gel phase CGF (gpCGF) exhibited superior bioactivity and osteoinductivity to liquid phase CGF (lpCGF) and liquid/gel mixed phase CGF (lgpCGF), and was further applied to construct CGF/MBG scaffolds. In vitro studies demonstrated that co-culture with gpCGF-conditioned medium further enhanced the biocompatibility of MBG, increasing cell adhesion and proliferation on the scaffold. On this basis, two compositing approaches to construct the scaffold by fibrin gel formation (CGF/FG/MBG) and freeze-drying (fdCGF/MBG) were applied, and the biological efficacy of CGFs was compared in vivo. In a rabbit mandibular defect model, higher osteogenic efficiency in in situ bone regeneration of CGF/FG/MBG composite scaffolds was proved, compared with fdCGF/MBG. Taken together, the CGF/FG/MBG composite scaffold is expected to be an efficient bone repairing therapy for clinical translation, and the CGF-composited scaffold using gpCGF and the fibrin gel formation method is a promising way to enhance the bioactivity and osteoinductivity of current clinical bone repairing materials, providing new thoughts on the development of future orthopedic biomaterials.
Collapse
Affiliation(s)
- Mengran Ma
- Department of Prosthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Wenjing Shen
- Department of Prosthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Beibei Li
- Department of Prosthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Mengwen Sun
- Department of Prosthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Dan Lin
- Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China.
| | - Lingqiang Meng
- Department of Prosthodontics, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
9
|
Atia GAN, Shalaby HK, Ali NG, Morsy SM, Ghobashy MM, Attia HAN, Barai P, Nady N, Kodous AS, Barai HR. New Challenges and Prospective Applications of Three-Dimensional Bioactive Polymeric Hydrogels in Oral and Craniofacial Tissue Engineering: A Narrative Review. Pharmaceuticals (Basel) 2023; 16:702. [PMID: 37242485 PMCID: PMC10224377 DOI: 10.3390/ph16050702] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Regenerative medicine, and dentistry offers enormous potential for enhancing treatment results and has been fueled by bioengineering breakthroughs over the previous few decades. Bioengineered tissues and constructing functional structures capable of healing, maintaining, and regenerating damaged tissues and organs have had a broad influence on medicine and dentistry. Approaches for combining bioinspired materials, cells, and therapeutic chemicals are critical in stimulating tissue regeneration or as medicinal systems. Because of its capacity to maintain an unique 3D form, offer physical stability for the cells in produced tissues, and replicate the native tissues, hydrogels have been utilized as one of the most frequent tissue engineering scaffolds during the last twenty years. Hydrogels' high water content can provide an excellent conditions for cell viability as well as an architecture that mimics real tissues, bone, and cartilage. Hydrogels have been used to enable cell immobilization and growth factor application. This paper summarizes the features, structure, synthesis and production methods, uses, new challenges, and future prospects of bioactive polymeric hydrogels in dental and osseous tissue engineering of clinical, exploring, systematical and scientific applications.
Collapse
Affiliation(s)
- Gamal Abdel Nasser Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Hany K. Shalaby
- Department of Oral Medicine, Periodontology and Oral Diagnosis, Faculty of Dentistry, Suez University, Suez P.O. Box 43512, Egypt
| | - Naema Goda Ali
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Shaimaa Mohammed Morsy
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia P.O. Box 41522, Egypt
| | - Mohamed Mohamady Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo P.O. Box 13759, Egypt
| | - Hager Abdel Nasser Attia
- Department of Molecular Biology and Chemistry, Faculty of Science, Alexandria University, Alexandria P.O. Box 21526, Egypt
| | - Paritosh Barai
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka 1213, Bangladesh
| | - Norhan Nady
- Polymeric Materials Research Department, Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg Elarab, Alexandria P.O. Box 21934, Egypt
| | - Ahmad S. Kodous
- Department of Radiation Biology, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo P.O. Box 13759, Egypt
| | - Hasi Rani Barai
- Department of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
10
|
Zhang Z, Bi F, Guo W. Research Advances on Hydrogel-Based Materials for Tissue Regeneration and Remineralization in Tooth. Gels 2023; 9:gels9030245. [PMID: 36975694 PMCID: PMC10048036 DOI: 10.3390/gels9030245] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Tissue regeneration and remineralization in teeth is a long-term and complex biological process, including the regeneration of pulp and periodontal tissue, and re-mineralization of dentin, cementum and enamel. Suitable materials are needed to provide cell scaffolds, drug carriers or mineralization in this environment. These materials need to regulate the unique odontogenesis process. Hydrogel-based materials are considered good scaffolds for pulp and periodontal tissue repair in the field of tissue engineering due to their inherent biocompatibility and biodegradability, slow release of drugs, simulation of extracellular matrix, and the ability to provide a mineralized template. The excellent properties of hydrogels make them particularly attractive in the research of tissue regeneration and remineralization in teeth. This paper introduces the latest progress of hydrogel-based materials in pulp and periodontal tissue regeneration and hard tissue mineralization and puts forward prospects for their future application. Overall, this review reveals the application of hydrogel-based materials in tissue regeneration and remineralization in teeth.
Collapse
Affiliation(s)
- Zhijun Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Fei Bi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Pediatric Dentistry, West China School of Stomatology, Sichuan University, Chengdu 610041, China
- Yunnan Key Laboratory of Stomatology, The Affiliated Hospital of Stomatology, School of Stomatology, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
11
|
Tahmasebi E, Mohammadi M, Alam M, Abbasi K, Gharibian Bajestani S, Khanmohammad R, Haseli M, Yazdanian M, Esmaeili Fard Barzegar P, Tebyaniyan H. The current regenerative medicine approaches of craniofacial diseases: A narrative review. Front Cell Dev Biol 2023; 11:1112378. [PMID: 36926524 PMCID: PMC10011176 DOI: 10.3389/fcell.2023.1112378] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Craniofacial deformities (CFDs) develop following oncological resection, trauma, or congenital disorders. Trauma is one of the top five causes of death globally, with rates varying from country to country. They result in a non-healing composite tissue wound as they degenerate in soft or hard tissues. Approximately one-third of oral diseases are caused by gum disease. Due to the complexity of anatomical structures in the region and the variety of tissue-specific requirements, CFD treatments present many challenges. Many treatment methods for CFDs are available today, such as drugs, regenerative medicine (RM), surgery, and tissue engineering. Functional restoration of a tissue or an organ after trauma or other chronic diseases is the focus of this emerging field of science. The materials and methodologies used in craniofacial reconstruction have significantly improved in the last few years. A facial fracture requires bone preservation as much as possible, so tiny fragments are removed initially. It is possible to replace bone marrow stem cells with oral stem cells for CFDs due to their excellent potential for bone formation. This review article discusses regenerative approaches for different types of craniofacial diseases.
Collapse
Affiliation(s)
- Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohammadi
- School of Dentistry, Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Gharibian Bajestani
- Student Research Committee, Dentistry Research Center, Research Institute of Dental Sciences, Dental School, Shahid Behesti University of Medical Sciences, Tehran, Iran
| | - Rojin Khanmohammad
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Haseli
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| |
Collapse
|
12
|
Zhao Z, Liu J, Weir MD, Schneider A, Ma T, Oates TW, Xu HHK, Zhang K, Bai Y. Periodontal ligament stem cell-based bioactive constructs for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:1071472. [PMID: 36532583 PMCID: PMC9755356 DOI: 10.3389/fbioe.2022.1071472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/17/2022] [Indexed: 09/29/2023] Open
Abstract
Objectives: Stem cell-based tissue engineering approaches are promising for bone repair and regeneration. Periodontal ligament stem cells (PDLSCs) are a promising cell source for tissue engineering, especially for maxillofacial bone and periodontal regeneration. Many studies have shown potent results via PDLSCs in bone regeneration. In this review, we describe recent cutting-edge researches on PDLSC-based bone regeneration and periodontal tissue regeneration. Data and sources: An extensive search of the literature for papers related to PDLSCs-based bioactive constructs for bone tissue engineering was made on the databases of PubMed, Medline and Google Scholar. The papers were selected by three independent calibrated reviewers. Results: Multiple types of materials and scaffolds have been combined with PDLSCs, involving xeno genic bone graft, calcium phosphate materials and polymers. These PDLSC-based constructs exhibit the potential for bone and periodontal tissue regeneration. In addition, various osteo inductive agents and strategies have been applied with PDLSCs, including drugs, biologics, gene therapy, physical stimulation, scaffold modification, cell sheets and co-culture. Conclusoin: This review article demonstrates the great potential of PDLSCs-based bioactive constructs as a promising approach for bone and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Jin Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Michael D. Weir
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Thomas W. Oates
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
| | - Hockin H. K. Xu
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Zhao H, Wang X, Jin A, Wang M, Wang Z, Huang X, Dai J, Wang X, Lin D, Shen SGF. Reducing relapse and accelerating osteogenesis in rapid maxillary expansion using an injectable mesoporous bioactive glass/fibrin glue composite hydrogel. Bioact Mater 2022; 18:507-525. [PMID: 35415307 PMCID: PMC8976096 DOI: 10.1016/j.bioactmat.2022.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022] Open
Abstract
Rapid maxillary expansion (RME), as a common treatment for craniomaxillofacial deformity, faces the challenge of high relapse rates and unsatisfactory therapeutic effects. In this study, a standardized Sprague-Dawley (SD) rat RME model was first established with a modified expander as well as retainer design and optimized anterior maxillary expanding force of 100 g which exerted the most synchronized mobility of mid-palatal suture and incisors. Via the standardized model, the high relapse rate was proven to be attributed to insufficient osteogenesis in expanded suture, requiring long-term retainer wearing in clinical situations. To reduce the relapse rate, mesoporous bioactive glass/fibrin glue (MBG/FG) composite hydrogels were developed for an in situ minimal invasive injection that enhance osteogenesis in the expanded palate. The component of 1 wt% MBG was adopted for enhanced mechanical strength, matched degradation rate and ion dissolution, excellent in vitro biocompatibility and osteoinductivity. Effects of 1%MBG/FG composite hydrogel on osteogenesis in expanded mid-palatal sutures with/without retention were evaluated in the standardized model. The results demonstrated that injection of 1%MBG/FG composite hydrogel significantly promoted bone formation within the expanded mid-palatal suture, inhibited osteoclastogenesis and benefited the balance of bone remodeling towards osteogenesis. Combination of retainer and injectable biomaterial was demonstrated as a promising treatment to reduce relapse rate and enhance osteogenesis after RME. The model establishment and the composite hydrogel development in this article might provide new insight to other craniomaxillofacial deformity treatment and design of bone-repairing biomaterials with higher regenerative efficiency. A standardized rat RME model was established with optimized parameters. Sufficient osteogenesis was the prerequisite of reducing relapse ratio. Design of an injectable MBG/FG composite hydrogel for osteogenic enhancement. Combinatory treatment of injection and retention was developed for relapse reduction.
Collapse
|
14
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
15
|
Shaikh MS, Shahzad Z, Tash EA, Janjua OS, Khan MI, Zafar MS. Human Umbilical Cord Mesenchymal Stem Cells: Current Literature and Role in Periodontal Regeneration. Cells 2022; 11:cells11071168. [PMID: 35406732 PMCID: PMC8997495 DOI: 10.3390/cells11071168] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
Periodontal disease can cause irreversible damage to tooth-supporting tissues such as the root cementum, periodontal ligament, and alveolar bone, eventually leading to tooth loss. While standard periodontal treatments are usually helpful in reducing disease progression, they cannot repair or replace lost periodontal tissue. Periodontal regeneration has been demonstrated to be beneficial in treating intraosseous and furcation defects to varied degrees. Cell-based treatment for periodontal regeneration will become more efficient and predictable as tissue engineering and progenitor cell biology advance, surpassing the limitations of present therapeutic techniques. Stem cells are undifferentiated cells with the ability to self-renew and differentiate into several cell types when stimulated. Mesenchymal stem cells (MSCs) have been tested for periodontal regeneration in vitro and in humans, with promising results. Human umbilical cord mesenchymal stem cells (UC-MSCs) possess a great regenerative and therapeutic potential. Their added benefits comprise ease of collection, endless source of stem cells, less immunorejection, and affordability. Further, their collection does not include the concerns associated with human embryonic stem cells. The purpose of this review is to address the most recent findings about periodontal regenerative mechanisms, different stem cells accessible for periodontal regeneration, and UC-MSCs and their involvement in periodontal regeneration.
Collapse
Affiliation(s)
- Muhammad Saad Shaikh
- Department of Oral Biology, Sindh Institute of Oral Health Sciences, Jinnah Sindh Medical University, Karachi 75510, Pakistan;
| | - Zara Shahzad
- Lahore Medical and Dental College, University of Health Sciences, Lahore 53400, Pakistan;
| | - Esraa Abdulgader Tash
- Department of Oral and Clinical Basic Science, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia;
| | - Omer Sefvan Janjua
- Department of Maxillofacial Surgery, PMC Dental Institute, Faisalabad Medical University, Faisalabad 38000, Pakistan;
| | | | - Muhammad Sohail Zafar
- Department of Restorative Dentistry, College of Dentistry, Taibah University, Al Madinah Al Munawarah 41311, Saudi Arabia
- Department of Dental Materials, Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan
- Correspondence: ; Tel.: +966-507544691
| |
Collapse
|
16
|
Arora S, Cooper PR, Ratnayake JT, Friedlander LT, Rizwan SB, Seo B, Hussaini HM. A critical review of in vitro research methodologies used to study mineralization in human dental pulp cell cultures. Int Endod J 2022; 55 Suppl 1:3-13. [PMID: 35030284 PMCID: PMC9303903 DOI: 10.1111/iej.13684] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/11/2022] [Indexed: 11/27/2022]
Abstract
Background The pulp contains a resident population of stem cells which can be stimulated to differentiate in order to repair the tooth by generating a mineralized extracellular matrix. Over recent decades there has been considerable interest in utilizing in vitro cell culture models to study dentinogenesis, with the aim of developing regenerative endodontic procedures, particularly where some vital pulp tissue remains. Objectives The purpose of this review is to provide a structured oversight of in vitro research methodologies which have been used to study human pulp mineralization processes. Method The literature was screened in the PubMed database up to March 2021 to identify manuscripts reporting the use of human dental pulp cells to study mineralization. The dataset identified 343 publications initially which were further screened and consequently 166 studies were identified and it was methodologically mined for information on: i) study purpose, ii) source and characterization of cells, iii) mineralizing supplements and concentrations, and iv) assays and markers used to characterize mineralization and differentiation, and the data was used to write this narrative review. Results Most published studies aimed at characterizing new biological stimulants for mineralization as well as determining the effect of scaffolds and dental (bio)materials. In general, pulp cells were isolated by enzymatic digestion, although the pulp explant technique was also common. For enzymatic digestion, a range of enzymes and concentrations were utilized, although collagenase type I and dispase were the most frequent. Isolated cells were not routinely characterized using either fluorescence‐activated cell sorting (FACS) and magnetic‐activated cell sorting (MACS) approaches and there was little consistency in terming cultures as dental pulp cells or dental pulp stem cells. A combination of media supplements, at a range of concentrations, of dexamethasone, ascorbic acid and beta‐glycerophosphate, were frequently applied as the basis for the experimental conditions. Alizarin Red S (ARS) staining was the method of choice for assessment of mineralization at 21‐days. Alkaline phosphatase assay was relatively frequently applied, solely or in combination with ARS staining. Further assessment of differentiation status was performed using transcript or protein markers, with dentine sialophosphoprotein (DSPP), osteocalcin and dentine matrix protein‐1 (DMP ‐1), the most frequent. Discussion While this review highlights variability among experimental approaches, it does however identify a consensus experimental approach. Conclusion Standardization of experimental conditions and sustained research will significantly benefit endodontic patient outcomes in the future.
Collapse
Affiliation(s)
- Shelly Arora
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Paul R Cooper
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Jithendra T Ratnayake
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Lara T Friedlander
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | | | - Benedict Seo
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Haizal M Hussaini
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| |
Collapse
|
17
|
Poorna MR, Jayakumar R, Chen JP, Mony U. Hydrogels: A potential platform for induced pluripotent stem cell culture and differentiation. Colloids Surf B Biointerfaces 2021; 207:111991. [PMID: 34333302 DOI: 10.1016/j.colsurfb.2021.111991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 01/02/2023]
Abstract
Induced pluripotent stem cells (iPSCs) can be used to generate desired types of cells that belong to the three germ layers (i.e., ectoderm, endoderm and mesoderm). These cells possess great potential in regenerative medicine. Before iPSCs are used in various biomedical applications, the existing xenogeneic culture methods must be improved to meet the technical standards of safety, cost effectiveness, and ease of handling. In addition to commonly used 2D substrates, a culture system that mimics the native cellular environment in tissues will be a good choice when culturing iPS cells and differentiating them into different lineages. Hydrogels are potential candidates that recapitulate the native complex three-dimensional microenvironment. They possess mechanical properties similar to those of many soft tissues. Moreover, hydrogels support iPSC adhesion, proliferation and differentiation to various cell types. They are xeno-free and cost-effective. In addition to other substrates, such as mouse embryonic fibroblast (MEF), Matrigel, and vitronectin, the use of hydrogel-based substrates for iPSC culture and differentiation may help generate large numbers of clinical-grade cells that can be used in potential clinical applications. This review mainly focuses on the use of hydrogels for the culture and differentiation of iPSCs into various cell types and their potential applications in regenerative medicine.
Collapse
Affiliation(s)
- M R Poorna
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - R Jayakumar
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan, ROC; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan, ROC; Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan, ROC.
| | - Ullas Mony
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India; Department of Biochemistry, Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India.
| |
Collapse
|
18
|
Song B, Fu H, Liu J, Ren K, Weir MD, Schneider A, Wang P, Song Y, Zhao L, Xu H. Bioactive small molecules in calcium phosphate scaffold enhanced osteogenic differentiation of human induced pluripotent stem cells. Dent Mater J 2021; 40:615-624. [PMID: 33814531 DOI: 10.4012/dmj.2019-263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are exciting for regenerative medicine due to their multi-potent differentiation. SB431542 bioactive molecule can activate bone morphogenetic protein-signalling in osteoblasts. The objectives were to: (1) develop a novel injectable calcium phosphate cement (CPC)-SB431542 scaffold for dental/craniofacial bone engineering; and (2) investigate cell proliferation and osteo-differentiation of hiPSC-derived mesenchymal stem cells (hiPSC-MSCs) on CPC-SB431542 scaffold. Three groups were tested: CPC control; CPC with SB431542 inside CPC (CPCSM); CPC with SB431542 in osteogenic medium (CPC+SMM). SB431542 in CPC promoted stem cell proliferation and viability. hiPSC-MSCs differentiated into osteogenic lineage and synthesized bone minerals. CPC with SB431542 showed much greater osteo-expressions and more bone minerals than those without SB431542. In conclusion, hiPSC-MSCs on CPC scaffold containing SB431542 showed excellent osteo-differentiation and bone mineral synthesis for the first time. CPC was a suitable scaffold for delivering stem cells and SB431542 to promote bone regeneration in dental/craniofacial applications.
Collapse
Affiliation(s)
- Bing Song
- Department of Orthopedic Surgery, Shunde Hospital of Southern Medical University.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Haijun Fu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology
| | - Jianwei Liu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology
| | - Ke Ren
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry
| | - Ping Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Yang Song
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry.,Department of Prosthodontics, Guanghua School of Stomatology, Sun Yat-sen University
| | - Liang Zhao
- Department of Orthopedic Surgery, Shunde Hospital of Southern Medical University.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Huakun Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine.,University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine
| |
Collapse
|
19
|
Wu S, Lei L, Bao C, Liu J, Weir MD, Ren K, Schneider A, Oates TW, Liu J, Xu HHK. An injectable and antibacterial calcium phosphate scaffold inhibiting Staphylococcus aureus and supporting stem cells for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111688. [PMID: 33545850 DOI: 10.1016/j.msec.2020.111688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Staphylococcus aureus (S. aureus) is the major pathogen for osteomyelitis, which can lead to bone necrosis and destruction. There has been no report on antibacterial calcium phosphate cement (CPC) against S. aureus. The aims of this study were to: (1) develop novel antibacterial CPC-chitosan-alginate microbead scaffold; (2) investigate mechanical and antibacterial properties of CPC-chitosan-penicillin-alginate scaffold; (3) evaluate the encapsulation and delivery of human umbilical cord mesenchymal stem cells (hUCMSCs). Flexural strength, elastic modulus and work-of-fracture of the CPC-chitosan-penicillin-alginate microbeads scaffold and CPC-chitosan scaffold were evaluated. Penicillin release profile and antibacterial effects on S. aureus were determined. The hUCMSC delivery and release from penicillin-alginate microbeads were investigated. Injectable CPC-chitosan-penicillin-alginate microbeads scaffold was developed for the first time. CPC-chitosan-penicillin-alginate microbeads scaffold had a flexural strength of 3.16 ± 0.55 MPa, matching that of cancellous bone. With sustained penicillin release, the new scaffold had strong antibacterial effects on S. aureus, with an inhibition zone diameter of 32.2 ± 2.5 mm, greater than that of penicillin disk control (15.1 ± 2.0 mm) (p < 0.05). Furthermore, this injectable and antibacterial scaffold had no toxic effects, yielding excellent hUCMSC viability, which was similar to that of CPC control without antibacterial activity (p > 0.05). CPC-chitosan-penicillin-microbeads scaffold had injectability, good strength, strong antibacterial effects, and good biocompatibility to support stem cell viability for osteogenesis. CPC-chitosan-penicillin-microbeads scaffold is promising for dental, craniofacial and orthopedic applications to combat infections and promote bone regeneration.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Key Laboratory of Shannxi for Craniofacial Precision Medicine Research, Clinical Research Center of Shannxi for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Shannxi 710004, China
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas W Oates
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
20
|
Nizami MZI, Nishina Y. Recent Advances in Stem Cells for Dental Tissue Engineering. ENGINEERING MATERIALS FOR STEM CELL REGENERATION 2021:281-324. [DOI: 10.1007/978-981-16-4420-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
21
|
Emara A, Shah R. Recent update on craniofacial tissue engineering. J Tissue Eng 2021; 12:20417314211003735. [PMID: 33959245 PMCID: PMC8060749 DOI: 10.1177/20417314211003735] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
The craniofacial region consists of several different tissue types. These tissues are quite commonly affected by traumatic/pathologic tissue loss which has so far been traditionally treated by grafting procedures. With the complications and drawbacks of grafting procedures, the emerging field of regenerative medicine has proved potential. Tissue engineering advancements and the application in the craniofacial region is quickly gaining momentum although most research is still at early in vitro/in vivo stages. We aim to provide an overview on where research stands now in tissue engineering of craniofacial tissue; namely bone, cartilage muscle, skin, periodontal ligament, and mucosa. Abstracts and full-text English articles discussing techniques used for tissue engineering/regeneration of these tissue types were summarized in this article. The future perspectives and how current technological advancements and different material applications are enhancing tissue engineering procedures are also highlighted. Clinically, patients with craniofacial defects need hybrid reconstruction techniques to overcome the complexity of these defects. Cost-effectiveness and cost-efficiency are also required in such defects. The results of the studies covered in this review confirm the potential of craniofacial tissue engineering strategies as an alternative to avoid the problems of currently employed techniques. Furthermore, 3D printing advances may allow for fabrication of patient-specific tissue engineered constructs which should improve post-operative esthetic results of reconstruction. There are on the other hand still many challenges that clearly require further research in order to catch up with engineering of other parts of the human body.
Collapse
Affiliation(s)
- Aala’a Emara
- OMFS Department, Faculty of Dentistry,
Cairo University, Cairo, Egypt
- Division of Craniofacial and Surgical
Care, University of North Carolina (UNC) School of Dentistry, Chapel Hill, NC,
USA
| | - Rishma Shah
- Division of Craniofacial and Surgical
Care, University of North Carolina (UNC) School of Dentistry, Chapel Hill, NC,
USA
| |
Collapse
|
22
|
Chen H, Yang H, Weir MD, Schneider A, Ren K, Homayounfar N, Oates TW, Zhang K, Liu J, Hu T, Xu HHK. An antibacterial and injectable calcium phosphate scaffold delivering human periodontal ligament stem cells for bone tissue engineering. RSC Adv 2020; 10:40157-40170. [PMID: 35520873 PMCID: PMC9057516 DOI: 10.1039/d0ra06873j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Osteomyelitis and post-operative infections are major problems in orthopedic, dental and craniofacial surgeries. It is highly desirable for a tissue engineering construct to kill bacteria, while simultaneously delivering stem cells and enhancing cell function and tissue regeneration. The objectives of this study were to: (1) develop a novel injectable calcium phosphate cement (CPC) scaffold containing antibiotic ornidazole (ORZ) while encapsulating human periodontal ligament stem cells (hPDLSCs), and (2) investigate the inhibition efficacy against Staphylococcus aureus (S. aureus) and the promotion of hPDLSC function for osteogenesis for the first time. ORZ was incorporated into a CPC-chitosan scaffold. hPDLSCs were encapsulated in alginate microbeads (denoted hPDLSCbeads). The ORZ-loaded CPCC+hPDLSCbeads scaffold was fully injectable, and had a flexural strength of 3.50 ± 0.92 MPa and an elastic modulus of 1.30 ± 0.45 GPa, matching those of natural cancellous bone. With 6 days of sustained ORZ release, the CPCC+10ORZ (10% ORZ) scaffold had strong antibacterial effects on S. aureus, with an inhibition zone of 12.47 ± 1.01 mm. No colonies were observed in the CPCC+10ORZ group from 3 to 7 days. ORZ-containing scaffolds were biocompatible with hPDLSCs. CPCC+10ORZ+hPDLSCbeads scaffold with osteogenic medium had 2.4-fold increase in alkaline phosphatase (ALP) activity and bone mineral synthesis by hPDLSCs, as compared to the control group (p < 0.05). In conclusion, the novel antibacterial construct with stem cell delivery had injectability, good strength, strong antibacterial effects and biocompatibility, supporting osteogenic differentiation and bone mineral synthesis of hPDLSCs. The injectable and mechanically-strong CPCC+10ORZ+hPDLSCbeads construct has great potential for treating bone infections and promoting bone regeneration.
Collapse
Affiliation(s)
- Hong Chen
- Department of Endodontics, College of Stomatological, Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education Chongqing China
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Hui Yang
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry Baltimore USA
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine Baltimore MD 21201 USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland Baltimore MD 21201 USA
| | - Negar Homayounfar
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University Beijing China
| | - Jin Liu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University Xi'an Shannxi China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine Baltimore MD 21201 USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine Baltimore MD 21201 USA
| |
Collapse
|
23
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
24
|
Wu S, Lei L, Zhang H, Liu J, Weir MD, Schneider A, Zhao L, Liu J, Xu HH. Nanographene oxide‐calcium phosphate to inhibit
Staphylococcus aureus
infection and support stem cells for bone tissue engineering. J Tissue Eng Regen Med 2020; 14:1779-1791. [PMID: 33025745 DOI: 10.1002/term.3139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital Sichuan University Chengdu China
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics University of Maryland Dental School Baltimore MD USA
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Hui Zhang
- Department of Orthopedic Surgery, West China Hospital Sichuan University Chengdu China
| | - Jin Liu
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics University of Maryland Dental School Baltimore MD USA
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shannxi Province for Dental and Maxillofacial Diseases, College of Stomatology Xi'an Jiaotong University Xi'an China
| | - Michael D. Weir
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics University of Maryland Dental School Baltimore MD USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences University of Maryland School of Dentistry Baltimore MD USA
| | - Liang Zhao
- Department of Orthopedic Surgery, Nanfang Hospital Southern Medical University Guangzhou China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Sichuan University Chengdu China
| | - Hockin H.K. Xu
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics University of Maryland Dental School Baltimore MD USA
- Marlene and Stewart Greenebaum Cancer Center University of Maryland School of Medicine Baltimore MD USA
- Center for Stem Cell Biology and Regenerative Medicine University of Maryland School of Medicine Baltimore MD USA
| |
Collapse
|
25
|
Mehrotra D, Dwivedi R, Nandana D, Singh RK. From injectable to 3D printed hydrogels in maxillofacial tissue engineering: A review. J Oral Biol Craniofac Res 2020; 10:680-689. [PMID: 33072505 DOI: 10.1016/j.jobcr.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/25/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction This review aims at describing different types of hydrogels in context to their composition, fabrication techniques and other specific features along with an insight into the latest advancements including smart hydrogels, 3D printed, programmable, shape memory and self-healing hydrogels for their applicability as scaffold in maxillofacial bone and cartilage tissue regeneration. Methods Electronic database searches were undertaken on PubMed, Ovid, Medline, Embase, ProQuest and science direct for English language literature, published for application of hydrogels in maxillofacial bone and cartilage tissue engineering. The search items used in this article were hydrogel, bone and cartilage tissue engineering, maxillofacial, clinical trials. Reviews and in vitro studies were excluded. Results Search for injectable hydrogel showed 4955 articles, when restricted to bone tissue engineering results were reduced to 463 and for cartilage engineering to 335; when we limited it to maxillofacial bone and cartilage tissue engineering, search results showed 49 articles to which 9 additional articles were included from references, after exclusion of in-vitro studies and duplicates 16 articles were obtained for our study. Similarly, for 3D printed hydrogels, result showed 1126 articles, which got restricted to 19 when searched for maxillofacial bone and cartilage engineering, then 2 additional articles were included directly from references, and finally after exclusion of the invitro studies and duplicates, a total of 5 articles were obtained. Conclusion Modifications in hydrogel can improve the mechanical properties, biocompatibility and unique chemistries for its use in bone and cartilage tissue engineering for future research.
Collapse
Affiliation(s)
- Divya Mehrotra
- Professor, Dept of Oral & Maxillofacial Surgery, King George's Medical University, Lucknow, India
| | - Ruby Dwivedi
- Research Student, Dept of Oral & Maxillofacial Surgery, King George's Medical University, Lucknow, India
| | - Deepti Nandana
- Research Student, Dept of Oral & Maxillofacial Surgery, King George's Medical University, Lucknow, India
| | - R K Singh
- Professor, Dept of Oral & Maxillofacial Surgery, King George's Medical University, Lucknow, India
| |
Collapse
|
26
|
Tran HD, Park KD, Ching YC, Huynh C, Nguyen DH. A comprehensive review on polymeric hydrogel and its composite: Matrices of choice for bone and cartilage tissue engineering. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
27
|
Chindamo G, Sapino S, Peira E, Chirio D, Gonzalez MC, Gallarate M. Bone Diseases: Current Approach and Future Perspectives in Drug Delivery Systems for Bone Targeted Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E875. [PMID: 32370009 PMCID: PMC7279399 DOI: 10.3390/nano10050875] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/14/2022]
Abstract
Bone diseases include a wide group of skeletal-related disorders that cause mobility limitations and mortality. In some cases, e.g., in osteosarcoma (OS) and metastatic bone cancer, current treatments are not fully effective, mainly due to low patient compliance and to adverse side effects. To overcome these drawbacks, nanotechnology is currently under study as a potential strategy allowing specific drug release kinetics and enhancing bone regeneration. Polymers, ceramics, semiconductors, metals, and self-assembled molecular complexes are some of the most used nanoscale materials, although in most cases their surface properties need to be tuned by chemical or physical reactions. Among all, scaffolds, nanoparticles (NPs), cements, and hydrogels exhibit more advantages than drawbacks when compared to other nanosystems and are therefore the object of several studies. The aim of this review is to provide information about the current therapies of different bone diseases focusing the attention on new discoveries in the field of targeted delivery systems. The authors hope that this paper could help to pursue further directions about bone targeted nanosystems and their application for bone diseases and bone regeneration.
Collapse
Affiliation(s)
- Giulia Chindamo
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (G.C.); (E.P.); (D.C.); (M.G.)
| | - Simona Sapino
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (G.C.); (E.P.); (D.C.); (M.G.)
| | - Elena Peira
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (G.C.); (E.P.); (D.C.); (M.G.)
| | - Daniela Chirio
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (G.C.); (E.P.); (D.C.); (M.G.)
| | - Mónica Cristina Gonzalez
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina;
| | - Marina Gallarate
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (G.C.); (E.P.); (D.C.); (M.G.)
| |
Collapse
|
28
|
Induced Pluripotent Stem Cells in Dental and Nondental Tissue Regeneration: A Review of an Unexploited Potential. Stem Cells Int 2020; 2020:1941629. [PMID: 32300365 PMCID: PMC7146092 DOI: 10.1155/2020/1941629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Cell-based therapies currently represent the state of art for tissue regenerative treatment approaches for various diseases and disorders. Induced pluripotent stem cells (iPSCs), reprogrammed from adult somatic cells, using vectors carrying definite transcription factors, have manifested a breakthrough in regenerative medicine, relying on their pluripotent nature and ease of generation in large amounts from various dental and nondental tissues. In addition to their potential applications in regenerative medicine and dentistry, iPSCs can also be used in disease modeling and drug testing for personalized medicine. The current review discusses various techniques for the production of iPSC-derived osteogenic and odontogenic progenitors, the therapeutic applications of iPSCs, and their regenerative potential in vivo and in vitro. Through the present review, we aim to explore the potential applications of iPSCs in dental and nondental tissue regeneration and to highlight different protocols used for the generation of different tissues and cell lines from iPSCs.
Collapse
|
29
|
Soares DG, Bordini EAF, Cassiano FB, Bronze-Uhle ES, Pacheco LE, Zabeo G, Hebling J, Lisboa-Filho PN, Bottino MC, de Souza Costa CA. Characterization of novel calcium hydroxide-mediated highly porous chitosan-calcium scaffolds for potential application in dentin tissue engineering. J Biomed Mater Res B Appl Biomater 2020; 108:2546-2559. [PMID: 32061059 DOI: 10.1002/jbm.b.34586] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022]
Abstract
The aim of this study was to develop a highly porous calcium-containing chitosan scaffold suitable for dentin regeneration. A calcium hydroxide (Ca[OH]2 ) suspension was used to modulate the degree of porosity and chemical composition of chitosan scaffolds. The chitosan solution concentration and freezing protocol were adjusted to optimize the porous architecture using the phase-separation technique. Scanning electron microscopy/energy-dispersive spectroscopy demonstrated the fabrication of a highly porous calcium-linked chitosan scaffold (CH-Ca), with a well-organized and interconnected porous network. Scaffolds were cross-linked on glutaraldehyde (GA) vapor. Following a 28-day incubation in water, cross-linked CH scaffold had no changes on humid mass, and CH-Ca featured a controlled degradability profile since the significant humid mass loss was observed only after 21 (26.0%) and 28 days (42.2%). Fourier-transform infrared spectroscopy indicated the establishment of Schiff base on cross-linked scaffolds, along with calcium complexation for CH-Ca. Cross-linked CH-Ca scaffold featured a sustained Ca2+ release up to 21 days in a humid environment. This porous and stable architecture allowed for human dental pulp cells (HDPCs) to spread throughout the scaffold, with cells exhibiting a widely stretched cytoplasm; whereas, the cells seeded onto CH scaffold were organized in clusters. HDPCs seeded onto CH-Ca featured significantly higher ALP activity, and gene expressions for ALP, Col1, DMP-1, and DSPP in comparison to CH, leading to a significant 3.5 times increase in calcium-rich matrix deposition. In sum, our findings suggest that CH-Ca scaffolds are attractive candidates for creating a highly porous and bioactive substrate for dentin tissue engineering.
Collapse
Affiliation(s)
- Diana Gabriela Soares
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, Sao Paulo University - USP, Bauru, SP, Brazil
| | - Ester Alves Ferreira Bordini
- Department of Physiology and Pathology, Araraquara School of Dentistry, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil
| | - Fernanda Balestrero Cassiano
- Department of Physiology and Pathology, Araraquara School of Dentistry, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil
| | - Erika Soares Bronze-Uhle
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, Sao Paulo University - USP, Bauru, SP, Brazil
| | - Leandro Edgar Pacheco
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, Sao Paulo University - USP, Bauru, SP, Brazil
| | - Giovana Zabeo
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, Sao Paulo University - USP, Bauru, SP, Brazil
| | - Josimeri Hebling
- Department of Orthodontics and Pediatric Dentistry, Araraquara School of Dentistry, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil
| | | | - Marco Cicero Bottino
- Department of Cariology, Restorative Sciences, Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan
| | - Carlos Alberto de Souza Costa
- Department of Physiology and Pathology, Araraquara School of Dentistry, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil
| |
Collapse
|
30
|
Zhao Z, Liu J, Schneider A, Gao X, Ren K, Weir MD, Zhang N, Zhang K, Zhang L, Bai Y, Xu HHK. Human periodontal ligament stem cell seeding on calcium phosphate cement scaffold delivering metformin for bone tissue engineering. J Dent 2019; 91:103220. [PMID: 31678476 DOI: 10.1016/j.jdent.2019.103220] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/16/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES (1) develop a CPC-metformin scaffold with hPDLSC seeding for bone tissue engineering; and (2) investigate the effects of CPC-metformin scaffold on hPDLSC proliferation, osteogenic differentiation and bone matrix mineralization for the first time. METHODS hPDLSCs were harvested from extracted teeth. CPC scaffolds (with or without metformin) were prepared. Three groups were tested: (1) control group (growth medium); (2) osteogenic group (osteogenic medium); (3) metformin + osteogenic group (CPC-metformin scaffold, cultured in osteogenic medium). hPDLSC viability, osteogenic differentiation and mineralization were measured. SEM was used to examine cell morphology. RESULTS After culturing for 14 days, all three groups demonstrated excellent hPDLSC attachment and viability, as shown in live-dead staining, CCK-8 assay, and SEM examinations. The osteogenic group had 3-8 folds, 5 folds and 6 folds of increases in osteogenic gene expressions, ALP activity and mineral synthesis, compared to control group. Furthermore, the metformin + osteogenic group had 3-fold to 4-fold increases over those of the osteogenic group in osteogenic gene expressions, ALP activity and mineral synthesis. CONCLUSIONS hPDLSCs were demonstrated to be a potent cell source for bone engineering. The novel CPC-metformin-hPDLSC construct is highly promising to enhance bone repair and regeneration efficacy in dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jin Liu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Key Laboratory of Shanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, China
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Xianling Gao
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Department of Endodontics, Guanghua School and Hospital of Stomatology & Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, & Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Ning Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
31
|
Xia Y, Guo Y, Yang Z, Chen H, Ren K, Weir MD, Chow LC, Reynolds MA, Zhang F, Gu N, Xu HHK. Iron oxide nanoparticle-calcium phosphate cement enhanced the osteogenic activities of stem cells through WNT/β-catenin signaling. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109955. [PMID: 31500064 DOI: 10.1016/j.msec.2019.109955] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 06/15/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
Abstract
Calcium phosphate cement (CPC), functionalized with iron oxide nanoparticles (IONP), is of great promise to promote osteoinduction and new bone formation. In this work, the IONP powder was added into the CPC powder to fabricate CPC + IONP scaffolds and the effects of the novel composite on bone matrix formation and osteogenesis of human dental pulp stem cells (hDPSCs) were explored. A series of CPC + IONP magnetic scaffolds with different IONP contents (1%, 3% and 6%) were fabricated using 5% chitosan solution as the cement liquid. Western blotting and RT-PCR were used to analyze the signaling pathway. The IONP incorporation substantially enhanced the performance of CPC + IONP, with increases in both mechanical strength and cellular activities. The IONP addition greatly promoted the osteogenesis of hDPSCs, elevating the ALP activity, the expression of osteogenic marker genes and bone matrix formation with 1.5-2-fold increases. The 3% IONP incorporation showed the most enhancement among all groups. Activation of the extracellular signal-related kinases WNT/β-catenin in DPSCs was observed, and this activation was attenuated by the WNT inhibitor DKK1. The results indicated that the osteogenic behavior of hDPSCs was likely driven by CPC + IONP via the WNT signaling pathway. In conclusion, incorporate IONP into CPC scaffold remarkably enhanced the spreading, osteogenic differentiation and bone mineral synthesis of stem cell. Therefore, this method had great potential for bone tissue engineering. The novel CPC + IONP composite scaffolds with stem cells are promising to provide an innovative strategy to enhance bone regenerative therapies.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, USA
| | - Yu Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zukun Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Huimin Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ke Ren
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, USA
| | - Laurence C Chow
- Volpe Research Center, American Dental Association Foundation, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, USA
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greene Baum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
32
|
Wang S, Xia Y, Ma T, Weir MD, Ren K, Reynolds MA, Shu Y, Cheng L, Schneider A, Xu HHK. Novel metformin-containing resin promotes odontogenic differentiation and mineral synthesis of dental pulp stem cells. Drug Deliv Transl Res 2019; 9:85-96. [PMID: 30465181 DOI: 10.1007/s13346-018-00600-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This represents the first report on the development of metformin-containing dental resins. The objectives were to use the resin as a carrier to deliver metformin locally to stimulate dental cells for dental tissue regeneration and to investigate the effects on odontogenic differentiation of dental pulp stem cells (DPSCs) and mineral synthesis. Metformin was incorporated into a resin at 20% by mass as a model system. DPSC proliferation attaching on resins was evaluated. Dentin sialophosphoprotein (DSPP), dentin matrix phosphoprotein 1 (DMP-1), alkaline phosphatase (ALP), and runt-related transcription factor 2 (Runx2) genes expressions were measured. ALP activity and alizarin red staining (ARS) of mineral synthesis by the DPSCs on resins were determined. DPSCs on metformin-containing resin proliferated well (mean ± SD; n = 6), and the number of cells increased by 4-fold from 1 to 14 days (p > 0.1). DSPP, ALP, and DMP-1 gene expressions of DPSCs on metformin resin were much higher than DPSCs on control resin without metformin (p < 0.05). ALP activity of metformin group was 70% higher than that without metformin at 14 days (p < 0.05). Mineral synthesis by DPSCs on metformin-containing resin at 21 days was 9-fold that without metformin (p < 0.05). A novel metformin-containing resin was developed, achieving substantial enhancement of odontoblastic differentiation of DPSCs and greater mineral synthesis. The metformin resin is promising for deep cavities and perforated cavities to stimulate DPSCs for tertiary dentin formation, for tooth root coatings with metformin release for periodontal regeneration, and for root canal fillings with apical lesions to stimulate bone regeneration.
Collapse
Affiliation(s)
- Suping Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral, Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Yang Xia
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA.,Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral, Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China. .,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA.
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA. .,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
33
|
Liu J, Ruan J, Weir MD, Ren K, Schneider A, Wang P, Oates TW, Chang X, Xu HHK. Periodontal Bone-Ligament-Cementum Regeneration via Scaffolds and Stem Cells. Cells 2019; 8:E537. [PMID: 31167434 PMCID: PMC6628570 DOI: 10.3390/cells8060537] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Periodontitis is a prevalent infectious disease worldwide, causing the damage of periodontal support tissues, which can eventually lead to tooth loss. The goal of periodontal treatment is to control the infections and reconstruct the structure and function of periodontal tissues including cementum, periodontal ligament (PDL) fibers, and bone. The regeneration of these three types of tissues, including the re-formation of the oriented PDL fibers to be attached firmly to the new cementum and alveolar bone, remains a major challenge. This article represents the first systematic review on the cutting-edge researches on the regeneration of all three types of periodontal tissues and the simultaneous regeneration of the entire bone-PDL-cementum complex, via stem cells, bio-printing, gene therapy, and layered bio-mimetic technologies. This article primarily includes bone regeneration; PDL regeneration; cementum regeneration; endogenous cell-homing and host-mobilized stem cells; 3D bio-printing and generation of the oriented PDL fibers; gene therapy-based approaches for periodontal regeneration; regenerating the bone-PDL-cementum complex via layered materials and cells. These novel developments in stem cell technology and bioactive and bio-mimetic scaffolds are highly promising to substantially enhance the periodontal regeneration including both hard and soft tissues, with applicability to other therapies in the oral and maxillofacial region.
Collapse
Affiliation(s)
- Jin Liu
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
- Clinical Research Center of Shannxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Jianping Ruan
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
- Clinical Research Center of Shannxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, & Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA.
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA.
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Ping Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Xiaofeng Chang
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
- Clinical Research Center of Shannxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
34
|
Enhanced efficacy of baicalin-loaded TPGS polymeric micelles against periodontitis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:387-395. [PMID: 31029332 DOI: 10.1016/j.msec.2019.03.103] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 05/14/2018] [Accepted: 03/27/2019] [Indexed: 02/03/2023]
Abstract
As a chronic infectious disease, periodontitis is the main cause of teeth exfoliation due to its severe inflammatory reaction and periodontal tissue destruction. Recent reports have shown that baicalin could inhibit the NF-κB signaling pathway in inflammatory activity of periodontitis, but the efficacy of baicalin is limited due to its poor water solubility. In this work, we report the fabrication and application of baicalin encapsulated D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) polymeric micelles (PMs) through thin-film hydration method. The monodispersed micelles showed a spherical shape in aqueous solution and a prolonged drug-release kinetic. After baicalin was loaded into PMs, cytotoxicity and apoptosis induction were both decreased. The expression of genes (including TNF-α, IL-1β, RANKL and NF-κB) and the phosphorylation level of NF-κB p65 protein in lipopolysaccharide (LPS)-induced rat gingival fibroblasts were also reduced. Further investigation of drug efficacy in a rat periodontal disease model confirmed that the use of baicalin-PMs could reduce the destruction of alveolar bone and gingival fiber. Moreover, the therapeutic effect of baicalin-PMs was significantly better than that of free baicalin. These results suggest that the direct injection of micelles containing water-insoluble drugs may become a simple but effective method for treating periodontitis.
Collapse
|
35
|
Lee E, Ko JY, Kim J, Park JW, Lee S, Im GI. Osteogenesis and angiogenesis are simultaneously enhanced in BMP2-/VEGF-transfected adipose stem cells through activation of the YAP/TAZ signaling pathway. Biomater Sci 2019; 7:4588-4602. [DOI: 10.1039/c9bm01037h] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
While bone has the capability to heal itself, there is a great difficulty in reconstituting large bone defects created by heavy trauma or the resection of malignant tumors.
Collapse
Affiliation(s)
- Eugene Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| | - Ji-Yun Ko
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Juyoung Kim
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Jeong-Won Park
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Songhee Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| |
Collapse
|
36
|
Xia Y, Chen H, Zhao Y, Zhang F, Li X, Wang L, Weir MD, Ma J, Reynolds MA, Gu N, Xu HHK. Novel magnetic calcium phosphate-stem cell construct with magnetic field enhances osteogenic differentiation and bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 98:30-41. [PMID: 30813031 DOI: 10.1016/j.msec.2018.12.120] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 01/09/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (IONPs) are promising bioactive additives to fabricate magnetic scaffolds for bone tissue engineering. To date, there has been no report on osteoinductivity of IONP-incorporated calcium phosphate cement (IONP-CPC) scaffold on stem cells using an exterior static magnetic field (SMF). The objectives of this study were to: (1) develop a novel magnetic IONP-CPC construct for bone tissue engineering, and (2) investigate the effects of IONP-incorporation and SMF application on the proliferation, osteogenic differentiation and bone mineral synthesis of human dental pulp stem cells (hDPSCs) seeded on IONP-CPC scaffold for the first time. The novel magnetic IONP-CPC under SMF enhanced the cellular performance of hDPSCs, yielding greater alkaline phosphatase activities (about 3-fold), increased expressions of osteogenic marker genes, and more cell-synthesized bone minerals (about 2.5-fold), compared to CPC control and nonmagnetic IONP-CPC. In addition, IONP-CPC induced more active osteogenesis than CPC control in rat mandible defects. These results were consistent with the enhanced cellular performance by magnetic IONP in media under SMF. Moreover, nano-aggregates were detected inside the cells by transmission electron microscopy (TEM). Therefore, the enhanced cell performance was attributed to the physical forces generated by the magnetic field together with cell internalization of the released magnetic nanoparticles from IONP-CPC constructs.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Huimin Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yantao Zhao
- Beijing Engineering Research Center of Orthopedic Implants, First Affiliated Hospital of CPLA General Hospital, Beijing 100048, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China
| | - Xiaodong Li
- Department of Oral Medicine, School of Stomatology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lin Wang
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu 215123, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greene Baum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
37
|
Bertucci TB, Dai G. Biomaterial Engineering for Controlling Pluripotent Stem Cell Fate. Stem Cells Int 2018; 2018:9068203. [PMID: 30627175 PMCID: PMC6304878 DOI: 10.1155/2018/9068203] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/11/2018] [Indexed: 01/02/2023] Open
Abstract
Pluripotent stem cells (PSCs) represent an exciting cell source for tissue engineering and regenerative medicine due to their self-renewal and differentiation capacities. The majority of current PSC protocols rely on 2D cultures and soluble factors to guide differentiation; however, many other environmental signals are beginning to be explored using biomaterial platforms. Biomaterials offer new opportunities to engineer the stem cell niches and 3D environments for exploring biophysical and immobilized signaling cues to further our control over stem cell fate. Here, we review the biomaterial platforms that have been engineered to control PSC fate. We explore how altering immobilized biochemical cues and biophysical cues such as dimensionality, stiffness, and topography can enhance our control over stem cell fates. Finally, we highlight biomaterial culture systems that assist in the translation of PSC technologies for clinical applications.
Collapse
Affiliation(s)
- Taylor B Bertucci
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
38
|
Jiao J, Huang J, Zhang Z. Hydrogels based on chitosan in tissue regeneration: How do they work? A mini review. J Appl Polym Sci 2018. [DOI: 10.1002/app.47235] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jiao Jiao
- Neuropsychiatric Institute; Medical School of Southeast University; Nanjing Jiangsu 210009 China
- Department of Neurology; Affiliated ZhongDa Hospital; Nanjing Jiangsu 210009 China
| | - Jinjian Huang
- Lab for Trauma and Surgical Infections, Department of Surgery; Jinling Hospital; Nanjing Jiangsu 210002 China
| | - Zhijun Zhang
- Neuropsychiatric Institute; Medical School of Southeast University; Nanjing Jiangsu 210009 China
- Department of Neurology; Affiliated ZhongDa Hospital; Nanjing Jiangsu 210009 China
| |
Collapse
|
39
|
Fliefel R, Ehrenfeld M, Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J Tissue Eng Regen Med 2018; 12:1780-1797. [DOI: 10.1002/term.2697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Riham Fliefel
- Experimental Surgery and Regenerative Medicine (ExperiMed), Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry; Alexandria University; Alexandria Egypt
| | - Michael Ehrenfeld
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| |
Collapse
|
40
|
Rheological and Mechanical Properties of Thermoresponsive Methylcellulose/Calcium Phosphate-Based Injectable Bone Substitutes. MATERIALS 2018; 11:ma11040604. [PMID: 29662018 PMCID: PMC5951488 DOI: 10.3390/ma11040604] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022]
Abstract
In this study, a novel injectable bone substitute (IBS) was prepared by incorporating a bioceramic powder in a polymeric solution comprising of methylcellulose (MC), gelatin and citric acid. Methylcellulose was utilized as the polymeric matrix due to its thermoresponsive properties and biocompatibility. 2.5 wt % gelatin and 3 wt % citric acid were added to the MC to adjust the rheological properties of the prepared IBS. Then, 0, 20, 30 and 50 wt % of the bioceramic component comprising tetracalcium phosphate/hydroxyapatite (TTCP/HA), dicalcium phosphate dehydrate (DCPD) and calcium sulfate dehydrate (CSD) were added into the prepared polymeric component. The prepared IBS samples had a chewing gum-like consistency. IBS samples were investigated in terms of their chemical structure, rheological characteristics, and mechanical properties. After that, in vitro degradation studies were carried out by measurement of pH and % remaining weight. Viscoelastic characteristics of the samples indicated that all of the prepared IBS were injectable and they hardened at approximately 37 °C. Moreover, with increasing wt % of the bioceramic component, the degradation rate of the samples significantly reduced and the mechanical properties were improved. Therefore, the experimental results indicated that the P50 mix may be a promising candidates to fill bone defects and assist bone recovery for non-load bearing applications.
Collapse
|
41
|
Xia Y, Chen H, Zhang F, Wang L, Chen B, Reynolds MA, Ma J, Schneider A, Gu N, Xu HHK. Injectable calcium phosphate scaffold with iron oxide nanoparticles to enhance osteogenesis via dental pulp stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:423-433. [PMID: 29355052 DOI: 10.1080/21691401.2018.1428813] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Literature search revealed no systematic report on iron oxide nanoparticle-incorporating calcium phosphate cement scaffolds (IONP-CPC). The objectives of this study were to: (1) use γFe2O3 nanoparticles (γIONPs) and αFe2O3 nanoparticles (αIONPs) to develop novel IONP-CPC scaffolds, and (2) investigate human dental pulp stem cells (hDPSCs) seeding on IONP-CPC for bone tissue engineering for the first time. IONP-CPC scaffolds were fabricated. Physiochemical properties of IONP-CPC scaffolds were characterized. hDPSC seeding on scaffolds, cell proliferation, osteogenic differentiation and bone matrix mineral synthesis by cells were measured. Our data demonstrated that the osteogenic differentiation of hDPSCs was markedly enhanced via IONP incorporation into CPC. Substantial increases (about three folds) in ALP activity and osteogenic gene expressions were achieved over those without IONPs. Bone matrix mineral synthesis by the cells was increased by two- to three folds over that without IONPs. The enhanced cellular osteogenesis was attributed to: (1) the surface nanotopography of IONP-CPC scaffold, and (2) the cell internalization of IONPs released from IONP-CPC scaffold. Our results demonstrate that the novel CPC functionalized with IONPs is promising to promote osteoinduction and bone regeneration. In conclusion, it is highly promising to incorporate γIONPs and αIONPs into CPC scaffold for bone tissue engineering, yielding substantially better stem cell attachment, spreading and osteogenic differentiation, and much greater bone mineral synthesis by the seeded cells. Therefore, novel CPC scaffolds containing γIONPs and αIONPs are promising for dental, craniofacial and orthopaedic applications to substantially enhance bone regeneration.
Collapse
Affiliation(s)
- Yang Xia
- a Jiangsu Key Laboratory of Oral Diseases , Nanjing Medical University , Nanjing , China.,b Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering , Southeast University , Nanjing , China.,c Department of Advanced Oral Sciences and Therapeutics , University of Maryland School of Dentistry , Baltimore , MD , USA
| | - Huimin Chen
- a Jiangsu Key Laboratory of Oral Diseases , Nanjing Medical University , Nanjing , China
| | - Feimin Zhang
- a Jiangsu Key Laboratory of Oral Diseases , Nanjing Medical University , Nanjing , China.,d Collaborative Innovation Center of Suzhou Nano Science and Technology , Suzhou , China
| | - Lin Wang
- c Department of Advanced Oral Sciences and Therapeutics , University of Maryland School of Dentistry , Baltimore , MD , USA.,e VIP Integrated Department, School and Hospital of Stomatology , Jilin University , Changchun , China
| | - Bo Chen
- b Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering , Southeast University , Nanjing , China
| | - Mark A Reynolds
- c Department of Advanced Oral Sciences and Therapeutics , University of Maryland School of Dentistry , Baltimore , MD , USA
| | - Junqing Ma
- a Jiangsu Key Laboratory of Oral Diseases , Nanjing Medical University , Nanjing , China
| | - Abraham Schneider
- f Department of Oncology and Diagnostic Sciences , University of Maryland School of Dentistry , Baltimore , MD , USA
| | - Ning Gu
- b Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering , Southeast University , Nanjing , China.,d Collaborative Innovation Center of Suzhou Nano Science and Technology , Suzhou , China
| | - Hockin H K Xu
- c Department of Advanced Oral Sciences and Therapeutics , University of Maryland School of Dentistry , Baltimore , MD , USA.,g Center for Stem Cell Biology and Regenerative Medicine , University of Maryland School of Medicine , Baltimore , MD , USA.,h University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine , Baltimore , MD , USA
| |
Collapse
|
42
|
Ercal P, Pekozer GG, Kose GT. Dental Stem Cells in Bone Tissue Engineering: Current Overview and Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:113-127. [PMID: 29498025 DOI: 10.1007/5584_2018_171] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment of bone that is impaired due to disease, trauma or tumor resection creates a challenge for both clinicians and researchers. Critical size bone defects are conventionally treated with autografts which are associated with risks such as donor site morbidity and limitations like donor shortage. Bone tissue engineering has become a promising area for the management of critical size bone defects by the employment of biocompatible materials and the discovery of novel stem cell sources. Mesenchymal stem cells (MSCs) can be isolated with ease from various dental tissues including dental pulp stem cells, stem cells from apical papilla, dental follicle stem cells, stem cells from human exfoliated deciduous teeth, periodontal ligament stem cells, gingival stem cells and tooth germ derived stem cells. Outcomes of dental MSC mediated bone tissue engineering is explored in various in vivo and in vitro preclinical studies. However, there are still obscurities regarding the mechanisms underlying in MSC mediated bone regeneration and challenges in applications of dental stem cells. In this review, we summarized dental stem cell sources and their characterizations, along with currently used biomaterials for cell delivery and future perspectives for dental MSCs in the field of bone tissue engineering. Further efforts are necessary before moving to clinical trials for future applications.
Collapse
|
43
|
Xia Y, Chen H, Zhang F, Bao C, Weir MD, Reynolds MA, Ma J, Gu N, Xu HHK. Gold nanoparticles in injectable calcium phosphate cement enhance osteogenic differentiation of human dental pulp stem cells. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:35-45. [PMID: 28887211 PMCID: PMC5803751 DOI: 10.1016/j.nano.2017.08.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/18/2017] [Accepted: 08/15/2017] [Indexed: 02/05/2023]
Abstract
In this study, a novel calcium phosphate cement containing gold nanoparticles (GNP-CPC) was developed. Its osteogenic induction ability on human dental pulp stem cells (hDPSCs) was investigated for the first time. The incorporation of GNPs improved hDPSCs behavior on CPC, including better cell adhesion (about 2-fold increase in cell spreading) and proliferation, and enhanced osteogenic differentiation (about 2-3-fold increase at 14 days). GNPs endow CPC with micro-nano-structure, thus improving surface properties for cell adhesion and subsequent behaviors. In addition, GNPs released from GNP-CPC were internalized by hDPSCs, as verified by transmission electron microscopy (TEM), thus enhancing cell functions. The culture media containing GNPs enhanced the cellular activities of hDPSCs. This result was consistent with and supported the osteogenic induction results of GNP-CPC. In conclusion, GNP-CPC significantly enhanced the osteogenic functions of hDPSCs. GNPs are promising to modify CPC with nanotopography and work as bioactive additives thus enhance bone regeneration.
Collapse
Affiliation(s)
- Yang Xia
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Huimin Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chongyun Bao
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Michael D Weir
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Xu HHK, Wang P, Wang L, Bao C, Chen Q, Weir MD, Chow LC, Zhao L, Zhou X, Reynolds MA. Calcium phosphate cements for bone engineering and their biological properties. Bone Res 2017; 5:17056. [PMID: 29354304 PMCID: PMC5764120 DOI: 10.1038/boneres.2017.56] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/13/2017] [Accepted: 08/09/2017] [Indexed: 02/08/2023] Open
Abstract
Calcium phosphate cements (CPCs) are frequently used to repair bone defects. Since their discovery in the 1980s, extensive research has been conducted to improve their properties, and emerging evidence supports their increased application in bone tissue engineering. Much effort has been made to enhance the biological performance of CPCs, including their biocompatibility, osteoconductivity, osteoinductivity, biodegradability, bioactivity, and interactions with cells. This review article focuses on the major recent developments in CPCs, including 3D printing, injectability, stem cell delivery, growth factor and drug delivery, and pre-vascularization of CPC scaffolds via co-culture and tri-culture techniques to enhance angiogenesis and osteogenesis.
Collapse
Affiliation(s)
- Hockin HK Xu
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
- Center for Stem Cell Biology and Regenerative
Medicine, University of Maryland School of Medicine, Baltimore,
MD
21201, USA
- University of Maryland Marlene and Stewart
Greenebaum Cancer Center, University of Maryland School of Medicine,
Baltimore, MD
21201, USA
- Mechanical Engineering Department, University
of Maryland Baltimore County, Baltimore, MD
21250, USA
| | - Ping Wang
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
- State Key Laboratory of Oral Diseases, West
China Hospital of Stomatology, Sichuan University, Chengdu,
Sichuan
610041, China
| | - Lin Wang
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
- VIP Integrated Department, Stomatological
Hospital of Jilin University, Changchun, Jilin
130011, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, West
China Hospital of Stomatology, Sichuan University, Chengdu,
Sichuan
610041, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West
China Hospital of Stomatology, Sichuan University, Chengdu,
Sichuan
610041, China
| | - Michael D Weir
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
| | - Laurence C Chow
- Volpe Research Center, American Dental
Association Foundation, National Institute of Standards & Technology,
Gaithersburg, MD
20899, USA
| | - Liang Zhao
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
- Department of Orthopaedic Surgery, Nanfang
Hospital, Southern Medical University, Guangzhou,
Guangdong
510515, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West
China Hospital of Stomatology, Sichuan University, Chengdu,
Sichuan
610041, China
| | - Mark A Reynolds
- Department of Endodontics, Periodontics and
Prosthodontics, University of Maryland School of Dentistry,
Baltimore, MD
21201, USA
| |
Collapse
|
45
|
Alcohol Inhibits Odontogenic Differentiation of Human Dental Pulp Cells by Activating mTOR Signaling. Stem Cells Int 2017; 2017:8717454. [PMID: 29062364 PMCID: PMC5618757 DOI: 10.1155/2017/8717454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/05/2017] [Accepted: 07/16/2017] [Indexed: 12/19/2022] Open
Abstract
Long-term heavy alcohol consumption could result in a range of health, social, and behavioral problems. People who abuse alcohol are at high risks of seriously having osteopenia, periodontal disease, and compromised oral health. However, the role of ethanol (EtOH) in the biological functions of human dental pulp cells (DPCs) is unknown. Whether EtOH affects the odontoblastic differentiation of DPCs through the mechanistic target of rapamycin (mTOR) remains unexplored. The objective of this study was to investigate the effects of EtOH on DPC differentiation and mineralization. DPCs were isolated and purified from human dental pulps. The proliferation and odontoblastic differentiation of DPCs treated with EtOH were subsequently investigated. Different doses of EtOH were shown to be cytocompatible with DPCs. EtOH significantly activated the mTOR pathway in a dose-dependent manner. In addition, EtOH downregulated the alkaline phosphatase activity, attenuated the mineralized nodule formation, and suppressed the expression of odontoblastic markers including ALP, DSPP, DMP-1, Runx2, and OCN. Moreover, the pretreatment with rapamycin, a specific mTOR inhibitor, markedly reversed the EtOH-induced odontoblastic differentiation and cell mineralization. Our findings show for the first time that EtOH can suppress DPC differentiation and mineralization in a mTOR-dependent manner, indicating that EtOH may be involved in negatively regulating the dental pulp repair.
Collapse
|
46
|
Fu L, Wang Z, Dong S, Cai Y, Ni Y, Zhang T, Wang L, Zhou Y. Bilayer Poly(Lactic-co-glycolic acid)/Nano-Hydroxyapatite Membrane with Barrier Function and Osteogenesis Promotion for Guided Bone Regeneration. MATERIALS 2017; 10:ma10030257. [PMID: 28772618 PMCID: PMC5503363 DOI: 10.3390/ma10030257] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 12/17/2022]
Abstract
Guided bone regeneration (GBR) is one such treatment that reconstructs neo-bone tissue by using a barrier membrane to prevent the invasion of soft tissue and to create a space for guiding new bone growth into the bone defect. Herein, we report a novel functionally graded bilayer membrane (FGBM) for GBR application. To fabricate the novel membrane, the composites of poly(lactic-co-glycolic acid) and nano-hydroxyapatite were prepared by phase inversion for the dense layer and by electrospinning for another porous layer, and their corresponding properties were evaluated including surface morphology, mechanics, degradability, cell barrier function, and in vitro osteogenic bioactivity. The results showed that PLGA with 5% nHA in dense layer could meet the requirement of mechanical strength and have excellent barrier function even on condition of post-degradation. Furthermore, PLGA with 30% nHA in porous layer could achieve the good physical and chemical properties. In addition, 30% nHA incorporation would enhance the in vitro mineralization, and have superior capabilities of cell adhesion, proliferation and differentiation compared to other groups. Therefore, the designed FGBM could potentially serve as a barrier for preferential tissue ingrowth and achieve a desirable therapeutic result for bone tissue regeneration.
Collapse
Affiliation(s)
- Li Fu
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Zhanfeng Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Shujun Dong
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Yan Cai
- Department of Oral Mucositis, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Yuxin Ni
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Tianshou Zhang
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Lin Wang
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Yanmin Zhou
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| |
Collapse
|