1
|
Frączek W, Kotela A, Kotela I, Grodzik M. Nanostructures in Orthopedics: Advancing Diagnostics, Targeted Therapies, and Tissue Regeneration. MATERIALS (BASEL, SWITZERLAND) 2024; 17:6162. [PMID: 39769763 PMCID: PMC11677186 DOI: 10.3390/ma17246162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Nanotechnology, delving into the realm of nanometric structures, stands as a transformative force in orthopedics, reshaping diagnostics, and numerous regenerative interventions. Commencing with diagnostics, this scientific discipline empowers accurate analyses of various diseases and implant stability, heralding an era of unparalleled precision. Acting as carriers for medications, nanomaterials introduce novel therapeutic possibilities, propelling the field towards more targeted and effective treatments. In arthroplasty, nanostructural modifications to implant surfaces not only enhance mechanical properties but also promote superior osteointegration and durability. Simultaneously, nanotechnology propels tissue regeneration, with nanostructured dressings emerging as pivotal elements in accelerating wound healing. As we navigate the frontiers of nanotechnology, ongoing research illuminates promising avenues for further advancements, assuring a future where orthopedic practices are not only personalized but also highly efficient, promising a captivating journey through groundbreaking innovations and tailored patient care.
Collapse
Affiliation(s)
- Wiktoria Frączek
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| | - Andrzej Kotela
- Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland
| | - Ireneusz Kotela
- National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| |
Collapse
|
2
|
Yuan Y, Hu J, Shen L, He L, Zhu Y, Meng D, Jiang Q. Injectable calcium phosphate cement integrated with BMSCs-encapsulated microcapsules for bone tissue regeneration. Biomed Mater 2024; 19:065034. [PMID: 39312953 DOI: 10.1088/1748-605x/ad7e69] [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: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
Injectable calcium phosphate cement (CPC) offers significant benefits for the minimally invasive repair of irregular bone defects. However, the main limitations of CPC, including its deficiency in osteogenic properties and insufficient large porosity, require further investigation and resolution. In this study, alginate-chitosan-alginate (ACA) microcapsules were used to encapsulate and deliver rat bone mesenchymal stem cells (rBMSCs) into CPC paste, while a porous CPC scaffold was established to support cell growth. Our results demonstrated that the ACA cell microcapsules effectively protect the cells and facilitate their transport into the CPC paste, thereby enhancing cell viability post-implantation. Additionally, the ACA + CPC extracts were found to stimulate osteogenic differentiation of rBMSCs. Furthermore, results from a rat cranial parietal bone defect model showed that ACA microcapsules containing exogenous rBMSCs initially improved thein situosteogenic potential of CPC within bone defects, providing multiple sites for bone growth. Over time, the osteogenic potential of the exogenous cells diminishes, yet the pores created by the microcapsules persist in supporting ongoing bone formation by recruiting endogenous cells to the osteogenic sites. In conclusion, the utilization of ACA loaded stem cell microcapsules satisfactorily facilitate osteogenesis and degradation of CPC, making it a promising scaffold for bone defect transplantation.
Collapse
Affiliation(s)
- Yafei Yuan
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Jiangqi Hu
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Lipei Shen
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Lin He
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Yixuan Zhu
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Dan Meng
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| | - Qingsong Jiang
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, People's Republic of China
| |
Collapse
|
3
|
Chen X, Li H, Ma Y, Jiang Y. Calcium Phosphate-Based Nanomaterials: Preparation, Multifunction, and Application for Bone Tissue Engineering. Molecules 2023; 28:4790. [PMID: 37375345 DOI: 10.3390/molecules28124790] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Calcium phosphate is the main inorganic component of bone. Calcium phosphate-based biomaterials have demonstrated great potential in bone tissue engineering due to their superior biocompatibility, pH-responsive degradability, excellent osteoinductivity, and similar components to bone. Calcium phosphate nanomaterials have gained more and more attention for their enhanced bioactivity and better integration with host tissues. Additionally, they can also be easily functionalized with metal ions, bioactive molecules/proteins, as well as therapeutic drugs; thus, calcium phosphate-based biomaterials have been widely used in many other fields, such as drug delivery, cancer therapy, and as nanoprobes in bioimaging. Thus, the preparation methods of calcium phosphate nanomaterials were systematically reviewed, and the multifunction strategies of calcium phosphate-based biomaterials have also been comprehensively summarized. Finally, the applications and perspectives of functionalized calcium phosphate biomaterials in bone tissue engineering, including bone defect repair, bone regeneration, and drug delivery, were illustrated and discussed by presenting typical examples.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, China
| | - Huizhang Li
- Department of Orthopedics, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, China
| | - Yinhua Ma
- Department of Orthopedics, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai 201800, China
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
4
|
Maji K, Pramanik K. Future of encapsulation in regenerative medicine. PRINCIPLES OF BIOMATERIALS ENCAPSULATION : VOLUME TWO 2023:749-772. [DOI: 10.1016/b978-0-12-824345-9.00003-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Yuan Y, Shen L, Liu T, He L, Meng D, Jiang Q. Physicochemical properties of bone marrow mesenchymal stem cells encapsulated in microcapsules combined with calcium phosphate cement and their ectopic bone formation. Front Bioeng Biotechnol 2022; 10:1005954. [PMID: 36277380 PMCID: PMC9582332 DOI: 10.3389/fbioe.2022.1005954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Calcium phosphate bone cement (CPC) serves as an excellent scaffold material for bone tissue engineering owing to its good biocompatibility, injectability, self-setting property and three-dimensional porous structure. However, its clinical use is limited due to the cytotoxic effect of its setting reaction on cells and difficulties in degradation into bone. In this study, bone marrow mesenchymal stem cells (BMSCs) were encapsulated in alginate chitosan alginate (ACA) microcapsules and compounded with calcium phosphate bone cement. Changes in the compressive strength, porosity, injectability and collapsibility of CPC at different volume ratios of microcapsules were evaluated. At a 40% volume ratio of microcapsules, the composite scaffold displayed high porosity and injectability with good collapsibility and compressive strength. Cell live/dead double staining, Cell Counting Kit-8 (CCK-8) assays and scanning electron microscopy were used to detect the viability, proliferation and adhesion of cells after cell microcapsules were combined with CPC. The results revealed that cells protected by microcapsules proliferated and adhered better than those that were directly combined with CPC paste, and cell microcapsules could effectively form macropores in scaffold material. The composite was subsequently implanted subcutaneously on the backs of nude mice, and ectopic osteogenesis of the scaffold was detected via haematoxylin-eosin (H&E), Masson’s trichrome and Goldner’s trichrome staining. CPC clearly displayed better new bone formation function and degradability after addition of pure microcapsules and cell microcapsules. Furthermore, the cell microcapsule treatment group showed greater osteogenesis than the pure microcapsule group. Collectively, these results indicate that BMSCs encapsulated in ACA microcapsules combined with CPC composite scaffolds have good application prospects as bone tissue engineering materials.
Collapse
Affiliation(s)
- Yafei Yuan
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Lipei Shen
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Tiankun Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Lin He
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Dan Meng
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Qingsong Jiang
- Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Metformin Facilitates Osteoblastic Differentiation and M2 Macrophage Polarization by PI3K/AKT/mTOR Pathway in Human Umbilical Cord Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:9498876. [PMID: 35761829 PMCID: PMC9233575 DOI: 10.1155/2022/9498876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are the most promising multipotent stem cells that can differentiate into osteoblasts, chondrocytes, and adipocytes. This cellular flexibility contributes to widespread clinical use of MSCs in tissue repair and regeneration. The immune system is a key player in regulating bone remodeling. In recent years, the association between the immune system and bone metabolism has become an increasing focus of interest. Metformin, a glucose-lowering drug, exerts powerful impact on metabolic signaling. However, whether metformin can modulate bone metabolism or whether metformin can influence immune milieu by regulation of macrophages has not been thoroughly elucidated. Herein, we specifically explored the complex interactions between macrophages and human umbilical cord mesenchymal stem cells (UC-MSCs) in the context of metformin. Our research demonstrated that metformin not only stimulated osteogenesis of UC-MSCs but also influenced the immune system via promoting M2 but reducing M1 macrophages. Mechanically, we found that metformin-treated M2 macrophages possessed more potent osteoinductive capacity in our coculture system. Molecularly, these metformin-stimulated M2 macrophages facilitated osteogenesis via activating the PI3K/AKT/mTOR pathway. As demonstrated by using PI3K-specific inhibitor LY294002, we found that the pathway inhibitor partly reversed osteoinductive activity which was activated by coculture of metformin-treated M2 macrophages. Overall, our novel research illuminated the cooperative and synergistic effects of metformin and M2 macrophages on the dynamic balance of bone metabolism.
Collapse
|
7
|
Polysaccharide hydrogels: Functionalization, construction and served as scaffold for tissue engineering. Carbohydr Polym 2022; 278:118952. [PMID: 34973769 DOI: 10.1016/j.carbpol.2021.118952] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/07/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023]
Abstract
Polysaccharide hydrogels have been widely utilized in tissue engineering. They interact with the organismal environments, modulating the cargos release and realizing of long-term survival and activations of living cells. In this review, the potential strategies for modification of polysaccharides were introduced firstly. It is not only used to functionalize the polysaccharides for the consequent formation of hydrogels, but also used to introduce versatile side groups for the regulation of cell behavior. Then, techniques and underlying mechanisms in inducing the formation of hydrogels by polysaccharides or their derivatives are briefly summarized. Finally, the applications of polysaccharide hydrogels in vivo, mainly focus on the performance for alleviation of foreign-body response (FBR) and as cell scaffolds for tissue regeneration, are exemplified. In addition, the perspectives and challenges for further research are addressed. It aims to provide a comprehensive framework about the potentials and challenges that the polysaccharide hydrogels confronting in tissue engineering.
Collapse
|
8
|
Park SS, Park M, Lee BT. Autologous stromal vascular fraction-loaded hyaluronic acid/gelatin-biphasic calcium phosphate scaffold for bone tissue regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 132:112533. [DOI: 10.1016/j.msec.2021.112533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022]
|
9
|
Bahraminasab M, Janmohammadi M, Arab S, Talebi A, Nooshabadi VT, Koohsarian P, Nourbakhsh MS. Bone Scaffolds: An Incorporation of Biomaterials, Cells, and Biofactors. ACS Biomater Sci Eng 2021; 7:5397-5431. [PMID: 34797061 DOI: 10.1021/acsbiomaterials.1c00920] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Large injuries to bones are still one of the most challenging musculoskeletal problems. Tissue engineering can combine stem cells, scaffold biomaterials, and biofactors to aid in resolving this complication. Therefore, this review aims to provide information on the recent advances made to utilize the potential of biomaterials for making bone scaffolds and the assisted stem cell therapy and use of biofactors for bone tissue engineering. The requirements and different types of biomaterials used for making scaffolds are reviewed. Furthermore, the importance of stem cells and biofactors (growth factors and extracellular vesicles) in bone regeneration and their use in bone scaffolds and the key findings are discussed. Lastly, some of the main obstacles in bone tissue engineering and future trends are highlighted.
Collapse
Affiliation(s)
- Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Mahsa Janmohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, Semnan University, Semnan 3513119111, Iran
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Athar Talebi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Parisa Koohsarian
- Department of Biochemistry and Hematology, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | | |
Collapse
|
10
|
Li G, Shen W, Tang X, Mo G, Yao L, Wang J. Combined use of calcium phosphate cement, mesenchymal stem cells and platelet-rich plasma for bone regeneration in critical-size defect of the femoral condyle in mini-pigs. Regen Med 2021; 16:451-464. [PMID: 34030462 DOI: 10.2217/rme-2020-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate the outcome of autologous bone marrow mesenchymal stem cells (BMMSCs) and platelet-rich plasma in combination with calcium phosphate cement (CPC) scaffold to reconstruct femoral critical bone defects in mini-pigs. Materials & methods: Scanning electron microscopy, micro-computed tomography evaluation and quantitative histological assessment were used. Results & conclusion: BMMSCs were attached to the CPC scaffold after 7 days of culture and decreased the residual CPC material in each group at 12 weeks compared with 6 weeks. The newly formed bone area was higher in the CPC+SC+P group than in the CPC group at each time point (all p < 0.05). The strategy of CPC combined with BMMSCs and platelet-rich plasma might be an effective method to repair bone defects.
Collapse
Affiliation(s)
- Guangjun Li
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Wen Shen
- Department of Radiology, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Xing Tang
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Guowei Mo
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Liqin Yao
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Jixing Wang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
11
|
Qiu G, Huang M, Liu J, Wang P, Schneider A, Ren K, Oates TW, Weir MD, Xu HHK, Zhao L. Antibacterial calcium phosphate cement with human periodontal ligament stem cell-microbeads to enhance bone regeneration and combat infection. J Tissue Eng Regen Med 2021; 15:232-243. [PMID: 33434402 DOI: 10.1002/term.3169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/14/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Infectious bone defects remain a significant challenge in orthopedics and dentistry. Calcium phosphate cement (CPC) have attracted significant interest in use as local drug delivery system, which with great potential to control release of antibiotics for the treatment of infectious bone defects. Within the current study, a novel antibacterial scaffold of chitosan-reinforced calcium phosphate cement delivering doxycycline hyclate (CPCC + DOX) was developed. Furthermore, the capacity of CPCC + DOX scaffolds for bone regeneration was enhanced by the human periodontal ligament stem cells (hPDLSCs) encapsulated in alginate beads. CPCC + DOX scaffolds were fabricated to contain different concentrations of DOX. Flexural strength of CPCC + DOX ranged from 5.56 ± 0.70 to 6.2 ± 0.72 MPa, which exceeded the reported strength of cancellous bone. Scaffolds exhibited continual DOX release, reaching 80% at 21 days. Scaffold with 5 mg/ml DOX (CPCC + DOX5mg) had a strong antibacterial effect, with a 4-log colony forming unit reduction against S. aureus and P. gingivalis. The proliferation and osteogenic differentiation of hPDLSCs encapsulated in alginate hydrogel microbeads were investigated in culture with CPCC + DOX scaffolds. CPCC + DOX5mg had no negative effect on proliferation of hPDLSCs. Alkaline phosphatase activity, mineral synthesis, and osteogenic gene expressions for CPCC + DOX5mg group were much higher than control group. DOX did not compromise the osteogenic induction. In summary, the novel CPCC + DOX scaffold exhibited excellent mechanical properties and strong antibacterial activity, while supporting the proliferation and osteogenic differentiation of hPDLSCs. The CPCC + DOX + hPDLSCs construct is promising to enhance bone regeneration and combat bone infections in dental, craniofacial, and orthopedic applications.
Collapse
Affiliation(s)
- Gengtao Qiu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China.,Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Mingguang Huang
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jin Liu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, Maryland, USA.,Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Ping Wang
- Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland, Baltimore, Maryland, USA
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Liang Zhao
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China.,Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
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 2020; 120:111688. [PMID: 33545850 DOI: 10.1016/j.msec.2020.111688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
13
|
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
|
14
|
Garzon I, Chato-Astrain J, Campos F, Fernandez-Valades R, Sanchez-Montesinos I, Campos A, Alaminos M, D'Souza RN, Martin-Piedra MA. Expanded Differentiation Capability of Human Wharton's Jelly Stem Cells Toward Pluripotency: A Systematic Review. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:301-312. [PMID: 32085697 DOI: 10.1089/ten.teb.2019.0257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human Wharton's jelly stem cells (HWJSC) can be efficiently isolated from the umbilical cord, and numerous reports have demonstrated that these cells can differentiate into several cell lineages. This fact, coupled with the high proliferation potential of HWJSC, makes them a promising source of stem cells for use in tissue engineering and regenerative medicine. However, their real potentiality has not been established to date. In the present study, we carried out a systematic review to determine the multilineage differentiation potential of HWJSC. After a systematic literature search, we selected 32 publications focused on the differentiation potential of these cells. Analysis of these studies showed that HWJSC display expanded differentiation potential toward some cell types corresponding to all three embryonic cell layers (ectodermal, mesodermal, and endodermal), which is consistent with their constitutive expression of key pluripotency markers such as OCT4, SOX2, and NANOG, and the embryonic marker SSEA4. We conclude that HWJSC can be considered cells in an intermediate state between multipotentiality and pluripotentiality, since their proliferation capability is not unlimited and differentiation to all cell types has not been demonstrated thus far. These findings support the clinical use of HWJSC for the treatment of diseases affecting not only mesoderm-type tissues but also other cell lineages. Impact statement Human Wharton's jelly stem cells (HWJSC) are mesenchymal stem cells that are easy to isolate and handle, and that readily proliferate. Their wide range of differentiation capabilities supports the view that these cells can be considered pluripotent. Accordingly, HWJSC are one of the most promising cell sources for clinical applications in advanced therapies.
Collapse
Affiliation(s)
- Ingrid Garzon
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| | - Jesus Chato-Astrain
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| | - Fernando Campos
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| | - Ricardo Fernandez-Valades
- ibs.GRANADA, Biohealth Institute, Granada, Spain.,Division of Pediatric Surgery, University of Granada Hospital Complex, Granada, Spain
| | - Indalecio Sanchez-Montesinos
- ibs.GRANADA, Biohealth Institute, Granada, Spain.,Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada, Spain
| | - Antonio Campos
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| | - Rena N D'Souza
- Department of Dentistry, School of Dentistry, University of Utah, Salt Lake City, Utah, USA
| | - Miguel A Martin-Piedra
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Granada, Spain.,ibs.GRANADA, Biohealth Institute, Granada, Spain
| |
Collapse
|
15
|
Zhao Z, Liu J, Weir MD, Zhang N, Zhang L, Xie X, Zhang C, Zhang K, Bai Y, Xu HHK. Human periodontal ligament stem cells on calcium phosphate scaffold delivering platelet lysate to enhance bone regeneration. RSC Adv 2019; 9:41161-41172. [PMID: 35540034 PMCID: PMC9076431 DOI: 10.1039/c9ra08336g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) are promising for tissue engineering applications but have received relatively little attention. Human platelet lysate (HPL) contains a cocktail of growth factors. To date, there has been no report on hPDLSC seeding on scaffolds loaded with HPL. The objectives of this study were to develop a calcium phosphate cement (CPC)-chitosan scaffold loaded with HPL and investigate their effects on hPDLSC viability, osteogenic differentiation and bone mineral synthesis for the first time. hPDLSCs were harvested from extracted human teeth. Scaffolds were formed by mixing CPC powder with a chitosan solution containing HPL. Four groups were tested: CPC-chitosan + 0% HPL (control); CPC-chitosan + 2.66% HPL; CPC-chitosan + 5.31% HPL; CPC-chitosan + 10.63% HPL. Scanning electron microscopy, live/dead staining, CCK-8, qRT-PCR, alkaline phosphatase and bone minerals assay were applied for hPDLSCs on scaffolds. hPDLSCs attached well on CPC-chitosan scaffold. Adding 10.63% HPL into CPC increased cell proliferation and viability (p < 0.05). ALP gene expression of CPC-chitosan + 10.63% HPL was 7-fold that of 0% HPL at 14 days. Runx2, OSX and Coll1 of CPC-chitosan + 10.63% HPL was 2-3 folds those at 0% HPL (p < 0.05). ALP activity of CPC-chitosan + 10.63% HPL was 2-fold that at 0% HPL (p < 0.05). Bone minerals synthesized by hPDLSCs for CPC-chitosan + 10.63% HPL was 3-fold that at 0% HPL (p < 0.05). This study showed that CPC-chitosan scaffold was a promising carrier for HPL delivery, and HPL in CPC exerted excellent promoting effects on hPDLSCs for bone tissue engineering for the first time. The novel hPDLSC-CPC-chitosan-HPL construct has great potential for orthopedic, dental and maxillofacial regenerative 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
| | - 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
| | - Li Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University Beijing China
| | - Xianju Xie
- Department of Orthodontics, School of Stomatology, Capital Medical University Beijing China
| | - Charles Zhang
- 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
| | - 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
|
16
|
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
|
17
|
Bone allografts combined with adipose-derived stem cells in an optimized cell/volume ratio showed enhanced osteogenesis and angiogenesis in a murine femur defect model. J Mol Med (Berl) 2019; 97:1439-1450. [PMID: 31367858 DOI: 10.1007/s00109-019-01822-9] [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: 09/27/2018] [Revised: 07/02/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Critical sized defects, especially in long bones, pose one of the biggest problems in orthopedic surgery. By definition, these defects do not heal without further treatment. Different therapeutic options range from autologous bone grafts, for example, free vascularized bone grafts, to commercially available bone allografts. Disadvantages of these bone allografts are related to reduced osteogenesis, since they are solely composed of cell-free bone matrix. The purpose of this study was to investigate the cell seeding efficiency of human adipose-derived stem cells (hASCs) on human bone allografts in vitro and furthermore analyze these optimized seeded allografts in a critical sized defect model in vivo. Cancellous human bone allografts were colonized with human ASCs in vitro. Cell seeding efficiency was evaluated by Cell Counting Kit-8 assay. Thereafter, optimized hASC-seeded bone scaffolds were examined in a murine femur defect model, stabilized with the MouseExFix system. Subsequently, x-ray analysis and histology were performed. Examination of cell seeding efficiency revealed an optimum starting population of 84,600 cells per 100 mm3 scaffold. In addition, scaffolds seeded with hASCs showed increased osteogenesis compared with controls. Histological analysis revealed increased remodeling and elevated new bone formation within hASC-seeded scaffolds. Moreover, immunohistochemical stainings revealed increased proliferation, osteogenesis, and angiogenesis. In this study, we systemically optimized cell/volume ratio of two promising components of tissue engineering: hASCs and human bone allografts. These findings may serve as a basis for future translational studies. KEY MESSAGES: Bone tissue engineering. Mesenchymal stem cells derived from human adipose tissue (hASCs). Optimal cell/volume ratio of cell-seeded scaffolds. Increased osteogenesis and angiogenesis in vivo.
Collapse
|
18
|
Comparative analysis of lncRNA and mRNA expression profiles between periodontal ligament stem cells and gingival mesenchymal stem cells. Gene 2019; 699:155-164. [PMID: 30876821 DOI: 10.1016/j.gene.2019.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 03/05/2019] [Accepted: 03/11/2019] [Indexed: 02/08/2023]
Abstract
Oral tissue-derived mesenchymal stem cells, such as periodontal ligament stem cells (PDLSCs) and gingival mesenchymal stem cells (GMSCs), possess different biological characteristics, but the molecular mechanism remains unclear, which restricts their application in tissue engineering. Long noncoding RNAs (lncRNAs) are known to be significant regulators of gene expression, but our knowledge about their roles in the regulation of stem cell biological properties is still limited. This study compared the lncRNA and mRNA expression profiles between PDLSCs and GMSCs through microarray analysis, and applied bioinformatics methods to analyze and predict the function and connection of differentially expressed genes, aiming to screen potential key regulators of diverse biological characteristics in PDLSCs and GMSCs. Microarray analysis showed that 2162 lncRNAs and 1347 mRNAs were significantly differentially expressed between PDLSCs and GMSCs. Gene ontology (GO) analysis and pathway analysis indicated that these differentially expressed genes were involved in diverse biological processes and signaling pathways. The gene signal network and pathway relation network predicted some potentially important regulators. The coding-noncoding gene coexpression network (CNC network) revealed many potential lncRNA-mRNA connection pairs that participated in the regulation of biological behaviors. These results stressed the roles of lncRNAs in controlling stem cell biological behaviors and provided guides for molecular mechanistic study of different biological characteristics in PDLSCs and GMSCs.
Collapse
|
19
|
Kang M, Huang J, Zhang L, Wang X, Guo H, He R. [Mechanical properties and effect on osteodifferentiation of induced pluripotent stem cells of chitosan/whisker/calcium phosphate cement composite biomaterial]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 32:959-967. [PMID: 30129324 DOI: 10.7507/1002-1892.201710028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective To investigate the mechanical properties of the novel compound calcium phosphate cement (CPC) biological material as well as the biological activity and osteogenesis effects of induced pluripotent stem cells (iPS) seeding on scaffold and compare their bone regeneration efficacy in cranial defects in rats. Methods Ac- cording to the different scaffold materials, the experiment was divided into 4 groups: pure CPC scaffold group (group A), CPC∶10% wt chitosan as 2∶1 ratio mixed scaffold group (group B), CPC∶10% wt chitosan∶whisker as 2∶1∶1 ratio mixed scaffold group (group C), and CPC∶10% wt chitosan∶whisker as 2∶1∶2 ratio mixed scaffold group (group D). Mechanical properties (bending strength, work-of-fracture, hardness, and modulus of elasticity) of each scaffold were detected. The scaffolds were cultured with fifth generation iPS-mesenchymal stem cells (MSCs), and the absorbance ( A) values of each group were detected at 1, 3, 7, and 14 days by cell counting kit 8 (CCK-8) method; the alkaline phosphatase (ALP) activity, Live/Dead fluorescence staining and quantitative detection, ALP, Runx2, collagen typeⅠ, osteocalcin (OC), and bone morphogenetic protein 2 (BMP-2) gene expressions by RT-PCR were detected at 1, 7, and 14 days; and the alizarin red staining were detected at 1, 7, 14, and 21 days. Twenty-four 3-month-old male Sprague Dawley rats were used to establish the 8 mm-long skull bone defect model, and were randomly divided into 4 groups ( n=6); 4 kinds of scaffold materials were implanted respectively. After 8 weeks, HE staining was used to observe the repair of bone defects and to detect the percentage of new bone volume and the density of neovascularization. Results The bending strength, work-of-fracture, hardness, and modulus of elasticity in groups B, C, and D were significantly higher than those in group A, and in groups C, D than in group B, and in group D than in group C ( P<0.05). CCK-8 assay showed that cell activity gradually increased with the increase of culture time, the A values in groups B, C, and D at 3, 7, 14 days were signifiantly higher than those in group A, and in groups C, D than in group B ( P<0.05), but no significant difference was found between groups C and D ( P>0.05). Live/Dead fluorescence staining showed that the proportion of living cells in groups B, C, and D at 7 and 14 days was significantly higher than that in group A ( P<0.05), and in groups C, D at 7 days than in group B ( P<0.05); but no significant difference was found between groups C and D ( P>0.05). RT-PCR showed that the relative expressions of genes in groups B, C, and D at 7 and 14 days were significantly higher than those in group A, and in groups C, D than in group B ( P<0.05); but no significant difference was found between groups C and D ( P>0.05). Alizarin red staining showed that the red calcium deposition on the surface of scaffolds gradually deepened and thickened with the prolongation of culture time; the A values in groups B, C, and D at 14 and 21 days were significantly higher than those in group A ( P<0.05), and in groups C and D than in group B ( P<0.05), but no significant difference was found between groups C and D ( P>0.05). In vivo repair experiments in animals showed that the new bone in each group was mainly filled with the space of scaffold material. Osteoblasts and neovascularization were surrounded by new bone tissue in the matrix, and osteoblasts were arranged on the new bone boundary. The new bone in groups B, C, and D increased significantly when compared with group A, and the new bone in groups C and D was significantly higher than that in group B. The percentage of new bone volume and the density of neovascularization in groups B, C, and D were significantly higher than those in group A, and in groups C and D than in group B ( P<0.05); but no significant difference was found between groups C and D ( P>0.05). Conclusion The mechanical properties of the new reinforced composite scaffold made from composite chitosan, whisker, and CPC are obviously better than that of pure CPC scaffold material, which can meet the mechanical properties of cortical bone and cancellous bone. iPS-MSCs is attaching and proliferating on the new reinforced composite scaffold material, and the repair effect of bone tissue is good. It can meet the biological and osteogenic activity requirements of the implant materials in the bone defect repair.
Collapse
Affiliation(s)
- Ming Kang
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001, P.R.China
| | - Jiehua Huang
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001, P.R.China
| | - Lixuan Zhang
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001, P.R.China
| | - Xinguang Wang
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001, P.R.China
| | - Hanming Guo
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001, P.R.China
| | - Ruixuan He
- Department of Joint Surgery, Huizhou Central People's Hospital, Huizhou Guangdong, 516001,
| |
Collapse
|
20
|
Acri TM, Shin K, Seol D, Laird NZ, Song I, Geary SM, Chakka JL, Martin JA, Salem AK. Tissue Engineering for the Temporomandibular Joint. Adv Healthc Mater 2019; 8:e1801236. [PMID: 30556348 DOI: 10.1002/adhm.201801236] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/17/2018] [Indexed: 12/24/2022]
Abstract
Tissue engineering potentially offers new treatments for disorders of the temporomandibular joint which frequently afflict patients. Damage or disease in this area adversely affects masticatory function and speaking, reducing patients' quality of life. Effective treatment options for patients suffering from severe temporomandibular joint disorders are in high demand because surgical options are restricted to removal of damaged tissue or complete replacement of the joint with prosthetics. Tissue engineering approaches for the temporomandibular joint are a promising alternative to the limited clinical treatment options. However, tissue engineering is still a developing field and only in its formative years for the temporomandibular joint. This review outlines the anatomical and physiological characteristics of the temporomandibular joint, clinical management of temporomandibular joint disorder, and current perspectives in the tissue engineering approach for the temporomandibular joint disorder. The tissue engineering perspectives have been categorized according to the primary structures of the temporomandibular joint: the disc, the mandibular condyle, and the glenoid fossa. In each section, contemporary approaches in cellularization, growth factor selection, and scaffold fabrication strategies are reviewed in detail along with their achievements and challenges.
Collapse
Affiliation(s)
- Timothy M. Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, Iowa 52242 USA
| | - Kyungsup Shin
- Department of Orthodontics; College of Dentistry and Dental Clinics; University of Iowa; Iowa City, Iowa 52242 USA
| | - Dongrim Seol
- Department of Orthopedics and Rehabilitation; Carver College of Medicine; University of Iowa; Iowa City, Iowa 52242 USA
| | - Noah Z. Laird
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, Iowa 52242 USA
| | - Ino Song
- Department of Orthopedics and Rehabilitation; Carver College of Medicine; University of Iowa; Iowa City, Iowa 52242 USA
| | - Sean M. Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, Iowa 52242 USA
| | - Jaidev L. Chakka
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, Iowa 52242 USA
| | - James A. Martin
- Department of Orthopedics and Rehabilitation; Carver College of Medicine; University of Iowa; Iowa City, Iowa 52242 USA
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics; College of Pharmacy; University of Iowa; Iowa City, Iowa 52242 USA
| |
Collapse
|
21
|
Liu T, Li J, Shao Z, Ma K, Zhang Z, Wang B, Zhang Y. Encapsulation of mesenchymal stem cells in chitosan/β-glycerophosphate hydrogel for seeding on a novel calcium phosphate cement scaffold. Med Eng Phys 2018; 56:9-15. [PMID: 29576458 DOI: 10.1016/j.medengphy.2018.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/04/2018] [Accepted: 03/13/2018] [Indexed: 01/09/2023]
Abstract
Due to its moldability, biocompatibility, osteoconductivity and resorbability, calcium phosphate cement (CPC) is a highly promising scaffold material for orthopedic applications. However, pH changes and ionic activity during the CPC setting reaction may adversely affect cells seeded directly on CPC. Moreover, a lack of macropores in CPC limits ingrowth of new bone. The objectives of this study were to prepare macroporous CPC scaffolds via porogen leaching, using mannitol crystals as the porogen and to evaluate the in vitro proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs) encapsulated in chitosan/β-glycerophosphate (C/GP) hydrogel prior to exposure to the novel CPC scaffold. MSCs were found to be adhered to the surfaces of CPC macropores via scanning electron microscopy. The viability and osteogenic differentiation of MSCs in C/GP hydrogel with or without exposure to CPC constructs containing mannitol crystals indicated that coating with C/GP hydrogel protected the cells during cement mixing and setting. In conclusion, novel, macroporous CPC scaffolds were prepared, and our data indicate that a hydrogel encapsulation-based strategy can be used to protect cells during scaffold formation. Thus, the MSC-laden CPC scaffolds show promise for the delivery of stem cells to promote bone regeneration.
Collapse
Affiliation(s)
- Tao Liu
- The 457th Hospital of PLA, Wuhan, 430012, Hubei, China
| | - Jian Li
- The 457th Hospital of PLA, Wuhan, 430012, Hubei, China
| | - Zengwu Shao
- Institute of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Kaige Ma
- Institute of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhicai Zhang
- Institute of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Baichuan Wang
- Institute of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yannan Zhang
- Institute of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| |
Collapse
|
22
|
Self-Setting Calcium Orthophosphate (CaPO4) Formulations. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2018. [DOI: 10.1007/978-981-10-5975-9_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Bijan Nejad D, Azandeh S, Habibi R, Mansouri E, Bayati V, Ahmadi Angali K. Investigation of the role of alginate containing high guluronic acid on osteogenic differentiation capacity of human umbilical cord Wharton’s jelly mesenchymal stem cells. J Microencapsul 2017; 34:732-743. [DOI: 10.1080/02652048.2017.1393115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Darioush Bijan Nejad
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Saeed Azandeh
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Rezvan Habibi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Kambiz Ahmadi Angali
- Department of Statistics and Epidemiology, Faculty of Public Health, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| |
Collapse
|
24
|
Enhanced bone formation in sheep vertebral bodies after minimally invasive treatment with a novel, PLGA fiber-reinforced brushite cement. Spine J 2017; 17:709-719. [PMID: 27871820 DOI: 10.1016/j.spinee.2016.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/21/2016] [Accepted: 11/09/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Injectable, brushite-forming calcium phosphate cements (CPC) show potential for bone replacement, but they exhibit low mechanical strength. This study tested a CPC reinforced with poly(l-lactide-co-glycolide) acid (PLGA) fibers in a minimally invasive, sheep lumbar vertebroplasty model. PURPOSE The study aimed to test the in vivo biocompatibility and osteogenic potential of a PLGA fiber-reinforced, brushite-forming CPC in a sheep large animal model. STUDY DESIGN/SETTING This is a prospective experimental animal study. METHODS Bone defects (diameter: 5 mm) were placed in aged, osteopenic female sheep, and left empty (L2) or injected with pure CPC (L3) or PLGA fiber-reinforced CPC (L4; fiber diameter: 25 µm; length: 1 mm; 10% [wt/wt]). Three and 9 months postoperation (n=20 each), the structural and functional CPC effects on bone regeneration were documented ex vivo by osteodensitometry, histomorphometry, micro-computed tomography (micro-CT), and biomechanical testing. RESULTS Addition of PLGA fibers enhanced CPC osteoconductivity and augmented bone formation. This was demonstrated by (1) significantly enhanced structural (bone volume/total volume, shown by micro-CT and histomorphometry; 3 or 9 months) and bone formation parameters (osteoid volume and osteoid surface; 9 months); (2) numerically enhanced bone mineral density (3 and 9 months) and biomechanical compression strength (9 months); and (3) numerically decreased bone erosion (eroded surface; 3 and 9 months). CONCLUSIONS The PLGA fiber-reinforced CPC is highly biocompatible and its PLGA fiber component enhanced bone formation. Also, PLGA fibers improve the mechanical properties of brittle CPC, with potential applicability in load-bearing areas.
Collapse
|
25
|
Minipig-BMSCs Combined with a Self-Setting Calcium Phosphate Paste for Bone Tissue Engineering. Mol Biotechnol 2017; 58:748-756. [PMID: 27683256 DOI: 10.1007/s12033-016-9974-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium phosphate cements (CPCs) are a new generation of bone repair materials with good biocompatibility for various stem cells. The minipig is a recommended large animal model for bone engineering research. This study aimed to evaluate the feasibility of utilizing CPC scaffolds for the adhesion, proliferation, and osteogenic differentiation of minipig's bone marrow mesenchymal stem cells (pBMSCs). Passage 3 pBMSCs were seeded on the CPC scaffold and cultured with osteogenic culture medium (osteogenic group) or normal medium (control group). The density of viable cells increased in both groups, and pBMSCs firmly attached and spread well on the CPC scaffold. The alkaline phosphatase (ALP) activity in the osteogenic group had significantly increased on day 7 (D7) and peaked on D14. qRT-PCR revealed that mRNA levels of ALP and three osteogenic marker genes were significantly higher on D4, D7, and D14 in the osteogenic group. Alizarin Red S staining showed a significantly higher degree of bone mineralization from D7, D14 to D21 in the osteogenic group. These results indicated that pBMSCs can attach, proliferate well on CPC scaffold, and be successfully induced to differentiate into osteogenic cells. Our findings may be helpful for bone tissue engineering and the studies of bone regeneration.
Collapse
|
26
|
Wang L, Wang P, Weir MD, Reynolds MA, Zhao L, Xu HHK. Hydrogel fibers encapsulating human stem cells in an injectable calcium phosphate scaffold for bone tissue engineering. ACTA ACUST UNITED AC 2016; 11:065008. [PMID: 27811389 DOI: 10.1088/1748-6041/11/6/065008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs), human embryonic stem cells (hESCs) and human umbilical cord mesenchymal stem cells (hUCMSCs) are exciting cell sources for use in regenerative medicine. There have been no reports on long hydrogel fibers encapsulating stem cells inside an injectable calcium phosphate cement (CPC) scaffold for bone tissue engineering. The objectives of this study were: (1) to develop a novel injectable CPC construct containing hydrogel fibers encapsulating cells for bone engineering, and (2) to investigate and compare cell viability, proliferation and osteogenic differentiation of hiPSC-MSCs, hESC-MSCs and hUCMSCs in injectable CPC. The pastes encapsulating the stem cells were fully injectable under a small injection force, and the injection did not harm the cells, compared with non-injected cells (p > 0.1). The mechanical properties of the stem cell-CPC construct were much better than those of previous injectable polymers and hydrogels for cell delivery. The hiPSC-MSCs, hESC-MSCs and hUCMSCs in hydrogel fibers in CPC had excellent proliferation and osteogenic differentiation. All three cell types yielded high alkaline phosphatase, runt-related transcription factor, collagen I and osteocalcin expression (mean ± SD; n = 6). Cell-synthesized minerals increased substantially with time (p < 0.05), with no significant difference among the three types of cells (p > 0.1). Mineralization by hiPSC-MSCs, hESC-MSCs and hUCMSCs in CPC at 14 d was 13-fold that at 1 d. In conclusion, all three types of cells (hiPSC-MSCs, hESC-MSCs and hUCMSCs) in a CPC scaffold showed high potential for bone tissue engineering, and the novel injectable CPC construct with cell-encapsulating hydrogel fibers is promising for enhancing bone regeneration in dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Lin Wang
- VIP Integrated Department, Stomatological Hospital of Jilin University, Changchun, Jilin 130011, People's Republic of China. Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
27
|
Wang L, Zhang C, Li C, Weir MD, Wang P, Reynolds MA, Zhao L, Xu HHK. Injectable calcium phosphate with hydrogel fibers encapsulating induced pluripotent, dental pulp and bone marrow stem cells for bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1125-36. [PMID: 27612810 DOI: 10.1016/j.msec.2016.08.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/29/2016] [Accepted: 08/07/2016] [Indexed: 12/21/2022]
Abstract
Human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs), dental pulp stem cells (hDPSCs) and bone marrow MSCs (hBMSCs) are exciting cell sources in regenerative medicine. However, there has been no report comparing hDPSCs, hBMSCs and hiPSC-MSCs for bone engineering in an injectable calcium phosphate cement (CPC) scaffold. The objectives of this study were to: (1) develop a novel injectable CPC containing hydrogel fibers encapsulating stem cells for bone engineering, and (2) compare cell viability, proliferation and osteogenic differentiation of hDPSCs, hiPSC-MSCs from bone marrow (BM-hiPSC-MSCs) and from foreskin (FS-hiPSC-MSCs), and hBMSCs in CPC for the first time. The results showed that the injection did not harm cell viability. The porosity of injectable CPC was 62%. All four types of cells proliferated and differentiated down the osteogenic lineage inside hydrogel fibers in CPC. hDPSCs, BM-hiPSC-MSCs, and hBMSCs exhibited high alkaline phosphatase, runt-related transcription factor, collagen I, and osteocalcin gene expressions. Cell-synthesized minerals increased with time (p<0.05), with no significant difference among hDPSCs, BM-hiPSC-MSCs and hBMSCs (p>0.1). Mineralization by hDPSCs, BM-hiPSC-MSCs, and hBMSCs inside CPC at 14d was 14-fold that at 1d. FS-hiPSC-MSCs were inferior in osteogenic differentiation compared to the other cells. In conclusion, hDPSCs, BM-hiPSC-MSCs and hBMSCs are similarly and highly promising for bone tissue engineering; however, FS-hiPSC-MSCs were relatively inferior in osteogenesis. The novel injectable CPC with cell-encapsulating hydrogel fibers may enhance bone regeneration in dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Lin Wang
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130011,China; Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Chi Zhang
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chunyan Li
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130011,China
| | - Michael D Weir
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ping Wang
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA.
| | - Mark A Reynolds
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, 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.
| | - Hockin H K Xu
- Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Mechanical Engineering, University of Maryland Baltimore County, Baltimore County, MD 21250, USA
| |
Collapse
|
28
|
Metoki N, Sadman K, Shull K, Eliaz N, Mandler D. Electro-Assisted Deposition of Calcium Phosphate on Self-Assembled Monolayers. Electrochim Acta 2016. [DOI: 10.1016/j.electacta.2016.04.143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Mussel-inspired alginate gel promoting the osteogenic differentiation of mesenchymal stem cells and anti-infection. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:496-504. [PMID: 27612740 DOI: 10.1016/j.msec.2016.06.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 05/30/2016] [Accepted: 06/13/2016] [Indexed: 11/23/2022]
Abstract
Alginate hydrogels have been used in cell encapsulation for many years but a prevalent issue with pure alginates is that they are unable to provide enough bioactive properties to interact with mammalian cells. This paper discusses the modification of alginate with mussel-inspired dopamine for cell loading and anti-infection. Mouse bone marrow stem cells were immobilized into alginate and alginate-dopamine beads and fibers. Through live-dead and MTT assay, alginates modified by dopamine promoted cell viability and proliferation. In vitro cell differentiation results showed that such an alginate-dopamine gel can promote the osteogenic differentiation of mesenchymal stem cell after PCR and ALP assays. In addition to that, the adhesive prosperities of dopamine allowed for coating the surface of alginate-dopamine gel with silver nanoparticles, which provided the gel with significant antibacterial characteristics. Overall, these results demonstrate that a dopamine-modified alginate gel can be a great tool for cell encapsulation to promote cell proliferation and can be applied to bone regeneration, especially in contaminated bone defects.
Collapse
|
30
|
Current View on Osteogenic Differentiation Potential of Mesenchymal Stromal Cells Derived from Placental Tissues. Stem Cell Rev Rep 2016; 11:570-85. [PMID: 25381565 PMCID: PMC4493719 DOI: 10.1007/s12015-014-9569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSC) isolated from human term placental tissues possess unique characteristics, including their peculiar immunomodulatory properties and their multilineage differentiation potential. The osteogenic differentiation capacity of placental MSC has been widely disputed, and continues to be an issue of debate. This review will briefly discuss the different MSC populations which can be obtained from different regions of human term placenta, along with their unique properties, focusing specifically on their osteogenic differentiation potential. We will present the strategies used to enhance osteogenic differentiation potential in vitro, such as through the selection of subpopulations more prone to differentiate, the modification of the components of osteo-inductive medium, and even mechanical stimulation. Accordingly, the applications of three-dimensional environments in vitro and in vivo, such as non-synthetic, polymer-based, and ceramic scaffolds, will also be discussed, along with results obtained from pre-clinical studies of placental MSC for the regeneration of bone defects and treatment of bone-related diseases.
Collapse
|
31
|
Maenz S, Hennig M, Mühlstädt M, Kunisch E, Bungartz M, Brinkmann O, Bossert J, Kinne RW, Jandt KD. Effects of oxygen plasma treatment on interfacial shear strength and post-peak residual strength of a PLGA fiber-reinforced brushite cement. J Mech Behav Biomed Mater 2016; 57:347-58. [DOI: 10.1016/j.jmbbm.2016.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 02/01/2023]
|
32
|
Moon HJ, Patel M, Chung H, Jeong B. Nanocomposite versus Mesocomposite for Osteogenic Differentiation of Tonsil-Derived Mesenchymal Stem Cells. Adv Healthc Mater 2016; 5:353-63. [PMID: 26634888 DOI: 10.1002/adhm.201500558] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/24/2015] [Indexed: 11/08/2022]
Abstract
Injectable inorganic/organic composite systems consisting of well-defined mesocrystals (4-8 μm) of calcium phosphate and polypeptide thermogel significantly enhance the osteogenic differentiation of the tonsil derived mesenchymal stem cells (TMSCs). Compared to composite systems incorporating nanoparticles (10-100 nm) or pure hydrogel systems, osteogenic biomarkers including alkaline phosphatase (ALP), bone morphogenetic protein 2, and osteocalcin are highly expressed at both the mRNA level and the protein level in the mesocrystal composite systems. ALP activity of differentiated cells is also significantly higher in the mesocomposite systems compared to the nanocomposite systems or the pure hydrogel systems. The mesocomposite systems provide not only hard surfaces for binding the cells/proteins by the inorganic mesocrystals but also a soft matrix for holding the cells by the hydrogel. Through the current research, (1) a novel method of preparing mesocrystals is developed, (2) TMSCs are proved as a new resource of stem cells, and (3) the mesocomposite systems are proved to be a promising tool in controlling stem cell differentiation. (4) Finally, the research emphasizes the significance of mesoscience as a new perspective of science in controlling cell and material interfaces.
Collapse
Affiliation(s)
- Hyo Jung Moon
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-guSeoul 120-750 South Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-guSeoul 120-750 South Korea
| | - Heejung Chung
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-guSeoul 120-750 South Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-guSeoul 120-750 South Korea
| |
Collapse
|
33
|
Saltz A, Kandalam U. Mesenchymal stem cells and alginate microcarriers for craniofacial bone tissue engineering: A review. J Biomed Mater Res A 2016; 104:1276-84. [PMID: 26826060 DOI: 10.1002/jbm.a.35647] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 12/29/2015] [Accepted: 01/07/2016] [Indexed: 01/01/2023]
Abstract
Craniofacial bone is a complex structure with an intricate anatomical and physiological architecture. The defects that exist in this region therefore require a precise control of osteogenesis in their reconstruction. Unlike traditional surgical intervention, tissue engineering techniques mediate bone development with limited postoperative risk and cost. Alginate stands as the premier polymer in bone repair because of its mild ionotropic gelation and excellent biocompatibility, biodegradability, and injectability. Alginate microcarriers are candidates of choice to mediate cells and accommodate into 3-D environment. Several studies reported the use of alginate microcarriers for delivering cells, drugs, and growth factors. This review will explore the potential use of alginate microcarrier for stem cell systems and its application in craniofacial bone tissue engineering.
Collapse
Affiliation(s)
- Adam Saltz
- Nova Southeastern University College of Dental Medicine, Fort Lauderdale, Florida, 33328
| | - Umadevi Kandalam
- Nova Southeastern University College of Dental Medicine, Fort Lauderdale, Florida, 33328
| |
Collapse
|
34
|
Hu J, Seeberger PH, Yin J. Using carbohydrate-based biomaterials as scaffolds to control human stem cell fate. Org Biomol Chem 2016; 14:8648-58. [DOI: 10.1039/c6ob01124a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review describes the current state and applications of several important and extensively studied natural polysaccharide and glycoprotein scaffolds that can control the stem cell fate.
Collapse
Affiliation(s)
- Jing Hu
- Wuxi Medical School
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education
- School of Biotechnology
- Jiangnan University
- Wuxi 214122
| | - Peter H. Seeberger
- Department of Biomolecular Systems
- Max Planck Institute of Colloids and Interfaces
- 14476 Potsdam
- Germany
| | - Jian Yin
- Wuxi Medical School
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education
- School of Biotechnology
- Jiangnan University
- Wuxi 214122
| |
Collapse
|
35
|
Hashemi M, Kalalinia F. Application of encapsulation technology in stem cell therapy. Life Sci 2015; 143:139-46. [DOI: 10.1016/j.lfs.2015.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/15/2015] [Accepted: 11/06/2015] [Indexed: 11/26/2022]
|
36
|
Sun H, Yang HL. Calcium phosphate scaffolds combined with bone morphogenetic proteins or mesenchymal stem cells in bone tissue engineering. Chin Med J (Engl) 2015; 128:1121-7. [PMID: 25881610 PMCID: PMC4832956 DOI: 10.4103/0366-6999.155121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: The purpose of this study was to review the current status of calcium phosphate (CaP) scaffolds combined with bone morphogenetic proteins (BMPs) or mesenchymal stem cells (MSCs) in the field of bone tissue engineering (BTE). Date Sources: Data cited in this review were obtained primarily from PubMed and Medline in publications from 1979 to 2014, with highly regarded older publications also included. The terms BTE, CaP, BMPs, and MSC were used for the literature search. Study Selection: Reviews focused on relevant aspects and original articles reporting in vitro and/or in vivo results concerning the efficiency of CaP/BMPs or CaP/MSCs composites were retrieved, reviewed, analyzed, and summarized. Results: An ideal BTE product contains three elements: Scaffold, growth factors, and stem cells. CaP-based scaffolds are popular because of their outstanding biocompatibility, bioactivity, and osteoconductivity. However, they lack stiffness and osteoinductivity. To solve this problem, composite scaffolds of CaP with BMPs have been developed. New bone formation by CaP/BMP composites can reach levels similar to those of autografts. CaP scaffolds are compatible with MSCs and CaP/MSC composites exhibit excellent osteogenesis and stiffness. In addition, a CaP/MSC/BMP scaffold can repair bone defects more effectively than an autograft. Conclusions: Novel BTE products possess remarkable osteoconduction and osteoinduction capacities, and exhibit balanced degradation with osteogenesis. Further work should yield safe, viable, and efficient materials for the repair of bone lesions.
Collapse
Affiliation(s)
| | - Hui-Lin Yang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Jiangsu 215006, China
| |
Collapse
|
37
|
Qiao PY, Li FF, Dong LM, Xu T, Xie QF. Delivering MC3T3-E1 cells into injectable calcium phosphate cement through alginate-chitosan microcapsules for bone tissue engineering. J Zhejiang Univ Sci B 2015; 15:382-92. [PMID: 24711359 DOI: 10.1631/jzus.b1300132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To deliver cells deep into injectable calcium phosphate cement (CPC) through alginate-chitosan (AC) microcapsules and investigate the biological behavior of the cells released from microcapsules into the CPC. METHODS Mouse osteoblastic MC3T3-E1 cells were embedded in alginate and AC microcapsules using an electrostatic droplet generator. The two types of cell-encapsulating microcapsules were then mixed with a CPC paste. MC3T3-E1 cell viability was investigated using a Wst-8 kit, and osteogenic differentiation was demonstrated by an alkaline phosphatase (ALP) activity assay. Cell attachment in CPC was observed by an environment scanning electron microscopy. RESULTS Both alginate and AC microcapsules were able to release the encapsulated MC3T3-E1 cells when mixed with CPC paste. The released cells attached to the setting CPC scaffolds, survived, differentiated, and formed mineralized nodules. Cells grew in the pores concomitantly created by the AC microcapsules in situ within the CPC. At Day 21, cellular ALP activity in the AC group was approximately four times that at Day 7 and exceeded that of the alginate microcapsule group (P<0.05). Pores formed by the AC microcapsules had a diameter of several hundred microns and were spherical compared with those formed by alginate microcapsules. CONCLUSIONS AC microcapsule is a promising carrier to release seeding cells deep into an injectable CPC scaffold for bone engineering.
Collapse
Affiliation(s)
- Peng-yan Qiao
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; Beijing Key Lab of Fine Ceramics, Institute of Nuclear and New Energy Technology, Tsinghua University, Beijing 100084, China; Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, China; National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | | | | | | | | |
Collapse
|
38
|
Wang KX, Xu LL, Rui YF, Huang S, Lin SE, Xiong JH, Li YH, Lee WYW, Li G. The effects of secretion factors from umbilical cord derived mesenchymal stem cells on osteogenic differentiation of mesenchymal stem cells. PLoS One 2015; 10:e0120593. [PMID: 25799169 PMCID: PMC4370627 DOI: 10.1371/journal.pone.0120593] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/24/2015] [Indexed: 12/11/2022] Open
Abstract
Factors synthesized by mesenchymal stem cells (MSCs) contain various growth factors, cytokines, exosomes and microRNAs, which may affect the differentiation abilities of MSCs. In the present study, we investigated the effects of secretion factors of human umbilical cord derived mesenchymal stem cells (hUCMSCs) on osteogenesis of human bone marrow derived MSCs (hBMSCs). The results showed that 20 μg/ml hUCMSCs secretion factors could initiate osteogenic differentiation of hBMSCs without osteogenic induction medium (OIM), and the amount of calcium deposit (stained by Alizarin Red) was significantly increased after the hUCMSCs secretion factors treatment. Real time quantitative reverse transcription-polymerase chain reaction (real time qRT-PCR) demonstrated that the expression of osteogenesis-related genes including ALP, BMP2, OCN, Osterix, Col1α and Runx2 were significantly up-regulated following hUCMSCs secretion factors treatment. In addition, we found that 10 μg hUCMSCs secretion factors together with 2×10(5) hBMSCs in the HA/TCP scaffolds promoted ectopic bone formation in nude mice. Local application of 10 μg hUCMSCs secretion factors with 50 μl 2% hyaluronic acid hydrogel and 1×10(5) rat bone marrow derived MSCs (rBMSCs) also significantly enhanced the bone repair of rat calvarial bone critical defect model at both 4 weeks and 8 weeks. Moreover, the group that received the hUCMSCs secretion factors treatment had more cartilage and bone regeneration in the defect areas than those in the control group. Taken together, these findings suggested that hUCMSCs secretion factors can initiate osteogenesis of bone marrow MSCs and promote bone repair. Our study indicates that hUCMSCs secretion factors may be potential sources for promoting bone regeneration.
Collapse
Affiliation(s)
- Kui-Xing Wang
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Liang-Liang Xu
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun-Feng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shuo Huang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Si-En Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang-Hui Xiong
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- State Key Laboratory of Space Medical Fundamentation and Application, Astronaut Research and Training Center of China (ACC), 26 Beiqing Road, 100094, Beijing, China
| | - Ying-Hui Li
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- State Key Laboratory of Space Medical Fundamentation and Application, Astronaut Research and Training Center of China (ACC), 26 Beiqing Road, 100094, Beijing, China
| | - Wayne Yuk-Wai Lee
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- * E-mail:
| |
Collapse
|
39
|
Zhang J, Ma X, Lin D, Shi H, Yuan Y, Tang W, Zhou H, Guo H, Qian J, Liu C. Magnesium modification of a calcium phosphate cement alters bone marrow stromal cell behavior via an integrin-mediated mechanism. Biomaterials 2015; 53:251-64. [PMID: 25890724 DOI: 10.1016/j.biomaterials.2015.02.097] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/17/2015] [Accepted: 02/21/2015] [Indexed: 12/24/2022]
Abstract
The chemical composition, structure and surface characteristics of biomaterials/scaffold can affect the adsorption of proteins, and this in turn influences the subsequent cellular response and tissue regeneration. With magnesium/calcium phosphate cements (MCPC) as model, the effects of magnesium (Mg) on the initial adhesion and osteogenic differentiation of bone marrow stromal cells (BMSCs) as well as the underlying mechanism were investigated. A series of MCPCs with different magnesium phosphate cement (MPC) content (0∼20%) in calcium phosphate cement (CPC) were synthesized. MCPCs with moderate proportion of MPC (5% and 10%, referred to as 5MCPC and 10MCPC) were found to effectively modulate the orientation of the adsorbed fibronectin (Fn) to exhibit enhanced receptor binding affinity, and to up-regulate integrin α5β1 expression of BMSCs, especially for 5MCPC. As a result, the attachment, morphology, focal adhesion formation, actin filaments assembly and osteogenic differentiation of BMSCs on 5MCPC were strongly enhanced. Further in vivo experiments confirmed that 5MCPC induced promoted osteogenesis in comparison to ot her CPC/MCPCs. Our results also suggested that the Mg on the underlying substrates but not the dissolved Mg ions was the main contributor to the above positive effects. Based on these results, it can be inferred that the specific interaction of Fn and integrin α5β1 had predominant effect on the MCPC-induced enhanced cellular response of BMSCs. These results provide a new strategy to regulate BMSCs adhesion and osteogenic differentiation by adjusting the Mg/Ca content and distribution in CPC, guiding the development of osteoinductive scaffolds for bone tissue regeneration.
Collapse
Affiliation(s)
- Jing Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Xiaoyu Ma
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Dan Lin
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hengsong Shi
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yuan Yuan
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Wei Tang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Huanjun Zhou
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Han Guo
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Jiangchao Qian
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
40
|
Li D, Wang C, Shan W, Zeng R, Fang Y, Wang P. Human amnion tissue injected with human umbilical cord mesenchymal stem cells repairs damaged sciatic nerves in rats. Neural Regen Res 2015; 7:1771-8. [PMID: 25624800 PMCID: PMC4302525 DOI: 10.3969/j.issn.1673-5374.2012.23.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/03/2012] [Indexed: 01/14/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells, incorporated into an amnion carrier tubes, were assessed for nerve regeneration potential in a rat nerve defect model. Damaged nerves were exposed to human amnion carriers containing either human umbilical cord mesenchymal stem cell (cell transplantation group) or saline (control group). At 8, 12, 16 and 20 weeks after cell implantation, the sciatic functional index was higher in the cell transplantation group compared with the control group. Furthermore, electrophysiological examination showed that threshold stimulus and maximum stimulus intensity gradually decreased while compound action potential amplitude gradually increased. Hematoxylin-eosin staining showed that regenerating nerve fibers were arranged in nerve tracts in the cell transplantation group and connective tissue between nerve tracts and amnion tissue reduced over time. Gastrocnemius muscle cell diameter, wet weight and restoration ratio were increased. These data indicate that transplanted human umbilical cord mesenchymal stem cells, using the amnion tube connection method, promote restoration of damaged sciatic nerves in rats.
Collapse
Affiliation(s)
- Dehua Li
- Department of Anatomy, Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| | - Changhui Wang
- Department of Neurology, Fushun Second Hospital, Fushun 113001, Liaoning Province, China
| | - Wei Shan
- Department of Anatomy, Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| | - Ruixia Zeng
- Department of Anatomy, Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| | - Yan Fang
- Department of Anatomy, Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| | - Pan Wang
- Department of Anatomy, Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| |
Collapse
|
41
|
|
42
|
Liu J, Chen W, Zhao Z, Xu HH. Effect of NELL1 gene overexpression in iPSC-MSCs seeded on calcium phosphate cement. Acta Biomater 2014; 10:5128-5138. [PMID: 25220281 DOI: 10.1016/j.actbio.2014.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/05/2014] [Accepted: 08/15/2014] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) are a promising source of patient-specific stem cells with great regenerative potential. There has been no report on NEL-like protein 1 (NELL1) gene modification of iPSC-MSCs. The objectives of this study were to genetically modify iPSC-MSCs with NELL1 overexpression for bone tissue engineering, and investigate the osteogenic differentiation of NELL1 gene-modified iPSC-MSCs seeded on Arg-Gly-Asp (RGD)-grafted calcium phosphate cement (CPC) scaffold. Cells were transduced with red fluorescence protein (RFP-iPSC-MSCs) or NELL1 (NELL1-iPSC-MSCs) by a lentiviral vector. Cell proliferation on RGD-grafted CPC scaffold, osteogenic differentiation and bone mineral synthesis were evaluated. RFP-iPSC-MSCs stably expressed high levels of RFP. Both the NELL1 gene and NELL1 protein levels were confirmed higher in NELL1-iPSC-MSCs than in RFP-iPSC-MSCs using RT-PCR and Western blot (P<0.05). Alkaline phosphatase activity was increased by 130% by NELL1 overexpression at 14days (P<0.05), indicating that NELL1 promoted iPSC-MSC osteogenic differentiation. When seeded on RGD-grafted CPC, NELL1-iPSC-MSCs attached and expanded similarly well to RFP-iPSC-MSCs. At 14days, the runt-related transcription factor 2 (RUNX2) gene level of NELL1-iPSC-MSCs was 2.0-fold that of RFP-iPSC-MSCs. The osteocalcin (OC) level of NELL1-iPSC-MSCs was 3.1-fold that of RFP-iPSC-MSCs (P<0.05). The collagen type I alpha 1 (COL1A1) gene level of NELL1-iPSC-MSCs was 1.7-fold that of RFP-iPSC-MSCs at 7days (P<0.05). Mineral synthesis was increased by 81% in NELL1-iPSC-MSCs at 21days. In conclusion, NELL1 overexpression greatly enhanced the osteogenic differentiation and mineral synthesis of iPSC-MSCs on RGD-grafted CPC scaffold for the first time. The novel NELL1-iPSC-MSC seeded RGD-CPC construct is promising for enhancing bone engineering.
Collapse
|
43
|
Maenz S, Kunisch E, Mühlstädt M, Böhm A, Kopsch V, Bossert J, Kinne RW, Jandt KD. Enhanced mechanical properties of a novel, injectable, fiber-reinforced brushite cement. J Mech Behav Biomed Mater 2014; 39:328-38. [DOI: 10.1016/j.jmbbm.2014.07.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/22/2014] [Accepted: 07/28/2014] [Indexed: 02/05/2023]
|
44
|
Li Y, Hou R, Wang Y, Lu B, Zhang J, Feng X, Liu Y, Cao Q. Fundamental study of application of umbilical cord mesenchymal stem cells to the periodontium to aid healing after autotransplantation of teeth. Br J Oral Maxillofac Surg 2014; 52:501-6. [DOI: 10.1016/j.bjoms.2014.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 04/02/2014] [Indexed: 10/25/2022]
|
45
|
Calcium phosphate cements for bone substitution: chemistry, handling and mechanical properties. Acta Biomater 2014; 10:1035-49. [PMID: 24231047 DOI: 10.1016/j.actbio.2013.11.001] [Citation(s) in RCA: 370] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 10/29/2013] [Accepted: 11/01/2013] [Indexed: 01/02/2023]
Abstract
Since their initial formulation in the 1980s, calcium phosphate cements (CPCs) have been increasingly used as bone substitutes. This article provides an overview on the chemistry, kinetics of setting and handling properties (setting time, cohesion and injectability) of CPCs for bone substitution, with a focus on their mechanical properties. Many processing parameters, such as particle size, composition of cement reactants and additives, can be adjusted to control the setting process of CPCs, concomitantly influencing their handling and mechanical performance. Moreover, this review shows that, although the mechanical strength of CPCs is generally low, it is not a critical issue for their application for bone repair--an observation not often realized by researchers and clinicians. CPCs with compressive strengths comparable to those of cortical bones can be produced through densification and/or homogenization of the cement matrix. The real limitation for CPCs appears to be their low fracture toughness and poor mechanical reliability (Weibull modulus), which have so far been only rarely studied.
Collapse
|
46
|
Thiele J, Ma Y, Bruekers SMC, Ma S, Huck WTS. 25th anniversary article: Designer hydrogels for cell cultures: a materials selection guide. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:125-47. [PMID: 24227691 DOI: 10.1002/adma.201302958] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/31/2013] [Indexed: 05/25/2023]
Abstract
Cell culturing, whether for tissue engineering or cell biology studies, always involves placing cells in a non-natural environment and no material currently exist that can mimic the entire complexity of natural tissues and variety of cell-matrix interactions that is found in vivo. Here, we review the vast range of hydrogels, composed of natural or synthetic polymers that provide a route to tailored microenvironments.
Collapse
Affiliation(s)
- Julian Thiele
- Institute for Molecules and Materials, Heyendaalseweg 135, 6525, AJ, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
47
|
Polo-Corrales L, Latorre-Esteves M, Ramirez-Vick JE. Scaffold design for bone regeneration. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2014; 14:15-56. [PMID: 24730250 PMCID: PMC3997175 DOI: 10.1166/jnn.2014.9127] [Citation(s) in RCA: 531] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The use of bone grafts is the standard to treat skeletal fractures, or to replace and regenerate lost bone, as demonstrated by the large number of bone graft procedures performed worldwide. The most common of these is the autograft, however, its use can lead to complications such as pain, infection, scarring, blood loss, and donor-site morbidity. The alternative is allografts, but they lack the osteoactive capacity of autografts and carry the risk of carrying infectious agents or immune rejection. Other approaches, such as the bone graft substitutes, have focused on improving the efficacy of bone grafts or other scaffolds by incorporating bone progenitor cells and growth factors to stimulate cells. An ideal bone graft or scaffold should be made of biomaterials that imitate the structure and properties of natural bone ECM, include osteoprogenitor cells and provide all the necessary environmental cues found in natural bone. However, creating living tissue constructs that are structurally, functionally and mechanically comparable to the natural bone has been a challenge so far. This focus of this review is on the evolution of these scaffolds as bone graft substitutes in the process of recreating the bone tissue microenvironment, including biochemical and biophysical cues.
Collapse
|
48
|
Injectable calcium phosphate–alginate–chitosan microencapsulated MC3T3-E1 cell paste for bone tissue engineering in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:4633-9. [DOI: 10.1016/j.msec.2013.07.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/26/2013] [Accepted: 07/17/2013] [Indexed: 11/22/2022]
|
49
|
RhBMP-2-loaded calcium silicate/calcium phosphate cement scaffold with hierarchically porous structure for enhanced bone tissue regeneration. Biomaterials 2013; 34:9381-92. [DOI: 10.1016/j.biomaterials.2013.08.059] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 08/19/2013] [Indexed: 12/17/2022]
|
50
|
Dorozhkin SV. Self-setting calcium orthophosphate formulations. J Funct Biomater 2013; 4:209-311. [PMID: 24956191 PMCID: PMC4030932 DOI: 10.3390/jfb4040209] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 01/08/2023] Open
Abstract
In early 1980s, researchers discovered self-setting calcium orthophosphate cements, which are bioactive and biodegradable grafting bioceramics in the form of a powder and a liquid. After mixing, both phases form pastes, which set and harden forming either a non-stoichiometric calcium deficient hydroxyapatite or brushite. Since both of them are remarkably biocompartible, bioresorbable and osteoconductive, self-setting calcium orthophosphate formulations appear to be promising bioceramics for bone grafting. Furthermore, such formulations possess excellent molding capabilities, easy manipulation and nearly perfect adaptation to the complex shapes of bone defects, followed by gradual bioresorption and new bone formation. In addition, reinforced formulations have been introduced, which might be described as calcium orthophosphate concretes. The discovery of self-setting properties opened up a new era in the medical application of calcium orthophosphates and many commercial trademarks have been introduced as a result. Currently such formulations are widely used as synthetic bone grafts, with several advantages, such as pourability and injectability. Moreover, their low-temperature setting reactions and intrinsic porosity allow loading by drugs, biomolecules and even cells for tissue engineering purposes. In this review, an insight into the self-setting calcium orthophosphate formulations, as excellent bioceramics suitable for both dental and bone grafting applications, has been provided.
Collapse
|