1
|
Drewry MD, Rothermund K, Syed-Picard FN. Topographical and Chemical Inductive Cues Synergistically Enhance the Schwann Cell Differentiation of Aligned Dental Pulp Stem Cell Sheets. J Tissue Eng Regen Med 2023; 2023:7958770. [PMID: 40226400 PMCID: PMC11918939 DOI: 10.1155/2023/7958770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 04/15/2025]
Abstract
Peripheral nerves have an inherent capacity for regeneration, but these Schwann cell-mediated mechanisms are insufficient for severe injuries. With current clinical treatments, slow regeneration and aberrant reinnervation result in poor functional outcomes. Dental pulp stem cells (DPSCs) offer a promising source of therapeutic neurotrophic factors (NTFs), growth factors that stimulate axon regeneration. Previously, we established that DPSCs can generate scaffold-free sheets with a linearly aligned extracellular matrix (ECM). These sheets provide trophic cues via the DPSCs and directional cues through the aligned ECM to both accelerate and orient axon outgrowth, thus providing a biomaterial capable of addressing the current clinical challenges. DPSCs have a propensity for differentiating into Schwann cells (SC-DPSCs), further enhancing their endogenous NTF expression. Here, we evaluated the effect of inducing SC differentiation on the neuroregenerative bioactivity of our DPSC sheets. These sheets were formed on substrates with linear microgrooves to direct the cells to deposit an aligned ECM. Inducing differentiation using an SC differentiation medium (SCDM) increased NTF expression 2-fold compared to unaligned uDPSC sheets, and this effect was amplified in linearly oriented SC-DPSC sheets by up to 8-fold. Furthermore, these aligned SC-DPSC sheets remodeled the sheet ECM to more closely emulate a regenerative neural microenvironment, expressing 8-fold and 2 × 107-fold more collagen IV and laminin, respectively, than unaligned uDPSC sheets. These data demonstrate that the chemical cues of the SCDM and the mechanotransductive cues of the aligned cell sheet synergistically enhanced the differentiation of DPSCs into repair SC-like cells. To evaluate their functional effects on neuritogenesis, the DPSC sheets were directly cocultured with neuronally differentiated neuroblastoma SH-SY5Y cells. In this in vitro culture system, the aligned SC-DPSC sheets promoted oriented neurite-like outgrowth similar to aligned uninduced DPSC sheets and increased collateral branching, which may emulate stages associated with natural SC-mediated repair processes. Therefore, linearly aligned SC-DPSC sheets have the potential to both promote nerve regeneration and reduce aberrant reinnervation, thus providing a promising biomaterial for applications to improve the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Michelle D. Drewry
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fatima N. Syed-Picard
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Tan S, Yang Y, Yang W, Han Y, Huang L, Yang R, Hu Z, Tao Y, Liu L, Li Y, Oyang L, Lin J, Peng Q, Jiang X, Xu X, Xia L, Peng M, Wu N, Tang Y, Cao D, Liao Q, Zhou Y. Exosomal cargos-mediated metabolic reprogramming in tumor microenvironment. J Exp Clin Cancer Res 2023; 42:59. [PMID: 36899389 PMCID: PMC9999652 DOI: 10.1186/s13046-023-02634-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of cancer. As nutrients are scarce in the tumor microenvironment (TME), tumor cells adopt multiple metabolic adaptations to meet their growth requirements. Metabolic reprogramming is not only present in tumor cells, but exosomal cargos mediates intercellular communication between tumor cells and non-tumor cells in the TME, inducing metabolic remodeling to create an outpost of microvascular enrichment and immune escape. Here, we highlight the composition and characteristics of TME, meanwhile summarize the components of exosomal cargos and their corresponding sorting mode. Functionally, these exosomal cargos-mediated metabolic reprogramming improves the "soil" for tumor growth and metastasis. Moreover, we discuss the abnormal tumor metabolism targeted by exosomal cargos and its potential antitumor therapy. In conclusion, this review updates the current role of exosomal cargos in TME metabolic reprogramming and enriches the future application scenarios of exosomes.
Collapse
Affiliation(s)
- Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wenjuan Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Lisheng Huang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Ruiqian Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Zifan Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Yi Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Lin Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yun Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xuemeng Xu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Deliang Cao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China. .,Hunan Key Laboratory of Translational Radiation Oncology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
3
|
The Implant Proteome—The Right Surgical Glue to Fix Titanium Implants In Situ. J Funct Biomater 2022; 13:jfb13020044. [PMID: 35466226 PMCID: PMC9036294 DOI: 10.3390/jfb13020044] [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: 03/14/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023] Open
Abstract
Titanium implants are frequently applied to the bone in orthopedic and trauma surgery. Although these biomaterials are characterized by excellent implant survivorship and clinical outcomes, there are almost no data available on the initial protein layer binding to the implant surface in situ. This study aims to investigate the composition of the initial protein layer on endoprosthetic surfaces as a key initiating step in osseointegration. In patients qualified for total hip arthroplasty, the implants are inserted into the femoral canal, fixed and subsequently explanted after 2 and 5 min. The proteins adsorbed to the surface (the implant proteome) are analyzed by liquid chromatography–tandem mass spectrometry (LC-MS/MS). A statistical analysis of the proteins’ alteration with longer incubation times reveals a slight change in their abundance according to the Vroman effect. The pathways involved in the extracellular matrix organization of bone, sterile inflammation and the beginning of an immunogenic response governed by neutrophils are significantly enriched based on the analysis of the implant proteome. Those are generally not changed with longer incubation times. In summary, proteins relevant for osseointegration are already adsorbed within 2 min in situ. A deeper understanding of the in situ protein–implant interactions in patients may contribute to optimizing implant surfaces in orthopedic and trauma surgery.
Collapse
|
4
|
Liu Y, Rui Z, Cheng W, Song L, Xu Y, Li R, Zhang X. Characterization and evaluation of a femtosecond laser-induced osseointegration and an anti-inflammatory structure generated on a titanium alloy. Regen Biomater 2021; 8:rbab006. [PMID: 33738120 PMCID: PMC7955712 DOI: 10.1093/rb/rbab006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Cell–material interactions during early osseointegration of the bone–implant interface are critical and involve crosstalk between osteoblasts and osteoclasts. The surface properties of titanium implants also play a critical role in cell–material interactions. In this study, femtosecond laser treatment and sandblasting were used to alter the surface morphology, roughness and wettability of a titanium alloy. Osteoblasts and osteoclasts were then cultured on the resulting titanium alloy disks. Four disk groups were tested: a polished titanium alloy (pTi) control; a hydrophilic micro-dislocation titanium alloy (sandblasted Ti (STi)); a hydrophobic nano-mastoid Ti alloy (femtosecond laser-treated Ti (FTi)); and a hydrophilic hierarchical hybrid micro-/nanostructured Ti alloy [femtosecond laser-treated and sandblasted Ti (FSTi)]. The titanium surface treated by the femtosecond laser and sandblasting showed higher biomineralization activity and lower cytotoxicity in simulated body fluid and lactate dehydrogenase assays. Compared to the control surface, the multifunctional titanium surface induced a better cellular response in terms of proliferation, differentiation, mineralization and collagen secretion. Further investigation of macrophage polarization revealed that increased anti-inflammatory factor secretion and decreased proinflammatory factor secretion occurred in the early response of macrophages. Based on the above results, the synergistic effect of the surface properties produced an excellent cellular response at the bone–implant interface, which was mainly reflected by the promotion of early ossteointegration and macrophage polarization.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Zhongying Rui
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei Cheng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Licheng Song
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China.,Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| | - Yunqiang Xu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ruixin Li
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin 300041, China
| | - Xizheng Zhang
- Institute of Medical Service Support, Institute of Systems Engineering, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
5
|
Chen ZY, Gao S, Zhang YW, Zhou RB, Zhou F. Antibacterial biomaterials in bone tissue engineering. J Mater Chem B 2021; 9:2594-2612. [PMID: 33666632 DOI: 10.1039/d0tb02983a] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bone infection is a devastating disease characterized by recurrence, drug-resistance, and high morbidity, that has prompted clinicians and scientists to develop novel approaches to combat it. Currently, although numerous biomaterials that possess excellent biocompatibility, biodegradability, porosity, and mechanical strength have been developed, their lack of effective antibacterial ability substantially limits bone-defect treatment efficacy. There is, accordingly, a pressing need to design antibacterial biomaterials for effective bone-infection prevention and treatment. This review focuses on antibacterial biomaterials and strategies; it presents recently reported biomaterials, including antibacterial implants, antibacterial scaffolds, antibacterial hydrogels, and antibacterial bone cement types, and aims to provide an overview of these antibacterial materials for application in biomedicine. The antibacterial mechanisms of these materials are discussed as well.
Collapse
Affiliation(s)
- Zheng-Yang Chen
- Orthopedic Department, Peking University Third Hospital, Beijing 100191, China.
| | | | | | | | | |
Collapse
|
6
|
Siqueira R, Ferreira JA, Rizzante FAP, Moura GF, Mendonça DBS, de Magalhães D, Cimões R, Mendonça G. Hydrophilic titanium surface modulates early stages of osseointegration in osteoporosis. J Periodontal Res 2020; 56:351-362. [PMID: 33368275 DOI: 10.1111/jre.12827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 10/16/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Using a mouse osteoporotic model, this study aimed to determine the influence of hydrophilic titanium surfaces on gene expression and bone formation during the osseointegration process. BACKGROUND Based on the previous evidence, it is plausible to assume that osteoporotic bone has a different potential of bone healing. Therefore, implant surface modification study that aims at enhancing bone formation to further improve short- and long-term clinical outcomes in osteoporosis is necessary. MATERIAL AND METHODS Fifty female, 3-month-old mice were included in this study. Osteoporosis was induced by ovariectomy (OVX, test group) in 25 mice. The further 25 mice had ovaries exposed but not removed (SHAM, control group). Seven weeks following the ovariectomy procedures, one customized implant (0.7 × 8 mm) of each surface was placed in each femur for both groups. Implants had either a hydrophobic surface (SAE) or a hydrophilic treatment surface (SAE-HD). Calcium (Ca) and phosphorus (P) content was measured by energy-dispersive X-ray spectroscopy (EDS) after 7 days. The femurs were analyzed for bone-to-implant contact (BIC) and bone volume fraction (BV) by nano-computed tomography (nano-CT) after 14 and 28 days. Same specimens were further submitted to histological analysis. Additionally, after 3 and 7 days, implants were removed and cells were collected around the implant to access gene expression profile of key osteogenic (Runx2, Alp, Sp7, Bsp, Sost, Ocn) and inflammatory genes (IL-1β, IL-10, Tnf-α, and Nos2) by qRT-PCR assay. Statistical analysis was performed by ANOVA and paired t test with significance at P < .05. RESULTS The amount of Ca and P deposited on the surface due to the mineralization process was higher for SAE-HD compared to SAE on the intra-group analysis. Nano-CT and histology revealed more BV and BIC for SAE-HD in SHAM and OVX groups compared to SAE. Analysis in OVX group showed that most genes (ie, ALP, Runx2) involved in the bone morphogenetic protein (BMP) signaling were significantly activated in the hydrophilic treatment. CONCLUSION Both surfaces were able to modulate bone responses toward osteoblast differentiation. SAE-HD presented a faster response in terms of bone formation and osteogenic gene expression compared to SAE. Hydrophilic surface in situations of osteoporosis seems to provide additional benefits in the early stages of osseointegration.
Collapse
Affiliation(s)
- Rafael Siqueira
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Afonso Ferreira
- Department of Periodontology and Implant Dentistry, School of Dentistry, Federal University of Uberlandia, Uberlândia, Brazil.,Department of Biological and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Fábio Antônio Piola Rizzante
- Department of Comprehensive Care, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Guilherme Faria Moura
- Department of Periodontology and Implant Dentistry, School of Dentistry, Federal University of Uberlandia, Uberlândia, Brazil.,Department of Biological and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | | | - Denildo de Magalhães
- Department of Periodontology and Implant Dentistry, School of Dentistry, Federal University of Uberlandia, Uberlândia, Brazil
| | - Renata Cimões
- Department of Prosthesis and Maxillofacial Surgery, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Gustavo Mendonça
- Department of Biological and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Physicochemical characterization of albumin immobilized on different TiO2 surfaces for use in implant materials. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2018.12.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Deng L, He X, Xie K, Xie L, Deng Y. Dual Therapy Coating on Micro/Nanoscale Porous Polyetheretherketone to Eradicate Biofilms and Accelerate Bone Tissue Repair. Macromol Biosci 2018; 19:e1800376. [PMID: 30549406 DOI: 10.1002/mabi.201800376] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/29/2018] [Indexed: 12/15/2022]
Abstract
Defective osteogenesis and latent infections continue to be two major issues in the therapy of bone tissue regeneration. In this study, a unique hierarchically micro/nanoscale-architecture is first proposed and produced on polyetheretherketone (PEEK). Besides, a "simvastatin-PLLA film-tobramycin microspheres" delivery system is subsequently fabricated to endow the PEEK implant with osteogenic and antibacterial capabilities. In vitro antibacterial evaluations confirm that the decorated PEEK scaffolds possess excellent resistance against planktonic/adherent bacteria. In vitro cell attachment/proliferation, lactate dehydrogenase (LDH) content, alkaline phosphatase (ALP) activity, calcium mineral deposition experiments, and real-time PCR analysis all exhibit that the superior proliferation rate and osteo-differentiation potential of MC3T3-E1 pre-osteoblasts are presented on the PEEK samples with dual functional decoration. In the mouse calvarial defect model, the micro-CT and histological results demonstrate that our scaffolds display a remarkable bone forming capability. Generally, the PEEK scaffolds co-endowed with simvastatin and tobramycin microspheres possess great potential in clinics.
Collapse
Affiliation(s)
- Lijun Deng
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Xianhua He
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Kenan Xie
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Lu Xie
- State Key Laboratory of Oral Diseases West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yi Deng
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China.,Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
9
|
Cai B, Zou Q, Zuo Y, Mei Q, Ma J, Lin L, Chen L, Li Y. Injectable Gel Constructs with Regenerative and Anti-Infective Dual Effects Based on Assembled Chitosan Microspheres. ACS APPLIED MATERIALS & INTERFACES 2018; 10:25099-25112. [PMID: 29952200 DOI: 10.1021/acsami.8b06648] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
There is increasing demand for biomaterials that both assist with bone regeneration and have anti-infection qualities in clinical applications. To achieve this goal, chitosan microspheres with either positive or negative charges were fabricated and then assembled as a gel for bone healing. The positively charged chitosan microspheres (CSM; ∼35.5 μm) and negatively charged O-carboxymethyl chitosan microspheres (CMCSM; ∼13.5 μm) were loaded, respectively, with bone morphogenetic protein (BMP-2) and berberine (Bbr) via swollen encapsulation and physical adsorption without a significant change in the electric charges. The release kinetics of BMP-2 and Bbr from the microspheres were also studied in vitro. The results showed that the Bbr/CMCSM microsphere group possessed high antibacterial activity against Staphylococcus aureus; the BMP-2/CSM microsphere group also had excellent cytocompatibility and improved osteoinductivity with the assistance of BMP-2. The assembled gel group consisting of Bbr/CMCSM and BMP-2/CSM had a porous structure that allowed biological signal transfer and tissue infiltration and exhibited significantly enhanced bone reconstruction compared with that of the respective microsphere groups, which should result from the osteoconductivity of the porous structure and the osteoinduction of the BMP-2 growth factor. The oppositely charged microspheres and their assembled gel provide a promising prospect for making injectable tissue-engineered constructs with regenerative and anti-infective dual effects for biomedical applications.
Collapse
Affiliation(s)
- Bin Cai
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Qin Zou
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Yi Zuo
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Quanjing Mei
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Jinqi Ma
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Lili Lin
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Li Chen
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| | - Yubao Li
- Research Center for Nano-Biomaterial, Analytical & Testing Center , Sichuan University , Chengdu 610064 , China
| |
Collapse
|
10
|
Vilardell AM, Cinca N, Garcia-Giralt N, Dosta S, Cano IG, Nogués X, Guilemany JM. Osteoblastic cell response on high-rough titanium coatings by cold spray. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:19. [PMID: 29392501 DOI: 10.1007/s10856-018-6026-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/10/2018] [Indexed: 06/07/2023]
Abstract
Highly rough and porous commercially pure titanium coatings have been directly produced for first time by the cold spray technology, which is a promising technology in front of the vacuum plasma spray for oxygen sensitive materials. The wettability properties as well as the biocompatibility evaluation have been compared to a simply sand blasted Ti6Al4V alloy substrate. Surface topographies were analysed using confocal microscopy. Next, osteoblast morphology (Phalloidin staining), proliferation (MTS assay), and differentiation (alkaline phosphatase activity) were examined along 1, 7 and 14 days of cell culture on the different surfaces. Finally, mineralization by alizarin red staining was quantified at 28 days of cell culture. The contact angle values showed an increased hydrophilic behaviour on the as-sprayed surface with a good correlation to the biological response. A higher cell viability, proliferation and differentiation were obtained for highly rough commercial pure titanium coatings in comparison with sand blasted substrates. Cell morphology was similar in all coatings tested; at 14 days both samples showed extended filopodia. A higher amount of calcium-rich deposits was detected on highly rough surfaces. In summary, in-vitro results showed an increase of biological properties when surface roughness increases.
Collapse
Affiliation(s)
- A M Vilardell
- Centre de Projecció Tèrmica (CPT), Dpt. Ciència dels Materials i Enginyeria Metal.lúrgica, Universitat de Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain.
| | - N Cinca
- Centre de Projecció Tèrmica (CPT), Dpt. Ciència dels Materials i Enginyeria Metal.lúrgica, Universitat de Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - N Garcia-Giralt
- IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), RETICEF, Doctor Aiguader 88, 08003, Barcelona, Spain
| | - S Dosta
- Centre de Projecció Tèrmica (CPT), Dpt. Ciència dels Materials i Enginyeria Metal.lúrgica, Universitat de Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - I G Cano
- Centre de Projecció Tèrmica (CPT), Dpt. Ciència dels Materials i Enginyeria Metal.lúrgica, Universitat de Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - X Nogués
- IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), RETICEF, Doctor Aiguader 88, 08003, Barcelona, Spain
| | - J M Guilemany
- Centre de Projecció Tèrmica (CPT), Dpt. Ciència dels Materials i Enginyeria Metal.lúrgica, Universitat de Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| |
Collapse
|
11
|
Nassif W, Rifai M. Surface Characterization and Cell Adhesion of Different Zirconia Treatments: An in vitro Study. J Contemp Dent Pract 2018; 19:181-188. [PMID: 29422468 DOI: 10.5005/jp-journals-10024-2234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIM The aim of this study was to characterize the surface of zirconia subjected to different treatments and evaluate its effect on cell adhesion and proliferation. MATERIALS AND METHODS A total of 80 zirconia disks were divided into four groups (n = 20) according to the surface treatments used: group I: as-sintered (AS), no surface treatment applied; group II: abrasion treatment applied using Rocatec (ROC; 3M ESPE) system with silica-coated alumina powder of grit size 110 μm; group III: erbium, chromium:yttrium, scandium, gallium, garnet (Er, Cr:YSGG) laser (LAS; BIOLASE) was used at a frequency of 20 Hz and output power of 3 W; and group IV: specimens were subjected to the selective infiltration etching (SIE) technique. Surface characterization was evaluated for the different groups (roughness, hardness, and morphology), and cell behavior (adhesion and proliferation) was tested (a = 0.05). RESULTS The ROC group reported a significant increase in surface roughness (2.201 ± 0.352) and Vickers hardness (1758 ± 16.6) compared with the other surface treatments. The SIE surface-treated group reported a significantly higher number of cells (64.5 ± 2.6 and 53.5 ± 2.2 respectively) compared with the other surface-treated groups. CONCLUSION The SIE is a promising surface treatment for zirconia that significantly enhances cell adhesion and osseointegration. CLINICAL SIGNIFICANCE The SIE treatment of zirconia implants may help in a faster and better osseointegration.
Collapse
Affiliation(s)
- Wadih Nassif
- Department of Prosthodontics, Faculty of Dental Medicine Lebanese University, Beirut, Lebanon, Phone: +9613686787, e-mail:
| | - Mohamad Rifai
- Department of Prosthodontics, Faculty of Dental Medicine Lebanese University, Beirut, Lebanon
| |
Collapse
|
12
|
Nguyen HG, Metavarayuth K, Wang Q. Upregulation of osteogenesis of mesenchymal stem cells with virus-based thin films. Nanotheranostics 2018; 2:42-58. [PMID: 29291162 PMCID: PMC5743837 DOI: 10.7150/ntno.19974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 10/15/2017] [Indexed: 01/16/2023] Open
Abstract
A major aim of tissue engineering is to develop biomimetic scaffolding materials that can guide the proliferation, self-renewal and differentiation of multipotent stem cells into specific lineages. Cellular functions can be controlled by the interactions between cells and biomaterials. Therefore, the surface chemistry and topography of support materials play a pivotal role in modulating cell behaviors at many stages of cell growth and development. Due to their highly ordered structure and programmable surface chemistries, which provide unique topography as biomaterials, viral nanoparticles have been utilized as building blocks for targeted cell growth and differentiation. This review article discusses the fabrication of two-dimensional virus-based thin film on substrates and highlights the study of the effect of chemical and physical cues introduced by plant virus nanoparticle thin films on the promotion of osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Huong Giang Nguyen
- Department of Chemistry and Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, SC 29208, USA
- National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899, USA
| | - Kamolrat Metavarayuth
- Department of Chemistry and Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, SC 29208, USA
| | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, 631 Sumter Street, Columbia, SC 29208, USA
| |
Collapse
|
13
|
Bressel TAB, de Queiroz JDF, Gomes Moreira SM, da Fonseca JT, Filho EA, Guastaldi AC, Batistuzzo de Medeiros SR. Laser-modified titanium surfaces enhance the osteogenic differentiation of human mesenchymal stem cells. Stem Cell Res Ther 2017; 8:269. [PMID: 29179738 PMCID: PMC5704576 DOI: 10.1186/s13287-017-0717-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/13/2017] [Accepted: 10/30/2017] [Indexed: 12/13/2022] Open
Abstract
Background Titanium surfaces have been modified by various approaches with the aim of improving the stimulation of osseointegration. Laser beam (Yb-YAG) treatment is a controllable and flexible approach to modifying surfaces. It creates a complex surface topography with micro and nano-scaled patterns, and an oxide layer that can improve the osseointegration of implants, increasing their usefulness as bone implant materials. Methods Laser beam irradiation at various fluences (132, 210, or 235 J/cm2) was used to treat commercially pure titanium discs to create complex surface topographies. The titanium discs were investigated by scanning electron microscopy, X-ray diffraction, and measurement of contact angles. The surface generated at a fluence of 235 J/cm2 was used in the biological assays. The behavior of mesenchymal stem cells from an umbilical cord vein was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, a mineralization assay, and an alkaline phosphatase activity assay and by carrying out a quantitative real-time polymerase chain reaction for osteogenic markers. CHO-k1 cells were also exposed to titanium discs in the MTT assay. Results The best titanium surface was that produced by laser beam irradiation at 235 J/cm2 fluence. Cell proliferation analysis revealed that the CHO-k1 and mesenchymal stem cells behaved differently. The laser-processed titanium surface increased the proliferation of CHO-k1 cells, reduced the proliferation of mesenchymal stem cells, upregulated the expression of the osteogenic markers, and enhanced alkaline phosphatase activity. Conclusions The laser-treated titanium surface modulated cellular behavior depending on the cell type, and stimulated osteogenic differentiation. This evidence supports the potential use of laser-processed titanium surfaces as bone implant materials, and their use in regenerative medicine could promote better outcomes.
Collapse
Affiliation(s)
- Tatiana A B Bressel
- Departamento de Biologia Celular e Genética, CB-UFRN, Universidade Federal do Rio Grande do Norte, Campus Universitário, Lagoa Nova, 59072-970, Natal, RN, Brazil
| | - Jana Dara Freires de Queiroz
- Departamento de Biologia Celular e Genética, CB-UFRN, Universidade Federal do Rio Grande do Norte, Campus Universitário, Lagoa Nova, 59072-970, Natal, RN, Brazil.,Programa de Pós Graduação em Ciências da Saúde, Natal, RN, Brazil
| | - Susana Margarida Gomes Moreira
- Departamento de Biologia Celular e Genética, CB-UFRN, Universidade Federal do Rio Grande do Norte, Campus Universitário, Lagoa Nova, 59072-970, Natal, RN, Brazil
| | - Jéssyca T da Fonseca
- Departamento de Biologia Celular e Genética, CB-UFRN, Universidade Federal do Rio Grande do Norte, Campus Universitário, Lagoa Nova, 59072-970, Natal, RN, Brazil
| | - Edson A Filho
- Departamento de Físico-Química, Instituto de Química de Araraquara-UNESP, Araraquara, SP, Brazil
| | - Antônio Carlos Guastaldi
- Departamento de Físico-Química, Instituto de Química de Araraquara-UNESP, Araraquara, SP, Brazil
| | - Silvia Regina Batistuzzo de Medeiros
- Departamento de Biologia Celular e Genética, CB-UFRN, Universidade Federal do Rio Grande do Norte, Campus Universitário, Lagoa Nova, 59072-970, Natal, RN, Brazil.
| |
Collapse
|
14
|
Zhang Z, Cheng X, Yao Y, Luo J, Tang Q, Wu H, Lin S, Han C, Wei Q, Chen L. Electrophoretic deposition of chitosan/gelatin coatings with controlled porous surface topography to enhance initial osteoblast adhesive responses. J Mater Chem B 2016; 4:7584-7595. [PMID: 32263815 DOI: 10.1039/c6tb02122k] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Electrophoretically deposited (EPD) coatings have often been employed recently for the addition of different new chemical compositions to classic chitosan coatings to improve the biocompatibility and therapeutic potential of coated implants. However, little attention has been paid to enhance the cell response to EPD coatings via integrating the effects of chemical components and surface topography. Here, we fabricated EPD chitosan/gelatin (CS/G) coatings with controlled porous surface topography by controlling bubble generation in the EPD process via changing the gelatin content in solution from 0, 0.01, 0.1, and 1 to 10 mg ml-1. The pure chitosan coating surface was characterized by homogeneous large pores of 500 μm. After 0.01 mg ml-1 gelatin was added, 180 μm small pores appeared on the walls of large pores. As the gelatin content increased to 0.1 mg ml-1, a number of small pores increased noticeably. When the gelatin content reached 1 mg ml-1, large pores disappeared, and the coating displayed homogeneous small pores. 10 mg ml-1 gelatin concentration led to coatings consisting of small pores with not integral and continuous structures. The initial osteoblastic responses, including cell adherence progress, spreading area, proliferation rate, and focal adhesion-related gene expression, gradually improved from 0 to 0.01, 0.1, and 1 mg ml-1 gelatin content, but decreased from 1 to 10 mg ml-1. All these results indicated that the initial cell responses to coatings reached a peak when it was 1 mg ml-1 gelatin and they had homogeneous small pores, which might contribute to the synergistic effects of the porous surface structure and components. This work would be beneficial for expanding the potential application of EPD coatings.
Collapse
Affiliation(s)
- Zhen Zhang
- Dept. Stomatol., Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Significance of osteogenic surface coatings on implants to enhance osseointegration under osteoporotic-like conditions. IMPLANT DENT 2016; 23:679-86. [PMID: 25290281 DOI: 10.1097/id.0000000000000161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim was to assess the significance of osteogenic surface coatings on implants to enhance osseointegration under osteoporotic-like (OP-like) conditions. METHODS To address the focused question "Do osteogenic surface coatings on implants enhance osseointegration under OP-like conditions?" PubMed/MEDLINE and Google-Scholar databases were searched from 1995 up to and including February 2014 using various keywords. Unpublished data, letters to the editor, review articles, and articles published in languages other than English were excluded. RESULTS Of the 28 studies identified, 11 experimental studies were included. These studies were performed on bilaterally ovariectomized animals. In all studies, implant surface roughness was increased by various osteogenetic surface coatings including alumina, hydroxyapatite, calcium phosphate, and zoledronic acid. Nine studies reported that compared with non-coated surfaces, osteogenic coatings on implant surfaces increases bone volume and bone-to-implant contact (BIC) under OP-like conditions. In 2 studies, there was no difference in BIC around hydroxyapatite-coated implants placed in animals with and without OP-like conditions. CONCLUSION Osteogenic coatings on implant surfaces enhanced osseointegration in animals with OP-like conditions. However, additional clinical studies are warranted to assess the role of osteogenic coatings in increasing osseointegration in patients with osteoporosis.
Collapse
|
16
|
Lee EM, Smith K, Gall K, Boyan BD, Schwartz Z. Change in surface roughness by dynamic shape-memory acrylate networks enhances osteoblast differentiation. Biomaterials 2016; 110:34-44. [PMID: 27710831 DOI: 10.1016/j.biomaterials.2016.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/11/2016] [Accepted: 08/03/2016] [Indexed: 12/17/2022]
Abstract
Microscale surface roughness has been shown to enhance osseointegration of titanium implants through increased osteoblast differentiation while osteoblast proliferation remains greater on smooth titanium. Taking advantage of these phenomena, we developed a shape memory (meth)acrylate copolymer with thermomechanical properties that created a time-dependent dynamic surface change from smooth to rough under in vitro cell culture conditions and evaluated the effect of the shape recovery on osteoblast response. Rough topographies were created using soft lithography techniques to mimic those found on clinically-used Ti surfaces (machined smooth; acid-etched; grit-blasted). The surface roughness was then reduced to smooth via compression and shown to fully recover within 24 h in culture conditions. When grown under static conditions, osteoblast number, alkaline phosphatase specific activity (ALP), and osteoprotegerin (OPG) and vascular endothelial growth factor (VEGF) production were unaffected by polymer surface roughness, but osteocalcin (OCN) was increased on the grit-blasted polymer mimic. Under dynamic conditions, DNA was reduced but OCN and OPG were increased on the compressed grit-blasted polymer at 3 days compared to static surfaces. The present study indicates that responses to polymer surface are sensitive to time-dependent changes in topography. The use of a shape memory polymer with dynamic surface roughness may improve osseointegration.
Collapse
Affiliation(s)
- Erin M Lee
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Ken Gall
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Barbara D Boyan
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology, Atlanta, GA, USA; Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
17
|
Blanquer A, Hynowska A, Nogués C, Ibáñez E, Sort J, Baró MD, Özkale B, Pané S, Pellicer E, Barrios L. Effect of Surface Modifications of Ti40Zr10Cu38Pd12 Bulk Metallic Glass and Ti-6Al-4V Alloy on Human Osteoblasts In Vitro Biocompatibility. PLoS One 2016; 11:e0156644. [PMID: 27243628 PMCID: PMC4887090 DOI: 10.1371/journal.pone.0156644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/17/2016] [Indexed: 01/03/2023] Open
Abstract
The use of biocompatible materials, including bulk metallic glasses (BMGs), for tissue regeneration and transplantation is increasing. The good mechanical and corrosion properties of Ti40Zr10Cu38Pd12 BMG and its previously described biocompatibility makes it a potential candidate for medical applications. However, it is known that surface properties like topography might play an important role in regulating cell adhesion, proliferation and differentiation. Thus, in the present study, Ti40Zr10Cu38Pd12 BMG and Ti6-Al-4V alloy were surface-modified electrochemically (nanomesh) or physically (microscratched) to investigate the effect of material topography on human osteoblasts cells (Saos-2) adhesion, proliferation and differentiation. For comparative purposes, the effect of mirror-like polished surfaces was also studied. Electrochemical treatments led to a highly interconnected hierarchical porous structure rich in oxides, which have been described to improve corrosion resistance, whereas microscratched surfaces showed a groove pattern with parallel trenches. Cell viability was higher than 96% for the three topographies tested and for both alloy compositions. In all cases, cells were able to adhere, proliferate and differentiate on the alloys, hence indicating that surface topography plays a minor role on these processes, although a clear cell orientation was observed on microscratched surfaces. Overall, our results provide further evidence that Ti40Zr10Cu38Pd12 BMG is an excellent candidate, in the present two topographies, for bone repair purposes.
Collapse
Affiliation(s)
- Andreu Blanquer
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Anna Hynowska
- Departament de Física, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Carme Nogués
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Elena Ibáñez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Jordi Sort
- Departament de Física, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Maria Dolors Baró
- Departament de Física, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Berna Özkale
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
| | - Salvador Pané
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zurich, Zurich, Switzerland
| | - Eva Pellicer
- Departament de Física, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| | - Leonardo Barrios
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Edifici Cc, Bellaterra, Spain
| |
Collapse
|
18
|
Metavarayuth K, Sitasuwan P, Luckanagul JA, Feng S, Wang Q. Virus Nanoparticles Mediated Osteogenic Differentiation of Bone Derived Mesenchymal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500026. [PMID: 27980904 PMCID: PMC5115314 DOI: 10.1002/advs.201500026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/21/2015] [Indexed: 05/29/2023]
Abstract
There are few methodologies that allow manipulating a biomaterial surface at nanometer scale, which controllably influence different cellular functions. In this study, virus nanoparticles with different structural features are selected to prepare 2D substrates with defined nanoscale topographies and the cellular responses are investigated. It is demonstrated that the viral nanoparticle based substrates could accelerate and enhance osteogenesis of bone derived mesenchymal stem cells as indicated by the upregulation of osteogenic markers, including bone morphogenetic protein-2, osteocalcin, and osteopontin, at both gene and protein expression levels. Moreover, alkaline phosphatase activity and calcium mineralization, both indicators for a -successful bone formation, are also increased in cells grown on these nanoscale possessed substrates. These discoveries and developments present a new paradigm for nanoscale engineering of a biomaterial surface.
Collapse
Affiliation(s)
- Kamolrat Metavarayuth
- Department of Chemistry and Biochemistry University of South Carolina 631 Sumter Street Columbia SC 29208 USA
| | - Pongkwan Sitasuwan
- Department of Chemistry and Biochemistry University of South Carolina 631 Sumter Street Columbia SC 29208 USA
| | - Jittima Amie Luckanagul
- Department of Food and Pharmaceutical Chemistry Faculty of Pharmaceutical Sciences Chulalongkorn University 254 Phayathai Rd., Wangmai Pathumwan Bangkok 10330 Thailand
| | - Sheng Feng
- Department of Chemistry and Biochemistry University of South Carolina 631 Sumter Street Columbia SC 29208 USA
| | - Qian Wang
- Department of Chemistry and Biochemistry University of South Carolina 631 Sumter Street Columbia SC 29208 USA
| |
Collapse
|
19
|
Mariscal-Muñoz E, Costa CAS, Tavares HS, Bianchi J, Hebling J, Machado JPB, Lerner UH, Souza PPC. Osteoblast differentiation is enhanced by a nano-to-micro hybrid titanium surface created by Yb:YAG laser irradiation. Clin Oral Investig 2015. [PMID: 26224513 DOI: 10.1007/s00784-015-1533-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The aim of this study was to analyze the capacity of a new modified laser surface to stimulate calvarial osteoblasts isolated from neonatal mouse bones to differentiate and form mineralized nodules. METHODS Titanium discs were subjectezd or not to laser irradiation according to specific parameters and characterized. Osteoblasts isolated from neonatal mouse calvaria were cultured over the discs, and the capacity of these cells to proliferate (MTT assay), form mineralized nodules (Alizarin red assay), and enhance alkaline phosphatase activity (ALPase activity) was analyzed. Real-time PCR was used for quantification of gene expression. RESULTS Laser-irradiated titanium discs (L) presented a rough nano-to-micrometric oxidized surface contrasting with the smooth pattern on polished discs (P). The Ra on the micrometric level increased from 0.32 ± 0.01 μm on P surfaces to 10.57 ± 0.39 μm on L surfaces. When compared with P, L promoted changes in osteoblast morphology, increased mineralized nodule formation in osteoblasts cultured on the surfaces for 14 days, and enhanced ALPase activity at days 7 and 14. Transcription factors triggering osteoblast differentiation (Runx2 and Sp7) and genes encoding the bone extracellular matrix proteins collagen type-1 (Col1a1), osteopontin (Spp1), and osteocalcin (Bglap) were upregulated in cells on L surfaces compared with those on P surfaces at days 1-14. CONCLUSION Laser treatment of titanium surfaces created a rough surface that stimulated osteoblast differentiation. CLINICAL RELEVANCE Laser treatment of titanium generates a reproducible and efficient surface triggering osteoblast differentiation that can be of importance for osteointegration.
Collapse
Affiliation(s)
- Eduardo Mariscal-Muñoz
- Department of Physiology and Pathology, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil
| | - Carlos A S Costa
- Department of Physiology and Pathology, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil
| | - Hewerson S Tavares
- Department of Physiology and Pathology, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil
| | - Jonas Bianchi
- Department of Physiology and Pathology, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil
| | - Josimeri Hebling
- Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil
| | - João P B Machado
- National Institute for Space Research-INPE, São José dos Campos, São Paulo, 12227-010, Brazil
| | - Ulf H Lerner
- Umeå University, Umeå, S-901 87, Sweden.,Sahlgrenska Academy at University of Gothenburg, Gothenburg, S-405 30, Sweden
| | - Pedro P C Souza
- Department of Physiology and Pathology, Faculty of Dentistry at Araraquara, Univ. Estadual Paulista-UNESP, Araraquara, São Paulo, 14801-903, Brazil.
| |
Collapse
|
20
|
Effect of Increasing Doses of γ-Radiation on Bone Marrow Stromal Cells Grown on Smooth and Rough Titanium Surfaces. Stem Cells Int 2015; 2015:359416. [PMID: 26257788 PMCID: PMC4518184 DOI: 10.1155/2015/359416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/28/2015] [Accepted: 07/01/2015] [Indexed: 02/05/2023] Open
Abstract
Radiation therapy for oral and maxillofacial tumors could damage bone marrow stromal cells (BMSCs) in jaw, which caused dental implant failure. However, how radiation affects BMSCs on SLA (sandblasted with large-grits, acid-etched) surfaces is still unknown. The aim of this study was to investigate effect of different dose of γ-radiation on BMSCs on SLA and PT (polished titanium) surfaces. Rat BMSCs were radiated with 2, 4, and 8 Gy γ-radiation and then seeded on both surfaces. Cell adhesion, spreading, and proliferation were tested. The osteogenesis and the adipogenesis ability were examined by Alizarin-Red and Oil-Red staining, respectively. Real-time PCR was performed to detect osteogenic (osteocalcin, OCN; runt-related transcription factor 2, Runx2) and adipogenic (peroxisome proliferator-activated receptor gamma, PPARγ) gene expression at days 7 and 14 postirradiation. Results showed that γ-radiation reduced cell proliferation, adhesion, spreading, and osteogenic differentiation. 2 Gy radiation promoted adipogenic differentiation, but it was significantly decreased when dosage reached 4 Gy. In conclusion, results suggest that γ-radiation influenced BMSCs behaviors in a dosage-dependent manner except adipogenic differentiation, low dose promoted it, and high dose inhibited it. This effect was influenced by surface characteristics, which may explain the different failure rate of various implants in patients after radiation.
Collapse
|
21
|
Matteson JL, Greenspan DC, Tighe TB, Gilfoy N, Stapleton JJ. Assessing the hierarchical structure of titanium implant surfaces. J Biomed Mater Res B Appl Biomater 2015; 104:1083-90. [PMID: 26034005 DOI: 10.1002/jbm.b.33462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 04/16/2015] [Accepted: 05/07/2015] [Indexed: 11/05/2022]
Abstract
The physical texture of implant surfaces are known to be one important factor in creating a stable bone-implant interface. Simple roughness parameters (for e.g., Sa or Sz) are not entirely adequate when characterizing surfaces possessing hierarchical structure (macro, micro, and nano scales). The aim of this study was to develop an analytical approach to quantify hierarchical surface structure of implant surfaces possessing nearly identical simple roughness. Titanium alloys with macro/micro texture (MM) and macro/micro/nano texture (MMN) were chosen as model surfaces to be evaluated. There was no statistical difference (p > 0.05) in either Sa (13.56 vs. 13.43 µm) or Sz (91.74 vs. 92.39 µm) for the MM and MMN surfaces, respectively. However, when advanced filtering algorithms were applied to these datasets, a statistical difference in roughness was found between MM (Sa = 0.54 µm) and MMN (Sa = 1.06 µm; p < 0.05). Additionally, a method was developed to specifically quantify the density of surface features appearing similar in geometry to natural osteoclastic pits. This analysis revealed a significantly greater numbers of these features (i.e., valleys) on the MMN surface as compared to the MM surface. Finally, atomic force microscopy showed a rougher nano-texture on the MMN surface compared with the MM surface (p < 0.05). The results support recent published studies that show a combination of appropriate micron and nano surface results in a more robust cellular response and increased osteoblast differentiation. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 104B: 1083-1090, 2016.
Collapse
Affiliation(s)
- Jesse L Matteson
- Department of Materials Science and Engineering, Pennsylvania State University, University Park, Pennsylvania, 16802
| | | | - Timothy B Tighe
- Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, 16802
| | | | - Joshua J Stapleton
- Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, 16802
| |
Collapse
|
22
|
Enhanced differentiation of human osteoblasts on Ti surfaces pre-treated with human whole blood. Acta Biomater 2015; 19:180-90. [PMID: 25818948 DOI: 10.1016/j.actbio.2015.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/02/2015] [Accepted: 03/19/2015] [Indexed: 12/20/2022]
Abstract
Early and effective integration of a metal implant into bone tissue is of crucial importance for its long-term stability. While different material properties including surface roughness and wettability but also initial blood-implant surface interaction are known to influence this osseointegration, implications of the latter process are still poorly understood. In this study, early interaction between blood and the implant surface and how this affects the mechanism of osseointegration were investigated. For this, blood coagulation on a micro-roughened hydrophobic titanium (Ti) surface (SLA-H(phob)) and on a hydrophilic micro-roughened Ti surface with nanostructures (SLActive-H(phil)NS), as well as the effects of whole human blood pre-incubation of these two surfaces on the differentiation potential of primary human bone cells (HBC) was assessed. Interestingly, pre-incubation with blood resulted in a dense fibrin network over the entire surface on SLActive-H(phil)NS but only in single patches of fibrin and small isolated fibre complexes on SLA-H(phob). On SLActive-H(phil)NS, the number of HBCs attaching to the fibrin network was greatly increased and the cells displayed enhanced cell contact to the fibrin network. Notably, HBCs displayed increased expression of the osteogenic marker proteins alkaline phosphatase and collagen-I when cultivated on both surfaces upon blood pre-incubation. Additionally, blood pre-treatment promoted an earlier and enhanced mineralization of HBCs cultivated on SLActive-H(phil)NS compared to SLA-H(phob). The results presented in this study therefore suggest that blood pre-incubation of implant surfaces mimics a more physiological situation, eventually providing a more predictive in vitro model for the evaluation of novel bone implant surfaces.
Collapse
|
23
|
Mashinchian O, Turner LA, Dalby MJ, Laurent S, Shokrgozar MA, Bonakdar S, Imani M, Mahmoudi M. Regulation of stem cell fate by nanomaterial substrates. Nanomedicine (Lond) 2015; 10:829-47. [DOI: 10.2217/nnm.14.225] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Stem cells are increasingly studied because of their potential to underpin a range of novel therapies, including regenerative strategies, cell type-specific therapy and tissue repair, among others. Bionanomaterials can mimic the stem cell environment and modulate stem cell differentiation and proliferation. New advances in these fields are presented in this review. This work highlights the importance of topography and elasticity of the nano-/micro-environment, or niche, for the initiation and induction of stem cell differentiation and proliferation.
Collapse
Affiliation(s)
- Omid Mashinchian
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, PO Box 14177–55469, Tehran, Iran
| | - Lesley-Anne Turner
- Centre for Cell Engineering, Joseph Black Building, Institute of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Matthew J Dalby
- Centre for Cell Engineering, Joseph Black Building, Institute of Biomedical & Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Sophie Laurent
- Department of General, Organic & Biomedical Chemistry, NMR & Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, B-7000 Mons, Belgium
- CMMI – Center for Microscopy & Molecular Imaging, Rue Adrienne Bolland, 8, B-6041 Gosselies, Belgium
| | | | - Shahin Bonakdar
- National Cell Bank, Pasteur Institute of Iran, PO Box 13169–43551, Tehran, Iran
| | - Mohammad Imani
- Novel Drug Delivery Systems Department, Iran Polymer & Petrochemical Institute (IPPI), PO Box 14965/115, Tehran, Iran
| | - Morteza Mahmoudi
- Department of Nanotechnology & Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, PO Box 14155–6451, Tehran, Iran
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305–5101, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305–5101, USA
| |
Collapse
|
24
|
Zhao N, Zhu D. Collagen self-assembly on orthopedic magnesium biomaterials surface and subsequent bone cell attachment. PLoS One 2014; 9:e110420. [PMID: 25303459 PMCID: PMC4193861 DOI: 10.1371/journal.pone.0110420] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/22/2014] [Indexed: 11/18/2022] Open
Abstract
Magnesium (Mg) biomaterials are a new generation of biodegradable materials and have promising potential for orthopedic applications. After implantation in bone tissues, these materials will directly interact with extracellular matrix (ECM) biomolecules and bone cells. Type I collagen, the major component of bone ECM, forms the architecture scaffold that provides physical support for bone cell attachment. However, it is still unknown how Mg substrate affects collagen assembly on top of it as well as subsequent cell attachment and growth. Here, we studied the effects of collagen monomer concentration, pH, assembly time, and surface roughness of two Mg materials (pure Mg and AZ31) on collagen fibril formation. Results showed that formation of fibrils would not initiate until the monomer concentration reached a certain level depending on the type of Mg material. The thickness of collagen fibril increased with the increase of assembly time. The structures of collagen fibrils formed on semi-rough surfaces of Mg materials have a high similarity to that of native bone collagen. Next, cell attachment and growth after collagen assembly were examined. Materials with rough surface showed higher collagen adsorption but compromised bone cell attachment. Interestingly, surface roughness and collagen structure did not affect cell growth on AZ31 for up to a week. Findings from this work provide some insightful information on Mg-tissue interaction at the interface and guidance for future surface modifications of Mg biomaterials.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
- NSF Engineering Research Center-Revolutionizing Metallic Biomaterials, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Donghui Zhu
- Department of Chemical, Biological and Bio-Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
- NSF Engineering Research Center-Revolutionizing Metallic Biomaterials, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
25
|
Ikeda T, Kasai M, Tatsukawa E, Kamitakahara M, Shibata Y, Yokoi T, Nemoto TK, Ioku K. A bone substitute with high affinity for vitamin D-binding protein--relationship with niche of osteoclasts. J Cell Mol Med 2013; 18:170-80. [PMID: 24286277 PMCID: PMC3916128 DOI: 10.1111/jcmm.12180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/07/2013] [Indexed: 12/20/2022] Open
Abstract
The biological activity of osteoblasts and osteoclasts is regulated not only by hormones but also by local growth factors, which are expressed in neighbouring cells or included in bone matrix. Previously, we developed hydroxyapatite (HA) composed of rod-shaped particles using applied hydrothermal methods (HHA), and it revealed mild biodegradability and potent osteoclast homing activity. Here, we compared serum proteins adsorbed to HHA with those adsorbed to conventional HA composed of globular-shaped particles (CHA). The two ceramics adsorbed serum albumin and γ-globulin to similar extents, but affinity for γ-globulin was much greater than that to serum albumin. The chemotactic activity for macrophages of serum proteins adsorbed to HHA was significantly higher than that of serum proteins adsorbed to CHA. Quantitative proteomic analysis of adsorbed serum proteins revealed preferential binding of vitamin D-binding protein (DBP) and complements C3 and C4B with HHA. When implanted with the femur of 8-week-old rats, HHA contained significantly larger amount of DBP than CHA. The biological activity of DBP was analysed and it was found that the chemotactic activity for macrophages was weak. However, DBP-macrophage activating factor, which is generated by the digestion of sugar chains of DBP, stimulated osteoclastogenesis. These results confirm that the microstructure of hydroxyapatite largely affects the affinity for serum proteins, and suggest that DBP preferentially adsorbed to HA composed of rod-shaped particles influences its potent osteoclast homing activity and local bone metabolism.
Collapse
Affiliation(s)
- Tohru Ikeda
- Department of Oral Pathology and Bone Metabolism, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Thalji GN, Nares S, Cooper LF. Early molecular assessment of osseointegration in humans. Clin Oral Implants Res 2013; 25:1273-1285. [DOI: 10.1111/clr.12266] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2013] [Indexed: 11/29/2022]
Affiliation(s)
| | - Salvador Nares
- Department of Periodontics; University of North Carolina; Chapel Hill NC USA
| | - Lyndon F. Cooper
- Department of Prosthodontics; University of North Carolina; Chapel Hill NC USA
| |
Collapse
|
27
|
Oliveira NCM, Moura CCG, Zanetta-Barbosa D, Mendonça DBS, Cooper L, Mendonça G, Dechichi P. Effects of titanium surface anodization with CaP incorporation on human osteoblastic response. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:1958-62. [PMID: 23498218 PMCID: PMC4504235 DOI: 10.1016/j.msec.2013.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/06/2013] [Indexed: 01/26/2023]
Abstract
In this study we investigated whether anodization with calcium phosphate (CaP) incorporation (Vulcano®) enhances growth factors' secretion, osteoblast-specific gene expression, and cell viability, when compared to acid etched surfaces (Porous®) and machined surfaces (Screw®) after 3 and 7days. Results showed significant cell viability for Porous and Vulcano at day 7, when compared with Screw (p=0.005). At the same time point, significant differences regarding runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and bone sialoprotein (BSP) expression were found for all surfaces (p<0.05), but with greater fold induction for Porous and Vulcano. The secretion of transforming growth factor β1 (TGF-β1) and bone morphogenetic protein 2 (BMP-2) was not significantly affected by surface treatment in any experimental time (p>0.05). Although no significant correlation was found for growth factors' secretion and Runx2 expression, a significant positive correlation between this gene and ALP/BSP expression showed that their strong association is independent on the type of surface. The incorporation of CaP affected the biological parameters evaluated similar to surfaces just acid etched. The results presented here support the observations that roughness also may play an important role in determining cell response.
Collapse
|
28
|
Aboushelib MN, Osman E, Jansen I, Everts V, Feilzer AJ. Influence of a Nanoporous Zirconia Implant Surface of on Cell Viability of Human Osteoblasts. J Prosthodont 2013; 22:190-5. [DOI: 10.1111/j.1532-849x.2012.00920.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
29
|
Koo KT, Lee SW, Lee MH, Kim KH, Jung SH, Kang YG. Time-dependent expression of osteoblast marker genes in human primary cells cultured on microgrooved titanium substrata. Clin Oral Implants Res 2013; 25:714-22. [DOI: 10.1111/clr.12131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Ki-Tae Koo
- Department of Periodontology and Dental Research Institute; School of Dentistry; Seoul National University; Seoul Republic of Korea
| | - Suk W. Lee
- Department of Biomaterials & Prosthodontics; Kyung Hee University Hospital at Gangdong; Institute of Oral Biology; School of Dentistry; Kyung Hee University; Seoul Republic of Korea
| | - Myung-Hyun Lee
- Green Ceramics Division; Korea Institute of Ceramic Engineering and Technology; Seoul Republic of Korea
| | - Kyung H. Kim
- Core Research Laboratory; Clinical Research Institute; Kyung Hee University Hospital at Gangdong; Seoul Republic of Korea
| | - Su H. Jung
- Core Research Laboratory; Clinical Research Institute; Kyung Hee University Hospital at Gangdong; Seoul Republic of Korea
| | - Yoon G. Kang
- Department of Orthodontics; Kyung Hee University Hospital at Gangdong; Institute of Oral Biology; School of Dentistry; Kyung Hee University; Seoul Republic of Korea
| |
Collapse
|
30
|
Lee MH, Kang JH, Lee SW. The effect of surface microgrooves and anodic oxidation on the surface characteristics of titanium and the osteogenic activity of human periodontal ligament cells. Arch Oral Biol 2013; 58:59-66. [PMID: 22717323 DOI: 10.1016/j.archoralbio.2012.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 05/18/2012] [Accepted: 05/23/2012] [Indexed: 01/18/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate the effect of titanium (Ti) surface microgrooves and anodic oxidation on the surface characteristics of titanium and the osteogenic activity of human periodontal ligament cells (PLCs) cultured on theses surfaces. DESIGN Mechanically ground Ti was used as the control substratum (NE0). Truncated V-shaped microgrooves, 60μm-wide and 10μm-deep in cross-sections, were created on the Ti substrata by photolithography (NE60/10). Anodically oxidized Ti (NE0AO) and anodically oxidized microgrooved Ti (NE60/10AO) were also prepared. Scanning electron microscopy, X-ray diffraction (XRD), and X-ray photoelectron spectroscopy (XPS) were performed for surface characterization. Cell proliferation assay, osteoblast differentiation assay, and quantitative real-time PCR analysis were performed to compare the osteogenic activity of PLCs on NE0, NE60/10, NEAO, and NE60/10AO. RESULTS A decrease in the microgroove-width of NE60/10AO compared to NE60/10 due to Ti oxide layer generation by anodic oxidation was detected with XRD and XPS. Cell proliferation, osteoblast differentiation, and osteo-related gene expression were enhanced on the NE60/10AO substrata compared with NE0, NE60/10, and NE0AO. CONCLUSIONS The combination of Ti surface microgrooves and subsequent anodic oxidation treatment synergistically upregulated osteo-related gene expression, despite showing limited ability to increase cell proliferation and osteoblast differentiation levels in PLCs.
Collapse
Affiliation(s)
- Myung Hyun Lee
- Green Ceramics Division, Korea Institute of Ceramic Engineering and Technology, 77 10-gil, Digital-ro, Geumcheon-gu, Seoul 153-801, Republic of Korea
| | | | | |
Collapse
|
31
|
Ikeda T, Gonda Y, Tatsukawa E, Shibata Y, Kamitakahara M, Okuda T, Yonezawa I, Kurosawa H, Ioku K. Stimulation of Osteogenesis in Bone Defects Implanted with Biodegradable Hydroxyapatite Composed of Rod-Shaped Particles under Mechanical Unloading. Acta Histochem Cytochem 2012; 45:283-92. [PMID: 23209337 PMCID: PMC3496864 DOI: 10.1267/ahc.12012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/13/2012] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to evaluate the influence of mechanical unloading on the repair of bone defects with implantation of biodegradable bone substitutes. Spherical granules of biodegradable hydroxyapatite composed of rod-shaped particles (RHA) or beta-tricalcium phosphate composed of rod-shaped particles (RTCP) were implanted into a bone defect created in the distal end of the right femur of 8-week-old Wistar rats. Two, 6, 10, and 22 weeks after implantation, part of the sciatic nerve in the thigh was resected and exposed to mechanical unloading for 2 weeks. Then, 4, 8, 12 and 24 weeks after implantation, repair of the bone defect was analyzed. As a control, the bone defect without implantation of ceramic granules was also analyzed. Both RHA and RTCP tended to be reduced, but the reduction was not obvious during the experimental period. At 12 and 24 weeks after implantation, the amount of newly formed bone in the animal implanted with RHA was significantly greater than that at 4 weeks after implantation, but that in the animal implanted with RTCP or without implantation was not significantly different. The number of osteoclasts in the region implanted with RHA was significantly larger than that of the region implanted with RTCP or without implantation at 12 and 24 weeks. The activities of alkaline phosphatase in osteoblasts and tartrate-resistant acid phosphatase in osteoclasts were remarkably increased in the bone defects with implantation compared with those in the bone defects without implantation. These results suggested that RHA stimulated osteogenesis and osteoclastogenesis even after 2 weeks of mechanical unloading, and that RHA could be expected to improve the repair of bone defects in patients under the condition of skeletal unloading.
Collapse
Affiliation(s)
- Tohru Ikeda
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
| | - Yoshinori Gonda
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
- Present address: Department of Orthopedic Surgery, Juntendo University Urayasu Hospital
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
| | - Eri Tatsukawa
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
| | - Yasuaki Shibata
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
- Department of Oral Pathology and Bone Metabolism, Basic Medical Sciences Unit, Nagasaki University Graduate School of Biomedical Sciences
| | - Masanobu Kamitakahara
- Graduate School of Environmental Studies, Tohoku University
- Graduate School of Environmental Studies, Tohoku University
| | - Takatoshi Okuda
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
| | - Ikuho Yonezawa
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
| | - Hisashi Kurosawa
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
- Present address: Department of Orthopedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center
- Department of Orthopedic Surgery, School of Medicine, Juntendo University
- Present address: Department of Orthopedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center
| | - Koji Ioku
- Graduate School of Environmental Studies, Tohoku University
- Graduate School of Environmental Studies, Tohoku University
| |
Collapse
|
32
|
Rebl H, Finke B, Lange R, Weltmann KD, Nebe JB. Impact of plasma chemistry versus titanium surface topography on osteoblast orientation. Acta Biomater 2012; 8:3840-51. [PMID: 22705633 DOI: 10.1016/j.actbio.2012.06.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 06/04/2012] [Accepted: 06/08/2012] [Indexed: 11/18/2022]
Abstract
Topographical and chemical modifications of biomaterial surfaces both influence tissue physiology, but unfortunately little knowledge exists as to their combined effect. There are many indications that rough surfaces positively influence osteoblast behavior. Having determined previously that a positively charged, smooth titanium surface boosts osteoblast adhesion, we wanted to investigate the combined effects of topography and chemistry and elucidate which of these properties is dominant. Polished, machined and corundum-blasted titanium of increasing microroughness was additionally coated with plasma-polymerized allylamine (PPAAm). Collagen I was then immobilized using polyethylene glycol diacid and glutar dialdehyde. On all PPAAm-modified surfaces (i) adhesion of human MG-63 osteoblastic cells increased significantly in combination with roughness, (ii) cells resemble the underlying structure and melt with the surface, and (iii) cells overcome the restrictions of a grooved surface and spread out over a large area as indicated by actin staining. Interestingly, the cellular effects of the plasma-chemical surface modification are predominant over surface topography, especially in the initial phase. Collagen I, although it is the gold standard, does not improve surface adhesion features comparably.
Collapse
Affiliation(s)
- Henrike Rebl
- Department of Cell Biology, Biomedical Research Centre, University of Rostock, Rostock 18057, Germany
| | | | | | | | | |
Collapse
|
33
|
Gittens RA, Olivares-Navarrete R, McLachlan T, Cai Y, Hyzy SL, Schneider JM, Schwartz Z, Sandhage KH, Boyan BD. Differential responses of osteoblast lineage cells to nanotopographically-modified, microroughened titanium-aluminum-vanadium alloy surfaces. Biomaterials 2012; 33:8986-94. [PMID: 22989383 DOI: 10.1016/j.biomaterials.2012.08.059] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/24/2012] [Indexed: 12/31/2022]
Abstract
Surface structural modifications at the micrometer and nanometer scales have driven improved success rates of dental and orthopaedic implants by mimicking the hierarchical structure of bone. However, how initial osteoblast-lineage cells populating an implant surface respond to different hierarchical surface topographical cues remains to be elucidated, with bone marrow mesenchymal stem cells (MSCs) or immature osteoblasts as possible initial colonizers. Here we show that in the absence of any exogenous soluble factors, osteoblastic maturation of primary human osteoblasts (HOBs) but not osteoblastic differentiation of MSCs is strongly influenced by nanostructures superimposed onto a microrough Ti6Al4V (TiAlV) alloy. The sensitivity of osteoblasts to both surface microroughness and nanostructures led to a synergistic effect on maturation and local factor production. Osteoblastic differentiation of MSCs was sensitive to TiAlV surface microroughness with respect to production of differentiation markers, but no further enhancement was found when cultured on micro/nanostructured surfaces. Superposition of nanostructures to microroughened surfaces affected final MSC numbers and enhanced production of vascular endothelial growth factor (VEGF) but the magnitude of the response was lower than for HOB cultures. Our results suggest that the differentiation state of osteoblast-lineage cells determines the recognition of surface nanostructures and subsequent cell response, which has implications for clinical evaluation of new implant surface nanomodifications.
Collapse
Affiliation(s)
- Rolando A Gittens
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Li Y, Ma W, Feng Z, Wang Z, Zha N, Deng B, Zhao Y. Effects of irradiation on osteoblast-like cells on different titanium surfaces in vitro. J Biomed Mater Res B Appl Biomater 2012; 101:9-17. [PMID: 22987814 DOI: 10.1002/jbm.b.32803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/29/2012] [Accepted: 07/19/2012] [Indexed: 12/15/2022]
Abstract
The aim of this study was to investigate the effects of irradiation on adhesion ability, proliferation, and differentiation of MC3T3-E1 cells on microarc oxidation (MAO) titanium surfaces and polished titanium (PT) surfaces. MC3T3-E1 cells were exposed to a single dose at 2, 4, 6, 8, or 10 Gy using a (60) Co source, with tissue culture polystyrene plates chosen as controls. On all surfaces, irradiation resulted in a dose-dependent decrease in cellular proliferation. At 4 Gy dose, the cell proliferation of cells decreased by 17.8% on MAO and 18.6% on PT surfaces, respectively, compared with nonirradiated controls. Cells exposed to 8 Gy dose showed significant inhibition in collagen secretion and osteogenesis-related genes expression (OSX, COL-Iα1, and OCN). In contrast, irradiation increased cell adhesion to three surfaces dose dependently. It was also demonstrated that cells on MAO surface showed higher adhesion and collagen secretion than on PT surface at different radiation doses. This study revealed the effects of irradiation on osteoblasts in vitro on two titanium surfaces. MAO surface could be used in dental implants in irradiated bone due to enhanced adhesion ability and collagen secretion in osteoblasts.
Collapse
Affiliation(s)
- Yumei Li
- Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Pisanti P, Yeatts AB, Cardea S, Fisher JP, Reverchon E. Tubular perfusion system culture of human mesenchymal stem cells on poly-L-lactic acid scaffolds produced using a supercritical carbon dioxide-assisted process. J Biomed Mater Res A 2012; 100:2563-72. [PMID: 22528808 DOI: 10.1002/jbm.a.34191] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/14/2012] [Accepted: 03/19/2012] [Indexed: 11/12/2022]
Abstract
In vitro human mesenchymal stem cell (hMSC) proliferation and differentiation is dependent on scaffold design parameters and specific culture conditions. In this study, we investigate how scaffold microstructure influences hMSC behavior in a perfusion bioreactor system. Poly-L-lactic acid (PLLA) scaffolds are fabricated using supercritical carbon dioxide (SC-CO(2)) gel drying. This production method results in scaffolds fabricated with nanostructure. To introduce a microporous structure, porogen leaching was used in addition to this technique to produce scaffolds of average pore size of 100, 250, and 500 μm. These scaffolds were then cultured in static culture in well plates or dynamic culture in the tubular perfusion system (TPS) bioreactor. Results indicated that hMSCs were able to attach and maintain viability on all scaffolds with higher proliferation in the 250 μm and 500 μm pore sizes of bioreactor cultured scaffolds and 100 μm pore size of statically cultured scaffolds. Osteoblastic differentiation was enhanced in TPS culture as compared to static culture with the highest alkaline phosphatase expression observed in the 250 μm pore size group. Bone morphogenetic protein-2 was also analyzed and expression levels were highest in the 250 μm and 500 μm pore size bioreactor cultured samples. These results demonstrate cellular response to pore size as well as the ability of dynamic culture to enhance these effects.
Collapse
Affiliation(s)
- Paola Pisanti
- Department of Industrial Engineering, University of Salerno, Fisciano, Salerno, Italy
| | | | | | | | | |
Collapse
|
36
|
Lee MH, Kang JH, Lee SW. The significance of differential expression of genes and proteins in human primary cells caused by microgrooved biomaterial substrata. Biomaterials 2012; 33:3216-34. [PMID: 22285466 DOI: 10.1016/j.biomaterials.2012.01.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 01/14/2012] [Indexed: 01/18/2023]
Abstract
We demonstrate that etched microgrooves, with truncated V-shape in cross-section and subsequent acid etching, on titanium substrata alter the expression of various genes and proteins in human primary cells. Etched microgrooves with 30 or 60 μm width and 10 μm depth promoted human gingival fibroblast proliferation and significantly enhanced the osteoblast differentiation of human bone marrow-derived mesenchymal stem cells and human periodontal ligament cells by inducing differential expression of various genes involved in cell adhesion, migration, proliferation, mitosis, cytoskeletal reorganization, translation initiation, vesicular trafficking, proton transportation, transforming growth factor-β signaling, mitogen-activated protein kinase signaling, simvastatin's anabolic effect on bone, inhibitory guanine nucleotide binding protein (G protein)'s action, sumoylation pathway, survival/apoptosis, mitochondrial distribution, type I collagen production, osteoblast differentiation, and bone remodeling that were verified by the differential display PCR and quantitative real-time PCR. The most influential genes on the enhancement of fibroblast proliferation or osteoblast differentiation were determined by multiple regression analysis, and the expression of relevant proteins was confirmed by western blotting and protein quantitation.
Collapse
Affiliation(s)
- Myung Hyun Lee
- Green Ceramics Division, Korea Institute of Ceramic Engineering and Technology, 77 10-gil, Digital-ro, Geumcheon-gu, Seoul 153-801, Republic of Korea
| | | | | |
Collapse
|
37
|
Sitasuwan P, Andrew Lee L, Bo P, Davis EN, Lin Y, Wang Q. A plant virus substrate induces early upregulation of BMP2 for rapid bone formation. Integr Biol (Camb) 2012; 4:651-60. [DOI: 10.1039/c2ib20041d] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
The effects of titanium nitride-coating on the topographic and biological features of TPS implant surfaces. J Dent 2011; 39:720-8. [PMID: 21856369 DOI: 10.1016/j.jdent.2011.08.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 08/03/2011] [Accepted: 08/05/2011] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES Titanium nitride (TiN) coating has been proposed as an adjunctive surface treatment aimed to increase the physico-mechanical and aesthetic properties of dental implants. In this study we investigated the surface characteristics of TiN-coated titanium plasma sprayed (TiN-TPS) and uncoated titanium plasma sprayed (TPS) surfaces and their biological features towards both primary human bone marrow mesenchymal stem cells (BM-MSC) and bacterial cultures. METHODS 15 mm×1 mm TPS and TiN-TPS disks (P.H.I. s.r.l., San Vittore Olona, Milano, Italy) were topographically analysed by confocal optical profilometry. Primary human BM-MSC were obtained from healthy donors, isolated and expanded. Cells were seeded on the titanium disks and cell adhesion, proliferation, protein synthesis and osteoblastic differentiation in terms of alkaline phosphatase activity, osteocalcin synthesis and extracellular mineralization, were evaluated. Furthermore, adhesion and proliferation of Streptococcus pyogenes and Streptococcus sanguinis on both surfaces were also analysed. RESULTS TiN-TPS disks showed a decreased roughness (about 50%, p < 0.05) and a decreased bacterial adhesion and proliferation compared to TPS ones. No difference (p > 0.05) in terms of BM-MSC adhesion, proliferation and osteoblastic differentiation between TPS and TiN-TPS surfaces was found. CONCLUSIONS TiN coating showed to modify the topographical characteristics of TPS titanium surfaces and to significantly reduce bacterial adhesion and proliferation, although maintaining their biological affinity towards bone cell precursors.
Collapse
|