1
|
Özkabadayı Y, Türk M, Kumandaş A, Karahan S. Amino acid surface modified bioglass: A candidate biomaterial for bone tissue engineering 1. Microsc Res Tech 2025; 88:26-41. [PMID: 39154380 PMCID: PMC11652810 DOI: 10.1002/jemt.24659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024]
Abstract
Bioglasses are solid materials consisted of sodium oxide, calcium oxide, silicon dioxide and phosphorus in various proportions and have used in bone tissue engineering. There have been ongoing efforts to improve the surface properties of bioglasses to increase biocompatibility and performance. The aim of the present study is to modify the bioglass surface with an amino acid mixture consisting of arginine, aspartic acid, phenylalanine, cysteine, histidine and lysine, to characterize the surface, and to evaluate the performance and biocompatibility in vitro and in vivo. The untreated bioglass, bioglass kept in simulated body fluid (SBF), and modified bioglass were used in further evaluation. After confirmation of the surface modification with FT-IR analyses and SEM analyses, MC3T3-E1 preosteoblasts adhesion on the surface was also revealed by SEM. The modified bioglass had significantly higher ALP activity in colorimetric measurement, rate of calcium accumulations in Alizarin red s staining, lower rate of cell death in Annexin-V/PI staining to determine apoptosis and necrosis. Having higher cell viability rate in MTT test and absence of genotoxicity in micronucleus test (OECD 487), the modified bioglass was further confirmed for biocompatibility in vitro. The results of the rat tibial defect model revealed that the all bioglass treatments had a significantly better bone healing score compared to the untreated negative control. However, the modified bioglass exhibited significantly better bone healing efforts especially during the first and the second months compared to the other bioglass treatment treatments. As a result, the amino acid surface modification of bioglasses improves the surface biocompatibility and osteogenic performance that makes the amino acid modified bioglass a better candidate for bone tissue engineering. RESEARCH HIGHLIGHTS: Bioglass surface modification with amino acids contributes to bioglass-tissue interaction with an improved cell attachment. Modified bioglass increases in vitro Alp activity and calcium accumulation, and also positively affects cell behavior by supporting cell adaptation. Bioglass exerts osteogenic potential in vivo especially during early bone healing.
Collapse
Affiliation(s)
- Yasin Özkabadayı
- Faculty of Veterinary Medicine, Department of Histology and EmbryologyKirikkale UniversityKirikkaleTurkey
| | - Mustafa Türk
- Faculty of Engineering and Natural Sciences, Department of BioengineeringKirikkale UniversityKirikkaleTurkey
| | - Ali Kumandaş
- Faculty of Veterinary Medicine, Department of SurgeryKirikkale UniversityKirikkaleTurkey
- Faculty of Veterinary MedicineKyrgyz‐Turkish Manas UniversityBishkekKyrgyzstan
| | - Siyami Karahan
- Faculty of Veterinary Medicine, Department of Histology and EmbryologyKirikkale UniversityKirikkaleTurkey
| |
Collapse
|
2
|
Piatti E, Miola M, Verné E. Tailoring of bioactive glass and glass-ceramics properties for in vitro and in vivo response optimization: a review. Biomater Sci 2024; 12:4546-4589. [PMID: 39105508 DOI: 10.1039/d3bm01574b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Bioactive glasses are inorganic biocompatible materials that can find applications in many biomedical fields. The main application is bone and dental tissue engineering. However, some applications in contact with soft tissues are emerging. It is well known that both bulk (such as composition) and surface properties (such as morphology and wettability) of an implanted material influence the response of cells in contact with the implant. This review aims to elucidate and compare the main strategies that are employed to modulate cell behavior in contact with bioactive glasses. The first part of this review is focused on the doping of bioactive glasses with ions and drugs, which can be incorporated into the bioceramic to impart several therapeutic properties, such as osteogenic, proangiogenic, or/and antibacterial ones. The second part of this review is devoted to the chemical functionalization of bioactive glasses using drugs, extra-cellular matrix proteins, vitamins, and polyphenols. In the third and final part, the physical modifications of the surfaces of bioactive glasses are reviewed. Both top-down (removing materials from the surface, for example using laser treatment and etching strategies) and bottom-up (depositing materials on the surface, for example through the deposition of coatings) strategies are discussed.
Collapse
Affiliation(s)
- Elisa Piatti
- Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.
| | - Marta Miola
- Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.
| | - Enrica Verné
- Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
3
|
Akoa DM, Hélary C, Foda A, Chaussain C, Poliard A, Coradin T. Silicon impacts collagen remodelling and mineralization by human dental pulp stem cells in 3D pulp-like matrices. Dent Mater 2024; 40:1390-1399. [PMID: 38908960 DOI: 10.1016/j.dental.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVES Silicon-releasing biomaterials are widely used in the field of dentistry. However, unlike bone, very little is known about the role of silicon on dental tissue formation and repair. This study investigates the influence of silicic acid on the survival, differentiation and mineralizing ability of human dental pulp stem cells (hDPSCs) in 3D pulp-like environments METHODS: Dense type I collagen hydrogels seeded with hDPSCs were cultured over 4 weeks in the presence of silicic acid at physiological (10 μM) and supraphysiological (100 μM) concentrations. Cell viability and proliferation were studied by Alamar Blue and live/dead staining. The collagen network was investigated using second harmonic generation imaging. Mineral deposition was monitored by histology and scanning electron microscopy. Gene expression of mineralization- and matrix remodeling-associated proteins was studied by qPCR. RESULTS Presence of silicic acid did not show any significant influence on cell survival, metabolic activity and gene expression of key mineralization-related proteins (ALP, OCN, BSP). However, it induced enhanced cell clustering and delayed expression of matrix remodeling-associated proteins (MMP13, Col I). OPN expression and mineral deposition were inhibited at 100 μM. It could be inferred that silicic acid has no direct cellular effect but rather interacts with the collagen network, leading to a modification of the cell-matrix interface. SIGNIFICANCE Our results offer advanced insights on the possible role of silicic acid, as released by pulp capping calcium silicates biomaterials, in reparative dentine formation. More globally, these results interrogate the possible role of Si in pulp pathophysiology.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Asmaa Foda
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Catherine Chaussain
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France; AP-HP Service de médecine bucco-dentaire, Hôpital Bretonneau, Paris, France
| | - Anne Poliard
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France.
| |
Collapse
|
4
|
Phull S, Marx D, Akens MK, Ghert M, Towler MR. In vitroassessment of a gallium-doped glass polyalkenoate cement: chemotherapeutic potential, cytotoxicity and osteogenic effects. Biomed Mater 2024; 19:055006. [PMID: 38917820 DOI: 10.1088/1748-605x/ad5ba5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024]
Abstract
Metastatic bone lesions are often osteolytic, which causes advanced-stage cancer sufferers to experience severe pain and an increased risk of developing a pathological fracture. Gallium (Ga) ion possesses antineoplastic and anti-bone resorption properties, suggesting the potential for its local administration to impede the growth of metastatic bone lesions. This study investigated the chemotherapeutic potential, cytotoxicity, and osteogenic effects of a Ga-doped glass polyalkenoate cement (GPC) (C-TA2) compared to its non-gallium (C-TA0) counterpart. Ion release profiles revealed a biphasic pattern characterized by an initial burst followed by a gradually declining release of ions. C-TA2 continued to release Ga steadily throughout the experimentation period (7 d) and exhibited prolonged zinc (Zn) release compared to C-TA0. Interestingly, the Zn release from both GPCs appeared to cause a chemotherapeutic effect against H1092 lung cancer cellsin vitro, with the prolonged Zn release from C-TA2 extending this effect. Unfortunately, both GPCs enhanced the viability of HCC2218 breast cancer cells, suggesting that the chemotherapeutic effects of Zn could be tied to cellular differences in preferred Zn concentrations. The utilization of SAOS-2 and MC3T3 cell lines as bone cell models yielded conflicting results, with the substantial decline in MC3T3 viability closely associated with silicon (Si) release, indicating cellular variations in Si toxicity. Despite this ambiguity, both GPCs exhibited harmful effects on the osteogenesis of primary rat osteoblasts, raising concerns about excessive burst Zn release. While Ga/Zn-doped GPCs hold promise for treating metastatic bone lesions caused by lung cancers, further optimization is required to mitigate cytotoxicity on healthy bone.
Collapse
Affiliation(s)
- Sunjeev Phull
- Department of Mechanical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Daniella Marx
- Department of Mechanical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Margarete K Akens
- University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Michelle Ghert
- Department of Surgery, McMaster University, Hamilton L8V 5C2, ON, Canada
| | - Mark R Towler
- Department of Chemical & Biochemical Engineering, Missouri S&T, Rolla, MO, United States of America
| |
Collapse
|
5
|
Xu H, Cui Y, Tian Y, Dou M, Sun S, Wang J, Wu D. Nanoparticle-Based Drug Delivery Systems for Enhancing Bone Regeneration. ACS Biomater Sci Eng 2024; 10:1302-1322. [PMID: 38346448 DOI: 10.1021/acsbiomaterials.3c01643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The treatment of bone defects has been a long-standing challenge in clinical practice. Among the various bone tissue engineering approaches, there has been substantial progress in the development of drug delivery systems based on functional drugs and appropriate carrier materials owing to technological advances in recent years. A large number of materials based on functional nanocarriers have been developed and applied to improve the complex osteogenic microenvironment, including for promoting osteogenic activity, inhibiting osteoclast activity, and exerting certain antibacterial effects. This Review discusses the physicochemical properties, drug loading mechanisms, advantages and disadvantages of nanoparticles (NPs) used for constructing drug delivery systems. In addition, we provide an overview of the osteogenic microenvironment regulation mechanism of drug delivery systems based on nanoparticle (NP) carriers and the construction strategies of drug delivery systems. Finally, the advantages and disadvantages of NP carriers are summarized along with their prospects and future research trends in bone tissue engineering. This Review thus provides advanced strategies for the design and application of drug delivery systems based on NPs in the treatment of bone defects.
Collapse
Affiliation(s)
- Hang Xu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yutao Cui
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yuhang Tian
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Minghan Dou
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Shouye Sun
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Jingwei Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Dankai Wu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| |
Collapse
|
6
|
Hashimoto K, Oiwa M, Shibata H. Effect of Silicon Dioxide Nanoparticles on the Sintering Properties of Beta-Tricalcium Phosphate Composites. MATERIALS (BASEL, SWITZERLAND) 2024; 17:797. [PMID: 38399047 PMCID: PMC10890285 DOI: 10.3390/ma17040797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Composite sintered bodies comprising silicon dioxide (SiO2) nanoparticles dispersed in β-tricalcium phosphate (β-TCP) were prepared. The addition of nano-sized colloidal SiO2 to the β-TCP produced well-dispersed secondary phase nanoparticles that promoted densification by suppressing grain growth and increasing linear shrinkage of the sintered bodies. The SiO2 was found not to react with the β-TCP at 1120 °C and the substitution of silicon for phosphorous to produce a solid solution did not occur. This lack of a reaction is ascribed to the absence of available calcium ions to compensate for the increase in charge associated with this substitution. The SiO2 nanoparticles were found to be present near the intersections of grain boundaries in the β-TCP. β-TCP composite sintered body containing 2.0 and 4.0 wt% SiO2 exhibited a bending strength comparable to that of cortical bone and hence could potentially be used as a bone filling material.
Collapse
Affiliation(s)
- Kazuaki Hashimoto
- Department of Applied Chemistry, Faculty of Engineering, Chiba Institute of Technology, 2-17-1 Tsudanuma, Narashino-shi 275-0016, Chiba, Japan; (M.O.); (H.S.)
| | | | | |
Collapse
|
7
|
Pritchard A, Nielsen BD. Silicon Supplementation for Bone Health: An Umbrella Review Attempting to Translate from Animals to Humans. Nutrients 2024; 16:339. [PMID: 38337624 PMCID: PMC10857027 DOI: 10.3390/nu16030339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
Studies have attempted to demonstrate the benefits of silicon on bone health using a wide range of Si amounts-provided in the diet or through supplementation-and several different animal species. Previous studies in humans have also demonstrated a positive correlation between Si intake and bone health measures. The aim of the current review is to determine the effective levels of Si intake or supplementation that influence bone health to better inform future study designs and guidelines. Articles were identified using one of two search terms: "silicon AND bone" or "sodium zeolite A AND bone". Articles were included if the article was a controlled research study on the effect of Si on bone health and/or mineral metabolism and was in English. Articles were excluded if the article included human subjects, was in vitro, or studied silica grafts for bone injuries. Silicon type, group name, Si intake from diet, Si supplementation amount, animal, and age at the start were extracted when available. Dietary Si intake, Si supplementation amount, and the amount of Si standardized on a kg BW basis were calculated and presented as overall mean ± standard deviations, medians, minimums, and maximums. Studies that left out animal weights, amount of food or water consumed, or nutrient profiles of the basal diet were excluded from these calculations. Standardized Si intakes ranged from 0.003 to 863 mg/kg BW, at times vastly exceeding current human Si intake recommendations (25 mg/d). The lack of data provided by the literature made definitively determining an effective threshold of supplementation for skeletal health difficult. However, it appears that Si consistently positively influences bone and mineral metabolism by around 139 mg Si/kg BW/d, which is likely unfeasible to attain in humans and large animal species. Future studies should examine this proposed threshold more directly and standardize supplemental or dietary Si intakes to kg BW for better study replication and translation.
Collapse
Affiliation(s)
- Abby Pritchard
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA;
- Regulatory and Nutritional Compliance, Mars Petcare, Franklin, TN 37067, USA
| | - Brian D. Nielsen
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
8
|
He J, Liu Y, Zeng X, Tong Y, Liu R, Wang K, Shangguan X, Qiu G, Sipaut CS. Silicon Nitride Bioceramics Sintered by Microwave Exhibit Excellent Mechanical Properties, Cytocompatibility In Vitro, and Anti-Bacterial Properties. J Funct Biomater 2023; 14:552. [PMID: 37998121 PMCID: PMC10671902 DOI: 10.3390/jfb14110552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Silicon nitride is a bioceramic with great potential, and multiple studies have demonstrated its biocompatibility and antibacterial properties. In this study, silicon nitride was prepared by a microwave sintering technique that was different from common production methods. SEM and pore distribution analysis revealed the microstructure of microwave-sintered silicon nitride with obvious pores. Mechanical performance analysis shows that microwave sintering can improve the mechanical properties of silicon nitride. The CCK-8 method was used to demonstrate that microwave-sintered silicon nitride has no cytotoxicity and good cytocompatibility. From SEM and CLSM observations, it was observed that there was good adhesion and cross-linking of cells during microwave-sintered silicon nitride, and the morphology of the cytoskeleton was good. Microwave-sintered silicon nitride has been proven to be non-cytotoxic. In addition, the antibacterial ability of microwave-sintered silicon nitride against Staphylococcus aureus and Escherichia coli was tested, proving that it has a good antibacterial ability similar to the silicon nitride prepared by commonly used processes. Compared with silicon nitride prepared by gas pressure sintering technology, microwave-sintered silicon nitride has excellent performance in mechanical properties, cell compatibility, and antibacterial properties. This indicates its enormous potential as a substitute material for manufacturing bone implants.
Collapse
Affiliation(s)
- Jiayu He
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Yuandong Liu
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Xiaofeng Zeng
- Hengyang Kaixin Special Material Technology Co., Ltd., Hengyang 421200, China;
- Faculty of Engineering, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Yan Tong
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Run Liu
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Kan Wang
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Xiangdong Shangguan
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | - Guanzhou Qiu
- Key Laboratory of Biohydrometallurgy of Ministry of Education, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China; (J.H.); (Y.T.); (R.L.); (K.W.); (X.S.); (G.Q.)
| | | |
Collapse
|
9
|
Nitu, Fopase R, Pandey LM, Seal P, Borah JP, Srinivasan A. Assessment of sol-gel derived iron oxide substituted 45S5 bioglass-ceramics for biomedical applications. J Mater Chem B 2023; 11:7502-7513. [PMID: 37458109 DOI: 10.1039/d3tb00287j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Magnetic bioactive glass-ceramic (MGC) powders with nominal compositions of (45 - x)SiO224.5CaO24.5Na2O6P2O5xFe2O3 (x = 2, 4, 6, 8, 10, and 15 wt%) have been synthesized by a sol-gel route by systematically substituting silicon dioxide with iron oxide in Hench's 45S5 glass composition. Powder X-ray diffraction studies revealed a variation in the percentage of combeite (Ca2Na2Si3O9), magnetite (Fe3O4), and hematite (Fe2O3) nanocrystalline phases in MGC powders as a function of composition. Zeta potential measurements showed that MGC containing up to 10 wt% iron oxide formed stable suspensions. The saturation magnetization and heat generation capacity of MGC fluids increased with an increase in iron oxide content. Degradation of MGC powders was investigated in phosphate buffered saline (PBS). The in vitro bioactivity of the MGC powders taken in pellet form was confirmed by observing the pH variation as well as hydroxyapatite layer (HAp) formation upon soaking in modified simulated body fluid. These studies showed a decrement in the overall bioactivity in samples with high iron oxide content due to the proportional decrease in the silanol group. Monitoring the proliferation of MG-63 osteoblast cells in Dulbecco's Modified Eagle Medium (DMEM) revealed that MGC with up to 10 wt% iron oxide exhibited acceptable viability. The systematic study revealed that the MGC with 10 wt% iron oxide exhibited optimal cell viability, magnetic properties and induction heating capacity, which were better than those of FluidMag-CT, which is used for hyperthermia treatment.
Collapse
Affiliation(s)
- Nitu
- Department of Physics, Indian Institute of Technology Guwahati, Guwahati 781039, India.
| | - Rushikesh Fopase
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Lalit Mohan Pandey
- Bio-interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Papori Seal
- Department of Science and Humanities, National Institute of Technology Nagaland, 797103, India
| | - Jyoti Prasad Borah
- Department of Science and Humanities, National Institute of Technology Nagaland, 797103, India
| | | |
Collapse
|
10
|
Skalny AV, Aschner M, Silina EV, Stupin VA, Zaitsev ON, Sotnikova TI, Tazina SI, Zhang F, Guo X, Tinkov AA. The Role of Trace Elements and Minerals in Osteoporosis: A Review of Epidemiological and Laboratory Findings. Biomolecules 2023; 13:1006. [PMID: 37371586 DOI: 10.3390/biom13061006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The objective of the present study was to review recent epidemiological and clinical data on the association between selected minerals and trace elements and osteoporosis, as well as to discuss the molecular mechanisms underlying these associations. We have performed a search in the PubMed-Medline and Google Scholar databases using the MeSH terms "osteoporosis", "osteogenesis", "osteoblast", "osteoclast", and "osteocyte" in association with the names of particular trace elements and minerals through 21 March 2023. The data demonstrate that physiological and nutritional levels of trace elements and minerals promote osteogenic differentiation through the up-regulation of BMP-2 and Wnt/β-catenin signaling, as well as other pathways. miRNA and epigenetic effects were also involved in the regulation of the osteogenic effects of trace minerals. The antiresorptive effect of trace elements and minerals was associated with the inhibition of osteoclastogenesis. At the same time, the effect of trace elements and minerals on bone health appeared to be dose-dependent with low doses promoting an osteogenic effect, whereas high doses exerted opposite effects which promoted bone resorption and impaired bone formation. Concomitant with the results of the laboratory studies, several clinical trials and epidemiological studies demonstrated that supplementation with Zn, Mg, F, and Sr may improve bone quality, thus inducing antiosteoporotic effects.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia
- Center of Bioelementology and Human Ecology, Institute of Biodesign and Modeling of Complex Systems, Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ekaterina V Silina
- Center of Bioelementology and Human Ecology, Institute of Biodesign and Modeling of Complex Systems, Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Victor A Stupin
- Department of Hospital Surgery No. 1, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Oleg N Zaitsev
- Department of Physical Education, Yaroslavl State Technical University, 150023 Yaroslavl, Russia
| | - Tatiana I Sotnikova
- Center of Bioelementology and Human Ecology, Institute of Biodesign and Modeling of Complex Systems, Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- City Clinical Hospital n. a. S.P. Botkin of the Moscow City Health Department, 125284 Moscow, Russia
| | - Serafima Ia Tazina
- Center of Bioelementology and Human Ecology, Institute of Biodesign and Modeling of Complex Systems, Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Health Science Center, School of Public Health, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiong Guo
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Health Science Center, School of Public Health, Xi'an Jiaotong University, Xi'an 710061, China
| | - Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia
- Center of Bioelementology and Human Ecology, Institute of Biodesign and Modeling of Complex Systems, Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
11
|
Moretti A, Liguori S, Paoletta M, Migliaccio S, Toro G, Gimigliano F, Iolascon G. Bone fragility during the COVID-19 pandemic: the role of macro- and micronutrients. Ther Adv Musculoskelet Dis 2023; 15:1759720X231158200. [PMID: 36937822 PMCID: PMC10015293 DOI: 10.1177/1759720x231158200] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/01/2023] [Indexed: 03/18/2023] Open
Abstract
Bone fragility is the susceptibility to fracture due to poor bone strength. This condition is usually associated with aging, comorbidities, disability, poor quality of life, and increased mortality. International guidelines for the management of patients with bone fragility include a nutritional approach, mainly aiming at optimal protein, calcium, and vitamin D intakes. Several biomechanical features of the skeleton, such as bone mineral density (BMD), trabecular and cortical microarchitecture, seem to be positively influenced by micro- and macronutrient intake. Patients with major fragility fractures are usually poor consumers of dairy products, fruit, and vegetables as well as of nutrients modulating gut microbiota. The COVID-19 pandemic has further aggravated the health status of patients with skeletal fragility, also in terms of unhealthy dietary patterns that might adversely affect bone health. In this narrative review, we discuss the role of macro- and micronutrients in patients with bone fragility during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Antimo Moretti
- Department of Medical and Surgical Specialties
and Dentistry, University of Campania “Luigi Vanvitelli”, 80138 Naples,
Italy
| | - Sara Liguori
- Department of Medical and Surgical Specialties
and Dentistry, University of Campania “Luigi Vanvitelli”, Naples,
Italy
| | - Marco Paoletta
- Department of Medical and Surgical Specialties
and Dentistry, University of Campania “Luigi Vanvitelli”, Naples,
Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health
Sciences, University of Rome “Foro Italico”, Rome, Italy
| | - Giuseppe Toro
- Department of Medical and Surgical Specialties
and Dentistry, University of Campania “Luigi Vanvitelli”, Naples,
Italy
| | - Francesca Gimigliano
- Department of Physical and Mental Health and
Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples,
Italy
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties
and Dentistry, University of Campania “Luigi Vanvitelli”, Naples,
Italy
| |
Collapse
|
12
|
Toro-Valdivieso C, Jugdaohsingh R, Powell JJ, Hoffman JI, Forcada J, Moore C, Blacklaws B. Heavy metal contamination in pristine environments: lessons from the Juan Fernandez fur seal ( Arctocephalus philippii philippii). ROYAL SOCIETY OPEN SCIENCE 2023; 10:221237. [PMID: 36998770 PMCID: PMC10049756 DOI: 10.1098/rsos.221237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/06/2023] [Indexed: 06/19/2023]
Abstract
Heavy metals, including mercury (Hg) and cadmium (Cd), occur naturally or anthropogenically and are considered toxic to the environment and human health. However, studies on heavy metal contamination focus on locations close to industrialized settlements, while isolated environments with little human activity are often ignored due to perceived low risk. This study reports heavy metal exposure in Juan Fernandez fur seals (JFFS), a marine mammal endemic to an isolated and relatively pristine archipelago off the coast of Chile. We found exceptionally high concentrations of Cd and Hg in JFFS faeces. Indeed, they are among the highest reported for any mammalian species. Following analysis of their prey, we concluded that diet is the most likely source of Cd contamination in JFFS. Furthermore, Cd appears to be absorbed and incorporated into JFFS bones. However, it was not associated with mineral changes observed in other species, suggesting Cd tolerance/adaptations in JFFS bones. The high levels of silicon found in JFFS bones may counteract the effects of Cd. These findings are relevant to biomedical research, food security and the treatment of heavy metal contamination. It also contributes to understanding the ecological role of JFFS and highlights the need for surveillance of apparently pristine environments.
Collapse
Affiliation(s)
| | - Ravin Jugdaohsingh
- Biominerals Research Laboratory, Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, UK
| | - Jonathan J. Powell
- Biominerals Research Laboratory, Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, UK
| | - Joseph I. Hoffman
- British Antarctic Survey, High Cross, Madingley Rd, Cambridge CB3 0ET, UK
- Department of Animal Behaviour, Bielefeld University, Bielefeld 33501, Germany
| | - Jaume Forcada
- British Antarctic Survey, High Cross, Madingley Rd, Cambridge CB3 0ET, UK
| | - Charles Moore
- Algalita Marine Research Foundation, 148N Marina Dr, Long Beach, CA 90803, USA
| | - Barbara Blacklaws
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, UK
| |
Collapse
|
13
|
Physicochemical and biological characterization of silica-coated alumina particles. Dent Mater 2022; 38:1878-1885. [PMID: 36207169 DOI: 10.1016/j.dental.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVES A tribochemical silica-coating (TSC) method has been developed to improve the adhesion of dental resin composites to various substrates. The method utilizes airborne-particle abrasion using particles having a silica surface and an alumina core. The impact of the TSC method has been extensively studied but less attention has been paid to the characterization of the silica-modified alumina particles. Due to the role of silicate ions in cell biology, e.g. osteoblast function and bone mineralization, silica-modified alumina particles could also be potentially used as a biomaterial in scaffolds of tissue regeneration. Thus, we carried out detailed physicochemical characterization of the silica-modified alumina particles. METHODS Silica-modified alumina particles (Rocatec, 3 M-ESPE) of an average particle size of 30 µm were studied for the phase composition, spectroscopic properties, surface morphology, dissolution, and the capability to modify the pH of an immersion solution. The control material was alumina without silica modification. Pre-osteoblastic MC3T3-E1 cells were used to assess cell viability in the presence of the particles. Cell viability was tested at 1, 3, 7 and 10 days of culture with various particle quantities. Multivariate ANOVA was used for statistical analyses. RESULTS Minor quantities of silica enrichment was verified on the surface of alumina particles and the silica did not evenly cover the alumina surface. In the dissolution test, no change in the pH of the immersion solution was observed in the presence of the particles. Minor quantities of silicate ions were dissolved from the particles to the cell culture medium but no major differences were observed in the viability of pre-osteoblastic cells, whether the cells were cultured with silica-modified or plain alumina particles. SIGNIFICANCE Characterization of silica-modified alumina particles demonstrated differences in the particle surface structure compared to control alumina. Dissolution of silica layer in Tris buffer or SBF solution varied from that of cell culture medium: minor quantities of dissolved Si were observed in cell culture test medium. The cell viability test did not shown significant differences between control alumina and its silica-modified counterpart.
Collapse
|
14
|
The Influence of the Matrix on the Apatite-Forming Ability of Calcium Containing Polydimethylsiloxane-Based Cements for Endodontics. Molecules 2022; 27:molecules27185750. [PMID: 36144487 PMCID: PMC9504520 DOI: 10.3390/molecules27185750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
This study aimed to characterize the chemical properties and bioactivity of an endodontic sealer (GuttaFlow Bioseal) based on polydimethylsiloxane (PDMS) and containing a calcium bioglass as a doping agent. Commercial PDMS-based cement free from calcium bioglass (GuttaFlow 2 and RoekoSeal) were characterized for comparison as well as GuttaFlow 2 doped with dicalcium phosphate dihydrate, hydroxyapatite, or a tricalcium silicate-based cement. IR and Raman analyses were performed on fresh materials as well as after aging tests in Hank’s Balanced Salt Solution (28 d, 37 °C). Under these conditions, the strengthening of the 970 cm−1 Raman band and the appearance of the IR components at 1455−1414, 1015, 868, and 600−559 cm−1 revealed the deposition of B-type carbonated apatite. The Raman I970/I638 and IR A1010/A1258 ratios (markers of apatite-forming ability) showed that bioactivity decreased along with the series: GuttaFlow Bioseal > GuttaFlow 2 > RoekoSeal. The PDMS matrix played a relevant role in bioactivity; in GuttaFlow 2, the crosslinking degree was favorable for Ca2+ adsorption/complexation and the formation of a thin calcium phosphate layer. In the less crosslinked RoekoSeal, such processes did not occur. The doped cements showed bioactivity higher than GuttaFlow 2, suggesting that the particles of the mineralizing agents are spontaneously exposed on the cement surface, although the hydrophobicity of the PDMS matrix slowed down apatite deposition. Relevant properties in the endodontic practice (i.e., setting time, radiopacity, apatite-forming ability) were related to material composition and the crosslinking degree.
Collapse
|
15
|
In Vivo Application of Silica-Derived Inks for Bone Tissue Engineering: A 10-Year Systematic Review. Bioengineering (Basel) 2022; 9:bioengineering9080388. [PMID: 36004914 PMCID: PMC9404869 DOI: 10.3390/bioengineering9080388] [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: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
As the need for efficient, sustainable, customizable, handy and affordable substitute materials for bone repair is critical, this systematic review aimed to assess the use and outcomes of silica-derived inks to promote in vivo bone regeneration. An algorithmic selection of articles was performed following the PRISMA guidelines and PICO method. After the initial selection, 51 articles were included. Silicon in ink formulations was mostly found to be in either the native material, but associated with a secondary role, or to be a crucial additive element used to dope an existing material. The inks and materials presented here were essentially extrusion-based 3D-printed (80%), and, overall, the most investigated animal model was the rabbit (65%) with a femoral defect (51%). Quality (ARRIVE 2.0) and risk of bias (SYRCLE) assessments outlined that although a large majority of ARRIVE items were “reported”, most risks of bias were left “unclear” due to a lack of precise information. Almost all studies, despite a broad range of strategies and formulations, reported their silica-derived material to improve bone regeneration. The rising number of publications over the past few years highlights Si as a leverage element for bone tissue engineering to closely consider in the future.
Collapse
|
16
|
Bavya Devi K, Lalzawmliana V, Saidivya M, Kumar V, Roy M, Kumar Nandi S. Magnesium Phosphate Bioceramics for Bone Tissue Engineering. CHEM REC 2022; 22:e202200136. [PMID: 35866502 DOI: 10.1002/tcr.202200136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/01/2022] [Indexed: 11/11/2022]
Abstract
Magnesium phosphate (MgP) is a family of newly developed resorbable bioceramics for bone tissue engineering. Although calcium phosphates (CaP) are the most commonly used bioceramics, low solubility, and slow degradation, when implanted in vivo, are their main drawbacks. Magnesium (Mg) is an essential element in the human body as it plays important role in bone metabolism, DNA stabilization, and skeletal development. Recent research on magnesium phosphates has established their higher degradability, in vitro, and in vivo biocompatibility. Compared to CaP, very limited research work has been found in the area of MgP. The prime goal of this review is to bring out the importance of magnesium phosphate ceramics for biomedical applications. In this review, we have discussed the synthesis methods, mechanical properties, in vitro and in vivo biocompatibility of MgP bioceramics. Moreover, we have highlighted the recent developments in metal ion-doped MgPs and MgP scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- K Bavya Devi
- Department of Chemistry, Thassim Beevi Abdul Kader College for Women, 623517, Kilakarai, Ramanathapuram, India
| | - V Lalzawmliana
- Department of Veterinary Surgery and Radiology, College of Veterinary Sciences and Animal Husbandry, 799008, R. K. Nagar, Tripura West, India
| | - Maktumkari Saidivya
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology-Kharagpur, 721302, Kharagpur, India
| | - Vinod Kumar
- Department of Veterinary Clinical Complex, Faculty of Veterinary & Animal Sciences, Banaras Hindu University, pin-221005, Mirzapur, India
| | - Mangal Roy
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology-Kharagpur, 721302, Kharagpur, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, 700037, Kolkata, India
| |
Collapse
|
17
|
Saghiri MA, Vakhnovetsky J, Vakhnovetsky A. Functional role of inorganic trace elements in dentin apatite-Part II: Copper, manganese, silicon, and lithium. J Trace Elem Med Biol 2022; 72:126995. [PMID: 35605438 DOI: 10.1016/j.jtemb.2022.126995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/12/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022]
Abstract
Trace elements are recognized as being essential in dentin and bone apatite. The effects of zinc, strontium, magnesium, and iron were discussed in part I. In part II, we evaluated the functional role of copper, manganese, silicon, and lithium on dentin apatite, with critical effects on morphology, crystallinity, and solubility. An electronic search was performed on the role of these trace elements in dentin apatite from January 2000 to January 2022. The recent aspects of the relationship between four different trace elements and their critical role in the structure and mechanics of dentin were assessed. These findings show that elements play a vital role in the human body, especially in the crystalline structure of dentin apatite. Copper presents immense benefits in dental restorative biomaterials because of its importance in enhancing odontogenesis. The biological role of manganese in dentin apatite is still largely unknown, but it has gained attention for many of its broad physiological functions such as modulating osteoblast proliferation, differentiation, and metabolism in bones. The functional role of silicon in dentin apatite is similarly lacking, but findings reveal its importance in mineralization and collagen formation, making it useful for the field of restorative dentistry. Likewise, lithium was found to have important roles in dentin mineralization as well as in the formation of dentin bridges and tissues. Therefore, there is growing importance in studying the aforementioned elements in the context of dentin apatite.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Director, Biomaterial and Prosthodontics Laboratory and Assistant Professor, Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, United States; Adjunct Assistant Professor, Department of Endodontics, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, United States.
| | - Julia Vakhnovetsky
- Visiting Researcher, Sector of Angiogenesis Regenerative Medicine, Dr. Hajar Afsar Lajevardi Research Cluster (DHAL), Hackensack, NJ, United States; Pre-Dental Student, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Anna Vakhnovetsky
- Pre-Medical Student, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
18
|
Yu Y, Li T, Wang X, Zhang M, Yu Q, Chen H, Zhang D, Yan C. Structural characterization and anti-osteoporosis activity of two polysaccharides extracted from the rhizome of Curculigo orchioides. Food Funct 2022; 13:6749-6761. [PMID: 35661847 DOI: 10.1039/d2fo00720g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Curculigo orchioides is widely used to treat osteoporosis in China. In this study, we identified the active substances in the crude polysaccharide (CO50) from C. orchioides that had anti-osteoporosis activity in vivo. Two polysaccharides, COP50-1 and COP50-4, were purified from CO50. Based on structural analysis, COP50-1 was composed of α-D-Glcp-(1→, β-D-Galp-(1→, →4)-α-D-Glcp-(1→, →3,4)-α-D-Glcp-(1→, →4,6)-α-D-Glcp-(1→, →4,6)-β-D-Manp-(1→, whereas COP50-4 was composed of α-L-Araf-(1→, →2)-α-L-Rhap-(1→, β-D-Manp-(1→, α-D-Galp-(1→, →2,4)-α-L-Rhap-(1→, →2)-β-D-Manp-(1→, →4)-α-D-GlcAp-(1→, →3)-α-D-GalAp-(1→, →4,6)-α-D-Galp-(1→, →2,3,6)-β-D-Manp-(1→, →2,3,5)-α-L-Araf-(1→, →2,5)-α-L-Araf-(1→, →4)-α-D-Glcp-(1→ and →3)-α-D-Galp-(1→. Pharmacological assessment revealed that COP50-1 had no obvious osteogenic activity. However, COP50-4 (0.5 μM) significantly enhanced the differentiation and mineralization of osteoblasts in vitro. Moreover, the effect of COP50-4 was greater than that of 17β-estradiol. Therefore, COP50-4 may be an effective component of CO50 that has great potential for development as an alternative drug for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yongbo Yu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Tianyu Li
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Xueqian Wang
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Mengliu Zhang
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qian Yu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dawei Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chunyan Yan
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
19
|
Effects of Diatomite Contents on Microstructure, Microhardness, Bioactivity and Biocompatibility of Gradient Bioceramic Coating Prepared by Laser Cladding. METALS 2022. [DOI: 10.3390/met12060931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Biometallic materials are widely used in medicine because of excellent mechanical properties. However, biometallic materials are limited in the application of biomaterials due to their lack of bioactivity. To solve this problem, a gradient bioceramic coating doped with diatomite (DE) was successfully fabricated on the surface of Ti6Al4V alloy by using the broadband-laser cladding process to improve the bioactivity of metal materials. As well as the DE contents on the microstructure, microhardness, bioactivity and biocompatibility were investigated. The experimental results demonstrate that the addition of moderate amounts of DE is effective in reducing the number of cracks. The X-ray diffraction (XRD) results reveal that the bioceramic coating doped with DE mainly consists of CaTiO3, hydroxyapatite (HA), tricalcium phosphate (TCP) and silicate, and that the amount of HA and TCP in the coating reached maximum when the bioceramic coating was doped with 10wt% DE. The bioceramic coating doped with 10wt% DE has favorable ability to deposit bone-like apatite. These results indicate that the addition of DE can improve cracking sensibility, bioactivity and biocompatibility of the coating.
Collapse
|
20
|
Kong Y, Zhang X, Ma X, Wu L, Chen D, Su B, Liu D, Wang X. Silicon-substituted calcium phosphate promotes osteogenic-angiogenic coupling by activating the TLR4/PI3K/AKT signaling axis. J Biomater Appl 2022; 37:459-473. [PMID: 35623361 DOI: 10.1177/08853282221105303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Silicon-substituted calcium phosphate (Si-CaP) is a promising bioactive material for bone tissue engineering. The mechanism of Si-CaP regulates osteogenic-angiogenic coupling during bone regeneration has not been fully elucidated. In this study, we screened the targets of Si-CaP and osteogenic-angiogenic coupling. 83 common genes were regarded as key targets for Si-CaP regulation of the osteogenic-angiogenic coupling. Then, we performed protein-protein interaction analysis, GO and KEGG enrichment analysis of these 83 targets to further predict their molecular mechanism. Our results showed that Si-CaP treatment could regulate the osteogenic-angiogenic coupling by up-regulating the expression of Toll-like receptor 4 (TLR4), and the phosphorylation of AKT which in turn activating the PI3K/AKT signaling pathway, promoting the expression of RUNX2, OPN, VEGF. In addition, we also found that TLR4 siRNA treatment could block the PI3K/AKT signaling pathway, while inhibiting the promoting effect of Si-CaP. However, although LY294002 can achieve the same inhibitory effect as TLR4 siRNA by blocking the PI3K/AKT signaling pathway, it could not affect the expression of TLR4. This indicates that TLR4 is an upstream activator of PI3K/AKT signaling pathway. These results are highly consistent with the prediction of bioinformatics. In conclusion, we have elucidated the role of TLR4/PI3K/AKT signaling axis in Si-CaP mediated osteogenic-angiogenic coupling for the first time. This study provides new data onto the regulatory role and molecular mechanism of Si-CaP in the process of osteogenic-angiogenic coupling, which strongly supports its wide application for bone tissue engineering.
Collapse
Affiliation(s)
- Yuanhang Kong
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Three authors contributed equally to this work as co-first author
| | - Xin Zhang
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Three authors contributed equally to this work as co-first author
| | - Xinnan Ma
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Three authors contributed equally to this work as co-first author
| | - Leilei Wu
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dechun Chen
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Su
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Daqian Liu
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintao Wang
- 34707Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Vrchovecká K, Pávková-Goldbergová M, Engqvist H, Pujari-Palmer M. Cytocompatibility and Bioactive Ion Release Profiles of Phosphoserine Bone Adhesive: Bridge from In Vitro to In Vivo. Biomedicines 2022; 10:biomedicines10040736. [PMID: 35453486 PMCID: PMC9044752 DOI: 10.3390/biomedicines10040736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/09/2022] [Accepted: 03/18/2022] [Indexed: 02/06/2023] Open
Abstract
One major challenge when developing new biomaterials is translating in vitro testing to in vivo models. We have recently shown that a single formulation of a bone tissue adhesive, phosphoserine modified cement (PMC), is safe and resorbable in vivo. Herein, we screened many new adhesive formulations, for cytocompatibility and bioactive ion release, with three cell lines: MDPC23 odontoblasts, MC3T3 preosteoblasts, and L929 fibroblasts. Most formulations were cytocompatible by indirect contact testing (ISO 10993-12). Formulations with larger amounts of phosphoserine (>50%) had delayed setting times, greater ion release, and cytotoxicity in vitro. The trends in ion release from the adhesive that were cured for 24 h (standard for in vitro) were similar to release from the adhesives cured only for 5−10 min (standard for in vivo), suggesting that we may be able to predict the material behavior in vivo, using in vitro methods. Adhesives containing calcium phosphate and silicate were both cytocompatible for seven days in direct contact with cell monolayers, and ion release increased the alkaline phosphatase (ALP) activity in odontoblasts, but not pre-osteoblasts. This is the first study evaluating how PMC formulation affects osteogenic cell differentiation (ALP), cytocompatibility, and ion release, using in situ curing conditions similar to conditions in vivo.
Collapse
Affiliation(s)
- Kateřina Vrchovecká
- Department of Pathology Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (K.V.); (M.P.-G.)
| | - Monika Pávková-Goldbergová
- Department of Pathology Physiology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (K.V.); (M.P.-G.)
| | - Håkan Engqvist
- Department of Materials Science and Engineering, Applied Material Science, Uppsala University, 75103 Uppsala, Sweden
- Correspondence: (H.E.); (M.P.-P.)
| | - Michael Pujari-Palmer
- Department of Materials Science and Engineering, Applied Material Science, Uppsala University, 75103 Uppsala, Sweden
- Correspondence: (H.E.); (M.P.-P.)
| |
Collapse
|
22
|
Hegedűs C, Czibulya Z, Tóth F, Dezső B, Hegedűs V, Boda R, Horváth D, Csík A, Fábián I, Tóth-Győri E, Sajtos Z, Lázár I. The Effect of Heat Treatment of β-Tricalcium Phosphate-Containing Silica-Based Bioactive Aerogels on the Cellular Metabolism and Proliferation of MG63 Cells. Biomedicines 2022; 10:662. [PMID: 35327463 PMCID: PMC8945762 DOI: 10.3390/biomedicines10030662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
β-Tricalcium phosphate was combined with silica aerogel in composites prepared using the sol-gel technique and supercritical drying. The materials were used in this study to check their biological activity and bone regeneration potential with MG63 cell experiments. The composites were sintered in 100 °C steps in the range of 500-1000 °C. Their mechanical properties, porosities, and solubility were determined as a function of sintering temperature. Dissolution studies revealed that the released Ca-/P molar ratios appeared to be in the optimal range to support bone tissue induction. Cell viability, ALP activity, and type I collagen gene expression results all suggested that the sintering of the compound at approximately 700-800 °C as a scaffold could be more powerful in vivo to facilitate bone formation within a bone defect, compared to that documented previously by our research team. We did not observe any detrimental effect on cell viability. Both the alkaline phosphatase enzyme activity and the type I collagen gene expression were significantly higher compared with the control and the other aerogels heat-treated at different temperatures. The mesoporous silica-based aerogel composites containing β-tricalcium phosphate particles treated at temperatures lower than 1000 °C produced a positive effect on the osteoblastic activity of MG63 cells. An in vivo 6 month-long follow-up study of the mechanically strongest 1000 °C sample in rat calvaria experiments provided proof of a complete remodeling of the bone.
Collapse
Affiliation(s)
- Csaba Hegedűs
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Zsuzsanna Czibulya
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Ferenc Tóth
- Department of Biomaterials and Prosthetic Dentistry, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (Z.C.); (F.T.)
| | - Balázs Dezső
- Department of Oral Pathology and Microbiology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary;
| | - Viktória Hegedűs
- Department of Pediatric Dentistry and Orthodontics, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary;
| | - Róbert Boda
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (R.B.); (D.H.)
| | - Dóra Horváth
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary; (R.B.); (D.H.)
| | - Attila Csík
- Laboratory of Materials Science, Institute for Nuclear Research, Eötvös Loránd Research Network, 4026 Debrecen, Hungary;
| | - István Fábián
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - Enikő Tóth-Győri
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - Zsófi Sajtos
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| | - István Lázár
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary; (I.F.); (E.T.-G.); (Z.S.)
| |
Collapse
|
23
|
Gritsch L, Granel H, Charbonnel N, Jallot E, Wittrant Y, Forestier C, Lao J. Tailored therapeutic release from polycaprolactone-silica hybrids for the treatment of osteomyelitis: antibiotic rifampicin and osteogenic silicates. Biomater Sci 2022; 10:1936-1951. [PMID: 35258044 DOI: 10.1039/d1bm02015c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The treatment of osteomyelitis, a destructive inflammatory process caused by bacterial infections to bone tissue, is one of the most critical challenges of orthopedics and bone regenerative medicine. The standard treatment consists of intense antibiotic therapies combined with tissue surgical debridement and the application of a bone defect filler material. Unfortunately, commercially available candidates, such as gentamicin-impregnated polymethylmethacrylate cements, possess very poor pharmacokinetics (i.e., 24 hours burst release) and little to no regenerative potential. Fostered by the intrinsic limitations associated with conventional treatments, alternative osteostimulative biomaterials with local drug delivery have recently started to emerge. In this study, we propose the use of a polycaprolactone-silica sol-gel hybrid material as carrier for the delivery of rifampicin, an RNA-polymerase blocker often used to treat bone infections, and of osteostimulative silicate ions. The release of therapeutic agents from the material is dual, offering two separate and simultaneous effects, and decoupled, meaning that the kinetics of rifampicin and silicate releases are independent from each other. A series of hybrid formulations with increasing amounts of rifampicin was prepared. The antibiotic loading efficacy, as well as the release profiles of rifampicin and silicates were measured. The characterization of cell viability and differentiation of rat primary osteoblasts and antibacterial performance were also performed. Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa and Escherichia coli were selected due to their high occurrence in bone infections. Results confirmed that rifampicin can be successfully loaded within the hybrids without significant degradation and that it is possible to tailor the antibiotic release according to need. Once in a physiological environment, the rapid release of silicates was associated with optimal cell proliferation and the overexpression of osteoblastic differentiation. Simultaneously, rifampicin is delivered over the course of several weeks with significant inhibition of all tested strains. In particular, the materials caused a growth reduction of 7-10 orders of magnitude in Staphylococcus aureus, the major strain responsible for osteomyelitis worldwide. Our data strongly suggest that PCL/silica hybrids are a very promising candidate to develop bone fillers with superior biological performance compared to currently available options. Thanks to their unique synthesis route and their dual tailored release they can promote bone regeneration while reducing the risk of infection for several weeks upon implantation.
Collapse
Affiliation(s)
- Lukas Gritsch
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| | - Henri Granel
- Unité de Nutrition Humaine UMR 1019 INRAE, Université Clermont Auvergne, 28 place Henri-Dunant, 63001 Clermont-Ferrand, France
| | - Nicolas Charbonnel
- Université Clermont Auvergne, CNRS, LMGE, 63000 Clermont-Ferrand, France
| | - Edouard Jallot
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| | - Yohann Wittrant
- Unité de Nutrition Humaine UMR 1019 INRAE, Université Clermont Auvergne, 28 place Henri-Dunant, 63001 Clermont-Ferrand, France
| | | | - Jonathan Lao
- Laboratoire de Physique de Clermont, UMR CNRS 6533, Université Clermont Auvergne, 4 avenue Blaise Pascal, 63178 Aubière, France.
| |
Collapse
|
24
|
Sakr MA, Sakthivel K, Hossain T, Shin SR, Siddiqua S, Kim J, Kim K. Recent trends in gelatin methacryloyl nanocomposite hydrogels for tissue engineering. J Biomed Mater Res A 2021; 110:708-724. [PMID: 34558808 DOI: 10.1002/jbm.a.37310] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022]
Abstract
Gelatin methacryloyl (GelMA), a photocrosslinkable gelatin-based hydrogel, has been immensely used for diverse applications in tissue engineering and drug delivery. Apart from its excellent functionality and versatile mechanical properties, it is also suitable for a wide range of fabrication methodologies to generate tissue constructs of desired shapes and sizes. Despite its exceptional characteristics, it is predominantly limited by its weak mechanical strength, as some tissue types naturally possess high mechanical stiffness. The use of high GelMA concentrations yields high mechanical strength, but not without the compromise in its porosity, degradability, and three-dimensional (3D) cell attachment. Recently, GelMA has been blended with various natural and synthetic biomaterials to reinforce its physical properties to match with the tissue to be engineered. Among these, nanomaterials have been extensively used to form a composite with GelMA, as they increase its biological and physicochemical properties without affecting the unique characteristics of GelMA and also introduce electrical and magnetic properties. This review article presents the recent advances in the formation of hybrid GelMA nanocomposites using a variety of nanomaterials (carbon, metal, polymer, and mineral-based). We give an overview of each nanomaterial's characteristics followed by a discussion of the enhancement in GelMA's physical properties after its incorporation. Finally, we also highlight the use of each GelMA nanocomposite for different applications, such as cardiac, bone, and neural regeneration.
Collapse
Affiliation(s)
- Mahmoud A Sakr
- School of Engineering, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Kabilan Sakthivel
- School of Engineering, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Towsif Hossain
- School of Engineering, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, Massachusetts, USA
| | - Sumi Siddiqua
- School of Engineering, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Jaehwan Kim
- Advanced Geo-materials Research Department, Korea Institute of Geosciece and Mineral Resources, Pohang-si, South Korea
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Ren L, Cong N, Han H, Zhang Z, Deng C, Zhang N, Li D. The effect of sodium metasilicate on the three-dimensional chondrogenesis of mesenchymal stem cells. Dent Mater J 2021; 40:853-862. [PMID: 34193723 DOI: 10.4012/dmj.2020-214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The benefits of different silicic concentrations on chondrogenesis of mesenchymal stem cell (MSC) are unclear. Here an in vitro scaffoldless model was used to determine the impact of different silicic concentrations on the three-dimensional chondrogenesis of MSCs. Sodium metasilicate solutions were used as the source of silica, and were added in the chondrogenic medium and replenished every 3 days. The thickness and area of cartilage; the expression of collagen II, aggrecan, and the collagen type II/I ratio; the glycosaminoglycan and cell contents; and the tangent modulus of the constructs were all significantly higher in 100 and 200 ng/mL groups compared with those in 0 and 10 ng/mL groups. All the above parameters, as well as several mechanical parameters of cartilage constructs were highest in 200 ng/mL group. Thus, 200 ng/mL sodium metasilicate could promote the chondrogenic differentiation of MSCs and the mechanical and biochemical properties of the cartilage constructs.
Collapse
Affiliation(s)
- Le Ren
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Nuonuo Cong
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University
| | - Hao Han
- Medical Emergency Center, Xi'an Xiangyang International Airport
| | - Zhe Zhang
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Chunni Deng
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Nan Zhang
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Daxu Li
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| |
Collapse
|
26
|
Gomes PS, Pinheiro B, Colaço B, Fernandes MH. The Osteogenic Assessment of Mineral Trioxide Aggregate-based Endodontic Sealers in an Organotypic Ex Vivo Bone Development Model. J Endod 2021; 47:1461-1466. [PMID: 34126159 DOI: 10.1016/j.joen.2021.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/27/2021] [Accepted: 06/05/2021] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Mineral trioxide aggregate (MTA)-based sealers are endodontic materials with widespread success in distinct clinical applications, potentially embracing direct contact with the bone tissue. Bone response to these materials has been traditionally addressed in vitro. Nonetheless, translational data are limited by the absence of native cell-to-cell and cell-to-matrix interactions that hinder the representativeness of the analysis. Ex vivo organotypic systems, relying on the culture of explanted biological tissues, preserve the cell/tissue composition, reproducing the spatial and organizational in situ complexity. This study was grounded on an innovative research approach, relying on the assessment of an ex vivo organotypic bone tissue culture system to address the osteogenic response to 3 distinct MTA-based sealers. METHODS Embryonic chick femurs were isolated and grown ex vivo for 11 days in the presence of MTA Plus (Avalon Biomed Inc, Bradenton, FL), ProRoot MTA (Dentsply Tulsa Dental, Hohnson City, Germany), Biodentine (Septodont, Saint Maurdes Fosses, France), or AH Plus (Dentsply Sirona, Konstanz, Germany); the latter was used as a control material. Femurs were characterized by histologic, histochemical, and histomorphometric analysis. Gene expression assessment of relevant osteogenic markers was conducted by quantitative polymerase chain reaction. RESULTS All MTA-based sealers presented an enhanced osteogenic performance compared with AH Plus. Histochemical and histomorphometric analyses support the increased activation of the osteogenic program by MTA-based sealers, with enhanced collagenous matrix deposition and tissue mineralization. Gene expression analysis supported the enhanced activation of the osteogenic program. Comparatively, ProRoot MTA induced the highest osteogenic functionality on the characterized femurs. CONCLUSIONS MTA-based sealers enhanced the osteogenic activity within the assayed organotypic bone model, which was found to be a sensitive system for the assessment of osteogenic modulation mediated by endodontic sealers.
Collapse
Affiliation(s)
- Pedro S Gomes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal; Associated Laboratory for Green Chemistry/Network of Chemistry and Technology (LAQV/REQUIMTE), University of Porto, Porto, Portugal.
| | - Bruna Pinheiro
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
| | - Bruno Colaço
- Department of Zootechnics, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal; Center for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Maria H Fernandes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal; Associated Laboratory for Green Chemistry/Network of Chemistry and Technology (LAQV/REQUIMTE), University of Porto, Porto, Portugal
| |
Collapse
|
27
|
Magnusson C, Uribe P, Jugdaohsingh R, Powell JJ, Johansson A, Ransjö M. Inhibitory effects of orthosilicic acid on osteoclastogenesis in RANKL-stimulated RAW264.7 cells. J Biomed Mater Res A 2021; 109:1967-1978. [PMID: 33817967 DOI: 10.1002/jbm.a.37189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/30/2021] [Accepted: 03/24/2021] [Indexed: 11/06/2022]
Abstract
Numerous studies have reported on the positive effects of silicon (Si) on bone metabolism, particularly on the stimulatory effects of Si on osteoblast cells and on bone formation. Inhibitory effects of Si on osteoclast formation and bone resorption have also been demonstrated in vitro and are suggested to be mediated indirectly via stromal and osteoblast cells. Direct effects of Si on osteoclasts have been less studied and mostly using soluble Si, but no characterisation of the Si treatment solutions are provided. The aims of the present study were to (a) further investigate the direct inhibitory effects of Si on osteoclastogenesis in RANKL-stimulated RAW264.7 cells, (b) determine at what stage during osteoclastogenesis Si acts upon, and (c) determine if these effects can be attributed to the biologically relevant soluble orthosilicic acid specie. Our results demonstrate that silicon, at 50 μg/ml (or 1.8 mM), does not affect cell viability but directly inhibits the formation of TRAP+ multinucleated cells and the expression of osteoclast phenotypic genes in RAW264.7 cells. The inhibitory effect of Si was clearly associated with the early stages (first 24 hr) of osteoclastogenesis. Moreover, these effects can be attributed to the soluble orthosilicic acid specie.
Collapse
Affiliation(s)
- Catarina Magnusson
- Department of Orthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pamela Uribe
- Department of Orthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ravin Jugdaohsingh
- Biomineral Research Group, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jonathan J Powell
- Biomineral Research Group, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Anders Johansson
- Unit of Molecular Periodontology, Department of Odontology, Umeå University, Umeå, Sweden
| | - Maria Ransjö
- Department of Orthodontics, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
Rondanelli M, Faliva MA, Peroni G, Gasparri C, Perna S, Riva A, Petrangolini G, Tartara A. Silicon: A neglected micronutrient essential for bone health. Exp Biol Med (Maywood) 2021; 246:1500-1511. [PMID: 33715532 DOI: 10.1177/1535370221997072] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bone matrix is predominantly made up of collagen, and in vitro and in animal models studies have shown that silicon is linked to glycosaminoglycans and plays an important role in the formation of cross-links between collagen and proteoglycans, determining the beneficial effects on strength, composition, and mechanical properties of bone. However, there are still no precise indications regarding a possible role of silicon on bone health in humans. Given this background, the aim of this narrative review was to consider the effectiveness of silicon dietary intake and silicon dietary supplementation (alone or with other micronutrients), in order to suggest a daily dosage of Si supplementation, on bone mineral density in humans. This review included eight eligible studies: four regarding dietary intake and four considering supplementation with silicon alone or with other nutrients. Despite the number of studies considered being low, the number of subjects studied is high (10012) and the results are interesting. Although to date the available scientific evidences are not considered valid enough to allow to establish an adequate level of Silicon intake, based on extrapolations from the data obtained with studies on animal and human models, it has been suggested that an adequate intake in order to promote beneficial effects for bone could be considered to be around 25 mg silicon/day. As for silicon dietary supplements, it has been shown that the combined treatment with orthosilicic acid (6 mg), calcium, and vitamin D has a potentially beneficial effect on femoral BMD compared to only use of calcium and vitamin D.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- IRCCS Mondino Foundation, Pavia 27100, Italy.,Department of Public Health, Experimental and Forensic Medicine, Unit of Human and Clinical Nutrition, University of Pavia, Pavia 27100, Italy
| | - Milena A Faliva
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Gabriella Peroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| | - Simone Perna
- Department of Biology, College of Science, University of Bahrain, Sakhir 32038, Bahrain
| | | | | | - Alice Tartara
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona ''Istituto Santa Margherita'', University of Pavia, Pavia 27100, Italy
| |
Collapse
|
29
|
Talabani RM, Garib BT, Masaeli R, Zandsalimi K, Ketabat F. Biomineralization of three calcium silicate-based cements after implantation in rat subcutaneous tissue. Restor Dent Endod 2021; 46:e1. [PMID: 33680890 PMCID: PMC7906840 DOI: 10.5395/rde.2021.46.e1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/22/2020] [Accepted: 08/07/2020] [Indexed: 01/07/2023] Open
Abstract
Objectives The aim of this study was to evaluate the dystrophic mineralization deposits from 3 calcium silicate-based cements (Micro-Mega mineral trioxide aggregate [MM-MTA], Biodentine [BD], and EndoSequence Root Repair Material [ESRRM] putty) over time after subcutaneous implantation into rats. Materials and Methods Forty-five silicon tubes containing the tested materials and 15 empty tubes (serving as a control group) were subcutaneously implanted into the backs of 15 Wistar rats. At 1, 4, and 8 weeks after implantation, the animals were euthanized (n = 5 animals/group), and the silicon tubes were removed with the surrounding tissues. Histopathological tissue sections were stained with von Kossa stain to assess mineralization. Scanning electron microscopy and energy-dispersive X-ray spectroscopy (SEM/EDX) were also used to assess the chemical components of the surface precipitates deposited on the implant and the pattern of calcium and phosphorus distribution at the material-tissue interface. The calcium-to-phosphorus ratios were compared using the non-parametric Kruskal-Wallis test at a significance level of 5%. Results The von Kossa staining showed that both BD and ESRRM putty induced mineralization starting at week 1; this mineralization increased further until the end of the study. In contrast, MM-MTA induced dystrophic calcification later, from 4 weeks onward. SEM/EDX showed no statistically significant differences in the calcium- and phosphorus-rich areas among the 3 materials at any time point (p > 0.05). Conclusions After subcutaneous implantation, biomineralization of the 3-calcium silicate-based cements started early and increased over time, and all 3 tested cements generated calcium- and phosphorus-containing surface precipitates.
Collapse
Affiliation(s)
| | | | - Reza Masaeli
- Department of Dental Biomaterial, Tehran University of Medical Sciences, Tehran, Iran
| | - Kavosh Zandsalimi
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Farinaz Ketabat
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
30
|
Pritchard A, Nielsen BD, Robison C, Manfredi JM. Low dietary silicon supplementation may not affect bone and cartilage in mature, sedentary horses. J Anim Sci 2021; 98:5996088. [PMID: 33216909 DOI: 10.1093/jas/skaa377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/16/2020] [Indexed: 11/14/2022] Open
Abstract
As osteoarthritis is a major cause of lameness in horses in the United States, improving collagen health prior to onset and increasing collagen turnover within affected joints could improve health- and welfare-related outcomes. Through its positive effects on bone mineral content and density and its role in increasing collagen synthesis, silicon (Si) may slow the development and progression of osteoarthritis, thereby reducing lameness. This study evaluated the hypothesis that Si supplementation would increase cartilage turnover through increased collagen degradation and formation markers, as well as bone formation markers, resulting in reduced lameness severity when compared with controls. Ten mature Standardbred geldings were assigned to either a Si-treated (SIL) or control (CON) group and group-housed on pasture for 84 d. Horses were individually fed to ensure no cross-contamination of Si other than what was present in the environment. For the duration of the study, SIL horses received a Si-collagen supplement at the rate of 0.3 g supplement/(100 kg body weight day). Serum samples were taken weekly for osteocalcin, and plasma samples were taken on days 0, 42, and 84 for plasma minerals. On days 0, 42, and 84, subjective and objective lameness exams were performed, and radiographs and synovial fluid samples were taken from reference and osteoarthritic joints. Plasma minerals were similar in both groups and were lower on day 84 than on day 0 (P < 0.05). Si supplementation, fed at the manufacturer's recommended rate, did not improve lameness or radiographs when compared with controls, and supplemented horses did not show greater collagen degradation and/or synthesis markers in synovial fluid than controls, indicating that cartilage turnover remained unaffected. However, a minimum beneficial threshold and range for Si supplementation standardized to body weight need to be established.
Collapse
Affiliation(s)
- Abby Pritchard
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI
| | - Brian D Nielsen
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI
| | - Cara Robison
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI
| | - Jane M Manfredi
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
31
|
Carter SSD, Atif AR, Kadekar S, Lanekoff I, Engqvist H, Varghese OP, Tenje M, Mestres G. PDMS leaching and its implications for on-chip studies focusing on bone regeneration applications. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.ooc.2020.100004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
Zhou P, Xia D, Ni Z, Ou T, Wang Y, Zhang H, Mao L, Lin K, Xu S, Liu J. Calcium silicate bioactive ceramics induce osteogenesis through oncostatin M. Bioact Mater 2020; 6:810-822. [PMID: 33024901 PMCID: PMC7528055 DOI: 10.1016/j.bioactmat.2020.09.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Immune reactions are a key factor in determining the destiny of bone substitute materials after implantation. Macrophages, the most vital factor in the immune response affecting implants, are critical in bone formation, as well as bone biomaterial-mediated bone repair. Therefore, it is critical to design materials with osteoimmunomodulatory properties to reduce host-to-material inflammatory responses by inducing macrophage polarization. Our previous study showed that calcium silicate (CS) bioceramics could significantly promote osteogenesis. Herein, we further investigated the effects of CS on the behavior of macrophages and how macrophages regulated osteogenesis. Under CS extract stimulation, the macrophage phenotype was converted to the M2 extreme. Stimulation by a macrophage-conditioned medium that was pretreated by CS extracts resulted in a significant enhancement of osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), indicating the important role of macrophage polarization in biomaterial-induced osteogenesis. Mechanistically, oncostatin M (OSM) in the macrophage-conditioned medium promoted osteogenic differentiation of BMSCs through the ERK1/2 and JAK3 pathways. This in vivo study further demonstrated that CS bioceramics could stimulate osteogenesis better than β-TCP implants by accelerating new bone formation at defective sites in the femur. These findings improve our understanding of immune modulation of CS bioactive ceramics and facilitate strategies to improve the in vitro osteogenesis capability of bone substitute materials. Calcium silicate (CS) bioceramics significantly promoted osteogenesis by the regulating of macrophage polarization. ERK1/2 and JAK3 pathways mediated the osteogenic differentiation stimulated by CS. CS played a promising osteoimmunomodulatory agent for bone induction.
Collapse
Affiliation(s)
- Panyu Zhou
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Demeng Xia
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhexin Ni
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tianle Ou
- Department of Clinical Medicine, the Naval Medical University, Shanghai, China
| | - Yang Wang
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hongyue Zhang
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Kaili Lin
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Corresponding author.
| | - Shuogui Xu
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
- Corresponding author.
| | - Jiaqiang Liu
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Corresponding author.
| |
Collapse
|
33
|
Frassica MT, Grunlan MA. Perspectives on Synthetic Materials to Guide Tissue Regeneration for Osteochondral Defect Repair. ACS Biomater Sci Eng 2020; 6:4324-4336. [PMID: 33455185 DOI: 10.1021/acsbiomaterials.0c00753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative engineering holds the potential to treat clinically pervasive osteochondral defects (OCDs). In a synthetic materials-guided approach, the scaffold's chemical and physical properties alone instruct cellular behavior in order to effect regeneration, referred to herein as "instructive" properties. While this alleviates the costs and off-target risks associated with exogenous growth factors, the scaffold must be potently instructive to achieve tissue growth. Moreover, toward achieving functionality, such a scaffold should also recapitulate the spatial complexity of the osteochondral tissues. Thus, in addition to the regeneration of the articular cartilage and underlying cancellous bone, the complex osteochondral interface, composed of calcified cartilage and subchondral bone, should also be restored. In this Perspective, we highlight recent synthetic-based, instructive osteochondral scaffolds that have leveraged new material chemistries as well as innovative fabrication strategies. In particular, scaffolds with spatially complex chemical and morphological features have been prepared with electrospinning, solvent-casting-particulate-leaching, freeze-drying, and additive manufacturing. While few synthetic scaffolds have advanced to clinical studies to treat OCDs, these recent efforts point to the promising use of the chemical and physical properties of synthetic materials for regeneration of osteochondral tissues.
Collapse
Affiliation(s)
- Michael T Frassica
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States.,Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843-3003, United States.,Department of Chemistry, Texas A&M University, College Station, Texas 77843-3255, United States
| |
Collapse
|
34
|
Soluble silica stimulates osteogenic differentiation and gap junction communication in human dental follicle cells. Sci Rep 2020; 10:9923. [PMID: 32555274 PMCID: PMC7303172 DOI: 10.1038/s41598-020-66939-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/29/2020] [Indexed: 12/16/2022] Open
Abstract
Several studies have indicated that dietary silicon (Si) is beneficial for bone homeostasis and skeletal health. Furthermore, Si-containing bioactive glass biomaterials have positive effects on bone regeneration when used for repair of bone defects. Si has been demonstrated to stimulate osteoblast differentiation and bone mineralisation in vitro. However, the mechanisms underlying these effects of Si are not well understood. The aim of the present study was to investigate the effects of soluble Si on osteogenic differentiation and connexin 43 (CX43) gap junction communication in cultured pluripotent cells from human dental follicles (hDFC). Neutral Red uptake assay demonstrated that 25 μg/ml of Si significantly stimulated hDFC cell proliferation. Dosages of Si above 100 μg/ml decreased cell proliferation. Alizarin Red staining showed that osteogenic induction medium (OIM) by itself and in combination with Si (25 μg/ml) significantly increased mineralisation in hDFC cultures, although Si alone had no such effect. The expression of osteoblast-related markers in hDFC was analysed with RT-qPCR. OSX, RUNX2, BMP2, ALP, OCN, BSP and CX43 genes were expressed in hDFC cultured for 1, 7, 14 and 21 days. Expression levels of BMP-2 and BSP were significantly upregulated by OIM and Si (25 μg/ml) and were also induced by Si alone. Notably, the expression levels of OCN and CX43 on Day 21 were significantly increased only in the Si group. Flow cytometric measurements revealed that Si (50 μg/ml) significantly increased CX43 protein expression and gap junction communication in hDFC. Next-generation sequencing (NGS) and bioinformatics processing were used for the identification of differentially regulated genes and pathways. The influence of OIM over the cell differentiation profile was more prominent than the influence of Si alone. However, Si in combination with OIM increased the magnitude of expression (up or down) of the differentially regulated genes. The gene for cartilage oligomeric matrix protein (COMP) was the most significantly upregulated. Genes for the regulator of G protein signalling 4 (RGS4), regulator of G protein signalling 2 (RGS2), and matrix metalloproteinases (MMPs) 1, 8, and 10 were also strongly upregulated. Our findings reveal that soluble Si stimulates Cx43 gap junction communication in hDFC and induces gene expression patterns associated with osteogenic differentiation. Taken together, the results support the conclusion that Si is beneficial for bone health.
Collapse
|
35
|
Fu X, Liu P, Zhao D, Yuan B, Xiao Z, Zhou Y, Yang X, Zhu X, Tu C, Zhang X. Effects of Nanotopography Regulation and Silicon Doping on Angiogenic and Osteogenic Activities of Hydroxyapatite Coating on Titanium Implant. Int J Nanomedicine 2020; 15:4171-4189. [PMID: 32606671 PMCID: PMC7297339 DOI: 10.2147/ijn.s252936] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/19/2020] [Indexed: 02/05/2023] Open
Abstract
Background Angiogenic and osteogenic activities are two major problems with biomedical titanium (Ti) and other orthopedic implants used to repair large bone defects. Purpose The aim of this study is to prepare hydroxyapatite (HA) coatings on the surface of Ti by using electrochemical deposition (ED), and to evaluate the effects of nanotopography and silicon (Si) doping on the angiogenic and osteogenic activities of the coating in vitro. Materials and Methods HA coating and Si-doped HA (HS) coatings with varying nanotopographies were fabricated using two ED modes, ie, the pulsive current (PC) and cyclic voltammetry (CV) methods. The coatings were characterized through scanning electron microscope (SEM), X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), X-ray photoelectron spectrometer (XPS), and atomic force microscopy (AFM), and their in vitro bioactivity and protein adsorption were assessed. Using MC3T3-E1 pre-osteoblasts and HUVECs as cell models, the osteogenic and angiogenic capabilities of the coatings were evaluated through in vitro cellular experiments. Results By controlling Si content in ~0.8 wt.%, the coatings resulting from the PC mode (HA-PC and HS-PC) and CV mode (HA-CV and HS-CV) had nanosheet and nanorod topographies, respectively. At lower crystallinity, higher ionic dissolution, smaller contact angle, higher surface roughness, and more negative zeta potential, the HS and PC samples exhibited quicker apatite deposition and higher BSA adsorption capacity. The in vitro cell study showed that Si doping was more favorable for enhancing the viability of the MC3T3-E1 cells, but nanosheet coating increased the area for cell spreading. Of the four coatings, HS-PC with Si doping and nanosheet topography exhibited the best effect in terms of up-regulating the expressions of the osteogenic genes (ALP, Col-I, OSX, OPN and OCN) in the MC3T3-E1 cells. Moreover, all leach liquors of the surface-coated Ti disks promoted the growth of the HUVECs, and the HS samples played a more significant role in promoting cell migration and tube formation than the HA samples. Of the four leach liquors, only the two HS samples up-regulated NO content and expressions of the angiogenesis-related genes (VEGF, bFGF and eNOS) in the HUVECs, and the HS-PC yielded a better effect. Conclusion The results show that Si doping while regulating the topography of the coating can help enhance the bone regeneration and vascularization of HA-coated Ti implants.
Collapse
Affiliation(s)
- Xi Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Pin Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Dingyun Zhao
- Department of Orthopaedics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Bo Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Zhanwen Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Yong Zhou
- Department of Orthopaedics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| | - Chongqi Tu
- Department of Orthopaedics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
| |
Collapse
|
36
|
Chen X, Li T, Qing D, Chen J, Zhang Q, Yan C. Structural characterization and osteogenic bioactivities of a novel Humulus lupulus polysaccharide. Food Funct 2020; 11:1165-1175. [PMID: 31872841 DOI: 10.1039/c9fo01918a] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Humulus lupulus is a perennial climbing plant of the subfamily Cannabioideae native to the Northern Hemisphere. The primary use of H. lupulus is in the brewing industry, where it is an essential ingredient for imparting a unique flavor (bitterness and aroma) to beer. The female flowers of H. lupulus are also used in traditional Chinese medicine, but the biologically active ingredients underlying its benefits remain unclear. China is the largest producer and consumer of H. lupulus in Asia. Using the waste from the beer-brewing process of H. lupulus as raw materials, the biologically active polysaccharides can be screened. This is useful for the full utilization of H. lupulus, potentially leading to disease prevention and treatment. In this study, we isolated a homogeneous polysaccharide (HLP50-1) with a molecular weight of 49.13 kDa from female flowers of H. lupulus via a DEAE-Cellulose 52 anion exchange column and a Sephadex G-75 gel filtration column. Methylation, GC-MS, and NMR analyses revealed that the HLP50-1 comprised →4)-α-d-Glcp-(1→, →6)-α-d-Manp-(1→, →3)-α-l-Rhap-(1→, β-d-Glcp-(1→, α-l-Araf-(1→, →4,6)-2-OAc-β-d-Galp-(1→, β-d-Galp-(1→, →3,6)-β-d-Glcp-(1→, →2,3,4)-α-d-Xylp-(1→, →6)-α-d-Glcp-(1→, →3)-α-d-Galp-(1→, →4)-α-d-Galp-(1→. Advanced structural analysis showed that the HLP50-1 contained irregular fragments of different sizes and shapes with a smooth surface. The aggregates appeared be composed of accumulated crystals. Furthermore, the osteogenic activities of the HLP50-1 were evaluated via MC3T3-E1 cells in vitro. The results showed that 0.13 μM HLP50-1 led to outstanding proliferation, differentiation, and mineralization of the MC3T3-E1 cells. Therefore, HLP50-1 has osteogenic effects, and it may be a candidate for the treatment of osteoporosis. It has broad application prospects in functional foods, health-care products, and pharmaceuticals.
Collapse
Affiliation(s)
- Xiaoxia Chen
- Center for Clinical Precision Medication, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China
| | | | | | | | | | | |
Collapse
|
37
|
Le TDH, Liaudanskaya V, Bonani W, Migliaresi C, Motta A. Diatom Particles: A Promising Osteoinductive Agent of Silk Fibroin-Based Scaffold for Bone Regeneration. IFMBE PROCEEDINGS 2020. [DOI: 10.1007/978-981-13-5859-3_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
38
|
Santos-Coquillat A, Esteban-Lucia M, Martinez-Campos E, Mohedano M, Arrabal R, Blawert C, Zheludkevich M, Matykina E. PEO coatings design for Mg-Ca alloy for cardiovascular stent and bone regeneration applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110026. [DOI: 10.1016/j.msec.2019.110026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/19/2019] [Accepted: 07/26/2019] [Indexed: 02/03/2023]
|
39
|
Incorporation of a silicon-based polymer to PEG-DA templated hydrogel scaffolds for bioactivity and osteoinductivity. Acta Biomater 2019; 99:100-109. [PMID: 31536841 DOI: 10.1016/j.actbio.2019.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/26/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022]
Abstract
A scaffold that is inherently bioactive, osteoinductive and osteoconductive may guide mesenchymal stem cells (MSCs) to regenerate bone tissue in the absence of exogenous growth factors. Previously, we established that hydrogel scaffolds formed by crosslinking methacrylated star poly(dimethylsiloxane) (PDMSstar-MA) with diacrylated poly(ethylene glycol) (PEG-DA) promote bone bonding by induction of hydroxyapatite formation ("bioactive") and promote MSC lineage progression toward osteoblast-like fate ("osteoinductive"). Herein, we have combined solvent induced phase separation (SIPS) with a fused salt template to create PDMSstar-PEG hydrogel scaffolds with controlled PDMSstar-MA distribution as well as interconnected macropores of a tunable size to allow for subsequent cell seeding and neotissue infiltration ("osteoconductive"). Scaffolds were prepared with PDMSstar-MA of two number average molecular weights (Mns) (2k and 7k) with varying PDMSstar-MA:PEG-DA ratios and template salt sizes. The distribution of PDMSstar-MA within the hydrogels was examined as well as pore size, percent interconnectivity, dynamic and static moduli, hydration, degradation and in vitro bioactivity (i.e. mineralization when exposed to simulated body fluid, SBF). Finally, cell culture with seeded human bone marrow-derived MSCs (hBMSCs) was used to confirm non-cytotoxicity and characterize osteoinductivity. Tunable, interconnected macropores were achieved by utilization of a fused salt template of a specified salt size during fabrication. Distribution of PDMSstar-MA within the PEG-DA matrix improved for the lower Mn and contributed to differences in specific material properties (e.g. local modulus) and cellular response. However, all templated SIPS PDMSstar-PEG hydrogels were confirmed to be bioactive, non-cytotoxic and displayed PDMSstar-MA dose-dependent osteogenesis. STATEMENT OF SIGNIFICANCE: A tissue engineering scaffold that can inherently guide mesenchymal stem cells (MSCs) to regenerate bone tissue without growth factors would be a more cost-effective and safe strategy for bone repair. Typically, glass/ceramic fillers are utilized to achieve this through their ability to induce hydroxyapatite formation ("bioactive") and promote MSC differentiation to an osteoblast-like fate ("osteoinductive"). Herein, we have fabricated an interconnected, macroporous PEG-DA hydrogel scaffold that utilizes PDMSstar-MA as a bioactive and osteoinductive scaffold component. We were able to show that these PDMSstar-PEG hydrogels maintain several key material characteristics for bone repair. Further, bioactivity and osteoinductivity were simultaneously achieved in human bone marrow-derived MSC culture, representing a notable achievement for an exclusively material-based strategy.
Collapse
|
40
|
Bossard C, Granel H, Jallot É, Montouillout V, Fayon F, Soulié J, Drouet C, Wittrant Y, Lao J. Mechanism of Calcium Incorporation Inside Sol–Gel Silicate Bioactive Glass and the Advantage of Using Ca(OH)2 over Other Calcium Sources. ACS Biomater Sci Eng 2019; 5:5906-5915. [PMID: 33405681 DOI: 10.1021/acsbiomaterials.9b01245] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Cédric Bossard
- Université Clermont Auvergne, CNRS/IN2P3, Laboratoire de Physique de Clermont, BP 10448, F-63000 Clermont-Ferrand, France
| | - Henri Granel
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRA, UNH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Édouard Jallot
- Université Clermont Auvergne, CNRS/IN2P3, Laboratoire de Physique de Clermont, BP 10448, F-63000 Clermont-Ferrand, France
| | - Valérie Montouillout
- CNRS, CEMHTI UPR3079, Université d’Orléans, 1D avenue de la Recherche Scientifique, 45071 Orléans, France
| | - Franck Fayon
- CNRS, CEMHTI UPR3079, Université d’Orléans, 1D avenue de la Recherche Scientifique, 45071 Orléans, France
| | - Jérémy Soulié
- CIRIMAT, Université de Toulouse, CNRS/INPT/UPS, Ensiacet, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France
| | - Christophe Drouet
- CIRIMAT, Université de Toulouse, CNRS/INPT/UPS, Ensiacet, 4 Allée Emile Monso, 31030 Toulouse Cedex 4, France
| | - Yohann Wittrant
- Unité de Nutrition Humaine, Université Clermont Auvergne, INRA, UNH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Jonathan Lao
- Université Clermont Auvergne, CNRS/IN2P3, Laboratoire de Physique de Clermont, BP 10448, F-63000 Clermont-Ferrand, France
| |
Collapse
|
41
|
Frassica MT, Jones SK, Diaz-Rodriguez P, Hahn MS, Grunlan MA. Incorporation of a silicon-based polymer to PEG-DA templated hydrogel scaffolds for bioactivity and osteoinductivity. Acta Biomater 2019. [PMID: 31536841 DOI: 10.1016/j.actbio.2019.09.018.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2022]
Abstract
A scaffold that is inherently bioactive, osteoinductive and osteoconductive may guide mesenchymal stem cells (MSCs) to regenerate bone tissue in the absence of exogenous growth factors. Previously, we established that hydrogel scaffolds formed by crosslinking methacrylated star poly(dimethylsiloxane) (PDMSstar-MA) with diacrylated poly(ethylene glycol) (PEG-DA) promote bone bonding by induction of hydroxyapatite formation ("bioactive") and promote MSC lineage progression toward osteoblast-like fate ("osteoinductive"). Herein, we have combined solvent induced phase separation (SIPS) with a fused salt template to create PDMSstar-PEG hydrogel scaffolds with controlled PDMSstar-MA distribution as well as interconnected macropores of a tunable size to allow for subsequent cell seeding and neotissue infiltration ("osteoconductive"). Scaffolds were prepared with PDMSstar-MA of two number average molecular weights (Mns) (2k and 7k) with varying PDMSstar-MA:PEG-DA ratios and template salt sizes. The distribution of PDMSstar-MA within the hydrogels was examined as well as pore size, percent interconnectivity, dynamic and static moduli, hydration, degradation and in vitro bioactivity (i.e. mineralization when exposed to simulated body fluid, SBF). Finally, cell culture with seeded human bone marrow-derived MSCs (hBMSCs) was used to confirm non-cytotoxicity and characterize osteoinductivity. Tunable, interconnected macropores were achieved by utilization of a fused salt template of a specified salt size during fabrication. Distribution of PDMSstar-MA within the PEG-DA matrix improved for the lower Mn and contributed to differences in specific material properties (e.g. local modulus) and cellular response. However, all templated SIPS PDMSstar-PEG hydrogels were confirmed to be bioactive, non-cytotoxic and displayed PDMSstar-MA dose-dependent osteogenesis. STATEMENT OF SIGNIFICANCE: A tissue engineering scaffold that can inherently guide mesenchymal stem cells (MSCs) to regenerate bone tissue without growth factors would be a more cost-effective and safe strategy for bone repair. Typically, glass/ceramic fillers are utilized to achieve this through their ability to induce hydroxyapatite formation ("bioactive") and promote MSC differentiation to an osteoblast-like fate ("osteoinductive"). Herein, we have fabricated an interconnected, macroporous PEG-DA hydrogel scaffold that utilizes PDMSstar-MA as a bioactive and osteoinductive scaffold component. We were able to show that these PDMSstar-PEG hydrogels maintain several key material characteristics for bone repair. Further, bioactivity and osteoinductivity were simultaneously achieved in human bone marrow-derived MSC culture, representing a notable achievement for an exclusively material-based strategy.
Collapse
Affiliation(s)
- Michael T Frassica
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120, USA
| | - Sarah K Jones
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120, USA
| | - Patricia Diaz-Rodriguez
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180-31590, USA
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180-31590, USA
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120, USA; Department of Materials Science & Engineering, Texas A&M University, College Station, TX 77843-3003, USA; Department of Chemistry, Texas A&M University, College Station, TX 77843-3255, USA.
| |
Collapse
|
42
|
Osada N, Terada A, Maeda H, Obata A, Nishikawa Y, Kasuga T. Tuning of ion-release capability from bio-ceramic-polymer composites for enhancing cellular activity. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190612. [PMID: 31598297 PMCID: PMC6774980 DOI: 10.1098/rsos.190612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
In our previous study, we investigated the synergetic effects of inorganic ions, such as silicate, Mg2+ and Ca2+ ions on the osteoblast-like cell behaviour. Mg2+ ions play an important role in cell adhesion. In the present study, we designed a new composite that releases a high concentration of Mg2+ ions during the early stage of the bone-forming process, and silicate and Ca2+ ions continuously throughout this process. Here, 40SiO2-40MgO-20Na2O glass (G) with high solubility and vaterite-based calcium carbonate (V) were selected as the source of silicate and Mg2+ and Ca2+ ions, respectively. These particles were mixed with poly(lactic-co-glycolic acid) (PLGA) using a kneading method at 110°C to prepare the composite (G-V/PLGA, G/V/PLGA = 4/56/40 (in weight ratio)). Most of the Mg2+ ions were released within 3 days of immersion at an important stage for cell adhesion, and silicate and Ca2+ ions were released continuously at rates of 70-80 and 180 ppm d-1, respectively, throughout the experiment (until day 7). Mouse-derived osteoblast-like MC3T3-E1 proliferated more vigorously on G-V/PLGA in comparison with V-containing PLGA without G particles; it is possible to control the ion-release behaviour by incorporating a small amount of glass particles.
Collapse
Affiliation(s)
- Naoki Osada
- Nagoya Institute of Technology, Gokiso cho, Showa ku, Nagoya 466-8555, Japan
- ORTHOREBIRTH Co. Ltd, 15-3-303 Chigasaki-Chuo, Tsuzuki-ku, Yokohama 224-0032, Japan
| | - Arisa Terada
- Nagoya Institute of Technology, Gokiso cho, Showa ku, Nagoya 466-8555, Japan
| | - Hirotaka Maeda
- Nagoya Institute of Technology, Gokiso cho, Showa ku, Nagoya 466-8555, Japan
| | - Akiko Obata
- Nagoya Institute of Technology, Gokiso cho, Showa ku, Nagoya 466-8555, Japan
| | - Yasutoshi Nishikawa
- ORTHOREBIRTH Co. Ltd, 15-3-303 Chigasaki-Chuo, Tsuzuki-ku, Yokohama 224-0032, Japan
| | - Toshihiro Kasuga
- Nagoya Institute of Technology, Gokiso cho, Showa ku, Nagoya 466-8555, Japan
| |
Collapse
|
43
|
The role of nitrogen off-stoichiometry in the osteogenic behavior of silicon nitride bioceramics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110053. [PMID: 31546420 DOI: 10.1016/j.msec.2019.110053] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/22/2019] [Accepted: 08/03/2019] [Indexed: 11/23/2022]
Abstract
The surface chemistry of silicon nitride plays an important role in stimulating osteoblasts to proliferate and produce bone tissue with improved efficiency. This property, which is advantageous in spinal fusion surgery has a chemical origin and is a direct consequence of the cleavage of covalent SN bonds in an aqueous environment. Building upon a wealth of published research on the stimulation of osteoblastic activity by silicon, the aim of this paper is to explore the role of nitrogen and, more specifically, the N/Si atomic ratio on the osteogenic response of Si3N4. The surface stoichiometry of Si3N4 was gradually altered toward a silicon-rich composition by systematically treating the Si3N4 surface with a high-power pulsed laser in an Ar gas atmosphere (i.e., operated at different pulse times, spot sizes, and voltages). Different analytical probes were used to characterize the surface including X-ray photoelectron spectroscopy (XPS), Raman spectroscopy, and energy dispersive X-ray spectroscopy (EDS). Osteoconductivity was tested in vitro using SaOS-2 osteosarcoma cells, and samples with different surface stoichiometry were compared for their osteogenic response. These experiments clearly indicated a fundamental role for nitrogen off-stoichiometry in osteogenesis, and showed that both cell proliferation and growth of bone tissue diminished with decreasing nitrogen content.
Collapse
|
44
|
Xu LH, Shao H, Ma YHV, You L. OCY454 Osteocytes as an in Vitro Cell Model for Bone Remodeling Under Mechanical Loading. J Orthop Res 2019; 37:1681-1689. [PMID: 30977540 DOI: 10.1002/jor.24302] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 02/04/2023]
Abstract
Osteocytes' mechano-regulation of bone formation and resorption is key to maintaining appropriate bone health. Although extensive in vitro studies have explored osteocyte mechanobiology using the well-established MLO-Y4 cell model, the low amount of sclerostin secreted by this cell line renders it inadequate for studying cross-talk between osteocytes and osteoblasts under mechanical loading. Here, we investigated the potential of the sclerostin-expressing OCY454 osteocyte cell model in fulfilling this role. Fully differentiated OCY454 cells were tested for mechano-sensitivity by measuring changes in protein secretion, total adenosine triphosphate (ATP) content, and intracellular calcium in response to oscillatory fluid flow. Increases in sclerostin expression and total ATP content were observed. However, very low levels of receptor activator of the nuclear factor κ-B ligand were detected, and there was a great inconsistency in calcium response. Conditioned medium (CM) from OCY454 cells was then used to culture osteoblast and osteoclast precursors. Osteoblast activity was quantified with alkaline phosphatase (ALP) and Alizarin Red S stain, while osteoclast differentiation was quantified with tartrate-resistant acid phosphatase (TRAP) staining. We demonstrated that mechanically stimulated OCY454 cells released soluble factors that increased osteoblasts' ALP activity (p < 0.05) and calcium deposition (p < 0.05). There was also a significant decrease of large-sized TRAP-positive osteoclasts when osteoclast precursors were treated with CM from flow-stimulated OCY454 cells (p < 0.05). Results from this study suggest that OCY454 cells respond to mechanical loading with the release of key factors such as sclerostin to regulate downstream bone cells, thus demonstrating its potential as a novel cell model for in vitro osteocyte mechanobiology studies. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1681-1689, 2019.
Collapse
Affiliation(s)
- Liangcheng Henry Xu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Han Shao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Yu-Heng V Ma
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Lidan You
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
45
|
Pezzotti G. Silicon Nitride: A Bioceramic with a Gift. ACS APPLIED MATERIALS & INTERFACES 2019; 11:26619-26636. [PMID: 31251018 DOI: 10.1021/acsami.9b07997] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In the closing decades of the 20th century, silicon nitride (Si3N4) was extensively developed for high-temperature gas turbine applications. Technologists attempted to take advantage of its superior thermal and mechanical properties to improve engine reliability and fuel economy. Yet, this promise was never realized in spite of the worldwide research, which was conducted at that time. Notwithstanding this disappointment, its use in medical applications in the early 21st century has been an unexpected gift. While retaining all of its engineered mechanical properties, it is now recognized for its peculiar surface chemistry. When immersed in an aqueous environment, the slow elution of silicon and nitrogen from its surface enhances healing of soft and osseous tissue, inhibits bacterial proliferation, and eradicates viruses. These benefits permit it to be used in a wide array of different disciplines inside and outside of the human body including orthopedics, dentistry, virology, agronomy, and environmental remediation. Given the global public health threat posed by mutating viruses and bacteria, silicon nitride offers a valid and straightforward alternative approach to fighting these pathogens. However, there is a conundrum behind these recent discoveries: How can this unique bioceramic be both friendly to mammalian cells while concurrently lysing invasive pathogens? This unparalleled characteristic can be explained by the pH-dependent kinetics of two ammonia species-NH4+ and NH3-both of which are leached from the wet Si3N4 surface.
Collapse
Affiliation(s)
- Giuseppe Pezzotti
- Ceramic Physics Laboratory , Kyoto Institute of Technology , Sakyo-ku, Matsugasaki , Kyoto 606-8585 , Japan
- Department of Orthopedic Surgery , Tokyo Medical University , 6-7-1 Nishi-Shinjuku , Shinjuku-ku, Tokyo 160-0023 , Japan
- The Center for Advanced Medical Engineering and Informatics , Osaka University , 2-2 Yamadaoka , Suita 565-0854 , Osaka , Japan
- Department of Immunology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kamigyo-ku, 465 Kajii-cho , Kyoto 602-8566 , Japan
| |
Collapse
|
46
|
Dalgic AD, Atila D, Karatas A, Tezcaner A, Keskin D. Diatom shell incorporated PHBV/PCL-pullulan co-electrospun scaffold for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:735-746. [DOI: 10.1016/j.msec.2019.03.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/23/2022]
|
47
|
Degradation Behaviour of Mg0.6Ca and Mg0.6Ca2Ag Alloys with Bioactive Plasma Electrolytic Oxidation Coatings. COATINGS 2019. [DOI: 10.3390/coatings9060383] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bioactive Plasma Electrolytic Oxidation (PEO) coatings enriched in Ca, P and F were developed on Mg0.6Ca and Mg0.6Ca2Ag alloys with the aim to impede their fast degradation rate. Different characterization techniques (SEM, TEM, EDX, SKPFM, XRD) were used to analyze the surface characteristics and chemical composition of the bulk and/or coated materials. The corrosion behaviour was evaluated using hydrogen evolution measurements in Simulated Body Fluid (SBF) at 37 °C for up to 60 days of immersion. PEO-coated Mg0.6Ca showed a 2–3-fold improved corrosion resistance compared with the bulk alloy, which was more relevant to the initial 4 weeks of the degradation process. In the case of the Mg0.6Ag2Ag alloy, the obtained corrosion rates were very high for both non-coated and PEO-coated specimens, which would compromise their application as resorbable implants. The amount of F− ions released from PEO-coated Mg0.6Ca during 24 h of immersion in 0.9% NaCl was also measured due to the importance of F− in antibacterial processes, yielding 33.7 μg/cm2, which is well within the daily recommended limit of F− consumption.
Collapse
|
48
|
Ahmadi SM, Behnamghader A, Asefnejaad A. Evaluation of hMSCs Response to Sodium Alginate / Bioactive Glass Composite Paste: Effect of CaO/P2O5, Sodium Alginate Concentration and P/L Ratios. Curr Stem Cell Res Ther 2019; 14:196-210. [DOI: 10.2174/1574888x13666180703141956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/29/2018] [Accepted: 06/12/2018] [Indexed: 11/22/2022]
Abstract
Background:
Bioactive glasses with different compositions have been extensively used as
bone tissue engineering. Preparation, development and characterization of alginate pastes containing
bioglass for bone repair applications were the purposes of this study.
Objective:
The injectable bone pastes were produced from sol-gel derived bioactive glass nanoparticles
with various CaO/P2O5 ratios of 19, 9.5 and 4.75 and sodium alginate solutions with different concentrations
of 1, 2 and 4 wt.%. The effect of CaO/P2O5 and powder to liquid (P/L) ratios and alginate concentration
on injectability, biodegradation, rheological properties, bioactivity and cellular behavior of
the pastes have been studied. The behavior of human mesenchymal stem cells (hMSCs) in the presence
of the pastes was assessed by MTT assay, biomineralization assay, ALP activity, Acridine orange
staining and Alizarin red staining tests.
Results:
By adding sodium alginate, the pastes exhibited a thixotropy behavior. The storage modulus
of all pastes was larger than the loss modulus in the frequency range of 0.1-100 s-1. Cytotoxicity
evaluation results revealed that there was a critical amount of bioactive glass in pastes which are above
the limit; the viability of hMSCs will be at risk. The pastes made of bioactive glass nanoparticles with
CaO/P2O5 = 9.5 and sodium alginate 1% with P/L ratio of 0.8 showed optimum behavior in terms of
mineral carrying capacity, injectability characteristics, accellular bioactivity in SBF, loss weight and
wash out behavior, proliferation and differentiation of hMSCs.
Conclusion:
According to the results, the pastes prepared with sodium alginate solution and bioactive
glass nanoparticles can be beneficial in bone tissue engineering.
Collapse
Affiliation(s)
- Seyed Mohammad Ahmadi
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aliasghar Behnamghader
- Biomaterials Group, Department of Nanotechnology and Advanced Materials, Materials & Energy Research Center, Karaj, Iran
| | - Azadeh Asefnejaad
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
49
|
Shang H, Zhou A, Jiang J, Liu Y, Xie J, Li S, Chen Y, Zhu X, Tan H, Li J. Inhibition of the fibrillation of highly amyloidogenic human calcitonin by cucurbit[7]uril with improved bioactivity. Acta Biomater 2018; 78:178-188. [PMID: 30076991 DOI: 10.1016/j.actbio.2018.07.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 02/08/2023]
Abstract
Protein/peptide fibrillation is an important challenge for biotechnological drug development. Salmon calcitonin (sCT) is currently used in the clinical treatment of bone-related diseases such as osteoporosis and hypercalcemia, but it still has the risk of immune responses. Although human calcitonin (hCT) would be a better choice in terms of immunogenicity, it has a strong tendency to irreversibly aggregate in aqueous solutions and form long amyloid fibrils, which significantly reduces its bioavailability and therapeutic potency. Here, we demonstrate that cucurbit[7]uril (CB[7]) can inhibit hCT fibrillation by supramolecular interaction with its aromatic groups (affinity: Phe16 > Tyr12 > Phe19 > Phe22). The hCT-CB[7] complex exhibits low cytotoxicity, even promotes osteoblast proliferation and osteogenic capacity of MC3T3 cells. Meanwhile the hCT-CB[7] complexes shows higher bioactivity compared to hCT in reducing blood calcium levels in rats, and also decreases the immunogenicity of hCT. These results suggest that CB[7] has the potential to improve the therapeutic potency of amyloidogenic protein/peptide drugs such as hCT.
Collapse
Affiliation(s)
- Hui Shang
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Anna Zhou
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jian Jiang
- College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Yanpeng Liu
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jing Xie
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yantao Chen
- Shenzhen Key Laboratory of Functional Polymer, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Xiaofeng Zhu
- College of Life Sciences, Sichuan University, Chengdu 610065, China.
| | - Hong Tan
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jianshu Li
- Department of Biomedical Polymers and Artificial Organs, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
50
|
Ding Y, Li W, Correia A, Yang Y, Zheng K, Liu D, Schubert DW, Boccaccini AR, Santos HA, Roether JA. Electrospun Polyhydroxybutyrate/Poly(ε-caprolactone)/Sol-Gel-Derived Silica Hybrid Scaffolds with Drug Releasing Function for Bone Tissue Engineering Applications. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14540-14548. [PMID: 29624366 PMCID: PMC6108537 DOI: 10.1021/acsami.8b02656] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 05/23/2023]
Abstract
Electrospun hybrid scaffolds are an effective platform to deliver drugs site specifically for the prevention and treatment of diseases in addition to promote tissue regeneration because of the flexibility to load drugs therein. In the present study, electrospun hybrid scaffolds containing antibiotics were developed to support cellular activities and eliminate potential postoperative inflammation and infection. As a model drug, levofloxacin (LFX) was successfully incorporated into pure polyhydroxybutyrate/poly(ε-caprolactone) (PHB/PCL) scaffolds and PHB/PCL/sol-gel-derived silica (SGS) scaffolds. The influence of LFX on the morphology, mechanical performance, chemical structure, drug release profile, and antibacterial effect of the scaffolds was thoroughly and comparatively investigated. MG-63 osteoblast-like cell cultivation on both scaffolds certified that LFX inclusion did not impair the biocompatibility. In addition to the favorable cellular proliferation and differentiation, scaffolds containing both LFX and SGS displayed highly increased mineralization content. Therefore, the present multifunctional hybrid scaffolds are promising in tissue engineering applications.
Collapse
Affiliation(s)
- Yaping Ding
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| | - Wei Li
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Correia
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Yuyun Yang
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
- Institute of Corrosion
Science and Surface Technology, Harbin Engineering
University, Nantong Street 145, 150001 Harbin, China
| | - Kai Zheng
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Dongfei Liu
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Dirk W. Schubert
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Hélder A. Santos
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Judith A. Roether
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| |
Collapse
|