1
|
Sun X, Lin Y, Zhong X, Fan C, Liu Z, Chen X, Luo Z, Wu J, Tima S, Zhang Z, Jiang J, Du X, Zhou X, Zhong Z. Alendronate-functionalized polymeric micelles target icaritin to bone for mitigating osteoporosis in a rat model. J Control Release 2024; 376:37-51. [PMID: 39368708 DOI: 10.1016/j.jconrel.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Formulating drugs into nanoparticles that target sites of disease can lead to strong therapeutic effects with lower doses of drugs and lower rates of off-target adverse effects. Few ways to target drugs to bone have been described, hampering the treatment of osteoporosis. Here we exploit the ability of alendronate to bind tightly to hydroxyapatite in bone as a tactic to target polymeric micelles loaded with the plant flavonoid icaritin to osteoporotic lesions. The traditional Chinese medicine icaritin, from Herba Epimedii, has previously been shown to inhibit adipogenesis and enhance osteogenesis by bone mesenchymal stem cells, but the compound on its own persists only briefly in the bloodstream. Our delivery system led to stronger inhibition of adipogenesis and activation of osteogenesis in a rat model of osteoporosis than when the icaritin-loaded micelles lacked alendronate. These results establish the feasibility of using alendronate to target osteogenic phytomolecules to sites of bone injury, which may guide the development of effective therapies against osteoporosis and, by extension, other bone disorders.
Collapse
Affiliation(s)
- Xiaoduan Sun
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chao Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhen Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zaiyi Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jili Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Zhirong Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingjie Du
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Wu Y, Wang Y, Chen F, Wang B. Loading rutin on surfaces by the layer-by-layer assembly technique to improve the oxidation resistance and osteogenesis of titanium implants in osteoporotic rats. Biomed Mater 2024; 19:045011. [PMID: 38740037 DOI: 10.1088/1748-605x/ad4aa8] [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: 12/22/2023] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
The purpose of this study was to construct a rutin-controlled release system on the surface of Ti substrates and investigate its effects on osteogenesis and osseointegration on the surface of implants. The base layer, polyethylenimine (PEI), was immobilised on a titanium substrate. Then, hyaluronic acid (HA)/chitosan (CS)-rutin (RT) multilayer films were assembled on the PEI using layer-by-layer (LBL) assembly technology. We used scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy and contact angle measurements to examine all Ti samples. The drug release test of rutin was also carried out to detect the slow-release performance. The osteogenic abilities of the samples were evaluated by experiments on an osteoporosis rat model and MC3T3-E1 cells. The results (SEM, FTIR and contact angle measurements) all confirmed that the PEI substrate layer and HA/CS-RT multilayer film were effectively immobilised on titanium. The drug release test revealed that a rutin controlled release mechanism had been successfully established. Furthermore, thein vitrodata revealed that osteoblasts on the coated titanium matrix had greater adhesion, proliferation, and differentiation capacity than the osteoblasts on the pure titanium surface. When MC3T3-E1 cells were exposed to H2O2-induced oxidative stressin vitro, cell-based tests revealed great tolerance and increased osteogenic potential on HA/CS-RT substrates. We also found that the HA/CS-RT coating significantly increased the new bone mass around the implant. The LBL-deposited HA/CS-RT multilayer coating on the titanium base surface established an excellent rutin-controlled release system, which significantly improved osseointegration and promoted osteogenesis under oxidative stress conditions, suggesting a new implant therapy strategy for patients with osteoporosis.
Collapse
Affiliation(s)
- Yinsheng Wu
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Yong Wang
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Fengyan Chen
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| | - Bingzhang Wang
- Department of Orthopedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, No. 75, JinXiu Road, Lucheng District, Wenzhou 325000, Zhejiang Province, People's Republic of China
| |
Collapse
|
3
|
Seki Y, Ohkuma RC, Miyakawa Y, Karakida T, Yamamoto R, Yamakoshi Y. Hyaluronan and chondroitin sulfate in chicken-vegetable bone broth delay osteoporosis progression. J Food Sci 2024; 89:1791-1803. [PMID: 38317402 DOI: 10.1111/1750-3841.16962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Bone broth has recently gained worldwide recognition as a superfood that supplements several nutrients lacking in modern human diets; however, little is known of its efficacy on osteoporosis. Therefore, we aimed to identify the components of chicken-vegetable bone broth (CVBB) that are associated with osteoporosis prevention and verified the efficacy of these components using in vivo studies. In biochemical and cell biological experiments, CVBB was fractionated using ion exchange chromatography (IEC), and the effect of each IEC fraction on osteoclast differentiation was evaluated based on tartrate-resistant acid phosphatase (TRAP) activity, TRAP staining, and quantitative polymerase chain reaction analysis using mouse macrophage-like cells (RAW264 cell). In animal experiments, an ovariectomized (OVX) rat model was generated, followed by whole bone broth (OVX/CVBB) or IEC fraction (OVX/CVBB-Ext) administration and bone structural parameter characterization of OVX rat tibia based on micro-CT. Four CVBB fractions were obtained using IEC, and the fraction containing both hyaluronan and chondroitin sulfate (CVBB-Ext) led to the maximum inhibition of RAW264 cell differentiation. CVBB-Ext downregulated the expression of osteoclast differentiation marker genes. In animal experiments, the OVX group showed a clear decrease in bone density compared to that in the Sham operation group. The OVX/CVBB and OVX/CVBB-Ext groups showed increased bone mineral density and bone volume/tissue volume values compared to those in the OVX/control group. These results suggested that CVBB and CVBB-Ext slowed osteoporosis progression. Therefore, we conclude that hyaluronan and chondroitin sulfate in CVBB are key substances that impede osteoporosis progression. PRACTICAL APPLICATION: This study provides practical information on the effects of bone broth ingredients on osteoporosis to expand the current knowledge on the efficacy of bone broth, which is a widely consumed food. These results may help in the future development of bone broth as a dietary supplement for managing osteoporosis.
Collapse
Affiliation(s)
- Yuka Seki
- Fourth Undergraduate Student, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| | - Risako Chiba Ohkuma
- Department of Biochemistry and Molecular Biology, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| | - Yuri Miyakawa
- Department of Pediatric Dentistry, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| | - Takeo Karakida
- Department of Biochemistry and Molecular Biology, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| | - Ryuji Yamamoto
- Department of Biochemistry and Molecular Biology, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| | - Yasuo Yamakoshi
- Department of Biochemistry and Molecular Biology, School of Dental Medicine, Tsurumi University, Tsurumi-ku, Yokohama, Japan
| |
Collapse
|
4
|
de Moraes R, Plepis AMDG, Martins VDCA, Garcia CF, Galdeano EA, Maia FLM, Machado EG, Munhoz MDAES, Buchaim DV, Fernandes VAR, Beraldo RA, Buchaim RL, da Cunha MR. Viability of Collagen Matrix Grafts Associated with Nanohydroxyapatite and Elastin in Bone Repair in the Experimental Condition of Ovariectomy. Int J Mol Sci 2023; 24:15727. [PMID: 37958710 PMCID: PMC10649653 DOI: 10.3390/ijms242115727] [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: 09/28/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Bone lesions have the capacity for regeneration under normal conditions of the bone metabolism process. However, due to the increasing incidence of major traumas and diseases that cause bone-mineral deficiency, such as osteoporosis, scaffolds are needed that can assist in the bone regeneration process. Currently, natural polymeric scaffolds and bioactive nanoparticles stand out. Therefore, the objective of the study was to evaluate the osteoregenerative potential in tibiae of healthy and ovariectomized rats using mineralized collagen and nanohydroxyapatite (nHA) scaffolds associated with elastin. The in-vivo experimental study was performed with 60 20-week-old Wistar rats, distributed into non-ovariectomized (NO) and ovariectomized (O) groups, as follows: Controls (G1-NO-C and G4-O-C); Collagen with nHA scaffold (G2-NO-MSH and G5-O-MSH); and Collagen with nHA and elastin scaffold (G3-NO-MSHC and G6-O-MSHC). The animals were euthanized 6 weeks after surgery and the samples were analyzed by macroscopy, radiology, and histomorphometry. ANOVA and Tukey tests were performed with a 95% CI and a significance index of p < 0.05. In the histological analyses, it was possible to observe new bone formed with an organized and compact morphology that was rich in osteocytes and with maturity characteristics. This is compatible with osteoconductivity in both matrices (MSH and MSHC) in rats with normal conditions of bone metabolism and with gonadal deficiency. Furthermore, they demonstrated superior osteogenic potential when compared to control groups. There was no significant difference in the rate of new bone formation between the scaffolds. Ovariectomy did not exacerbate the immune response but negatively influenced the bone-defect repair process.
Collapse
Affiliation(s)
- Renato de Moraes
- Interunit Postgraduate Program in Bioengineering (EESC/FMRP/IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil; (R.d.M.); (A.M.d.G.P.); (C.F.G.); (M.R.d.C.)
| | - Ana Maria de Guzzi Plepis
- Interunit Postgraduate Program in Bioengineering (EESC/FMRP/IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil; (R.d.M.); (A.M.d.G.P.); (C.F.G.); (M.R.d.C.)
- São Carlos Institute of Chemistry, University of São Paulo, USP, São Carlos 13566-590, Brazil;
| | | | - Claudio Fernandes Garcia
- Interunit Postgraduate Program in Bioengineering (EESC/FMRP/IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil; (R.d.M.); (A.M.d.G.P.); (C.F.G.); (M.R.d.C.)
| | - Ewerton Alexandre Galdeano
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| | | | - Eduardo Gomes Machado
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| | - Marcelo de Azevedo e Souza Munhoz
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| | - Daniela Vieira Buchaim
- Medical School, University Center of Adamantina (UniFAI), Adamantina 17800-000, Brazil;
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Marilia 17525-902, Brazil
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo 05508-270, Brazil
| | - Victor Augusto Ramos Fernandes
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| | - Rodrigo Alves Beraldo
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo 05508-270, Brazil
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of Sao Paulo, Bauru 17012-901, Brazil
| | - Marcelo Rodrigues da Cunha
- Interunit Postgraduate Program in Bioengineering (EESC/FMRP/IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil; (R.d.M.); (A.M.d.G.P.); (C.F.G.); (M.R.d.C.)
- Department of Morphology and Pathology, Medical College of Jundiai, Jundiaí 13202-550, Brazil; (E.A.G.); (E.G.M.); (M.d.A.e.S.M.); (V.A.R.F.); (R.A.B.)
| |
Collapse
|
5
|
Yang X, Yang X, Luo P, Zhong Y, Zhang B, Zhu W, Liu M, Zhang X, Lai Q, Wei Y. Novel one-pot strategy for fabrication of a pH-Responsive bone-targeted drug self-frame delivery system for treatment of osteoporosis. Mater Today Bio 2023; 20:100688. [PMID: 37441135 PMCID: PMC10333685 DOI: 10.1016/j.mtbio.2023.100688] [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: 03/28/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023] Open
Abstract
Osteoporosis (OP) is a systemic metabolic orthopedic disorder prevalent in elderly people, that is characterized by a decrease in bone mass. Although many therapeutics have been adopted for OP treatment, many of them are still not well satisfied clinical requirements and therefore development of novel therapeutics is of great significance. In this work, a novel bone-targeting drug self-frame delivery system (DSFDS) with high drug loading efficiency and pH responsive drug release was fabricated by condensation of curcumin (Cur), amino group terminated polyethylene glycol (NH2-PEG), and alendronate (ALN) using hexachlorocyclotriphosphonitrile (HCCP) as the linker. The final product named as HCCP-Cur-PEG-ALN (HCPA NPs) displayed excellent water dispersity with small size (181.9 ± 25.9 nm). Furthermore, the drug loading capacity of Cur can reach 25.8%, and Cur can be released from HCPA NPs under acidic environment. Owing to the introduction of ALN, HCPA NPs exhibited strong binding to HAp in vitro and excellent bone-targeting effect in vivo. Results from cellular and biochemical analyses revealed that HCPA NPs could effectively inhibit the formation and differentiation function of osteoclasts. More importantly, we also demonstrated that HCPA NPs could effectively reduce bone loss in OVX mice with low toxicity to major organs. The above results clearly demonstrated that HCPA NPs are promising for OP treatment. Given the simplicity and well designability of fabrication strategy, explicit therapy efficacy and low toxicity of HCPA NPs, we believe that this work should be of great interest for fabrication of various DSFDS to deal with many diseases.
Collapse
Affiliation(s)
- Xinmin Yang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, China
| | - Xiaowei Yang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, China
| | - Peng Luo
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, China
| | - Yanlong Zhong
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Chinese Medicine Preparation of Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Meiying Liu
- Key Laboratory of Modern Chinese Medicine Preparation of Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Xiaoyong Zhang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang, 330031, China
| | - Qi Lai
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
6
|
Bian Y, Hu T, Lv Z, Xu Y, Wang Y, Wang H, Zhu W, Feng B, Liang R, Tan C, Weng X. Bone tissue engineering for treating osteonecrosis of the femoral head. EXPLORATION (BEIJING, CHINA) 2023; 3:20210105. [PMID: 37324030 PMCID: PMC10190954 DOI: 10.1002/exp.20210105] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/12/2022] [Indexed: 06/16/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and complicated disease with an unclear etiology. Femoral head-preserving surgeries have been devoted to delaying and hindering the collapse of the femoral head since their introduction in the last century. However, the isolated femoral head-preserving surgeries cannot prevent the natural progression of ONFH, and the combination of autogenous or allogeneic bone grafting often leads to many undesired complications. To tackle this dilemma, bone tissue engineering has been widely developed to compensate for the deficiencies of these surgeries. During the last decades, great progress has been made in ingenious bone tissue engineering for ONFH treatment. Herein, we comprehensively summarize the state-of-the-art progress made in bone tissue engineering for ONFH treatment. The definition, classification, etiology, diagnosis, and current treatments of ONFH are first described. Then, the recent progress in the development of various bone-repairing biomaterials, including bioceramics, natural polymers, synthetic polymers, and metals, for treating ONFH is presented. Thereafter, regenerative therapies for ONFH treatment are also discussed. Finally, we give some personal insights on the current challenges of these therapeutic strategies in the clinic and the future development of bone tissue engineering for ONFH treatment.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Tingting Hu
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yingjie Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Wei Zhu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Bin Feng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Chaoliang Tan
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Słota D, Piętak K, Jampilek J, Sobczak-Kupiec A. Polymeric and Composite Carriers of Protein and Non-Protein Biomolecules for Application in Bone Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2235. [PMID: 36984115 PMCID: PMC10059071 DOI: 10.3390/ma16062235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Conventional intake of drugs and active substances is most often based on oral intake of an appropriate dose to achieve the desired effect in the affected area or source of pain. In this case, controlling their distribution in the body is difficult, as the substance also reaches other tissues. This phenomenon results in the occurrence of side effects and the need to increase the concentration of the therapeutic substance to ensure it has the desired effect. The scientific field of tissue engineering proposes a solution to this problem, which creates the possibility of designing intelligent systems for delivering active substances precisely to the site of disease conversion. The following review discusses significant current research strategies as well as examples of polymeric and composite carriers for protein and non-protein biomolecules designed for bone tissue regeneration.
Collapse
Affiliation(s)
- Dagmara Słota
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Karina Piętak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| | - Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Department of Chemical Biology, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 783 71 Olomouc, Czech Republic
| | - Agnieszka Sobczak-Kupiec
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland
| |
Collapse
|
8
|
Lei C, Song JH, Li S, Zhu YN, Liu MY, Wan MC, Mu Z, Tay FR, Niu LN. Advances in materials-based therapeutic strategies against osteoporosis. Biomaterials 2023; 296:122066. [PMID: 36842238 DOI: 10.1016/j.biomaterials.2023.122066] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
Osteoporosis is caused by the disruption in homeostasis between bone formation and bone resorption. Conventional management of osteoporosis involves systematic drug administration and hormonal therapy. These treatment strategies have limited curative efficacy and multiple adverse effects. Biomaterials-based therapeutic strategies have recently emerged as promising alternatives for the treatment of osteoporosis. The present review summarizes the current status of biomaterials designed for managing osteoporosis. The advantages of biomaterials-based strategies over conventional systematic drug treatment are presented. Different anti-osteoporotic delivery systems are concisely addressed. These materials include injectable hydrogels and nanoparticles, as well as anti-osteoporotic bone tissue engineering materials. Fabrication techniques such as 3D printing, electrostatic spinning and artificial intelligence are appraised in the context of how the use of these adjunctive techniques may improve treatment efficacy. The limitations of existing biomaterials are critically analyzed, together with deliberation of the future directions in biomaterials-based therapies. The latter include discussion on the use of combination strategies to enhance therapeutic efficacy in the osteoporosis niche.
Collapse
Affiliation(s)
- Chen Lei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing-Han Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Song Li
- School of Stomatology, Xinjiang Medical University. Urumqi 830011, China
| | - Yi-Na Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming-Yi Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Mei-Chen Wan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhao Mu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Franklin R Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Li-Na Niu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
9
|
Kim K, Kim MG, Lee GM. Improving bone morphogenetic protein (BMP) production in CHO cells through understanding of BMP synthesis, signaling and endocytosis. Biotechnol Adv 2023; 62:108080. [PMID: 36526238 DOI: 10.1016/j.biotechadv.2022.108080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) are a group of growth factors with the clinical potential to regulate cartilage and bone formation. Functionally active mature recombinant human BMPs (rhBMPs), produced primarily in Chinese hamster ovary (CHO) cells for clinical applications, are considered difficult to express because they undergo maturation processes, signaling pathways, or endocytosis. Although BMPs are a family of proteins with similar mature domain sequence identities, their individual properties are diverse. Thus, understanding the properties of individual rhBMPs is essential to improve rhBMP production in CHO cells. In this review, we discuss various approaches to improve rhBMP production in CHO cells by understanding the overall maturation process, signaling pathways and endocytosis of individual rhBMPs.
Collapse
Affiliation(s)
- Kyungsoo Kim
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Mi Gyeom Kim
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
10
|
Reversing the imbalance in bone homeostasis via sustained release of SIRT-1 agonist to promote bone healing under osteoporotic condition. Bioact Mater 2023; 19:429-443. [PMID: 35574058 PMCID: PMC9079176 DOI: 10.1016/j.bioactmat.2022.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
The imbalance of bone homeostasis is the root cause of osteoporosis. However current therapeutic approaches mainly focus on either anabolic or catabolic pathways, which often fail to turn the imbalanced bone metabolism around. Herein we reported that a SIRT-1 agonist mediated molecular therapeutic strategy to reverse the imbalance in bone homeostasis by simultaneously regulating osteogenesis and osteoclastogenesis via locally sustained release of SRT2104 from mineral coated acellular matrix microparticles. Immobilization of SRT2104 on mineral coating (MAM/SRT) harnessing their electrostatic interactions resulted in sustained release of SIRT-1 agonist for over 30 days. MAM/SRT not only enhanced osteogenic differentiation and mineralization, but also attenuated the formation and function of excessive osteoclasts via integrating multiple vital upstream signals (β-catenin, FoxOs, Runx2, NFATc1, etc.) in vitro. Osteoporosis animal model also validated that it accelerated osteoporotic bone healing and improved osseointegration of the surrounding bone. Overall, our work proposes a promising strategy to treat osteoporotic bone defects by reversing the imbalance in bone homeostasis using designated small molecule drug delivery systems. A mineral coated acellular matrix microcarriers sustainably release SIRT2104 more than 30 days. This drug delivery system regulates osteogenesis and osteoclastogenesis. It can accelerate osteoporotic bone healing by reversing the imbalance in bone homeostasis.
Collapse
|
11
|
Xi Y, Wang W, Ma L, Xu N, Shi C, Xu G, He H, Pan W. Alendronate modified mPEG-PLGA nano-micelle drug delivery system loaded with astragaloside has anti-osteoporotic effect in rats. Drug Deliv 2022; 29:2386-2402. [PMID: 35869674 PMCID: PMC9310824 DOI: 10.1080/10717544.2022.2086942] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Astragaloside (AS) has an anti-osteoporotic effect, but its poor water solubility and low bioavailability limit its application. In this study, a novel nano-carrier with bone targeting was prepared by modifying mPEG-PLGA with alendronate (AL) before incorporation into astragaloside nano-micelles (AS-AL-mPEG-PLGA) to enhance the oral bioavailability, bone targeting and anti-osteoporosis effect of AS. The release behavior of AS-AL-mPEG-PLGA in vitro was investigated via dialysis. The pharmacokinetics of AS-AL-mPEG-PLGA was studied in Sprague-Dawley (SD) rats. The cytotoxicity of AS-AL-mPEG-PLGA in vitro (via MTT method), coupled with bone targeting ability in vitro and in vivo were evaluated. The therapeutic effects of free AS and AS-AL-mPEG-PLGA (ELISA, micro-CT, H&E staining) were compared in osteoporotic rats. AS-AL-mPEG-PLGA with smaller particle size (45.3 ± 3.8 nm) and high absolute zeta potential (−23.02 ± 0.51 mV) were successfully prepared, wherein it demonstrated higher entrapment efficiency (96.16 ± 0.18%), a significant sustained-release effect for 96 h and acceptable safety within 10–200 μg/mL. AS-AL-mPEG-PLGA could enhance the hydroxyapatite affinity and bone tissue concentration of AS. The relative bioavailability of AS-AL-mPEG-PLGA was 233.90% compared with free AS. In addition, the effect of AS in reducing serum levels of bone metabolism-related indicators, restoring the bone microarchitecture and improving bone injury could be enhanced by AS-AL-mPEG-PLGA. AS-AL-mPEG-PLGA with small particle size, good stability, remarkable sustained-release effect, safety and bone targeting was successfully constructed in this experiment to potentially improve the oral bioavailability and anti-osteoporosis effect of AS. Thus, AS-AL-mPEG-PLGA may be a promising strategy to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Yanhai Xi
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weiheng Wang
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Liang Ma
- Minimally invasive Spinal Surgery department, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ning Xu
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Changgui Shi
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guohua Xu
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hailong He
- Department of Orthopedics, Spine Surgery, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wenming Pan
- Department of Spine Surgery, The Affiliated Changshu Hospital of Xuzhou Medical School, The Second People's Hospital of Changshu, Changshu, China
| |
Collapse
|
12
|
Advances in bone regeneration with growth factors for spinal fusion: A literature review. NORTH AMERICAN SPINE SOCIETY JOURNAL 2022; 13:100193. [PMID: 36605107 PMCID: PMC9807829 DOI: 10.1016/j.xnsj.2022.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Bone tissue is regenerated via the spatiotemporal involvement of various cytokines. Among them, the bone morphogenetic protein (BMP), which plays a vital role in the bone regeneration process, has been applied clinically for the treatment of refractory orthopedic conditions. Although BMP therapy using a collagen carrier has shown efficiency in bone regeneration over the last two decades, a major challenge-considerable side effects associated with the acute release of high doses of BMPs-has also been revealed. To improve BMP efficiency, the development of new carriers and biologics that can be used in conjunction with BMPs is currently underway. In this review, we describe the current status and future prospects of bone regeneration therapy, with a focus on BMPs. Furthermore, we outline the characteristics and molecular signaling pathways involving BMPs, clinical applications of BMPs in orthopedics, clinical results of BMP use in human spinal surgeries, drugs combined with BMPs to provide synergistic effects, and novel BMP carriers.
Collapse
|
13
|
Wu J, Wang S, Zheng Z, Li J. Fabrication of Biologically Inspired Electrospun Collagen/Silk fibroin/bioactive glass composited nanofibrous scaffold to accelerate the treatment efficiency of bone repair. Regen Ther 2022; 21:122-138. [PMID: 35844293 PMCID: PMC9253997 DOI: 10.1016/j.reth.2022.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/15/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022] Open
Abstract
Bone disease and disorder treatment might be difficult because of its complicated nature. Millions of patients each year need bone substitutes that may help them recover quickly from a variety of illnesses. Synthetic bone replacements that mirror the structural, chemical, and biological features of bone matrix structure will be very helpful and in high demand. In this research, the inorganic bioactive glass nanoparticles matrixed with organic collagen and silk fibroin structure (COL/SF/CaO-SiO2) were used to create multifunctional bone-like fibers in this study, which we describe here. The fiber structure is organized in a layered fashion comparable to the sequence in which apatite and neo tissue are formed. The amino groups in COL and SF combined with CaO-SiO2 to stabilize the resulting composite nanofiber. Morphological and functional studies confirmed that crystalline CaO-SiO2 nanoparticles with average sizes of 20 ± 5 nm are anchored on a 115 ± 10 nm COL/SF nanofiber matrix. X-ray photoelectron spectroscopic (XPS) results confirmed the presence of C, N, O, Ca, and Si in the composite fiber with an atomic percentage of 59.46, 3.30, 20.25, 3.38 and 13.61%. respectively. The biocompatibility examination with osteoblast cells (Saos-2) revealed that the CAL/SF/CaO-SiO2 composite nanofiber had enhanced osteogenic activity. Finally, when the CAL/SF/CaO-SiO2 composite nanofiber scaffolds were used to treat an osteoporotic bone defect in a rat model, the composite nanofiber scaffolds significantly promoted bone regeneration and vascularization. This novel fibrous scaffold class represents a potential breakthrough in the design of advanced materials for complicated bone regeneration.
Collapse
Affiliation(s)
- Jianjun Wu
- Department of Spine Surgery, The Third Clinical Medical College, Fujian Medical University
- Department of Spine Surgery, Fuzhou Second Hospital, PR China
- Corresponding author. No. 47, Shangteng Road, Cangshan District, Fuzhou 350007, Fujian Province, China.
| | - Shengxuan Wang
- Department of Spine Surgery, The Third Clinical Medical College, Fujian Medical University
- Department of Spine Surgery, Fuzhou Second Hospital, PR China
| | - Zhong Zheng
- Department of Spine Surgery, The Third Clinical Medical College, Fujian Medical University
- Department of Spine Surgery, Fuzhou Second Hospital, PR China
| | - Jianbao Li
- Department of Spine Surgery, The Third Clinical Medical College, Fujian Medical University
- Department of Spine Surgery, Fuzhou Second Hospital, PR China
| |
Collapse
|
14
|
Badiee M, Hassanzadeh Nemati N, Khorasani MT, Shokrgozar MA. Highly porous bio-glass scaffolds fabricated by polyurethane template method with hydrothermal treatment for tissue engineering uses. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1513-1521. [PMID: 36544527 PMCID: PMC9742566 DOI: 10.22038/ijbms.2022.67272.14746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/15/2022] [Indexed: 12/24/2022]
Abstract
Objectives Bioglass scaffolds, which contain a significant percentage of porosity for tissue engineering purposes, have low strength. For increasing the strength and efficiency of such structures for use in tissue engineering, fabrication of hierarchical meso/macro-porous bioglass scaffolds, developing their mechanical strength by hydrothermal treatment and adjusting pH method, and achieving the appropriate mesopore size for loading large biomolecules, were considered in this study. Materials and Methods Mesoporous bioglass (MBG) powders were synthesized using cetyltrimethylammonium bromide as a surfactant, with different amounts of calcium sources to obtain the appropriate size of the mesoporous scaffolds. Then MBG scaffolds were fabricated by a polyurethane foam templating method, and for increasing scaffold strength hydrothermal treatment (90 °C, for 5 days) and adjustment pH (pH=9) method was used to obtain hierarchical meso/macro-porous structures. The sample characterization was done by Simultaneous thermal analysis, Fourier transform infrared spectroscopy, Field Emission Scanning electron microscopy, small and wide-angle X-ray powder diffractions, transmission electron microscopy, and analysis of nitrogen adsorption-desorption isotherm. The mechanical strength of scaffolds was also determined. Results The MBG scaffolds based on 80.28 (wt.) % SiO2- 17.89 (wt.) % CaO- 1.81 (wt.) % P2O5 presented interconnected large pores and pores in the range of 100-150 μm and 6-18 nm, respectively and 0.4 MPa compressive strength. Conclusion The total pore volume and specific surface area were obtained from the Brunauer-Emmett-Teller theory, 0.709 cm3 g-1 and 213.83 m2 g-1, respectively. These findings could be considered in bone-cartilage tissue engineering.
Collapse
Affiliation(s)
- Maryam Badiee
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 14778-93885, Iran
| | - Nahid Hassanzadeh Nemati
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 14778-93885, Iran ,Corresponding author: Nahid Hassanzadeh Nemati. Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 14778-93885, Iran. Tel/Fax: +98-21-44861815;
| | | | | |
Collapse
|
15
|
Zhang HJ, Li FS, Wang F, Wang H, He TC, Reid RR, He BC, Xia Q. Transgenic PDGF-BB sericin hydrogel potentiates bone regeneration of BMP9-stimulated mesenchymal stem cells through a crosstalk of the Smad-STAT pathways. Regen Biomater 2022; 10:rbac095. [PMID: 36683747 PMCID: PMC9847547 DOI: 10.1093/rb/rbac095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Silk as a natural biomaterial is considered as a promising bone substitute in tissue regeneration. Sericin and fibroin are the main components of silk and display unique features for their programmable mechanical properties, biocompatibility, biodegradability and morphological plasticity. It has been reported that sericin recombinant growth factors (GFs) can support cell proliferation and induce stem cell differentiation through cross-talk of signaling pathways during tissue regeneration. The transgenic technology allows the productions of bioactive heterologous GFs as fusion proteins with sericin, which are then fabricated into solid matrix or hydrogel format. Herein, using an injectable hydrogel derived from transgenic platelet-derived GF (PDGF)-BB silk sericin, we demonstrated that the PDGF-BB sericin hydrogel effectively augmented osteogenesis induced by bone morphogenetic protein (BMP9)-stimulated mesenchymal stem cells (MSCs) in vivo and in vitro, while inhibiting adipogenic differentiation. Further gene expression and protein-protein interactions studies demonstrated that BMP9 and PDGF-BB synergistically induced osteogenic differentiation through the cross-talk between Smad and Stat3 pathways in MSCs. Thus, our results provide a novel strategy to encapsulate osteogenic factors and osteoblastic progenitors in transgenic sericin-based hydrogel for robust bone tissue engineering.
Collapse
Affiliation(s)
- Hui-Jie Zhang
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Fu-Shu Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Feng Wang
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| | - Han Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacy, Panzhou People’s Hospital, Guizhou 553599, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bai-Cheng He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qingyou Xia
- Biological Science Research Center, Chongqing Key Laboratory of Sericultural Science, Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400715, China
| |
Collapse
|
16
|
Wang R, Wang Y, Zai W, Xu N. Bibliometric and visual analysis of mesenchymal stem cells in the treatment of osteoporosis based on CiteSpace software. Medicine (Baltimore) 2022; 101:e31859. [PMID: 36401376 PMCID: PMC9678533 DOI: 10.1097/md.0000000000031859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The focus of research in the treatment of osteoporosis (OP) has evolved from promoting bone formation and inhibiting bone resorption to current stem cell therapy. Due to their multipotent differentiation properties, mesenchymal stem cells (MSCs) can repair degenerated bones through transplantation, and have become a new method for the treatment of OP. METHODS Relevant literatures included in the Web of Science database core collection database from 2012 to 2021 were retrieved. CiteSpace software was used to analyze the cooperative relationship among authors, journals, institutions, and countries, and to analyze the co-citation situation of the literature. And performed co-occurrence analysis, cluster analysis and burst analysis of keywords, draw visual maps and analyzed the results. RESULTS A total of 2100 papers were included, and the number of papers published from 2012 to 2021 was on the rise. A total of 484 authors were included, and 176 authors published more than 3 papers. The high-yield authors were mainly represented by YAN JIN and BO GAO. A total of 99 journals were included, and the journal with the most publications was J BONE MINER RES. A total of 787 institutions were included, and the institution with the largest number of publications was Shanghai Jiao Tong University. A total of 65 countries were included. The country with the largest number of publications was China, and the United States had the highest centrality. The co-citation analysis of the literature found 2 articles with high citation frequency and high centrality. The main research direction was the mechanism of MSCs in the treatment of osteoporosis. A total of 133 keywords were included, and the hot keywords were osteogenic differentiation, expression, proliferation, bone marrow, etc. CONCLUSIONS The research hotspots in this field mainly focused on the mechanism of bone regeneration, proliferation and osteogenic differentiation of bone marrow MSCs, and the expression of osteogenic-related genes. The future research trends in this field are predicted to be the mechanism of action of microRNA and long non-coding RNA on MSCs and their relationship with OP, the mechanism of MSCs adipogenic and osteogenic differentiation, and tissue engineering scaffolds applications.
Collapse
Affiliation(s)
- Runfang Wang
- Medical School of Rehabilitation, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yueying Wang
- Medical School of Rehabilitation, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weiyi Zai
- Medical School of Rehabilitation, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ning Xu
- Medical School of Rehabilitation, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Ning Xu, Medical School of Rehabilitation, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China (e-mail: )
| |
Collapse
|
17
|
High-efficient engineering of osteo-callus organoids for rapid bone regeneration within one month. Biomaterials 2022; 288:121741. [PMID: 36031458 DOI: 10.1016/j.biomaterials.2022.121741] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/19/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023]
Abstract
Large bone defects that cannot form a callus tissue are often faced with long-time recovery. Developmental engineering-based strategies with mesenchymal stem cell (MSC) aggregates have shown enhanced potential for bone regeneration. However, MSC aggregates are different from the physiological callus tissues, which limited the further endogenous osteogenesis. This study aims to achieve engineering of osteo-callus organoids for rapid bone regeneration in cooperation with bone marrow-derived stem cell (BMSC)-loaded hydrogel microspheres (MSs) by digital light-processing (DLP) printing technology and stepwise-induction. The printed MSC-loaded MSs aggregated into osteo-callus organoids after chondrogenic induction and showed much higher chondrogenic efficiency than that of traditional MSC pellets. Moreover, the osteo-callus organoids exhibited stage-specific gene expression pattern that recapitulated endochondral ossification process, as well as a synchronized state of cell proliferation and differentiation, which highly resembled the diverse cell compositions and behaviors of developmentally endochondral ossification. Lastly, the osteo-callus organoids efficiently led to rapid bone regeneration within only 4 weeks in a large bone defect in rabbits which need 2-3 months in previous tissue engineering studies. The findings suggested that in vitro engineering of osteo-callus organoids with developmentally osteogenic properties is a promising strategy for rapid bone defect regeneration and recovery.
Collapse
|
18
|
Camacho-Alonso F, Tudela-Mulero MR, Navarro JA, Buendía AJ, Mercado-Díaz AM. Use of buccal fat pad-derived stem cells cultured on bioceramics for repair of critical-sized mandibular defects in healthy and osteoporotic rats. Clin Oral Investig 2022; 26:5389-5408. [PMID: 35524820 PMCID: PMC9381637 DOI: 10.1007/s00784-022-04506-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/14/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To compare new bone formation in mandibular symphysis critical-sized bone defects (CSBDs) in healthy and osteoporotic rats filled with bioceramics (BCs) with or without buccal fat pad mesenchymal stem cells (BFPSCs). MATERIALS AND METHODS Thirty-two adult female Sprague-Dawley rats were randomized to two groups (n = 16 per group): group 1 healthy and group 2 osteoporotic (with bilateral ovariectomy). The central portion of the rat mandibular symphysis was used as a physiological CSBD. In each group, eight defects were filled with BC (hydroxyapatite 60% and β-tricalcium phosphate 40%) alone and eight with BFPSCs cultured on BC. The animals were sacrificed at 4 and 8 weeks, and the mandibles were processed for micro-computed tomography to analyze radiological union and bone mineral density (BMD); histological analysis of the bone union; and immunohistochemical analysis, which included immunoreactivity of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2). RESULTS In both groups, CSBDs filled with BC + BFPSCs showed greater radiological bone union, BMD and histological bone union, and more VEGF and BMP-2 positivity, compared with CSBDs treated with BC alone at 4 and 8 weeks. CONCLUSIONS The application of BFPSCs cultured on BCs improves bone regeneration in CSBDs compared with BCs alone in healthy and osteoporotic rats. CLINICAL RELEVANCE Our results may aid bone regeneration of maxillofacial CSBDs of both healthy and osteoporotic patients, but further studies are necessary.
Collapse
Affiliation(s)
- Fabio Camacho-Alonso
- Department of Oral Surgery, University of Murcia, Murcia, Spain.
- Oral Surgery Teaching Unit, University Dental Clinic, Morales Meseguer Hospital (2Nd Floor), Marqués de los Vélez s/n, 30008, Murcia, Spain.
| | | | - J A Navarro
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - A J Buendía
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | | |
Collapse
|
19
|
Camacho-Alonso F, Tudela-Mulero MR, Buendía AJ, Navarro JA, Pérez-Sayáns M, Mercado-Díaz AM. Bone regeneration in critical-sized mandibular symphysis defects using bioceramics with or without bone marrow mesenchymal stem cells in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats. Dent Mater 2022; 38:1283-1300. [PMID: 35717229 DOI: 10.1016/j.dental.2022.06.019] [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: 12/11/2021] [Revised: 05/14/2022] [Accepted: 06/05/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To compare new bone formation in mandibular critical-sized bone defects (CSBDs) in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats filled with bioceramics (BCs) with or without bone marrow mesenchymal stem cells (BMSCs). METHODS A total of 64 adult female Sprague-Dawley rats were randomized into four groups (n = 16 per group): Group 1 healthy, Group 2 diabetic, Group 3 osteoporotic, and Group 4 diabetic-osteoporotic rats. Streptozotocin was used to induce type 1 diabetes in Group 2 and 4, while bilateral ovariectomy was used to induce osteoporosis in Group 3 and 4. The central portion of the rat mandibular symphysis was used as a physiological CSBD. In each group, eight defects were filled with BC (hydroxypatatite 60% and β-tricalcium phosphate 40%) alone and eight with BMSCs cultured on BC. The animals were sacrificed at 4 and 8 weeks, and the mandibles were processed for micro-computed tomography to analyze radiological union and bone mineral density (BMD); histological analysis of the bone union; and immunohistochemical analysis, which included immunoreactivity of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2). RESULTS In all groups (healthy, diabetics, osteoporotics, and diabetics-osteoporotics), the CSBDs filled with BC + BMSCs showed greater radiological bone union, BMD, histological bone union, and more VEGF and BMP-2 positivity, in comparison with CSBDs treated with BC alone (at 4 and 8 weeks). CONCLUSIONS Application of BMSCs cultured on BCs improves bone regeneration in CSBDs compared with application of BCs alone in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats.
Collapse
Affiliation(s)
- F Camacho-Alonso
- Department of Oral Surgery, University of Murcia, Murcia, Spain.
| | | | - A J Buendía
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - J A Navarro
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - M Pérez-Sayáns
- Department of Oral Medicine, Oral Surgery and Implantology, University of Santiago de Compostela, Spain. MedOralRes Group, Health Research Institute of Santiago de Compostela (IDIS). Santiago de Compostela, Spain
| | | |
Collapse
|
20
|
Shar A, Aboutalebianaraki N, Misiti K, Sip YYL, Zhai L, Razavi M. A novel ultrasound-mediated nanodroplet-based gene delivery system for osteoporosis treatment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102530. [PMID: 35104672 DOI: 10.1016/j.nano.2022.102530] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/30/2021] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
This project aimed to develop, optimize, and test an ultrasound-responsive targeted nanodroplet system for the delivery of osteoporosis-related silencing gene Cathepsin K small interfering RNA (CTSK siRNA) for osteoporosis treatment. The nanodroplet (ND) is composed of a gas core made from perfluorocarbon, stabilized with albumin, encapsulated with CTSK siRNA, and embedded with alendronate (AL) for bone targeting (CTSK siRNA-ND-AL). Following the development, the responsiveness of CTSK siRNA-ND-AL to a therapeutic ultrasound probe was examined. The results of biocompatibility tests with human bone marrow-derived mesenchymal stem cells proved no significant cell death (P > 0.05). When the CTSK siRNA-ND-AL was supplemented with human osteoclast precursors, they suppressed osteoclastogenesis. Thus, this project establishes the potential of nanotechnology and ultrasound to deliver genes into the osteoclasts. This research also presents a novel ultrasound responsive and targeted nanodroplet platform that can be used as a gene and drug delivery system for various diseases including cancer.
Collapse
Affiliation(s)
- Angela Shar
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida, USA; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Nadia Aboutalebianaraki
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida, USA; Department of Material Sciences and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Kaylee Misiti
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Yuen Yee Li Sip
- Department of Material Sciences and Engineering, University of Central Florida, Orlando, Florida, USA; Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Lei Zhai
- Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA; Department of Chemistry, University of Central Florida, Orlando, Florida, USA
| | - Mehdi Razavi
- Biionix™ (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, Florida, USA; Department of Material Sciences and Engineering, University of Central Florida, Orlando, Florida, USA.
| |
Collapse
|
21
|
Deng Y, Wei W, Tang P. Applications of Calcium-Based Nanomaterials in Osteoporosis Treatment. ACS Biomater Sci Eng 2022; 8:424-443. [PMID: 35080365 DOI: 10.1021/acsbiomaterials.1c01306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With rapidly aging populations worldwide, osteoporosis has become a serious global public health problem. Caused by disordered systemic bone remodeling, osteoporosis manifests as progressive loss of bone mass and microarchitectural deterioration of bone tissue, increasing the risk of fractures and eventually leading to osteoporotic fragility fractures. As fracture risk increases, antiosteoporosis treatments transition from nonpharmacological management to pharmacological intervention, and finally to the treatment of fragility fractures. Calcium-based nanomaterials (CBNMs) have unique advantages in osteoporosis treatment because of several characteristics including similarity to natural bone, excellent biocompatibility, easy preparation and functionalization, low pH-responsive disaggregation, and inherent pro-osteogenic properties. By combining additional ingredients, CBNMs can play multiple roles to construct antiosteoporotic biomaterials with different forms. This review covers recent advances in CBNMs for osteoporosis treatment. For ease of understanding, CBNMs for antiosteoporosis treatment can be classified as locally applied CBNMs, such as implant coatings and filling materials for osteoporotic bone regeneration, and systemically administered CBNMs for antiosteoporosis treatment. Locally applied CBNMs for osteoporotic bone regeneration develop faster than the systemically administered CBNMs, an important consideration given the serious outcomes of fragility fractures. Nevertheless, many innovations in construction strategies and preparation methods have been applied to build systemically administered CBNMs. Furthermore, with increasing interest in delaying osteoporosis progression and avoiding fragility fracture occurrence, research into systemic administration of CBNMs for antiosteoporosis treatment will have more development prospects. Deep understanding of the CBNM preparation process and optimizing CBNM properties will allow for increased application of CBNMs in osteoporosis treatments in the future.
Collapse
Affiliation(s)
- Yuan Deng
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences No. 1 Bei-Er-Tiao, Beijing 100190, P. R. China
| | - Peifu Tang
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| |
Collapse
|
22
|
Zheng S, Zhou C, Yang H, Li J, Feng Z, Liao L, Li Y. Melatonin Accelerates Osteoporotic Bone Defect Repair by Promoting Osteogenesis-Angiogenesis Coupling. Front Endocrinol (Lausanne) 2022; 13:826660. [PMID: 35273570 PMCID: PMC8902312 DOI: 10.3389/fendo.2022.826660] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/18/2022] [Indexed: 01/10/2023] Open
Abstract
Previous studies have revealed that melatonin could play a role in anti-osteoporosis and promoting osteogenesis. However, the effects of melatonin treatment on osteoporotic bone defect and the mechanism underlying the effects of melatonin on angiogenesis are still unclear. Our study was aimed to investigate the potential effects of melatonin on angiogenesis and osteoporotic bone defect. Bone marrow mesenchymal stem cells (BMSCs) were isolated from the femur and tibia of rats. The BMSC osteogenic ability was assessed using alkaline phosphatase (ALP) staining, alizarin red S staining, qRT-PCR, western blot, and immunofluorescence. BMSC-mediated angiogenic potentials were determined using qRT-PCR, western blot, enzyme-linked immunosorbent assay, immunofluorescence, scratch wound assay, transwell migration assay, and tube formation assay. Ovariectomized (OVX) rats with tibia defect were used to establish an osteoporotic bone defect model and then treated with melatonin. The effects of melatonin treatment on osteoporotic bone defect in OVX rats were analyzed using micro-CT, histology, sequential fluorescent labeling, and biomechanical test. Our study showed that melatonin promoted both osteogenesis and angiogenesis in vitro. BMSCs treated with melatonin indicated higher expression levels of osteogenesis-related markers [ALP, osteocalcin (OCN), runt-related transcription factor 2, and osterix] and angiogenesis-related markers [vascular endothelial growth factor (VEGF), angiopoietin-2, and angiopoietin-4] compared to the untreated group. Significantly, melatonin was not able to facilitate human umbilical vein endothelial cell angiogenesis directly, but it possessed the ability to promote BMSC-mediated angiogenesis by upregulating the VEGF levels. In addition, we further found that melatonin treatment increased bone mineralization and formation around the tibia defect in OVX rats compared with the control group. Immunohistochemical staining indicated higher expression levels of osteogenesis-related marker (OCN) and angiogenesis-related markers (VEGF and CD31) in the melatonin-treated OVX rats. Then, it showed that melatonin treatment also increased the bone strength of tibia defect in OVX rats, with increased ultimate load and stiffness, as performed by three-point bending test. In conclusion, our study demonstrated that melatonin could promote BMSC-mediated angiogenesis and promote osteogenesis-angiogenesis coupling. We further found that melatonin could accelerate osteoporotic bone repair by promoting osteogenesis and angiogenesis in OVX rats. These findings may provide evidence for the potential application of melatonin in osteoporotic bone defect.
Collapse
Affiliation(s)
- Sheng Zheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chunhao Zhou
- Department of Orthopedics-Spine Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Han Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Junhua Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ziyu Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Liqing Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Yikai Li,
| |
Collapse
|
23
|
Silicified collagen scaffold induces semaphorin 3A secretion by sensory nerves to improve in-situ bone regeneration. Bioact Mater 2021; 9:475-490. [PMID: 34820584 PMCID: PMC8586786 DOI: 10.1016/j.bioactmat.2021.07.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/14/2022] Open
Abstract
Sensory nerves promote osteogenesis through the release of neuropeptides. However, the potential application and mechanism in which sensory nerves promote healing of bone defects in the presence of biomaterials remain elusive. The present study identified that new bone formation was more abundantly produced after implantation of silicified collagen scaffolds into defects created in the distal femur of rats. The wound sites were accompanied by extensive nerve innervation and angiogenesis. Sensory nerve dysfunction by capsaicin injection resulted in significant inhibition of silicon-induced osteogenesis in the aforementioned rodent model. Application of extracellular silicon in vitro induced axon outgrowth and increased expression of semaphorin 3 A (Sema3A) and semaphorin 4D (Sema4D) in the dorsal root ganglion (DRG), as detected by the upregulation of signaling molecules. Culture medium derived from silicon-stimulated DRG cells promoted proliferation and differentiation of bone marrow mesenchymal stem cells and endothelial progenitor cells. These effects were inhibited by the use of Sema3A neutralizing antibodies but not by Sema4D neutralizing antibodies. Knockdown of Sema3A in DRG blocked silicon-induced osteogenesis and angiogenesis almost completely in a femoral defect rat model, whereas overexpression of Sema3A promoted the silicon-induced phenomena. Activation of “mechanistic target of rapamycin” (mTOR) pathway and increase of Sema3A production were identified in the DRG of rats that were implanted with silicified collagen scaffolds. These findings support the role of silicon in inducing Sema3A production by sensory nerves, which, in turn, stimulates osteogenesis and angiogenesis. Taken together, silicon has therapeutic potential in orthopedic rehabilitation. Nerve innervation, vascularization and tissue mineralization integrated into a single scaffold. Silicified collagen scaffolds has therapeutic potential in orthopedic rehabilitation. Silicified collagen scaffolds promote in-situ bone regeneration via sensory nerve innervation and semaphorin 3A production.
Collapse
|
24
|
Li TL, Tao ZS, Wu XJ, Yang M, Xu HG. Selenium-modified calcium phosphate cement can accelerate bone regeneration of osteoporotic bone defect. J Bone Miner Metab 2021; 39:934-943. [PMID: 34189659 DOI: 10.1007/s00774-021-01240-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The purpose is to observe whether local administration with selenium (Se) can enhance the efficacy of calcium phosphate cement (CPC) in the treatment of osteoporotic bone defects. METHODS Thirty ovariectomized (OVX) rats with two defects were generated and randomly allocated into the following graft study groups: (1) OVX group (n = 10), (2) CPC group (n = 10); and (3) Se-CPC group (n = 10). Then, these selenium-modified calcium phosphate cement (Se-CPC) scaffolds were implanted into the femoral epiphysis bone defect model of OVX rats for 12 weeks. Micro-CT, history, western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis were used to observe the therapeutic effect and to explore the possible mechanism. RESULT Micro-CT and histological analysis evaluation showed that the Se-CPC group presented the strongest effect on bone regeneration and bone mineralization when compared with the CPC group and the OVX group. Protein expressions showed that the oxidative stress protein expressions, such as SOD2 and GPX1 of the Se-CPC group, are significantly higher than those of the OVX group and the CPC group, while Se-CPC remarkably reduced the expression of CAT. RT-qPCR analysis showed that the Se-CPC group displayed more OPG than the OVX and CPC groups (p < 0.05), while Se-CPC exhibited less RANKL than the OVX and CPC groups (p < 0.05). CONCLUSION Our current study demonstrated that Se-CPC is a scheme for rapid repair of femoral condylar defects, and these effects may be achieved by inhibiting local oxidative stress and through OPG/RANKL signaling pathway.
Collapse
Affiliation(s)
- Tian-Lin Li
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe shan Xi Road, Wuhu, 241001, Anhui, People's Republic of China
| | - Zhou-Shan Tao
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe shan Xi Road, Wuhu, 241001, Anhui, People's Republic of China.
| | - Xing-Jing Wu
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe shan Xi Road, Wuhu, 241001, Anhui, People's Republic of China
| | - Min Yang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe shan Xi Road, Wuhu, 241001, Anhui, People's Republic of China
| | - Hong-Guang Xu
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, No. 2, Zhe shan Xi Road, Wuhu, 241001, Anhui, People's Republic of China
| |
Collapse
|
25
|
Wu T, Liu W, Huang S, Chen J, He F, Wang H, Zheng X, Li Z, Zhang H, Zha Z, Lin Z, Chen Y. Bioactive strontium ions/ginsenoside Rg1-incorporated biodegradable silk fibroin-gelatin scaffold promoted challenging osteoporotic bone regeneration. Mater Today Bio 2021; 12:100141. [PMID: 34632364 PMCID: PMC8488313 DOI: 10.1016/j.mtbio.2021.100141] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022] Open
Abstract
Autogenous healing of osteoporotic fractures is challenging, as the regenerative capacity of bone tissues is impaired by estrogen reduction and existed pro-inflammatory cytokines. In this study, a biofunctional ginsenoside Rg1 and strontium-containing mineral (SrHPO4, SrP)-incorporated biodegradable silk fibroin-gelatin (SG) scaffold (Rg1/SrP/SG) was developed to stimulate the osteoporotic bone repair. The incorporation of 15 wt% SrP significantly enhanced the mechanical strength, stimulated the osteogenic differentiation of mouse bone marrow mesenchymal stem cells, and suppressed the osteoclastogenesis of RAW264.7 in a concentration-related manner. The loading of Rg1 in SG and 15SrP/SG scaffolds obviously promoted the angiogenesis of human umbilical vein endothelial cells via activating the expression of vascular endothelial growth factor and basic fibroblast growth factor genes and proteins. The bioactive strontium ions (Sr2+) and Rg1 released from the scaffolds together mediated lipopolysaccharide-treated macrophages polarizing into M2 type. They downregulated the expression of inflammatory-related genes (interleukin (IL)-1β, tumor necrosis factor α, and IL-6) and stimulated the expression of genes related to anti-inflammation (Arginase and IL-10) as well as bone repair (BMP-2 and PDGF-BB) in the macrophages. The in vivo results also displayed that SrP and Rg1 significantly promoted the bone repair effect of SG scaffolds in osteoporotic critical-sized calvarial defects. Besides, the degradation rate of the scaffolds was close to the bone regeneration rate. Therefore, the simultaneous addition of SrP and Rg1 is a promising way for facilitating the osteoporotic bone repair activity of SG scaffolds via promoting the osteogenesis and angiogenesis, as well as inhibiting the osteoclastogenesis and inflammation.
Collapse
Affiliation(s)
- Tingting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Institute of Medicine and Health, Guangdong Academy of Sciences, Guangzhou, 510500, China.,Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Wenping Liu
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Shusen Huang
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Jiwen Chen
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Fupo He
- School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China
| | - Huajun Wang
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xiaofei Zheng
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zhenyan Li
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Huantian Zhang
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zhengang Zha
- Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Zefeng Lin
- Guangdong Key Lab of Orthopedic Technology and Implant, General Hospital of Southern Theater Command of PLA, Guangzhou, 510010, China.,School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Yuanfeng Chen
- Research Center of Medical Science, Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.,Institute of Orthopedic Diseases, Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
26
|
Microenvironment Influences on Human Umbilical Cord Mesenchymal Stem Cell-Based Bone Regeneration. Stem Cells Int 2021; 2021:4465022. [PMID: 34447439 PMCID: PMC8384552 DOI: 10.1155/2021/4465022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
The microenvironment, or niche, regulates stem cell fate and improves differentiation efficiency. Human umbilical cord mesenchymal stem cells (hUC-MSCs) are ideal cell source for bone tissue engineering. However, the role of the microenvironments in hUC-MSC-based bone regeneration is not yet fully understood. This study is aimed at investigating the effects of the in vitro culture microenvironment (hUC-MSCs, nano-hydroxyapatite/collagen/poly (L-lactide) (nHAC/PLA), osteogenic media (OMD), and recombinant human bone morphogenetic protein-7 (rhBMP-7)) and the in vivo transplanted microenvironment (ectopic and orthotopic) on bone regeneration ability of hUC-MSCs. The isolated hUC-MSCs showed self-renewal potential and MSCs' characteristics. In the in vitro two-dimensional culture microenvironment, OMD or OMD with rhBMP-7 significantly enhanced hUC-MSCs' osteocalcin immunofluorescence staining, alkaline phosphatase, and Alizarin red staining; OMD with rhBMP-7 exhibited the highest ALP secretion and mineralized matrix formation. In the in vitro three-dimensional culture microenvironment, nHAC/PLA supported hUC-MSCs' adhesion, proliferation, and differentiation; the microenvironment containing OMD or OMD and rhBMP-7 shortened cell proliferation progression and made osteogenic differentiation progression advance; rhBMP-7 significantly attenuated the inhibiting effect of OMD on hUC-MSCs' proliferation and significantly enhanced the promoting effect of OMD on gene expression and protein secretion of osteogenic differentiation markers, calcium and phosphorous concentration, and mineralized matrix formation. The in vitro three-dimensional culture microenvironment containing OMD and rhBMP-7 induced hUC-MSCs to form the most new bones in ectopic or orthotopic microenvironment as proved by microcomputed tomography and hematoxylin and eosin staining, but bone formation in orthotopic microenvironment was significantly higher than that in ectopic microenvironment. The results indicated that the combination of in vitro hUC-MSCs+nHAC/PLA+OMD+rhBMP-7 microenvironment and in vivo orthotopic microenvironment provided a more optimized niche for bone regeneration of hUC-MSCs. This study elucidates that hUC-MSCs and their local microenvironment, or niche, play an important role in hUC-MSC-based bone regeneration. The endogenously produced BMP may serve an important regulatory role in the process.
Collapse
|
27
|
Ye J, Jiang J, Zhou Z, Weng Z, Xu Y, Liu L, Zhang W, Yang Y, Luo J, Wang X. Near-Infrared Light and Upconversion Nanoparticle Defined Nitric Oxide-Based Osteoporosis Targeting Therapy. ACS NANO 2021; 15:13692-13702. [PMID: 34328303 DOI: 10.1021/acsnano.1c04974] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Osteoporosis is one of the most common diseases affecting bone metabolism. Nitric oxide (NO), an endogenous gas molecule involved in osteogenesis, can effectively promote the proliferation and differentiation of osteoblasts. Although exogenous NO can reverse osteoporosis to a certain extent, the transitory half-life and short diffusion radius of NO severely limit its application. In this work, a gas generation nanoplatform of NO with bone targeting property (UCPA) is developed based on the upconversion nanoparticles (UCNPs) that can convert 808 nm near-infrared (NIR) light into UV/blue light, and further stimulate the NO donor (BNN) to release NO. With an adjustment of the output power of the 808 nm NIR, the amount of released NO can be precisely controlled. Both in vitro and in vivo experiments demonstrate the favorable affinity of UCPA to bone due to the modification of alendronate; thus, it can directly release NO in bone and reverse osteoporosis. In addition, the cellular uptake of nanocomposites and intracellular NO release can be observed in preosteoblasts, thereby promoting their differentiation efficiently.
Collapse
Affiliation(s)
- Jing Ye
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Junkai Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Zhirui Zhou
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Zhenzhen Weng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Yingying Xu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Lubing Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Wei Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Yifei Yang
- School of Public Health, Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Jun Luo
- The Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| |
Collapse
|
28
|
Xu J, Su W, Chen J, Ye Z, Wu C, Jiang J, Yan X, Cai J, Zhao J. The Effect of Antiosteoporosis Therapy With Risedronate on Rotator Cuff Healing in an Osteoporotic Rat Model. Am J Sports Med 2021; 49:2074-2084. [PMID: 33998839 DOI: 10.1177/03635465211011748] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteoporosis increases the revision rate of rotator cuff repair (RCR). Weak fixation might not be the only cause of high RCR failure rates. The biological mechanism associated with tendon-to-bone healing after RCR in osteoporosis should be investigated. HYPOTHESIS (1) Osteoporosis would impair rotator cuff healing through the high osteoclastic activity at the repaired interface. (2) Risedronate would promote rotator cuff healing by reducing osteoclastic activity at the repaired interface. STUDY DESIGN Controlled laboratory study. METHODS A total of 84 female Sprague Dawley rats were randomly treated using ovariectomy or sham surgeries to establish osteoporotic and nonosteoporotic rat models. After confirming osteoporosis, a chronic rotator cuff tear model was created and RCR was performed. Postoperatively, osteoporotic rats were randomly divided into osteoporosis (OP) and osteoporosis with risedronate administration (OP+RIS) groups. Nonosteoporotic rats were used as the control (CON) group. Osteoclastic activity was measured at 1 and 3 weeks after RCR, and histologic analysis of the tendon-to-bone interface, bone morphometric evaluation, and biomechanical tests were performed at 4 and 8 weeks. RESULTS At the early healing stages of 1 and 3 weeks after RCR, the OP group showed the highest osteoclast density at the repaired interface. Compared with the OP group, risedronate administration significantly decreased osteoclast density in the OP+RIS group. At 8 weeks, histologic scores were greater in the OP+RIS group than in the OP group but still lower than in the CON group. Histologic scores at 8 weeks were negatively correlated with osteoclast density at the early healing stage. Additionally, the OP+RIS group showed better bone morphometric parameters and biomechanical properties than did the OP group. CONCLUSION Osteoporosis impaired rotator cuff healing, which might be related to the high osteoclast density at the repaired interface at the early healing stage. Postoperative risedronate administration decreased osteoclast density and enhanced rotator cuff healing in osteoporotic rats, although the effect was inferior to that in nonosteoporotic rats. CLINICAL RELEVANCE Postoperative risedronate administration can be considered a potential therapy to enhance rotator cuff healing in patients with postmenopausal osteoporosis. However, this needs to be verified in a clinical setting.
Collapse
Affiliation(s)
- Junjie Xu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Su
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiebo Chen
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zipeng Ye
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chenliang Wu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jia Jiang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaoyu Yan
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiangyu Cai
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
29
|
Xie C, Ye J, Liang R, Yao X, Wu X, Koh Y, Wei W, Zhang X, Ouyang H. Advanced Strategies of Biomimetic Tissue-Engineered Grafts for Bone Regeneration. Adv Healthc Mater 2021; 10:e2100408. [PMID: 33949147 DOI: 10.1002/adhm.202100408] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/16/2021] [Indexed: 12/21/2022]
Abstract
The failure to repair critical-sized bone defects often leads to incomplete regeneration or fracture non-union. Tissue-engineered grafts have been recognized as an alternative strategy for bone regeneration due to their potential to repair defects. To design a successful tissue-engineered graft requires the understanding of physicochemical optimization to mimic the composition and structure of native bone, as well as the biological strategies of mimicking the key biological elements during bone regeneration process. This review provides an overview of engineered graft-based strategies focusing on physicochemical properties of materials and graft structure optimization from macroscale to nanoscale to further boost bone regeneration, and it summarizes biological strategies which mainly focus on growth factors following bone regeneration pattern and stem cell-based strategies for more efficient repair. Finally, it discusses the current limitations of existing strategies upon bone repair and highlights a promising strategy for rapid bone regeneration.
Collapse
Affiliation(s)
- Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
| | - Jinchun Ye
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Xudong Yao
- The Fourth Affiliated Hospital Zhejiang University School of Medicine Yiwu 322000 China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Yiwen Koh
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Wei Wei
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine and Department of Orthopedic Surgery of the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310058 China
- Zhejiang University‐University of Edinburgh Institute Zhejiang University School of Medicine and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province Zhejiang University School of Medicine Hangzhou 314499 China
- Department of Sports Medicine Zhejiang University School of Medicine Hangzhou 310058 China
- China Orthopedic Regenerative Medicine Group (CORMed) Hangzhou 310058 China
| |
Collapse
|
30
|
Hejazi F, Ebrahimi V, Asgary M, Piryaei A, Fridoni MJ, Kermani AA, Zare F, Abdollahifar MA. Improved healing of critical-size femoral defect in osteoporosis rat models using 3D elastin/polycaprolactone/nHA scaffold in combination with mesenchymal stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:27. [PMID: 33683483 PMCID: PMC7940275 DOI: 10.1007/s10856-021-06495-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Osteoporosis is a common bone disease that results in elevated risk of fracture, and delayed bone healing and impaired bone regeneration are implicated by this disease. In this study, Elastin/Polycaprolactone/nHA nanofibrous scaffold in combination with mesenchymal stem cells were used to regenerate bone defects. Cytotoxicity, cytocompatibility and cellular morphology were evaluated in vitro and observations revealed that an appropriate environment for cellular attachment, growth, migration, and proliferation is provided by this scaffold. At 3 months following ovariectomy (OVX), the rats were used as animal models with an induced critical size defect in the femur to evaluate the therapeutic potential of osteogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs) seeded on 3 dimension (3D) scaffolds. In this experimental study, 24 female Wistar rats were equally divided into three groups: Control, scaffold (non-seeded BM-MSC), and scaffold + cell (seeded BM-MSC) groups. 30 days after surgery, the right femur was removed, and underwent a stereological analysis and RNA extraction in order to examine the expression of Bmp-2 and Vegf genes. The results showed a significant increase in stereological parameters and expression of Bmp-2 and Vegf in scaffold and scaffold + cell groups compared to the control rats. The present study suggests that the use of the 3D Elastin/Polycaprolactone (PCL)/Nano hydroxyapatite (nHA) scaffold in combination with MSCs may improve the fracture regeneration and accelerates bone healing at the osteotomy site in rats.
Collapse
Affiliation(s)
- Fatemeh Hejazi
- Department of Advanced Technology, Shiraz University, Shiraz, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Asgary
- Department of Biology and Anatomical Sciences, School of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran, Tehran, Iran
| | - Mohammad Javad Fridoni
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Asghar Kermani
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Fatemeh Zare
- Department of Biology and Anatomical Sciences, School of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Kim H, Kumbar SG, Nukavarapu SP. Biomaterial-directed cell behavior for tissue engineering. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 17:100260. [PMID: 33521410 PMCID: PMC7839921 DOI: 10.1016/j.cobme.2020.100260] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful tissue regeneration strategies focus on the use of novel biomaterials, structures, and a variety of cues to control cell behavior and promote regeneration. Studies discovered how biomaterial/ structure cues in the form of biomaterial chemistry, material stiffness, surface topography, pore, and degradation properties play an important role in controlling cellular events in the contest of in vitro and in vivo tissue regeneration. Advanced biomaterials structures and strategies are developed to focus on the delivery of bioactive factors, such as proteins, peptides, and even small molecules to influence cell behavior and regeneration. The present article is an effort to summarize important findings and further discuss biomaterial strategies to influence and control cell behavior directly via physical and chemical cues. This article also touches on various modern methods in biomaterials processing to include bioactive factors as signaling cues to program cell behavior for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Hyun Kim
- Biomedical Engineering, University of Connecticut, Storrs-06269
| | - Sangamesh G. Kumbar
- Biomedical Engineering, University of Connecticut, Storrs-06269
- Materials Science & Engineering, University of Connecticut, Storrs-06269
- Orthopaedic Surgery, University of Connecticut Health, Farmington-06030
| | - Syam P. Nukavarapu
- Biomedical Engineering, University of Connecticut, Storrs-06269
- Materials Science & Engineering, University of Connecticut, Storrs-06269
- Orthopaedic Surgery, University of Connecticut Health, Farmington-06030
| |
Collapse
|
32
|
Inoue S, Hatakeyama J, Aoki H, Kuroki H, Niikura T, Oe K, Fukui T, Kuroda R, Akisue T, Moriyama H. Effects of ultrasound, radial extracorporeal shock waves, and electrical stimulation on rat bone defect healing. Ann N Y Acad Sci 2021; 1497:3-14. [PMID: 33619772 DOI: 10.1111/nyas.14581] [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: 12/06/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 11/27/2022]
Abstract
Fractures associated with osteoporosis are a major public health concern. Current treatments for fractures are limited to surgery or fixation, leading to long-term bedrest, which is linked to increased mortality. Alternatively, utilization of physical agents has been suggested as a promising therapeutic approach for fractures. Here, we examined the effects of ultrasound, radial extracorporeal shock waves, and electrical stimulation on normal or osteoporotic fracture healing. Femoral bone defects were created in normal or ovariectomized rats. Rats were divided into four groups: untreated, and treated with ultrasound, shock waves, or electrical stimulation after surgery. Samples were collected at 2 or 4 weeks after surgery, and the healing process was evaluated with micro-CT, histological, and immunohistochemical analyses. Ultrasound at intensities of 0.5 and 1.0 W/cm2 , but not 0.05 W/cm2 , accelerated new bone formation. Shock wave exposure also increased newly formed bone, but formed abnormal periosteal callus around the defect site. Conversely, electrical stimulation did not affect the healing process. Ultrasound exposure increased osteoblast activity and cell proliferation and decreased sclerostin-positive osteocytes. We demonstrated that higher-intensity ultrasound and radial extracorporeal shock waves accelerate fracture healing, but shock wave treatment may increase the risk of periosteal callus formation.
Collapse
Affiliation(s)
- Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Junpei Hatakeyama
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Hitoshi Aoki
- OG Wellness Technologies Co., Ltd., Okayama, Japan
| | - Hiroshi Kuroki
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshihiro Akisue
- Life and Medical Sciences Area, Health Sciences Discipline, Kobe University, Kobe, Japan
| | - Hideki Moriyama
- Life and Medical Sciences Area, Health Sciences Discipline, Kobe University, Kobe, Japan
| |
Collapse
|
33
|
Zhu J, Zhang C, Jia J, Wang H, Leng H, Xu Y, Wu C, Zhang Q, Song C. Osteogenic effects in a rat osteoporosis model and femur defect model by simvastatin microcrystals. Ann N Y Acad Sci 2020; 1487:31-42. [PMID: 33098131 DOI: 10.1111/nyas.14513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022]
Abstract
Simvastatin is a translational drug that may be used to induce local bone formation. In this study, simvastatin microcrystals were made by a wet media milling method, and then we verified the osteogenic effect of the microcrystals in rat ovariectomy (OVX)-induced osteoporosis and femur defect models. For the osteoporosis model, we delivered simvastatin microcrystals to the tibia with poloxamer hydrogels via an intraosseous injection. Bone mineral density and the ultimate force of the treated tibia were significantly improved after injection of simvastatin microcrystals at 0.5 and 1 mg compared with the OVX or 0-mg control groups. For the femur defect model, simvastatin microcrystals were incorporated in clinically used calcium phosphate cements (CPCs) as an implant. Quantitative analysis of bone regeneration by microcomputed tomography (μCT) showed improved bone morphology with simvastatin microcrystals at 50 and 100 μg, compared with the CPC vehicle. A semiquantitative scale for histology assessment further demonstrated a higher bone regeneration score in the drug-loaded groups. Our study shows that simvastatin microcrystals can promote bone formation by local delivery using a poloxamer hydrogel or CPC, which may be translationally useful.
Collapse
Affiliation(s)
- Junxiong Zhu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Chenggui Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Jialin Jia
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Hong Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Huijie Leng
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Cuishuan Wu
- Beijing Delivery Pharmaceutical Technology Co., Ltd, Beijing, China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Diseases, Beijing, China
| |
Collapse
|
34
|
Miao Q, Yang S, Ding H, Liu J. Controlled degradation of chitosan-coated strontium-doped calcium sulfate hemihydrate composite cement promotes bone defect repair in osteoporosis rats. Biomed Mater 2020; 15:055039. [DOI: 10.1088/1748-605x/ab9fcf] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Zeng Y, Zhou M, Mou S, Yang J, Yuan Q, Guo L, Zhong A, Wang J, Sun J, Wang Z. Sustained delivery of alendronate by engineered collagen scaffold for the repair of osteoporotic bone defects and resistance to bone loss. J Biomed Mater Res A 2020; 108:2460-2472. [PMID: 32419333 DOI: 10.1002/jbm.a.36997] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 04/09/2020] [Accepted: 04/19/2020] [Indexed: 12/17/2022]
Abstract
Researches of biomaterials for osteoporotic bone defects focus on the improvement of its anti-osteoporosis ability, due to osteoporosis is a kind of systemic and long-range bone metabolism disorder. Nevertheless, how to steadily deliver anti-osteoporosis drugs in osteoporotic bone defects is rarely studied. Reported evidences have shown that alendronate (Aln) is known to not only restrain osteoclasts from mediating bone resorption but also stimulate osteoblasts to regenerate bone tissue. Here, we developed an engineered implantable scaffold that could sustainably release Aln for osteoporotic bone defects. Briefly, Aln was added into 2% collagen (Col) solution to form a 5 mg/ml mixture. Then the mixture was filled into pre-designed round models (diameter: 5 mm, height: 2 mm) and crosslinked to obtain engineered Col-Aln scaffolds. The release kinetics showed that Aln was released at an average rate of 2.99 μg/d in the initial 8 days and could sustainably release for 1 month. To detect the repair effects of the Col-Aln scaffolds for osteoporotic defects, the Col and Col-Aln scaffolds were implanted into 5 mm cranial defects in ovariectomized rats. After 3 months, the cranial defects implanted with Col-Aln scaffolds achieved more bone regeneration in defect area (11.74 ± 3.82%) than Col scaffold (5.12 ± 1.15%) (p < .05). Moreover, ovariectomized rats in Col-Aln scaffold group possessed more trabecular bone in femur metaphysis than Col scaffold group as analyzed by Micro-CT. This study demonstrated the engineered Col-Aln scaffold has the potential to repair osteoporotic bone defects and resist bone loss in osteoporosis.
Collapse
Affiliation(s)
- Yuyang Zeng
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Muran Zhou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Shan Mou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Jie Yang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Quan Yuan
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Liang Guo
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Aimei Zhong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Jiecong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, China
| |
Collapse
|
36
|
Ion R, Necula MG, Mazare A, Mitran V, Neacsu P, Schmuki P, Cimpean A. Drug Delivery Systems Based on Titania Nanotubes and Active Agents for Enhanced Osseointegration of Bone Implants. Curr Med Chem 2020; 27:854-902. [PMID: 31362646 DOI: 10.2174/0929867326666190726123229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 01/16/2019] [Accepted: 05/04/2019] [Indexed: 12/31/2022]
Abstract
TiO2 nanotubes (TNTs) are attractive nanostructures for localized drug delivery. Owing to their excellent biocompatibility and physicochemical properties, numerous functionalizations of TNTs have been attempted for their use as therapeutic agent delivery platforms. In this review, we discuss the current advances in the applications of TNT-based delivery systems with an emphasis on the various functionalizations of TNTs for enhancing osteogenesis at the bone-implant interface and for preventing implant-related infection. Innovation of therapies for enhancing osteogenesis still represents a critical challenge in regeneration of bone defects. The overall concept focuses on the use of osteoconductive materials in combination with the use of osteoinductive or osteopromotive factors. In this context, we highlight the strategies for improving the functionality of TNTs, using five classes of bioactive agents: growth factors (GFs), statins, plant derived molecules, inorganic therapeutic ions/nanoparticles (NPs) and antimicrobial compounds.
Collapse
Affiliation(s)
- Raluca Ion
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Anca Mazare
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patricia Neacsu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Patrik Schmuki
- University of Erlangen-Nuremberg, Department of Materials Science, Erlangen, Germany
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
37
|
Martínez-Martínez A, Muñoz-Islas E, Ramírez-Rosas MB, Acosta-González RI, Torres-Rodríguez HF, Jiménez-Andrade JM. Blockade of the colony-stimulating factor-1 receptor reverses bone loss in osteoporosis mouse models. Pharmacol Rep 2020; 72:1614-1626. [PMID: 32222915 DOI: 10.1007/s43440-020-00091-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 02/19/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mice lacking either colony-stimulating factor-1 (CSF-1) or its receptor, CSF-1R, display osteopetrosis. Accordingly, genetic deletion or pharmacological blockade of CSF-1 prevents the bone loss associated with estrogen deficiency. However, the role of CSF-1R in osteoporosis models of type-1 diabetes (T1D) and ovariectomy (OVX) has not been examined. Thus, we evaluated whether CSF-1R blockade would relieve the bone loss in a model of primary osteoporosis (female mice with OVX) and a model of secondary osteoporosis (female with T1D) using micro-computed tomography. METHODS Female ICR mice at 10 weeks underwent OVX or received five daily administrations of streptozotocin (ip, 50 mg/kg) to induce T1D. Four weeks after OVX and 14 weeks after first injection of streptozotocin, mice received an anti-CSF-1R (2G2) antibody (10 mg/kg, ip; once/week for 6 weeks) or vehicle. At the last day of antibody administration, mice were sacrificed and femur and tibia were harvested for micro-computed tomography analysis. RESULTS Mice with OVX had a significant loss of trabecular bone at the distal femoral and proximal tibial metaphysis. Chronic treatment with anti-CSF-1R significantly reversed the trabecular bone loss at these anatomical sites. Streptozotocin-induced T1D resulted in significant loss of trabecular bone at the femoral neck and cortical bone at the femoral mid-diaphysis. Chronic treatment with anti-CSF-1R antibody significantly reversed the bone loss observed in mice with T1D. CONCLUSION Our results demonstrate that blockade of CSF-1R signaling reverses bone loss in two different mouse models of osteoporosis.
Collapse
Affiliation(s)
- Arisaí Martínez-Martínez
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México
| | - Enriqueta Muñoz-Islas
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México
| | - Martha B Ramírez-Rosas
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México
| | - Rosa I Acosta-González
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México
| | - Héctor F Torres-Rodríguez
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México
| | - Juan M Jiménez-Andrade
- Laboratorio de Farmacología, Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, 88740, Reynosa, TAMPS, México.
| |
Collapse
|
38
|
Wang H, Qu X, Zhang Z, Lei M, Tan H, Bao C, Lin S, Zhu L, Kohn J, Liu C. Tag-Free Site-Specific BMP-2 Immobilization with Long-Acting Bioactivities via a Simple Sugar-Lectin Interaction. ACS Biomater Sci Eng 2020; 6:2219-2230. [PMID: 33455345 DOI: 10.1021/acsbiomaterials.9b01730] [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: 12/24/2022]
Abstract
The construction of a biomaterial matrix with biological properties is of great importance to developing functional materials for clinical use. However, the site-specific immobilization of growth factors to endow materials with bioactivities has been a challenge to date. Considering the wide existence of glycosylation in mammalian proteins or recombinant proteins, we establish a bioaffinity-based protein immobilization strategy (bioanchoring method) utilizing the native sugar-lectin interaction between concanavalin A (Con A) and the oligosaccharide chain on glycosylated bone morphogenetic protein-2 (GBMP-2). The interaction realizes the site-specific immobilization of GBMP-2 to a substrate modified with Con A while preserving its bioactivity in a sustained and highly efficient way, as evidenced by its enhanced ability to induce osteodifferentiation compared with that of the soluble GBMP-2. Moreover, the surface with Con A-bioanchored GBMP-2 can be reused to stimulate multiple batches of C2C12 cells to differentiate almost to the same degree. Even after 4 month storage at 4 °C in phosphate-buffered saline (PBS), the Con A-bioanchored GBMP-2 still maintains the bioactivity to stimulate the differentiation of C2C12 cells. Furthermore, the ectopic ossification test proves the in vivo bioactivity of bioanchored GBMP-2. Overall, our results demonstrate that the tag-free and site (i.e., sugar chain)-specific protein immobilization strategy represents a simple and generic alternative, which is promising to apply for other glycoprotein immobilization and application. It should be noted that although the lectin we utilized can only bind to d-mannose/d-glucose, the diversity of the lectin family assures that a specific lectin could be offered for other sugar types, thus expanding the applicable scope further.
Collapse
Affiliation(s)
| | | | - Zheng Zhang
- Department of Chemistry and Chemical Biology and New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | | | | | | | | | | | - Joachim Kohn
- Department of Chemistry and Chemical Biology and New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | | |
Collapse
|
39
|
Combined antisclerostin antibody and parathyroid hormone (1–34) synergistically enhance the healing of bone defects in ovariectomized rats. Z Gerontol Geriatr 2020; 53:163-170. [DOI: 10.1007/s00391-019-01685-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/04/2019] [Indexed: 01/24/2023]
|
40
|
Yin C, Jia X, Zhao Q, Zhao Z, Wang J, Zhang Y, Li Z, Sun H, Li Z. Transcription factor 7-like 2 promotes osteogenic differentiation and boron-induced bone repair via lipocalin 2. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110671. [PMID: 32204099 DOI: 10.1016/j.msec.2020.110671] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 01/07/2023]
Abstract
Boron-containing mesoporous bioactive glass (B-MBG) scaffolds could be capable of promoting osteogenesis by activating Wnt/β-catenin signaling pathway during the process of bone defect repair. Despite this, more involving molecular controls are still largely unclear. In the present study, we identified that the downstream of Wnt/β-catenin signaling pathway named transcription factor 7-like 2 (TCF7L2) served as a key effector to promote boron-induced bone regeneration and osteogenesis through lipocalin 2 (LCN2). TCF7L2 was highly expressed in osteoblasts when treated with B-MBG scaffold extraction than MBG. LCN2, as a secreted bone factor, positively affected osteogenic differentiation of MC3T3-E1 and osteogenesis in vivo, which could be induced by TCF7L2. In addition, interference of TCF7L2 decreased the osteogenic differentiation of osteoblasts. Finally, we identified that rLCN2 could rescue the poor ability of osteogenic differentiation of MC3T3-E1 whose Tcf7l2 gene was knocked down by lentiviral transfection of shRNA. Our findings provide some new insights into the molecular controls of boron-associated bone regeneration and potential therapeutic targets for the treatment of bone defects.
Collapse
Affiliation(s)
- Chengcheng Yin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China
| | - Xiaoshi Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qin Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zifan Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jinyang Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhi Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Hongchen Sun
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110002, China.
| | - Zubing Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
41
|
Camacho-Alonso F, Martínez-Ortiz C, Plazas-Buendía L, Mercado-Díaz AM, Vilaplana-Vivo C, Navarro JA, Buendía AJ, Merino JJ, Martínez-Beneyto Y. Bone union formation in the rat mandibular symphysis using hydroxyapatite with or without simvastatin: effects on healthy, diabetic, and osteoporotic rats. Clin Oral Investig 2020; 24:1479-1491. [PMID: 31925587 DOI: 10.1007/s00784-019-03180-9] [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: 09/03/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The objective is to compare new bone formation in critical defects in healthy, diabetic, and osteoporotic rats filled with hydroxyapatite (HA) alone and HA combined with simvastatin (SV). MATERIALS AND METHODS A total of 48 adult female Sprague-Dawley rats were randomized into three groups (n = 16 per group): Group, 1 healthy; Group 2, diabetics; and Group 3, osteoporotics. Streptozotocin was used to induce type 1 diabetes in Group 2, while bilateral ovariectomy was used to induce osteoporosis in Group 3. The central portion of the rat mandibular symphysis was used as a physiological critical bone defect. In each group, eight defects were filled with HA alone and eight with HA combined with SV. The animals were sacrificed at 4 and 8 weeks, and the mandibles were processed for micro-computed tomography to analyze radiological union and bone mineral density (BMD); histological analysis of the bone union; and immunohistochemical analysis, which included immunoreactivity of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2). RESULTS In all groups (healthy, diabetics, and osteoporotics), the defects filled with HA + SV presented greater radiological bone union, BMD, histological bone union, and more VEGF and BMP-2 positivity, in comparison with bone defects treated with HA alone. CONCLUSIONS Combined application of HA and SV improves bone regeneration in mandibular critical bone defects compared with application of HA alone in healthy, diabetic, and osteoporotic rats. CLINICAL RELEVANCE This study might help to patients with osteoporosis or uncontrolled diabetes type 1, but future studies should be done.
Collapse
Affiliation(s)
- F Camacho-Alonso
- Department of Oral Surgery, University of Murcia, Murcia, Spain.
| | | | | | | | | | - J A Navarro
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - A J Buendía
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - J J Merino
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Y Martínez-Beneyto
- Department of Preventive and Community Dentistry, University of Murcia, Murcia, Spain
| |
Collapse
|
42
|
Shintani K, Uemura T, Takamatsu K, Yokoi T, Onode E, Okada M, Tabata Y, Nakamura H. Evaluation of dual release of stromal cell-derived factor-1 and basic fibroblast growth factor with nerve conduit for peripheral nerve regeneration: An experimental study in mice. Microsurgery 2019; 40:377-386. [PMID: 31868964 DOI: 10.1002/micr.30548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/30/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The development of drug delivery systems has enabled the release of multiple bioactive molecules. The efficacy of nerve conduits coated with dual controlled release of stromal cell-derived factor-1 (SDF-1) and basic fibroblast growth factor (bFGF) for peripheral nerve regeneration was investigated. MATERIALS AND METHODS Sixty-two C57BL6 mice were used for peripheral nerve regeneration with a nerve conduit (inner diameter, 1 mm, and length, 7 mm) and an autograft. The mice were randomized into five groups based on the different repairs of nerve defects. In the group of repair with conduits alone (n = 9), a 5-mm sciatic nerve defect was repaired by the nerve conduit. In the group of repair with conduits coated with bFGF (n = 10), SDF-1 (n = 10), and SDF-1/bFGF (n = 10), it was repaired by the nerve conduit with bFGF gelatin, SDF-1 gelatin, and SDF-1/bFGF gelatin, respectively. In the group of repair with autografts (n = 10), it was repaired by the resected nerve itself. The functional recovery, nerve regeneration, angiogenesis, and TGF-β1 gene expression were assessed. RESULTS In the conduits coated with SDF-1/bFGF group, the mean sciatic functional index value (-88.68 ± 10.64, p = .034) and the axon number (218.8 ± 111.1, p = .049) were significantly higher than the conduit alone group, followed by the autograft group; in addition, numerous CD34-positive cells and micro vessels were observed. TGF-β1 gene expression relative values in the conduits with SDF-1/bFGF group at 3 days (7.99 ± 5.14, p = .049) significantly increased more than the conduits alone group. CONCLUSION Nerve conduits coated with dual controlled release of SDF-1 and bFGF promoted peripheral nerve regeneration.
Collapse
Affiliation(s)
- Kosuke Shintani
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Pediatric Orthopaedic Surgery, Children's Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Takuya Uemura
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | - Kiyohito Takamatsu
- Department of Orthopaedic Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Takuya Yokoi
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ema Onode
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mitsuhiro Okada
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiko Tabata
- Department of Regeneration Science and Engineering, Institute for Frontier Life and MedicalSciences, Kyoto University, Kyoto, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
43
|
Tao Z, Zhou W, Wu X, Lu H, Ma N, Li Y, Zhang R, Yang M, Xu HG. Local administration of aspirin improves osseointegration of hydroxyapatite-coated titanium implants in ovariectomized rats through activation of the Notch signaling pathway. J Biomater Appl 2019; 34:1009-1018. [PMID: 31757183 DOI: 10.1177/0885328219889630] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zhoushan Tao
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Wanshu Zhou
- Department of Geriatrics, The Second Affiliated Hospital of Wannan Medical College, Anhui, People's Republic of China
| | - Xingjing Wu
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Hanli Lu
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Nengfeng Ma
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Yang Li
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Ruotian Zhang
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Min Yang
- Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Hong-Guang Xu
- Department of Spine Surgery, Spine Research Center of Wannan Medical College, Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Yijishan hospital of Wannan Medical College, Anhui, People's Republic of China
| |
Collapse
|
44
|
Vargas‐Muñoz VM, Martínez‐Martínez A, Muñoz‐Islas E, Ramírez‐Rosas MB, Acosta‐González RI, Jiménez‐Andrade JM. Chronic administration of Cl‐amidine, a pan‐peptidylarginine deiminase inhibitor, does not reverse bone loss in two different murine models of osteoporosis. Drug Dev Res 2019; 81:93-101. [DOI: 10.1002/ddr.21608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Virginia M. Vargas‐Muñoz
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| | - Arisai Martínez‐Martínez
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| | - Enriqueta Muñoz‐Islas
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| | - Martha B. Ramírez‐Rosas
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| | - Rosa I. Acosta‐González
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| | - Juan M. Jiménez‐Andrade
- Unidad Académica Multidisciplinaria Reynosa‐Aztlán, Universidad Autónoma de Tamaulipas Reynosa Tamaulipas Mexico
| |
Collapse
|
45
|
Yao S, Lin X, Xu Y, Chen Y, Qiu P, Shao C, Jin B, Mu Z, Sommerdijk NAJM, Tang R. Osteoporotic Bone Recovery by a Highly Bone-Inductive Calcium Phosphate Polymer-Induced Liquid-Precursor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900683. [PMID: 31592093 PMCID: PMC6774089 DOI: 10.1002/advs.201900683] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/28/2019] [Indexed: 05/17/2023]
Abstract
Osteoporosis is an incurable chronic disease characterized by a lack of mineral mass in the bones. Here, the full recovery of osteoporotic bone is achieved by using a calcium phosphate polymer-induced liquid-precursor (CaP-PILP). This free-flowing CaP-PILP material displays excellent bone inductivity and is able to readily penetrate into collagen fibrils and form intrafibrillar hydroxyapatite crystals oriented along the c-axis. This ability is attributed to the microstructure of the material, which consists of homogeneously distributed ultrasmall (≈1 nm) amorphous calcium phosphate clusters. In vitro study shows the strong affinity of CaP-PILP to osteoporotic bone, which can be uniformly distributed throughout the bone tissue to significantly increase the bone density. In vivo experiments show that the repaired bones exhibit satisfactory mechanical performance comparable with normal ones, following a promising treatment of osteoporosis by using CaP-PILP. The discovery provides insight into the structure and property of biological nanocluster materials and their potential for hard tissue repair.
Collapse
Affiliation(s)
- Shasha Yao
- Center for Biomaterials and BiopathwaysDepartment of ChemistryZhejiang UniversityHangzhouZhejiang310027China
| | - Xianfeng Lin
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouZhejiang310016China
| | - Yifei Xu
- Laboratory of Materials and Interface Chemistry and Center for Multiscale Electron MicroscopyDepartment of Chemical Engineering and ChemistryEindhoven University of Technology, EindhovenPO box 5135600 MBEindhovenThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of Technology, EindhovenPO box 5135600 MBEindhovenThe Netherlands
| | - Yangwu Chen
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouZhejiang310016China
- Department of Orthopedic SurgerySecond Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310012China
| | - Pengcheng Qiu
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310016China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouZhejiang310016China
| | - Changyu Shao
- Center for Biomaterials and BiopathwaysDepartment of ChemistryZhejiang UniversityHangzhouZhejiang310027China
| | - Biao Jin
- Center for Biomaterials and BiopathwaysDepartment of ChemistryZhejiang UniversityHangzhouZhejiang310027China
| | - Zhao Mu
- Center for Biomaterials and BiopathwaysDepartment of ChemistryZhejiang UniversityHangzhouZhejiang310027China
| | - Nico A. J. M. Sommerdijk
- Laboratory of Materials and Interface Chemistry and Center for Multiscale Electron MicroscopyDepartment of Chemical Engineering and ChemistryEindhoven University of Technology, EindhovenPO box 5135600 MBEindhovenThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of Technology, EindhovenPO box 5135600 MBEindhovenThe Netherlands
| | - Ruikang Tang
- Center for Biomaterials and BiopathwaysDepartment of ChemistryZhejiang UniversityHangzhouZhejiang310027China
| |
Collapse
|
46
|
Janani G, Kumar M, Chouhan D, Moses JC, Gangrade A, Bhattacharjee S, Mandal BB. Insight into Silk-Based Biomaterials: From Physicochemical Attributes to Recent Biomedical Applications. ACS APPLIED BIO MATERIALS 2019; 2:5460-5491. [DOI: 10.1021/acsabm.9b00576] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Miao G, Li Z, Meng Y, Wu J, Li Y, Hu Q, Chen X, Yang X, Chen X. Preparation, characterization, in vitro bioactivity and protein loading/release property of mesoporous bioactive glass microspheres with different compositions. ADV POWDER TECHNOL 2019. [DOI: 10.1016/j.apt.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
49
|
Tao ZS, Wu XJ, Yang M, Xu HG. Local administration with silymarin could increase osseointegration of hydroxyapatite-coated titanium implants in ovariectomized rats. J Biomater Appl 2019; 34:664-672. [PMID: 31342833 DOI: 10.1177/0885328219863290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Zhou-Shan Tao
- 1 Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Xing-Jing Wu
- 1 Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Min Yang
- 1 Department of Trauma orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Anhui, People's Republic of China
| | - Hong-Guang Xu
- 2 Department of Spine Surgery, Spine Research Center of Wannan Medical College, Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Yijishan hospital of Wannan Medical College. Anhui, People's Republic of China
| |
Collapse
|
50
|
Lehman LFC, de Noronha MS, Diniz IMA, da Costa E Silva RMF, Andrade ÂL, de Sousa Lima LF, de Alcântara CEP, Domingues R, Ferreira AJ, da Silva TA, Mesquita RA. Bioactive glass containing 90% SiO 2 in hard tissue engineering: An in vitro and in vivo characterization study. J Tissue Eng Regen Med 2019; 13:1651-1663. [PMID: 31218837 DOI: 10.1002/term.2919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/07/2019] [Accepted: 06/07/2019] [Indexed: 01/10/2023]
Abstract
Bioactive glass has been proved to have many applications in bioengineering due to its bone regenerative properties. In this work, an innovative, highly resorbable bioactive glass containing 90% SiO2 (BG90) to be used as a bone substitute was developed. The BG90 was synthetized by the sol-gel process with the dry step at room temperature. The biomaterial showed in vitro and in vivo bioactivities even with silica content up to 90%. Moreover, the BG90 presented high porosity and surface area due to its homogenously interconnected porous network. In vitro, it was observed to have high cell viability and marked osteoblastic differentiation of rat bone marrow-derived cells when in contact with BG90 ion extracts. The BG90 transplantation into rat tibia defects was analysed at 1, 2, 3, 4, 7, and 10 weeks post-operatively and compared with the defects of negative (no graft) and positive (autogenous bone graft) controls. After 4 weeks of grafting, the BG90 was totally resorbed and induced higher bone formation than did the positive control. Bone morphogenetic protein 2 (BMP-2) expression at the grafting site peaked at 1 week and decreased similarly after 7 weeks for all groups. Only the BG90 group was still exhibiting BMP-2 expression in the last experimental time. Our data demonstrated that the BG90 could be an attractive candidate to provide useful approaches in hard-tissue bioengineering.
Collapse
Affiliation(s)
- Luiz Felipe Cardoso Lehman
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana Saturnino de Noronha
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ivana Márcia Alves Diniz
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ângela Leão Andrade
- Department of Chemistry, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | | | - Rosana Domingues
- Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anderson José Ferreira
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tarcília Aparecida da Silva
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Alves Mesquita
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|